Вы находитесь на странице: 1из 2710

ellis d.

avner
william e. harmon
patrick niaudet
norishige yoshikawa
francesco emma
stuart l. goldstein
Editors

Pediatric
Nephrology
Seventh Edition

OFFICIALLY
ENDORSED BY 1 3Reference
Pediatric Nephrology
Ellis D. Avner • William E. Harmon
Patrick Niaudet • Norishige Yoshikawa
Francesco Emma • Stuart L. Goldstein
Editors

Pediatric Nephrology
Seventh Edition

With 506 Figures and 310 Tables


Editors
Ellis D. Avner William E. Harmon
Department of Pediatrics Boston Children’s Hospital
Medical College of Wisconsin Harvard Medical School
Children’s Research Institute Boston, MA, USA
Children’s Hospital
Health System of Wisconsin
Milwaukee, WI, USA
Patrick Niaudet Norishige Yoshikawa
Service de Néphrologie Pédiatrique Department of Pediatrics
Hôpital Necker-Enfants Malades Wakayama Medical University
Université Paris-Descartes Wakayama City, Japan
Paris, France

Francesco Emma Stuart L. Goldstein


Division of Nephrology Division of Nephrology and
Bambino Gesù Children’s Hospital – IRCCS Hypertension
Rome, Italy The Heart Institute
Cincinnati Children’s Hospital
Medical Center, College of
Medicine
Cincinnati, OH, USA

ISBN 978-3-662-43595-3 ISBN 978-3-662-43596-0 (eBook)


ISBN 978-3-662-43597-7 (print and electronic bundle)
DOI 10.1007/978-3-662-43596-0
Library of Congress Control Number: 2015954467

Springer Heidelberg New York Dordrecht London


# Springer-Verlag Berlin Heidelberg 2009, 2016
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or
part of the material is concerned, specifically the rights of translation, reprinting, reuse of
illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way,
and transmission or information storage and retrieval, electronic adaptation, computer software, or
by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are exempt
from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this
book are believed to be true and accurate at the date of publication. Neither the publisher nor the
authors or the editors give a warranty, express or implied, with respect to the material contained
herein or for any errors or omissions that may have been made.

Printed on acid-free paper

Springer-Verlag GmbH Berlin Heidelberg is part of Springer Science+Business Media (www.


springer.com)
Preface

Through its past six editions, Pediatric Nephrology has become the standard
medical reference for health care professionals treating children with kidney
disease. This new edition, published 6 years since the previous version,
reflects the tremendous increase in critical information required to translate
molecular and cellular pathophysiology into the prevention, diagnosis, and
therapy of childhood renal disorders. This text is particularly targeted to
pediatricians, pediatric nephrologists, pediatric urologists, and physicians in
training. It is also targeted to the increased number of health care professionals
comprising the multidisciplinary teams required to provide comprehensive
care for children with kidney disease and their families: geneticists, genetic
counselors, nurse specialists, dialysis personnel, nutritionists, social workers,
and mental health professionals. Finally, this reference is designed to serve the
needs of primary care physicians (internists and family practitioners) as well as
internist nephrologists who are increasingly involved in the initial evaluation
and/or the longitudinal care of children with renal disease under different
health care delivery systems evolving throughout the world.
To keep pace with the dramatic changes in pediatric renal medicine since
the publication of the previous edition, the content of the seventh edition of
Pediatric Nephrology has been completely revised, updated, and enlarged.
The seventh edition contains 83 chapters in 3 volumes, which are organized
into 12 main sections. The text begins with an overview of the basic develop-
mental anatomy, biology, and physiology of the kidney, which provides the
basic information necessary to understand the developmental nature of pedi-
atric renal diseases. This is followed by a comprehensive coverage of the
evaluation, diagnosis, and therapy of specific childhood kidney diseases,
including the extensive use of clinical algorithms. Of special note is a section
focused on rapidly evolving research methodologies, which are being trans-
lated into new clinical approaches and therapies for many childhood renal
diseases. The final sections focus on comprehensive, state-of-the-art reviews
of acute and chronic renal failure in childhood.
Many chapters of the seventh edition have been completely rewritten by
new authors, all recognized as global authorities in their respective areas. The
remainder of the text has been totally revised, with junior authors often joining
senior authors from the previous edition. The number of contributors has
increased by 20 %. In addition to the new section on Global Pediatric
Nephrology, which focuses on unique aspects of pediatric nephrology practice

v
vi Preface

and the epidemiology of pediatric renal disease in different regions of the


world, all of the chapters reflect a global perspective. This has been achieved
through a dynamic, evolving relationship between the Editors and the Inter-
national Pediatric Nephrology Association (IPNA). This has led to the con-
tinued endorsement of Pediatric Nephrology as the standard global
reference text in the field of childhood kidney disease. We are proud
that the IPNA logo adorns the cover of Pediatric Nephrology in recognition
of this endorsement. The Editors look forward to this dynamic interaction
with IPNA to take advantage of future opportunities that such collaboration
may provide in the areas of education and outreach.
Other major changes are also evidenced by the new publication of the
seventh edition of Pediatric Nephrology as a basic reference handbook in the
SpringerReference series. Representing advances in state-of-the-art electronic
publishing, there are regularly updated online versions of each chapter of this
text at SpringerLink.com. The new publishing format has led to a welcome
expansion of the published text to three volumes and the increased utilization
of high-resolution, color figures. Further, two new Editors, Professor
Francesco Emma of Ospedale Pediatrico Bambino Gesu in Rome, Italy, and
Professor Stuart L. Goldstein of the University of Cincinnati, USA, have
joined the Editorial Team. The addition of new Editors continues to provide
a dynamic mixture of continuity, new ideas, new perspectives, and globalism,
which has distinguished each new edition of the text. Professors Emma and
Goldstein join the four Editors from the sixth edition: Senior Editor
and Emeritus Professor Ellis D. Avner, from the Medical College of Wiscon-
sin, USA, and Professors William E. Harmon M.D. of Harvard University,
USA, Patrick Niaudet, from the Hôpital Necker-Enfants Malades in
Paris, France; and Norishige Yoshikawa from Wakayama, Japan. The current
Editors are internationally recognized leaders in complementary areas of
pediatric nephrology and along with the more than 150 contributors reflect
the global nature of the text and the subspecialty it serves.
The Editors wish to thank a number of individuals whose efforts were
critical in the success of this project. The book would never have reached
this seventh edition without the dedication of our professional colleagues at
Springer, Gabriele Schroder, Sandra Lesny, Gregory Sutorius, and particularly
Mr. Andrew Spencer, Senior Editor of Major Reference Works, who served as
our “guide for the perplexed” in all aspects of project management. We thank
our families, and particularly our wives (Jane, Diane, Claire, Hiro, and
Elizabeth) for their support and understanding. In particular, the Senior Editor
wishes to recognize his lifetime partner in all endeavors, Jane A. Avner, Ph.D.,
for her extraordinary editorial assistance. And finally, we thank our mentors,
our students, and most importantly, our patients and their families. Without
them, our work would lack purpose.
Preface vii

Finally, the Editors wish to dedicate this seventh edition of Pediatric Nephrol-
ogy to three former Editors who passed away in 2014. Professors Martin Barratt,
Malcolm A. “Mac” Holiday, and Robert Vernier were extraordinary physician-
scientists who served as mentors to a generation of pediatric nephrologists. This
text would not exist without their efforts, commitment, and selfless contributions.

Ellis D. Avner
William E. Harmon
Patrick Niaudet
Norishige Yoshikawa
Francesco Emma
Stuart L. Goldstein
Contents

Volume 1

Part I Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1

1 Embryonic Development of the Kidney . . . . . . . . . . . . . . . . . 3


Carlton Bates, Jacqueline Ho, and Sunder Sims-Lucas
2 Development of Glomerular Circulation and Function .... 37
Alda Tufro and Ashima Gulati
3 Renal Tubular Development . . . . . . . . . . . . . . . . . . . . . . . . . . 61
Michel Baum
4 Clinical Perinatal Urology . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
David A. Diamond and Richard S. Lee
5 Renal Dysplasia/Hypoplasia . . . . . . . . . . . . . . . . . . . . . . . . . . 115
Paul Goodyer and Indra R. Gupta
6 Developmental Syndromes and Malformations of the
Urinary Tract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Chanin Limwongse

Part II Homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179

7 Physiology of the Developing Kidney: Sodium and Water


Homeostasis and Its Disorders . . . . . . . . . . . . . . . . . . . . . . . . 181
Nigel Madden and Howard Trachtman
8 Physiology of the Developing Kidney: Potassium
Homeostasis and Its Disorder . . . . . . . . . . . . . . . . . . . . . . . . . 219
Lisa M. Satlin and Detlef Bockenhauer
9 Physiology of the Developing Kidney: Acid-Base
Homeostasis and Its Disorders . . . . . . . . . . . . . . . . . . . . . . . . 247
Peter D. Yorgin, Elizabeth G. Ingulli, and Robert H. Mak
10 Bone Developmental Physiology . . . . . . . . . . . . . . . . . . . . . . . 279
MH Lafage-Proust

ix
x Contents

11 Physiology of the Developing Kidney: Disorders and


Therapy of Calcium and Phosphorous Homeostasis . . . . . . . 291
Amita Sharma, Rajesh V. Thakker, and Harald J€uppner

12 Nutrition Management in Childhood Kidney Disease:


An Integrative and Lifecourse Approach . . . . . . . . . . . . . . . . 341
Lauren Graf, Kimberly Reidy, and Frederick J. Kaskel

13 Physiology of the Developing Kidney: Fluid and


Electrolyte Homeostasis and Therapy of Basic
Disorders (Na/H2O/K/Acid Base) . . . . . . . . . . . . . . . . . . . . . . 361
Isa F. Ashoor and Michael J. G. Somers

Part III Translational Research Methods .................... 423

14 Translational Research Methods: Basics of Renal Molecular


Biology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 425
Gian Marco Ghiggeri, Maurizio Bruschi, and
Simone Sanna-Cherchi

15 Translational Research Methods: The Value of Animal


Models in Renal Research . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
Jordan Kreidberg

16 Basics of Clinical Investigation . . . . . . . . . . . . . . . . . . . . . . . . 473


Susan L. Furth and Jeffrey J. Fadrowski

17 Genomic Methods in the Diagnosis and Treatment of


Pediatric Kidney Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 499
Karen Maresso and Ulrich Broeckel

18 Translational Research Methods: Renal Stem Cells . . . . . . . 525


Kenji Osafune

19 Translational Research Methods: Tissue Engineering of the


Kidney and Urinary Tract . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
Austin G. Hester and Anthony Atala

Part IV Clinical Approach to the Child with Suspected Renal


Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 593

20 Clinical Evaluation of the Child with Suspected


Renal Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 595
Mohan A. Shenoy and Nicholas J. A. Webb

21 Laboratory Investigation of the Child with Suspected


Renal Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 613
George van der Watt, Fierdoz Omar, Anita Brink, and
Mignon McCulloch
Contents xi

22 Growth and Development of the Child with


Renal Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 637
Bethany Foster

23 Diagnostic Imaging of the Child with Suspected


Renal Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 667
Jonathan Loewen and Larry A. Greenbaum

24 Pediatric Renal Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . 705


Agnes B. Fogo

Part V Glomerular Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 751

25 Congenital Nephrotic Syndrome . . . . . . . . . . . . . . . . . . . . . . . 753


Hannu Jalanko and Christer Holmberg

26 Inherited Glomerular Diseases . . . . . . . . . . . . . . . . . . . . . . . . 777


Michelle N. Rheault and Clifford E. Kashtan

27 Idiopathic Nephrotic Syndrome in Children: Genetic


Aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 805
Olivia Boyer, Kálmán Tory, Eduardo Machuca, and
Corinne Antignac

28 Idiopathic Nephrotic Syndrome in Children: Clinical


Aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 839
Patrick Niaudet and Olivia Boyer

29 Immune-Mediated Glomerular Injury in Children . . . . . . . . 883


Michio Nagata

30 Complement-Mediated Glomerular Injury in


Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 927
Zoltán Prohászka, Marina Vivarelli, and
George S. Reusz

31 Acute Postinfectious Glomerulonephritis in Children . . . . . . 959


Bernardo Rodríguez-Iturbe, Behzad Najafian,
Alfonso Silva, and Charles E. Alpers

32 Immunoglobulin A Nephropathies in Children


(Includes HSP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 983
Koichi Nakanishi and Norishige Yoshikawa

33 Membranoproliferative and C3-Mediated


GN in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1035
Christoph Licht, Magdalena Riedl, Matthew C. Pickering, and
Michael Braun

34 Membranous Nephropathy in Children . . . . . . . . . . . . . . . . . 1055


Rudolph P. Valentini
xii Contents

Volume 2

Part VI Tubular Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1077

35 Nephronophthisis and Medullary Cystic Kidney Disease in


Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1079
Friedhelm Hildebrandt
36 Childhood Polycystic Kidney Disease . . . . . . . . . . . . . . . . . . . 1103
William E. Sweeney Jr., Meral Gunay-Aygun,
Ameya Patil, and Ellis D. Avner
37 Aminoaciduria and Glycosuria in Children . . . . . . . . . . . . . . 1155
Israel Zelikovic
38 Renal Tubular Disorders of Electrolyte Regulation
in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1201
Olivier Devuyst, Hendrica Belge, Martin Konrad,
Xavier Jeunemaitre, and Maria-Christina Zennaro
39 Renal Tubular Acidosis in Children . . . . . . . . . . . . . . . . . . . . 1273
Raymond Quigley and Matthias T. F. Wolf
40 Nephrogenic Diabetes Insipidus in Children . . . . . . . . . . . . . 1307
Nine V. A. M. Knoers and Elena N. Levtchenko
41 Cystinosis and Its Renal Complications in Children . . . . . . . 1329
William A. Gahl and Galina Nesterova
42 Pediatric Fanconi Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . 1355
Takashi Igarashi
43 Primary Hyperoaxaluria in Children . . . . . . . . . . . . . . . . . . . 1389
Pierre Cochat, Neville Jamieson, and
Cecile Acquaviva-Bourdain
44 Pediatric Tubulointerstitial Nephritis . . . . . . . . . . . . . . . . . . . 1407
Uri S. Alon

Part VII Kidney Involvement in Systemic Diseases . . . . . . . . . . . 1429

45 Renal Involvement in Children with Vasculitis . . . . . . . . . . . 1431


Seza Ozen and Diclehan Orhan
46 Renal Involvement in Children with Systemic Lupus
Erythematosus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1449
Patrick Niaudet, Brigitte Bader-Meunier, and Rémi Salomon
47 Renal Involvement in Children with HUS . . . . . . . . . . . . . . . 1489
Carla M. Nester and Sharon P. Andreoli
Contents xiii

48 Sickle Cell Nephropathy in Children . . . . . . . . . . . . . . . . . . . 1523


Connie Piccone and Katherine MacRae Dell

49 Diabetic Nephropathy in Children . . . . . . . . . . . . . . . . . . . . . 1545


M. Loredana Marcovecchio and Francesco Chiarelli

50 Renal Manifestations of Metabolic Disorders


in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1569
Francesco Emma, William G. van’t Hoff, and
Carlo Dionisi Vici

51 Infectious Diseases and the Kidney in Children . . . . . . . . . . 1609


Jennifer Stevens, Jethro A. Herberg, and Michael Levin

52 Nephrotoxins and Pediatric Kidney Injury . . . . . . . . . . . . . . 1655


Takashi Sekine

Part VIII Urinary Tract Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1693

53 Urinary Tract Infections in Children . . . . . . . . . . . . . . . . . . . 1695


Elisabeth M. Hodson and Jonathan C. Craig

54 Vesicoureteral Reflux and Renal Scarring in Children . . . . . 1715


Tej K. Mattoo, Ranjiv Mathews, and Indra R. Gupta

55 Pediatric Obstructive Uropathy . . . . . . . . . . . . . . . . . . . . . . . 1749


Bärbel Lange-Sperandio

56 Pediatric Bladder Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . 1779


Etienne Berard

57 Urolithiasis in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1821


Vidar Edvardsson

58 Pediatric Renal Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1869


Elizabeth Mullen, Jordan Kreidberg, and
Christopher B. Weldon

Volume 3

Part IX Hypertension . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1905

59 Epidemiology of Hypertension in Children . . . . . . . . . . . . . . 1907


Midori Awazu

60 Pathophysiology of Pediatric Hypertension . . . . . . . . . . . . . . 1951


Ikuyo Yamaguchi and Joseph T. Flynn

61 Evaluation of Hypertension in Childhood Diseases . . . . . . . . 1997


Eileen D. Brewer and Sarah J. Swartz
xiv Contents

62 Management of the Hypertensive Child . . . . . . . . . . . . . . . . . 2023


Demetrius Ellis and Yosuke Miyashita

Part X Acute Renal Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2099

63 Pathogenesis of Acute Kidney Injury . . . . . . . . . . . . . . . . . . . 2101


David P. Basile, Rajasree Sreedharan, and
Scott K. Van Why
64 Evaluation and Management of Acute Kidney Injury in
Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2139
Stuart L. Goldstein and Michael Zappitelli

Part XI Chronic Renal Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2169

65 Pathophysiology of Progressive Renal Disease


in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2171
H. William Schnaper
66 Management of Chronic Kidney Disease in Children . . . . . . 2207
Rene G. VanDeVoorde, Craig S. Wong, and
Bradley A. Warady
67 Handling of Drugs in Children with Abnormal
Renal Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2267
Guido Filler, Amrit Kirpalani, and Bradley L. Urquhart
68 Endocrine and Growth Abnormalities in Chronic
Kidney Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2295
Franz Schaefer
69 Mineral and Bone Disorders in Children with Chronic
Kidney Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2349
Katherine Wesseling-Perry and Isidro B. Salusky
70 Peritoneal Dialysis in Children . . . . . . . . . . . . . . . . . . . . . . . . 2381
Enrico Verrina and Claus Peter Schmitt
71 Hemodialysis in Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2433
Lesley Rees
72 Immunology of Pediatric Renal Transplantation . . . . . . . . . 2457
Elizabeth G. Ingulli, Stephen I. Alexander, and David M. Briscoe
73 Pediatric Renal Transplantation . . . . . . . . . . . . . . . . . . . . . . . 2501
Nancy M. Rodig, Khashayar Vakili, and William E. Harmon
74 Immunosuppression for Pediatric Renal
Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2553
Jodi M. Smith, Thomas L. Nemeth, and Ruth A. McDonald
75 Complications of Pediatric Renal Transplantation . . . . . . . . 2573
Vikas R. Dharnidharka and Carlos E. Araya
Contents xv

Part XII IPNA: Global Pediatric Nephrology . . . . . . . . . . . . . . . . . 2605

76 IPNA: Global Pediatric Nephrology, Introduction and


Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2607
Pierre Cochat and Isidro B. Salusky
77 AFPNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2613
Mignon McCulloch, Hesham Safouh, Amal Bourquia, and
Priya Gajjar
78 ALANEPE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2631
Vera Koch, Nelson Orta, and Ramon Exeni
79 AsPNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2639
Hui-Kim Yap, Man-Chun Chiu, Arvind Bagga, and
Hesham Safouh
80 Pediatric Nephrology in North America . . . . . . . . . . . . . . . . 2665
Victoria F. Norwood and Maury Pinsk
81 ANZPNA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2673
Deborah Lewis
82 ESPN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2681
Rosanna Coppo
83 JSPN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2687
Kazumoto Iijima
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2697
Contributors

Cecile Acquaviva-Bourdain Centre de référence des maladies rénales rares


Néphrogones, Hôpital Femme Mère Enfant, Hospices Civils de Lyon &
Université de Lyon, Lyon, France
Service Maladies Héréditaires du Métabolisme et Dépistage Néonatal, Centre
de Biologie et Pathologie Est, Hospices Civils de Lyon, Lyon, France

Stephen I. Alexander Discipline of Paediatrics and Child Health, The


University of Sydney, Sydney, New South Wales, Australia
Division of Nephrology, Children’s Hospital at Westmead, Sydney, New
South Wales, Australia

Uri S. Alon Section of Pediatric Nephrology, The Children’s Mercy Hospital


and Clinics, University of Missouri at Kansas City, School of Medicine,
Kansas City, MO, USA

Charles E. Alpers Department of Pathology, University of Washington,


Seattle, WA, USA

Sharon P. Andreoli Division of Nephrology, Indianapolis, IN, USA

Corinne Antignac Laboratory of Hereditary Kidney Diseases, Inserm UMR


1163, Paris, France
Paris Descartes, Sorbonne Paris Cité University, Imagine Institute, Paris,
France
Department of Genetics, MARHEA reference center, Necker – Enfants
Malades Hospital, Paris, France

Carlos E. Araya University of Central Florida and Nemours Children’s


Hospital, Orlando, FL, USA

Isa F. Ashoor Division of Nephrology, Children’s Hospital, New Orleans,


LA, USA

Anthony Atala School of Medicine, Department of Urology, Wake Forest


Institute for Regenerative Medicine, Wake Forest University, Winston Salem,
NC, USA

xvii
xviii Contributors

Ellis D. Avner Department of Pediatrics, Medical College of Wisconsin,


Children’s Research Institute, Children’s Hospital Health System of Wiscon-
sin, Milwaukee, WI, USA

Midori Awazu Department of Pediatrics, Keio University School of Medi-


cine, Shinjuku-ku, Tokyo, Japan

Brigitte Bader-Meunier Service d’Immunologie et Rhumatologie


Pédiatrique, Hôpital Necker-Enfants Malades, Paris, France

Arvind Bagga Division of Nephrology, All India Institute of Medical


Sciences, New Delhi, India

David P. Basile Indiana University School of Medicine, Indianapolis, IN, USA

Carlton Bates Department of Pediatrics, Division of Pediatric Nephrology,


Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA

Michel Baum Departments of Pediatrics and Internal Medicine, University


of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA

Hendrica Belge Zurich Institute for Human Physiology, University of


Zurich, Institute of Physiology, Zurich Center for Integrative Human Physiol-
ogy (ZIHP), Z€ urich, Switzerland

Etienne Berard Pediatric Nephrology Unit, Universitary Hospital of Nice


(France), Nice, France

Detlef Bockenhauer Great Ormond Street Hospital, Institute of Child


Health, University College London, London, UK

Amal Bourquia Paediatric Nephology, Red Cross War Memorial Children’s


Hospital, Dept. of Paediatric Medicine, University of Cape Town, Western
Cape, South Africa

Olivia Boyer Service de Néphrologie Pédiatrique, Hôpital Necker-Enfants


Malades, Université Paris-Descartes, Paris, France

Michael Braun Renal Section, Department of Pediatrics, Texas Children’s


Hospital, Balyor College of Medicine, Houston, TX, USA

Eileen D. Brewer Department of Pediatrics Renal Section, Baylor College of


Medicine and Texas Children’s Hospital, Houston, TX, USA

Anita Brink Department of Pediatrics and Child Health (Nuclear Medicine),


University of Cape Town, Red Cross War Memorial Children’s Hospital, Cape
Town, South Africa

David M. Briscoe Department of Pediatrics, Harvard Medical School,


Boston, MA, USA
Division of Nephrology, Transplant Research Program, Boston Children’s
Hospital, Boston, MA, USA
Contributors xix

Ulrich Broeckel Department of Pediatrics, Medical College of Wisconsin


and Children’s Hospital of Wisconsin, Milwaukee, WI, USA
Maurizio Bruschi Laboratory of Physiopathology of Uremia, Division of
Nephrology, Dialysis and Transplantation, Istituto Giannina Gaslini, Genoa,
Italy
Francesco Chiarelli University of Chieti, Chieti, Italy
Man-Chun Chiu Department of Pediatrics and Adolescent Medicine, Prin-
cess Margaret Hospital, Hong Kong University, Kowloon, Hong Kong
Pierre Cochat Centre de référence des maladies rénales rares Néphrogones,
Hôpital Femme Mère Enfant, Hospices Civils de Lyon & Université de Lyon,
Lyon, France
IBCP-UMR 5305 CNRS, Université Claude-Bernard Lyon 1, Lyon, France
Rosanna Coppo Nephrology Dialysis and Transplantation Unit, Azienda
Ospedaliera-Universitaria Città della Salute e della Scienza di Torino, Regina
Margherita Children’s University Hospital, Turin, Italy
Jonathan C. Craig Centre for Kidney Research, The Children’s Hospital at
Westmead, Westmead, Sydney, NSW, Australia
Sydney School of Public Health, University of Sydney, Sydney, NSW,
Australia
Katherine MacRae Dell Center for Pediatric Nephrology, Department of
Pediatrics, Cleveland Clinic Children’s and Case Western Reserve University,
Cleveland, OH, USA
Olivier Devuyst Zurich Institute for Human Physiology, University of
Zurich, Institute of Physiology, Zurich Center for Integrative Human Physiol-
ogy (ZIHP), Z€urich, Switzerland
Vikas R. Dharnidharka Pediatric Nephrology, Washington University
School of Medicine and St. Louis Children’s Hospital, St. Louis, MO, USA
David A. Diamond Department of Urology, Harvard Medical School,
Boston Children’s Hospital, Boston, MA, USA
Carlo Dionisi Vici Division of Metabolic Diseases, Bambino Gesù
Children’s Hospital – IRCCS, Rome, Italy
Vidar Edvardsson Landspitali – The National University Hospital of Ice-
land, Reykjavik, Iceland and Faculty of Medicine, School of Health Sciences,
University of Iceland, Reykjavik, Iceland
Demetrius Ellis Department of Pediatrics, Division of Pediatric Nephrology,
Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA
Francesco Emma Division of Nephrology, Bambino Gesù Children’s
Hospital – IRCCS, Rome, Italy
xx Contributors

Ramon Exeni Department of Nephrology, Children’s Hospital “San Justo”,


Buenos Aires, Argentina

Jeffrey J. Fadrowski Department of Pediatrics, John Hopkins University


School of Medicine, Baltimore, MD, USA

Guido Filler Departments of Pediatrics, Medicine, and Pathology Laboratory


Medicine, Schulich School of Medicine and Dentistry, Western University,
London, ON, Canada
Children’s Hospital of Western Ontario, Children’s Health Research Institute
(CHRI), London, ON, Canada

Joseph T. Flynn Division of Nephrology, Seattle Children’s Hospital;


Department of Pediatrics, University of Washington School of Medicine,
Seattle, WA, USA

Agnes B. Fogo Department of Pathology, Microbiology and Immunology,


Vanderbilt University Medical Center, Nashville, TN, USA

Bethany Foster The Research Institute of the McGill University Health


Centre, Montreal, QC, Canada

Susan L. Furth Departments of Pediatrics and Epidemiology, Perelman


School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA

William A. Gahl Section on Human Biochemical Genetics, Medical Genet-


ics Branch, National Human Genome Research Institute, National Institutes of
Health, Bethesda, MD, USA

Priya Gajjar Paediatric Nephology, Red Cross War Memorial Children’s


Hospital, Dept. of Paediatric Medicine, University of Cape Town, Western
Cape, South Africa

Gian Marco Ghiggeri Laboratory of Physiopathology of Uremia, Division


of Nephrology, Dialysis and Transplantation, Istituto Giannina Gaslini,
Genoa, Italy

Stuart L. Goldstein Division of Nephrology and Hypertension, The Heart


Institute, Cincinnati Children’s Hospital Medical Center, College of Medicine,
Cincinnati, OH, USA

Paul Goodyer Division of Pediatric Nephrology, Montreal Children’s


Hospital, McGill University, Montreal, QC, Canada

Lauren Graf Children’s Hospital at Montefiore, Albert Einstein College of


Medicine, Bronx, NY, USA

Larry A. Greenbaum Department of Pediatric Radiology, Emory University


School of Medicine, Atlanta, GA, USA

Ashima Gulati Department of Pediatrics, Nephrology Section, Yale School


of Medicine, New Haven, CT, USA
Contributors xxi

Meral Gunay-Aygun Medical Genetics Branch, The Intramural Program of


the Office of Rare Diseases, National Human Genome Research Institute,
Bethesda, MD, USA
Department of Pediatrics, McKusick-Nathans Institute of Genetic Medicine,
Johns Hopkins University School of Medicine, Baltimore, MD, USA
Indra R. Gupta Department of Pediatrics and Department of Human Genet-
ics, Division of Pediatric Nephrology, Montreal Children’s Hospital, McGill
University, Montréal, QC, Canada
William E. Harmon Boston Children’s Hospital, Harvard Medical School,
Boston, MA, USA
Jethro A. Herberg Imperial College London, London, UK
Austin G. Hester School of Medicine, Department of Urology, Wake Forest
Institute for Regenerative Medicine, Wake Forest University, Winston Salem,
NC, USA
Friedhelm Hildebrandt Harvard Medical School, Boston, MA, USA
Jacqueline Ho Department of Pediatrics, Division of Pediatric Nephrology,
Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh School
of Medicine, Pittsburgh, PA, USA
Elisabeth M. Hodson Centre for Kidney Research, The Children’s Hospital
at Westmead, Westmead, Sydney, NSW, Australia
Sydney School of Public Health, University of Sydney, Sydney, NSW,
Australia
Christer Holmberg Hospital for Children and Adolescents, University of
Helsinki, Helsinki, Finland
Takashi Igarashi National Center for Child Health and Development
(NCCHD), Tokyo, Japan
Kazumoto Iijima Department of Pediatrics, Kobe University Graduate
School of Medicine, Kobe, Japan
Elizabeth G. Ingulli Department of Pediatrics, University of California,
San Diego, CA, USA
Division of Nephrology, Rady Children’s Hospital San Diego, San Diego,
CA, USA
Kidney Transplant Program, Rady Children’s Hospital, San Diego, CA, USA
Hannu Jalanko Hospital for Children and Adolescents, University of
Helsinki, Helsinki, Finland
Neville Jamieson Department of Surgery, Addenbrookes Hospital,
Cambridge University Teaching Hospitals, Cambridge, UK
Xavier Jeunemaitre Department of Molecular Genetics, Hôpital Européen
George Pompidou, Paris, France
xxii Contributors

Harald J€uppner Departments of Medicine and Pediatrics, Endocrine Unit


and Pediatric Nephrology Unit, Massachusetts General Hospital and Harvard
Medical School, Boston, MA, USA
Clifford E. Kashtan Department of Pediatrics, Division of Pediatric
Nephrology, University of Minnesota Masonic Children’s Hospital, Minneap-
olis, MN, USA
Frederick J. Kaskel Division of Pediatric Nephrology, Children’s Hospital
at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
Amrit Kirpalani Departments of Pediatrics, Schulich School of Medicine
and Dentistry, Western University, London, ON, Canada
Nine V. A. M. Knoers Departments of Medical Genetics, University Medical
Centre Utrecht, Utrecht, The Netherlands
Vera Koch Instituto da Criança- Pediatric Nephrology Unit, Department of
Pediatrics, University of Sao Paulo Medical School, Sao Paulo, Brazil
Martin Konrad Department of General Pediatrics, Pediatric Nephrology,
University Hospital, M€unster, Germany
Jordan Kreidberg Children’s Hospital Boston, Boston, MA, USA
MH Lafage-Proust INSERM U 1059, Université de Lyon, Saint-Etienne,
France
Bärbel Lange-Sperandio Dr. v. Hauner Children’s Hospital, Department of
Pediatric Nephrology, LMU, Munich, Germany
Richard S. Lee Department of Urology, Harvard Medical School, Boston
Children’s Hospital, Boston, MA, USA
Michael Levin Department of Medicine, Imperial College London, London,
UK
Elena N. Levtchenko Department of Pediatric Nephrology, Department of
Growth and Regeneration, University Hospitals Leuven, Katholieke
Universiteit Leuven, Leuven, Belgium
Deborah Lewis Sydney, NSW, Australia
Christoph Licht Division of Nephrology, The Hospital for Sick Children,
University of Toronto, Toronto, ON, Canada
Research Institute, Cell Biology Program, The Hospital for Sick Children,
Toronto, ON, Canada
Department of Paediatrics, University of Toronto, Toronto, ON, Canada
Chanin Limwongse Department of Medicine, Division of Medical Genetics,
Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkoknoi,
Bangkok, Thailand
Jonathan Loewen Department of Pediatric Radiology, Emory University
School of Medicine, Atlanta, GA, USA
Contributors xxiii

Eduardo Machuca Department of Nephrology, Medical School, Pontificia


Universidad Católica de Chile, Santiago, Chile
Nigel Madden NYU Langone Medical Center and NYU School of
Medicine, New York, NY, USA
Robert H. Mak Pediatric Nephrology, University of California, San Diego,
CA, USA
Pediatric Nephrology Division, Rady Children’s Hospital, San Diego, CA, USA
M. Loredana Marcovecchio University of Chieti, Chieti, Italy
Karen Maresso Section of Genomic Pediatrics, Medical College of
Wisconsin, Milwaukee, WI, USA
Ranjiv Mathews The Nevada School of Medicine, The Johns Hopkins
School of Medicine, Las Vegas, NV, USA
Tej K. Mattoo Pediatric Nephrology and Hypertension, Wayne State
University School of Medicine, Children’s Hospital of Michigan, Detroit,
MI, USA
Mignon McCulloch Department of Paediatric Intensive Care/Nephrology,
University of Cape Town, Red Cross War Memorial Children’s Hospital, Cape
Town, Western Cape, South Africa
Ruth A. McDonald Division of Nephrology, Seattle Children’s, University
of Washington, Seattle, WA, USA
Yosuke Miyashita Department of Pediatrics, Division of Pediatric Nephrol-
ogy, Children’s Hospital of Pittsburgh of UPMC, University of Pittsburgh
School of Medicine, Pittsburgh, PA, USA
Elizabeth Mullen Hematology Oncology, Dana-Farber/Boston Children’s
Blood Disorders and Cancer Center, Boston, MA, USA
Michio Nagata Graduate School of Comprehensive Human Sciences,
University of Tsukuba, Tsukuba, Japan
Behzad Najafian Department of Pathology, University of Washington,
Seattle, WA, USA
Koichi Nakanishi Department of Pediatrics, Wakayama Medical University,
Wakayama City, Japan
Thomas L. Nemeth Department of Pharmacy, Seattle Children’s, University
of Washington, Seattle, WA, USA
Carla M. Nester Stead Family Department of Pediatrics, Department of
Internal Medicine, Divisions of Nephrology, University of Iowa Children’s
Hospital, Carver College of Medicine, Iowa City, IA, USA
Galina Nesterova Section on Human Biochemical Genetics, Medical Genet-
ics Branch, National Human Genome Research Institute, National Institutes of
Health, Bethesda, MD, USA
xxiv Contributors

Patrick Niaudet Service de Néphrologie Pédiatrique, Hôpital Necker-


Enfants Malades, Université Paris-Descartes, Paris, France
Victoria F. Norwood University of Virginia, Charlottesville, VA, USA
Fierdoz Omar Chemical Pathology, University of Cape Town and National
Health Laboratory Service, Red Cross Children’s Hospital and Groote Schuur
Hospital, Cape Town, South Africa
Diclehan Orhan Department of Pediatric Pathology, Hacettepe University,
Sihhiye, Ankara, Turkey
Nelson Orta Service of Pediatric Nephrology, Children’s Hospital “Jorge
Lizarraga”, University of Carabobo, Valencia, Venezuela
Kenji Osafune Center for iPS Cell Research and Application (CiRA), Kyoto
University, Sakyo-ku, Kyoto, Japan
Seza Ozen Department of Pediatrics, Faculty of Medicine, Hacettepe
University, Sihhiye, Ankara, Turkey
Ameya Patil Department of Pediatrics, Medical College of Wisconsin, Chil-
dren’s Research Institute, Children’s Hospital Health System of Wisconsin,
Milwaukee, WI, USA
Connie Piccone Rainbow Babies and Children’s Hospital, Cleveland,
OH, USA
Matthew C. Pickering Centre for Complement and Inflammation Research,
Imperial College, London, UK
Maury Pinsk University of Alberta, Edmonton, AB, Canada
Zoltán Prohászka 3rd Department of Medicine, Faculty of Medicine,
Semmelweis University, Budapest, Hungary
Raymond Quigley Department of Pediatrics, University of Texas South-
western Medical Center at Dallas, Dallas, TX, USA
Lesley Rees Department of Nephrology, Great Ormond Street Hospital for
Children NHS Trust, London, UK
Kimberly Reidy Division of Pediatric Nephrology, Children’s Hospital at
Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
George S. Reusz 1st Department of Pediatrics, Faculty of Medicine,
Semmelweis University, Budapest, Hungary
Michelle N. Rheault Department of Pediatrics, Division of Pediatric
Nephrology, University of Minnesota Masonic Children’s Hospital, Minneap-
olis, MN, USA
Magdalena Riedl Research Institute, Cell Biology Program, The Hospital
for Sick Children, Toronto, ON, Canada
Department of Paediatrics, Innsbruck Medical University, Innsbruck, Tyrol,
Austria
Contributors xxv

Nancy M. Rodig Boston Children’s Hospital, Harvard Medical School,


Boston, MA, USA
Bernardo Rodríguez-Iturbe Nephrology Service, Hospital Universitario,
Maracaibo, Estado Zulia, Venezuela
Hesham Safouh Faculty of Medicine, Center for Pediatric Nephrology and
Transplantation (CPNT), Cairo University, Orman, Giza, Egypt
Rémi Salomon Service de Néphrologie Pédiatrique, Hôpital Necker-Enfants
Malades, Université Paris-Descartes, Paris, France
Isidro B. Salusky Division of Pediatric Nephrology, Clinical Translational
Research Center, David Geffen School of Medicine at UCLA, Los Angeles,
CA, USA
Simone Sanna-Cherchi Division of Nephrology, Columbia University,
College of Physicians and Surgeons, New York, NY, USA
Lisa M. Satlin Department of Pediatrics, Division of Pediatric Nephrology,
Icahn School of Medicine at Mount Sinai, New York, NY, USA
Franz Schaefer Division of Pediatric Nephrology, University Children’s
Hospital, Heidelberg, Germany
Claus Peter Schmitt Centre for Pediatric and Adolescent Medicine, Heidel-
berg, Germany
H. William Schnaper Division of Kidney Diseases, Ann and Robert H. Lurie
Children’s Hospital of Chicago, Department of Pediatrics, Northwestern
University Feinberg School of Medicine, Chicago, IL, USA
Takashi Sekine Department of Pediatrics, Toho University Faculty of
Medicine, Meguro-ku, Tokyo, Japan
Amita Sharma Department of Pediatrics, Pediatric Nephrology Unit, Mas-
sachusetts General Hospital and Harvard Medical School, Boston, MA, USA
Mohan A. Shenoy Department of Pediatric Nephrology, Royal Manchester
Children’s Hospital, Manchester, UK
Alfonso Silva Nephrology Service, Hospital Universitario, Maracaibo,
Estado Zulia, Venezuela
Sunder Sims-Lucas Department of Pediatrics, Division of Pediatric
Nephrology, Children’s Hospital of Pittsburgh of UPMC, University of Pitts-
burgh School of Medicine, Pittsburgh, PA, USA
Jodi M. Smith Division of Nephrology, Seattle Children’s, University of
Washington, Seattle, WA, USA
Michael J. G. Somers Division of Nephrology, Boston Children’s Hospital,
Harvard Medical School, Boston, MA, USA
Rajasree Sreedharan Medical College of Wisconsin, Milwaukee, WI, USA
Jennifer Stevens University Hospital Wales, Cardiff, S. Wales, UK
xxvi Contributors

Sarah J. Swartz Department of Pediatrics Renal Section, Baylor College of


Medicine and Texas Children’s Hospital, Houston, TX, USA
William E. Sweeney Jr. Department of Pediatrics, Medical College of Wis-
consin, Children’s Research Institute, Children’s Hospital Health System of
Wisconsin, Milwaukee, WI, USA
Rajesh V. Thakker Radcliffe Department of Medicine, Academic Endocrine
Unit, University of Oxford, OCDEM (Oxford Centre for Diabetes, Endocri-
nology and Metabolism), The Churchill Hospital Headington, Oxford, UK
Kálmán Tory Laboratory of Hereditary Kidney Diseases, Inserm UMR
1163, Paris, France
Department of Pediatrics, Semmelweis University, Budapest, Hungary
Howard Trachtman NYU Langone Medical Center and NYU School of
Medicine, New York, NY, USA
Alda Tufro Department of Pediatrics, Nephrology Section, Yale School of
Medicine, New Haven, CT, USA
Bradley L. Urquhart Departments of Pediatrics, Department of Medicine,
Physiology and Pharmacology, Western University, London, ON, Canada
Children’s Health Research Institute (CHRI), London, ON, Canada
Khashayar Vakili Boston Children’s Hospital, Harvard Medical School,
Boston, MA, USA
Rudolph P. Valentini Pediatric Nephrology, Children’s Hospital of Michi-
gan, Detroit, MI, USA
Wayne State University School of Medicine, Detroit, MI, USA
George van der Watt Chemical Pathology, University of Cape Town and
National Health Laboratory Service, Red Cross Children’s Hospital and
Groote Schuur Hospital, Cape Town, South Africa
Scott K. Van Why Medical College of Wisconsin, Milwaukee, WI, USA
William G. van’t Hoff Great Ormond Street Hospital, London, UK
Rene G. VanDeVoorde Cincinnati Children’s Hospital Medical Center,
Cincinnati, OH, USA
Enrico Verrina Nephrology, Dialysis and Transplantation Unit, Giannina
Gaslini Childrens Hospital, Genoa, Italy
Marina Vivarelli Division of Nephrology and Dialysis, Children’s Hospital
Bambino Gesù-IRCCS, Rome, Italy
Bradley A. Warady Pediatric Nephrology, Children’s Mercy Hospital,
Kansas City, MO, USA
Nicholas J. A. Webb Department of Pediatric Nephrology, Royal Manches-
ter Children’s Hospital, Manchester, UK
Contributors xxvii

Christopher B. Weldon Department of Surgery, Boston Children’s Hospital


and Harvard Medical School, Boston, MA, USA
Katherine Wesseling-Perry Division of Pediatric Nephrology, David
Geffen School of Medicine at UCLA, Los Angeles, CA, USA
Matthias T. F. Wolf Department of Pediatrics, University of Texas South-
western Medical Center at Dallas, Dallas, TX, USA
Craig S. Wong Pediatric Nephrology, University of New Mexico Health
Sciences Center, Albuquerque, NM, USA
Ikuyo Yamaguchi Division of Pediatric Nephrology, University of Texas
School of Medicine at San Antonio, University of Texas Health Science Center
at San Antonio, San Antonio, TX, USA
Hui-Kim Yap Yong Loo Lin School of Medicine, National University of
Singapore, Singapore, Singapore
Peter D. Yorgin Pediatric Nephrology, University of California, San Diego,
CA, USA
Pediatric Nephrology Division, Rady Children’s Hospital, San Diego,
CA, USA
Norishige Yoshikawa Department of Pediatrics, Wakayama Medical
University, Wakayama City, Japan
Michael Zappitelli Pediatrics, Division of Nephrology, Montreal Children’s
Hospital, McGill University Health Center, Montreal, QC, Canada
Israel Zelikovic Department of Physiology and Biophysics, Faculty of
Medicine, Technion – Israel Institute of Technology, Haifa, Israel
Division of Pediatric Nephrology, Rambam Medical Center, Haifa, Israel
Maria-Christina Zennaro Inserm U970, Paris Cardiovascular Research
Center, Paris, France
Part I
Development
Embryonic Development of the Kidney
1
Carlton Bates, Jacqueline Ho, and Sunder Sims-Lucas

Contents Vascular Development of the Kidney . . . . . . . . . . . . . . 14


Angiogenesis Versus Vasculogenesis . . . . . . . . . . . . . . . . . 15
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Origins of the Peritubular Capillary Endothelia . . . . . . 16
Studying the Kidney and Urinary Tract Molecular Control of Renal Vascular
Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5 Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17

Development of the Metanephros . . . . . . . . . . . . . . . . . . 7 Collecting System Development . . . . . . . . . . . . . . . . . . . . 17


Ureteric Bud Induction and Outgrowth . . . . . . . . . . . . . . . 17
Nephron Formation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8 Renal Branching Morphogenesis . . . . . . . . . . . . . . . . . . . . . 19
Specification of Nephron Progenitors/Cap Patterning of the Medullary and Cortical Collecting
Mesenchyme . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 Ducts . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
Nephron Induction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Nephron Segmentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 Lower Urinary Tract Development . . . . . . . . . . . . . . . . 21
Glomerulogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12 Anatomic and Functional Development . . . . . . . . . . . . . . 21
Molecular Control of Podocyte Terminal Molecular Control of Ureter and Bladder
Differentiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
Molecular Control of Glomerular Capillary Tuft Bladder . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 Ureter–Bladder Anastomosis . . . . . . . . . . . . . . . . . . . . . . . . . 25

Renal Stroma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

C. Bates (*) • J. Ho • S. Sims-Lucas


Department of Pediatrics, Division of Pediatric
Nephrology, Children’s Hospital of Pittsburgh of UPMC,
University of Pittsburgh School of Medicine, Pittsburgh,
PA, USA
e-mail: batescm@upmc.edu; jacqueline.ho2@chp.edu;
sunder.sims-lucas@chp.edu

# Springer-Verlag Berlin Heidelberg 2016 3


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_1
4 C. Bates et al.

Introduction morphogenesis, and regulation of cell number.


Differentiation is the process by which precursor
The mammalian kidney functions as a key regu- cells or tissues mature into more specialized cells.
lator of water balance, acid–base homeostasis, During kidney development, renal mesenchymal
maintenance of electrolytes, and waste excretion. cells have the potential to differentiate into
The performance of these activities depends on nephron epithelia or stromal cells and interstitial
the development of specific cell types in a precise fibroblasts. Morphogenesis describes the
temporal and spatial pattern, to produce a suffi- process whereby cells and tissues acquire three-
cient number of nephrons. Over the past several dimensional patterns. This is particularly impor-
decades, considerable advances have been made tant in the kidney, as the three-dimensional rela-
in understanding the molecular basis for this tionship between the nephrons, the vasculature,
developmental program. Defects in this program and the collecting system is critical for
result in congenital anomalies of the kidney and normal kidney structural function. Finally, the
urinary tract, which are the leading causes of regulation of cell number at different stages of
chronic kidney disease and renal failure in chil- development is crucial. Such regulation maintains
dren. These developmental disorders range from a balance between cellular proliferation and
renal malformations, such as renal aplasia programmed cell death or apoptosis. All of these
(absence of the kidney), dysplasia (failure of nor- processes are integrated tightly and regulated both
mal renal differentiation), and hypoplasia (smaller spatially and temporally in normal renal
kidneys), to urinary tract abnormalities such as development.
vesicoureteral reflux and duplicated collecting The molecular control of these developmental
systems. This chapter describes the embryology processes has been the subject of several recent
of the kidney and urinary tract, as a means to comprehensive reviews [8–14]. Genetic, epige-
understand the developmental origins of these netic, and environmental factors all regulate dif-
disorders. ferentiation, morphogenesis, and cell number
Human kidney development starts in the fifth within the developing kidney. Mutational ana-
week of gestation, and new nephrons are formed lyses in animal models have provided significant
until approximately 32–34 weeks gestation insights into the genetic control of renal develop-
[1–3]. Remarkably, nephron endowment is quite ment. Genes critical for kidney development in
variable ranging from 200,000 to 1.8 million animal models include transcription factors that
nephrons per person [4]. While the human kidney act as master regulators of other genes, growth
continues to grow after 34 weeks gestation, this factors that signal to other cells, and adhesion
occurs due to the growth and maturation of molecules that regulate how cells interact with
existing nephrons, rather than the formation of each other and with the extracellular matrix.
new nephrons. The mature mammalian kidney Increasingly, analyses of humans with congenital
cannot compensate for nephron loss due to renal renal malformations (such as renal aplasia or
injury by the de novo generation of nephrons duplex kidneys) have identified gene mutations
[2, 5]. Therefore, the number of nephrons present originally described in animal models [15]. Recent
at birth in an individual is an important determi- studies have also implicated epigenetic mecha-
nant of long-term kidney health. Therefore, nisms (defined as heritable changes in gene activ-
reduced nephron number is associated with hyper- ity that are not caused by changes in DNA
tension and chronic kidney disease [6, 7]. sequence) in regulating nephron formation. Two
Critical determinants of nephron endowment major classes of epigenetic molecules that appear
are structural development and three-dimensional to regulate renal development include chromatin
nephron patterning. The formation of kidneys in remodeling proteins and regulatory RNAs such as
utero involves the coordinated regulation of miRNAs [16–22]. Finally, environmental influ-
critical developmental processes: differentiation, ences that may interact with genetic and
1 Embryonic Development of the Kidney 5

epigenetic factors are also important in determin- growth limitations from diffusion of the culture
ing nephron number and patterning. For example, media across the air–media interface, and distor-
vitamin A deficiency leads to decreased nephron tions in the three-dimensional kidney architecture
number in rodents and has been implicated in as explants flatten in culture.
decreased renal size in humans [23, 24]. Recently, several methods to generate more
physiological and quantifiable three-dimensional
reconstructions of developing kidneys and urinary
Studying the Kidney and Urinary Tract tracts have been developed. One method utilizes
Development exhaustive serial sectioning through developing
kidneys, histological staining, projection of each
The methods for studying the molecular and serial image onto a monitor to identify each tissue
genetic control of kidney development have con- lineage, and rendering of the serial images into a
tinued to evolve over the past several decades. three-dimensional image (Fig. 1e) [31, 32]. This
Visualization of tissue morphology and expres- technique allows for the quantification of both
sion of individual genes and proteins in the devel- developing nephron structures and the branching
oping kidney have traditionally been performed ureteric tree. Another method uses optical projec-
on tissue sections by general staining (e.g., hema- tion tomography to image through the full thick-
toxylin and eosin), detecting messenger RNA ness of a developing kidney that has been whole
(mRNA) via in situ hybridization or protein via mount stained for a specific tissue and also per-
immunohistochemistry (Fig. 1a–c). The advent of mits the quantification of these elements [33].
high-throughput technologies to detect gene Physical, chemical, and genetic strategies can
expression with microarrays and more recently be used to manipulate developing kidneys in vivo.
by high-throughput RNA sequencing has resulted For example, ureteric obstruction in utero in sheep
in the ability to assay the transcriptome of the and monkeys results in hydronephrotic kidneys
developing kidney and/or different kidney tissue with renal dysplasia [34, 35]. In addition, dietary
compartments in an unbiased fashion. These tech- manipulations including high doses of vitamin A or
niques have led to large public databases that dietary protein restriction result in kidney and uri-
describe the gene expression of the developing nary tract defects [36, 37]. Transgenic approaches
kidney, including the GenitoUrinary Develop- have also been used to drive gene expression in
ment Molecular Anatomy Project (URL: www. specific spatiotemporal patterns, usually in the
gudmap.org) and Eurexpress (URL: www. mouse. In these experiments, a transgenic construct
eurexpress.org) [25–27]. consisting of a tissue-specific promoter and the
A classical technique to analyze kidney devel- gene of interest is randomly inserted into the
opment is to culture rodent embryonic kidneys genome, leading to expression of that gene in a
in vitro as explants. Studies using these methods tissue-specific pattern. The limitations of this
were the first to show that reciprocal interactions approach include: (1) that the random insertion
between the metanephric mesenchyme and the can result in unintended changes in gene expres-
ureteric bud are critical to induce the formation sion (due to other nearby promoters/enhancers near
of new nephrons and ureteric branching (Fig. 1d) the site of integration), (2) that the insertion of the
[28]. Moreover, kidney explants still allow one to transgene into the genome may lead to loss of
modulate the expression and function of specific function of an endogenous gene, and that (3) epige-
genes and proteins using reagents such as anti- netic factors may silence the construct. The
sense oligonucleotides or blocking antibodies increased utilization of bacterial artificial chromo-
[29, 30]. While these experiments have been illu- some (BAC) constructs, which contain more of
minating, the growth of embryonic kidney endogenous promoter elements than are found in
explants differs from kidney development traditional plasmid constructs, has led to more
in vivo in several key ways: lack of blood flow, faithful and reliable transgene expression.
6 C. Bates et al.

Fig. 1 Experimental methods utilized to study kidney demonstrating branching ureteric structures (green) after
development. (a) Hematoxylin and eosin (H&E)-stained 5 days of growth. (e) 3D reconstruction of an embryonic
tissue section of a control postnatal day 0 mouse kidney. day 13.5 mouse kidney with the ureteric epithelium
The ureteric bud is outlined in yellow, and the arrow points depicted in pink and developing nephron types including
to the cap metanephric mesenchyme. (b) In situ hybridiza- renal vesicles (blue), comma-shaped bodies (red),
tion in a control mouse embryonic day 16.5 tissue section S-shaped bodies (purple), and glomeruli (green)
for the transcription factor, Wt1, which stains the meta- (Reproduced with kind permission from Springer Science
nephric mesenchyme and developing glomeruli. (c) Immu- +Business Media: Sims-Lucas S. Kidney Development:
nofluorescent staining in a control embryonic day 14.5 Methods and Protocols, Methods in Molecular Biology,
mouse tissue section for the transcription factor, Wt1 vol. 886, 2012, pp 81, Figure 3F) (f) H&E-stained tissue
(red), and lotus tetragonolobus lectin (LTL, green), which section of a postnatal day 0 mouse kidney lacking
stains the proximal tubule. (d) Embryonic culture of a microRNAs in the ureteric lineage using a conditional
transgenic embryonic day 11.5 HoxB7GFP mouse kidney, knockout approach (HoxB7Cre; Dicerflx/flx)
1 Embryonic Development of the Kidney 7

As opposed to transgenic approaches, homol- Development of the Metanephros


ogous recombination is the method whereby a
gene is “knocked-out of” or “knocked-into” the The mesoderm forms as one of the three embryonic
mouse genome. Using these methods, a gene of germ layers during gastrulation. The mammalian
interest is deleted so that it becomes kidney develops from the intermediate mesoderm,
nonfunctional, or a gene (such as a green fluores- lying between the somites and lateral plate meso-
cent reporter) is added to the genome at a specific derm, on the posterior abdominal wall of the devel-
locus. A limitation to traditional knockout tech- oping embryo. In mammals, three pairs of
niques is that global loss of function of the gene embryonic kidneys develop from the intermediate
may result in extrarenal effects (such as early mesoderm: the pronephros, the mesonephros, and
embryonic lethality), which can impact or the metanephros (Fig. 2). At their maximal devel-
severely limit the study of the gene’s function in opment, the pronephros and mesonephros extend
the kidney. Given these limitations, it has become from the cervical to the lumbar levels of the devel-
more common to perform conditional gene oping embryo. The pronephric and mesonephric
targeting (e.g., with the Cre–loxP system) nephrons are induced to differentiate by signals
(Fig. 1f) and/or inducible gene targeting (e.g., from the adjacent pronephric/mesonephric ducts,
with tamoxifen), allowing for kidney- and/or uri- paired epithelial tubules running in a longitudinal
nary tract-specific gene deletion (using a kidney course along the embryo on either side of the
zebrafish specific Cre-) and/or at a particular time midline. The mesonephric duct (also known as
(driving induction of gene targeting with a drug). the Wolffian duct) continues to grow caudally in
While most investigators using genetically the embryos to eventually fuse with the cloaca,
modified animals utilize mice, a growing number which eventually gives rise to the bladder. The
of scientists study kidney development in other pronephros is not functional in mammals, but is
model systems, such as avians, zebrafish, and the functional kidney in larval fish [40] and frogs
Xenopus. These simple systems are obviously [41]. The mesonephros becomes the mature kidney
limited by their inability to recapitulate the com- in these lower species and is functional in mam-
plex regulation of development previously mals during embryogenesis. Ultimately, the pro-
described in three-dimensional mammalian kid- nephros and mesonephros largely degenerate in
ney formation. However, their simplicity has mammals. Portions of the mesonephros and meso-
advantages in isolating possible molecular path- nephric duct persist in mammals as the rete testis,
ways involved in specific renal developmental efferent ducts, epididymis, vas deferens, seminal
processes. These systems produce larger number vesicle, and prostate in males [42]. In mammalian
of embryos in a shorter period of time than in females, remnants of the mesonephric tubules per-
mammals. Many of these models, such as sist as the epoophroron and paroophoron.
zebrafish and Xenopus, have only a The mature mammalian kidney, the metaneph-
one-dimensional pronephric tubule as a kidney. ros, is derived from two tissues, the ureteric bud
However, many of the genes which pattern such and metanephric mesenchyme (see a recent
simple structures have important roles in mamma- review in Ref. [13]). Starting at approximately
lian metanephric kidney development. Increas- embryonic day 10.5 in the mouse and the 5th
ingly, investigators using have utilized more week of gestation in humans, reciprocal inductive
sophisticated techniques such as transgenic fish signals cause the ureteric bud to grow out from the
to examine the roles of genes or drugs in modify- caudal portion of the mesonephric duct and the
ing nephron progenitor populations [38]. Finally, metanephric mesenchyme to condense around the
the advent of clustered regularly interspaced short ureteric bud to form nephron progenitors. The
palindromic repeats (CRISPR)/CRISPR- ureteric bud ultimately gives rise to the collecting
associated protein (Cas) techniques should even- system, including the collecting ducts, renal
tually allow relatively quick and easy genetic calyces, renal pelvis, and ureters [2, 43]. In turn,
modifications of any animal model desired [39]. the nephrogenic metanephric mesenchyme
8 C. Bates et al.

limbs of the loops of Henle, and the distal convo-


luted tubule [2, 43]. Just after the condensation of
nephrogenic mesenchyme around the ureteric
bud, stromal metanephric mesenchyme (or renal
stroma) develops adjacent to the nephrogenic
mesenchyme. The renal stroma develops into
perivascular cells, vascular smooth muscle, fibro-
blasts, mesangial cells, renin-producing cells, and
even some peritubular endothelial cells (see
below).
The transcription factor, Odd-skipped related 1
(Osr1), is one of the several key molecules neces-
sary to specify portions of the posterior intermediate
mesoderm to become the mesonephric duct, ureteric
bud, and metanephric mesenchyme (nephrogenic
and stromal) [44]. Osr1-expressing cells in the inter-
mediate mesoderm have been shown to give rise to
most of the cellular components of the metanephric
kidney, including the nephron, vascular cells, inter-
stitial cells, and the mesonephric duct (including its
derivatives, viz., the ureteric bud/collecting system)
[45]. Other molecules critical for specification and
development of the mesonephric duct include the
transcription factors Paired box 2 (Pax2) [46], Pax8
[47], Lim homeobox 1 (Lhx1) [48], Gata binding
protein 3 (Gata3) [49], and the receptor tyrosine
kinase, Ret [50].
Many key signaling pathways have been shown
to drive reciprocal interactions between the ureteric
epithelium, the renal mesenchymal lineages, and
Fig. 2 Schematic overview of kidney development.
Mammalian kidney development begins with the forma- the renal vasculature. Ureteric bud outgrowth
tion of the nephric duct, which is divided into three seg- depends on inductive signals from nephron pro-
ments: pronephros, mesonephros, and metanephros. The genitors [51–53], stromal cells [54–58], and
pronephros degenerates in mammals, whereas the meso- angioblasts [59, 60], as well as from itself
nephros forms the male reproductive organs (rete testis,
efferent ducts, epididymis, vas deferens, seminal vesicles, [61]. The nephrogenic mesenchyme relies in part
and prostate). The metanephros becomes the mature mam- on signaling from the ureteric bud and renal stroma
malian kidney and is derived from inductive interactions for self-renewal and for initiation of nephron for-
between the metanephric mesenchyme and the ureteric bud mation [8, 11, 62–64]. Subsequent nephrogenesis
(Reproduced with kind permission from Springer Science
+Business Media: Moritz K, et al. Factors Influencing (i.e., patterning and differentiation of nephron epi-
Mammalian Kidney Development: Implications for Health thelia) is highly dependent on factors from both
in Adult Life, Morphological Development of the Kidney, ureteric epithelial and stromal cells [57, 65, 66].
Advances in Anatomy and Cell Biology, volume
196, 2008, pp 9–16, figure number 1)

Nephron Formation
differentiates into the epithelial cells that comprise
the mature nephron, including the parietal cells The metanephric mesenchyme gives rise to
and podocytes of the glomerulus, the proximal nephrogenic mesenchyme or nephron progeni-
convoluted tubule, the ascending and descending tors, which self-renew and have the capacity to
1 Embryonic Development of the Kidney 9

form the multiple epithelial cell types of the neph- Concurrently, the middle and upper limbs of the
ron. Much research is geared toward understand- S-shaped body elongate and differentiate into
ing this progenitor cell population, which is nephron tubules including proximal tubules,
critical for determining nephron endowment and loops of Henle (including descending and ascend-
thus long-term kidney health. ing limbs), and distal convoluted tubules. The
Anatomically, there are several steps that take terminal ends of the distal convoluted tubules
place in nephron formation. After the ureteric bud eventually connect to ureteric epithelia, which
has penetrated the metanephric mesenchyme, ultimately form the collecting system (collecting
nephron progenitors condense around the first ducts, renal pelvis, and ureters) (Fig. 3).
ureteric ampulla, forming “cap mesenchyme.” Nephrogenesis repeats in a radial fashion with
As the ureteric bud continues to branch and elon- the first nephrons forming in the juxtamedullary
gate, the nephrogenic mesenchyme continues to regions and last in the peripheral cortex, until the
form new caps surrounding each ureteric tip. After full complement of nephrons is reached.
the initial few ureteric branches, the earliest cap During prenatal and/or postnatal life, each
mesenchymal cells receive spatiotemporal cues to nephron increases in size and complexity as it
begin the differentiation process to form matures. Starting in the first month of life, matur-
epithelialized renal vesicles [67]. Subsequent ing proximal tubules transition from a columnar to
growth and differentiation of the renal vesicle cuboidal epithelium, develop microvilli, and
results in formation of the comma-shaped body, increase their tubular dimensions [70, 71]. While
which then lengthens to form the S-shaped body. the earliest limbs of the Henle loop are located in
The lower limb of the S-shaped body begins to the renal cortex, subsequent maturation and elon-
differentiate into glomerular podocytes. During gation of these limbs in utero results in the loops
this time, endothelial cells migrate into the cleft pushing through the corticomedullary boundary
of the lower limb of the S-shaped body and will in term infants [72–74]. Postnatal maturation
ultimately form the glomerular capillary loops results in the Henle loops eventually reaching
[68, 69]. Simultaneously, nascent mesangial the inner renal medulla in the mature kidney.
cells, derived from renal stroma (see below), also Thus, the urinary concentrating capacity of new-
migrate into this cleft. Thus, the lower limb of the born infants is limited by a reduced medullary
S-shaped body forms the immature glomerulus. tonicity gradient, due to the relatively shorter

Fig. 3 H&E-stained sections showing the four stages of differentiated from the lower limb of the S-shaped body
nephron formation in mice. (a) Image of a renal vesicle, (Reproduced with kind permission from Springer Science
the first stage of nephron formation. (b) Image of a comma- +Business Media: Sims-Lucas S. Analysis of 3D
shaped body that has differentiated from a vesicle. (c) Branching Pattern: Hematoxylin and Eosin Method,
Image of an S-shaped body, the third stage of nephron Methods in Molecular Biology, volume 886, pp 73–86,
formation. (d) Image of an immature glomerulus that 2012, Figure 3, panels A–D)
10 C. Bates et al.

loops of Henle. Finally, as the distal convoluted mesenchyme that is unable to differentiate into
tubule matures, a portion of the cells are found in nephrons and fail to form the mesonephric duct,
close proximity to the future vascular pole of the which is required for ureteric bud induction
developing glomerulus, where they develop into [88]. In Lhx1 [86, 89] and Sall1 [75] mutants,
the macula densa [72]. the metanephric mesenchyme does induce ure-
teric bud formation, but it fails to elongate and
branch, and the mesenchyme is once again unable
Specification of Nephron Progenitors/ to differentiate into nephrons. In Wt1 mutants, a
Cap Mesenchyme defective metanephric mesenchyme is formed,
but rapidly undergoes apoptosis [80]. Six2, a spe-
Differentiation of the intermediate mesoderm and cific marker of nephron progenitors, is required
metanephric mesenchyme into nephron progeni- for the maintenance of progenitor cells, but not for
tors and their derivatives is genetically defined by nephron differentiation; deletion of Six2 in mice
the sequential upregulation of several transcrip- results in the formation of ectopic nephron tubules
tion factors, cell adhesion molecules, and growth and the rapid depletion of nephron progenitor
factors. The intermediate mesoderm and early cells [82]. Similarly, the p53–E3 ubiquitin ligase,
metanephric mesenchyme express the transcrip- murine double minute 2 (Mdm2), appears critical
tion factors Sal-like 1 (Sall1) [75], Sine oculis for the maintenance of nephron progenitor
homeobox homolog 1 (Six1) [76], Eyes absent cells [91].
homolog 1 (Eya1) [77, 78], and the secreted pep- Recent studies have identified factors that gov-
tide growth factor, Glial-derived neurotrophic ern the delicate balance of self-renewal and dif-
factor (Gdnf) [79]. Induction of cap mesen- ferentiation of nephron progenitors, including
chyme/nephron progenitors by the ureteric bud WNT genes. Studies from the mid-1990s showed
tips is marked by expression of transcription fac- that lithium chloride, a potent inducer of Wnt
tors such as Wilms tumor 1 (Wt1) [80]; Cbp/p300- signaling, was able to drive tubulogenesis in iso-
interacting transactivator, with Glu/Asp-rich lated rodent metanephric mesenchyme cultures
carboxy-terminal domain, 1 (Cited1) [81]; and [92–94]. More recently Wnt9b, secreted from ure-
Sine oculis homeobox homolog 2 (Six2) [82], as teric bud cells, was shown to be required for the
well as the transmembrane molecules cadherin-11 differentiation of nephron progenitor cells.
[83] and α8 integrin [84]. The earliest epithelial Genetic deletion of Wnt9b resulted in a failure of
derivative of the cap mesenchyme, the renal ves- nephron progenitors to undergo the mesenchymal
icle, is marked by the transcription factors Pax2 to epithelial transition that is required to form the
[85] and Lhx1 [86]. renal vesicle [95]. A subsequent study revealed
The use of tissue-specific gene knockouts gen- that Wnt9b, along with signals from the renal
erally via conditional transgenic techniques has stroma (see below), plays an essential role in
revealed the importance of several transcription mediating the decision of nephron progenitors to
factors (including many mentioned above) in the self-renew or differentiate [64, 96].
specification of the cap mesenchyme. Conditional Another major signaling pathway that has been
homozygous deletion of Eya1 [78], Six1 [76], shown to mediate nephron progenitor survival is
Pax2 [87, 88], Wt1 [80], Sall1 [75], Six2 [82], or the fibroblast growth factor (FGF) signaling path-
Lhx1 [86, 89] leads to bilateral renal aplasia or way. FGF ligands are secreted peptides that bind
severe renal dysgenesis from defects in cap mes- and signal through their receptor tyrosine kinases,
enchyme specification and/or differentiation; FGF receptors (FGFRs). Isolated nephrogenic
these mesenchymal defects are often accompa- zone cell culture studies revealed that addition of
nied by ureteric induction and/or branching FGF1, 2, 9, and 20 ligands drives the expression
abnormalities due in large part to loss of GDNF of nephron progenitor markers and progenitor
signaling from the metanephric mesenchyme. proliferation [97]. More recently, in vivo mouse
Pax2 mutant mice generate a metanephric studies showed that Fgf9 and Fgf20 are critical for
1 Embryonic Development of the Kidney 11

maintaining nephron progenitor survival, prolif- including Osr1, Eya1, Pax2, Wt1, and Wnt9b
eration, and competence to respond to inductive (among others) [16]. MicroRNAs (miRNAs) are
signals; furthermore, FGF20 mutations in humans small noncoding RNAs that bind to specific
were shown to be associated with severe renal mRNA targets to block translation and promote
dysplasia [98]. Other work in mice has identified mRNA degradation. Conditional targeting of
that the Fgfrs critical for metanephric mesen- dicer, an enzyme required for the processing of
chyme development are Fgfr1 and Fgfr2. Condi- all miRNAs, in mouse nephron progenitors, led to
tional deletion of Fgfr1 and Fgfr2 in the a loss of the progenitors due to excessive apopto-
metanephric mesenchyme leads to severe renal sis, likely from upregulation of the proapoptotic
dysgenesis [99–101]. protein, Bim [22]. A more recent study revealed
Several studies have shown that a balance that conditional deletion of a specific miRNA
between cell survival and apoptosis is necessary cluster, the miR-17~92 complex, in nephron pro-
for normal nephron progenitor function. Classic genitors led to decreases in progenitor prolifera-
in vitro studies using isolated metanephric mes- tion, fewer numbers of nephrons, proteinuria, and
enchyme have shown that nephron progenitors podocyte damage. Moreover, this report was the
undergo massive apoptosis when cultured without first to identify a specific miRNA cluster essential
an inducer [102]. Coculture with isolated ureteric for kidney development [110].
buds or heterologous inducers, such as embryonic
spinal cords, dampens apoptosis and drives pro-
genitor survival [43, 52]. Other studies have iden- Nephron Induction
tified several factors that when added to
metanephric mesenchyme or nephrogenic zone The initial differentiation step of nephron progen-
cell cultures drive progenitor survival, including itors is to undergo a mesenchymal to epithelial
transforming growth factor-β2 (TGF-β2), TGFα, transition to form the renal vesicle. In vitro exper-
leukemia inhibitory factor (LIF), epidermal iments utilizing isolated rodent metanephric mes-
growth factor (EGF), FGF2, and bone morphoge- enchymal rudiments (similar to and including
netic protein 7 (BMP7) [62, 103–105]. Whether some of the survival studies noted above) have
some or all of these factors act as endogenous identified exogenous factors that stimulate neph-
nephron progenitor inducers remains to be deter- ron progenitors to undergo tubulo-epithelial dif-
mined. Counterbalancing cell survival, apoptosis ferentiation [52]. Some of these growth factors
is required for normal nephron progenitor func- can act alone or in concert with others and include
tion. Moreover, suppression of apoptosis via phar- FGF2 [111], LIF [63, 105, 112], TGFβ2 [105,
maceutical or genetic means leads to kidney 113], growth/differentiation factor-11 (GDF-11)
malformations including abnormal ureteric [105, 114], and WNT1/4 [115, 116]. Sequestration
branching and defective nephrogenesis [106, of Wnt ligands in intact rodent kidney explants by
107]. Relative “overabundance” of nephron pro- addition of secreted frizzled-related proteins
genitors also leads to epithelial and/or stromal cell (sFrps) leads to decreases in mesenchyme-derived
defects [108, 109]. tubulogenesis [117]. More recent mouse genetic
Recent studies have revealed the importance of experiments have identified at least some of the
epigenetic mechanisms that regulate nephron pro- critical endogenous pathways and growth factors
genitor specification, survival, and potential for necessary for the induction of the mesenchymal to
differentiation. Histone deacetylases (HDACs) epithelial transition. For example, global deletion
are enzymes that remove acetyl groups from his- of Wnt4, normally expressed in renal vesicles, did
tones, which then modulate (usually stimulate) not perturb cap mesenchyme formation; however,
gene transcription. Recent work has revealed the mutant nephron progenitors were completely
that Class I HDACs are highly expressed in neph- unable to form renal vesicles [95]. Conditional
ron progenitors and are required for the proper deletion of Fgf8 in the metanephric mesenchyme
expression of several key developmental genes leads to a block of nephrogenesis beyond the renal
12 C. Bates et al.

vesicle stage. Fgf8 likely normally acts with Wnt4 glomeruli and proximal tubules [127]. In vivo,
to drive Lhx1 expression [118, 119]. Interestingly, conditional deletion of Notch2 or Rbpsuh in
global deletion of Fgfr-like 1, a membrane-bound mouse metanephric mesenchyme leads to an
Fgf receptor that lacks an intracellular tyrosine absence of proximal tubules and glomerular epi-
kinase domain, also leads to a block in thelium [128]. Finally, ectopic Notch expression
nephrogenesis similar to Fgf8 conditional in nephron progenitor cells results in the prema-
mutants [120]. ture differentiation of the progenitors into proxi-
mal nephron epithelia [130].

Nephron Segmentation
Glomerulogenesis
Establishment of a proper proximal–distal axis is
critical for normal nephron segmentation. Nega- Glomerulogenesis is initiated when the comma-
tive reciprocal interactions between Wt1 and shaped body differentiates into the S-shaped body
Pax2 at early stages of nephron development [2, 131]. During this time, immature podocytes
appear vital to proximal–distal axis patterning along the lower limb are highly proliferative and
[121–123]. In the S-shaped body, Wt1 is localized have a columnar shape with apical cell attach-
to the lower limb and inhibits Pax2 expression, ments and a single-layer basement membrane
which together drives cells toward podocyte fates [131]. Concurrently, endothelial and mesangial
[124]. Transgenic mice with overexpression of cell progenitors are recruited into the lower cleft
Pax2 throughout the embryo including develop- of the S-shaped body, which will become the
ing nephrons develop glomerular defects and vascular pole [132]. While mesangial cells origi-
renal cystic dysplasia [125]. In contrast Pax2 is nate from the renal stroma (see below), the devel-
expressed in the upper limb of the S-shaped body opmental origin of the glomerular endothelium is
and represses Wt1 expression, stimulating these still unclear. Transplantation of avascular rodent
cells to become proximal and distal tubular neph- embryonic kidney rudiments under neonatal kid-
ron segments [118, 126]. ney capsule led to the formation of endothelial
Two other transcription factors critical for precursors or angioblasts originating from the
proximal–distal axis patterning of the nephron graft metanephric mesenchyme [132–134]. How-
include Lhx1 and Brain specific homeobox 1 ever, engraftment of embryonic rat kidney rudi-
(Brn1), both of which are expressed at the renal ments onto avian chorioallantoic membrane led to
vesicle stage. Conditional deletion of Lhx1 vascular ingrowth of avian vessels into the rat
throughout the metanephric mesenchyme blocks glomeruli [135]. As will be expanded on in the
nephrogenesis at the renal vesicle stage and also Vascular Development section below, it is likely
leads to a loss of Brn1 expression [86]. Condi- that both processes/sources contribute to the for-
tional targeting of Brn1 in the metanephric mes- mation of glomerular capillaries. As the S-shaped
enchyme does not block proximal nephrogenesis; body matures, the lower cleft transforms into a
however, the loop of Henle fails to form, and cup shape configuration.
distal convoluted tubules fail to terminally differ- At this time the podocytes lose their prolifera-
entiate [73]. These results suggest that Lhx1 acts tive ability [136] and differentiate, forming foot
earlier in nephron patterning than Brn1, which is a processes and slit diaphragms, specialized intra-
critical distal nephron patterning. cellular junctions critical for proper glomerular
Notch receptor signaling, mediated largely by filtration [137, 138]. Concurrently, the composi-
Recombining binding protein suppressor of hair- tion of the glomerular basement membrane
less (Rbpsuh), appears critical for proximal neph- changes from laminin-1 to laminin-11 and from
ron patterning [127–129]. Use of a Notch α -1 and α -2 type IV collagen chains to α -3, α -4,
inhibitor in mouse metanephric kidney explants and α -5 type IV collagen chains [139]. Several
led to a loss of proximal cell fates, including mouse knockout mice models have shown how
1 Embryonic Development of the Kidney 13

failure in these transitions leads to structural and with glomerular basement membrane thickening
functional glomerular basement membrane and proteinuria that can progress to chronic kid-
defects [140–142]. At this stage of development, ney disease [156, 157]. Three studies recently
the nascent mesangial cells act as a scaffold for the showed the importance of microRNAs in
formation of the glomerular capillary loops and maintaining differentiated podocytes in the
ultimately form the supportive core for the entire mouse. Targeted ablation of dicer in murine
glomerulus via the deposition of extracellular podocytes, resulting in a loss of all miRNAs, led
matrix [143, 144]. Developing glomerular endo- to podocyte injury, severe proteinuria, and tubular
thelial cells branch extensively during this time damage starting 2 weeks after birth [20, 158, 159].
and begin differentiating into fenestrated endothe-
lia [2] (see Vascular section below).
By 32–34 weeks gestation, glomerulogenesis/ Molecular Control of Glomerular
nephrogenesis ceases in humans, whereas it per- Capillary Tuft Development
sists in mice and rats for 7–10 days following full
gestation [2]. In newborn humans, the superficial There are several signaling cascades that have
glomeruli are the most immature and are smaller been implicated in the homing and maturation of
than the deeper juxtamedullary glomeruli the endothelial and mesangial precursors to form
[71]. While no new glomeruli are formed after the glomerular capillary tuft. Vascular endothelial
birth, they continue to grow and mature postna- growth factor (VEGF), which is secreted from the
tally, reaching their adult size at approximately podocytes at the S-shaped body stage, promotes
three and a half years of age [71]. recruitment of endothelial precursors to the vas-
cular cleft [160, 161]. Angiopoietin-1 and -2,
growth factors expressed by podocytes and
Molecular Control of Podocyte mesangial cells, respectively, are also critical for
Terminal Differentiation normal glomerular capillary development
[162]. Mesangial cell recruitment into the cleft is
Transcription factors and epigenetic factors, largely mediated by the secretion of platelet-
including microRNAs, have been shown to be derived growth factor (PDGF)-B by endothelial
critical for podocyte differentiation. Examples of cells, which binds PDGF receptor-β (PDGFRβ)
essential transcription factors include Wt1, on the mesangial cell progenitors [163]. Mice that
podocyte expressed 1 (Pod1), Lim homeobox 1b lack either Pdgfβ or Pdgfrβ fail to form glomeru-
(Lmx1b), and Mafb. Several studies utilizing lar capillary tufts demonstrating the importance of
murine genetic knockout models of Wt1 have mesangial cell recruitment [164, 165]. Finally,
demonstrated its critical roles in mediating Notch2 and its ligand Jagged1 are critical for
podocyte differentiation [145–148]. In humans, glomerular endothelial and mesangial cell devel-
WT1 mutations can lead to diffuse mesangial scle- opment. Notch2 hypomorphic mice and Notch2/
rosis, characterized by podocyte differentiation Jagged1 compound heterozygous mice develop
defects resulting in varied glomerular lesions and glomerular aneurysms and possess no mesangial
proteinuria, and can occur as an isolated disease or cells [166].
in association with Denys–Drash or Frasier syn-
dromes [149–152]. Deletion of Pod1, expressed
in stromal cells, leads to nonautonomous Renal Stroma
podocyte defects at the capillary loop stage in
mice [153]. Genetic deletion of Lmx1b or Mafb The renal stroma, like the nephrogenic mesen-
leads to podocyte differentiation defects past the chyme, is derived from Osr1-positive intermedi-
capillary loop stage [154, 155]. Furthermore, ate mesoderm and the metanephric mesenchyme
mutations in the LMX1b gene in humans lead to [167, 168]. A hallmark of the initial renal stroma
nail–patella syndrome, which is often associated is the expression of the transcription factor,
14 C. Bates et al.

Foxd1, which is seen as early as E11.5 in the and the renal capsule) or Pod1 (found in medul-
mouse. The renal stroma is initially located at lary stroma) appears to appropriately restrict Ret
the periphery of the kidney and interdigitates expression to ureteric tips; genetic deletion of
between the developing nephron units and ure- either gene leads to mis-expression of Ret
teric tips. One function of the early renal stroma throughout the entire ureteric tree and subsequent
is to support framework for the developing ves- ureteric branching defects [54, 57, 153, 173].
sels, nephron progenitors, and ureteric epithelia. Cross talk from the stroma is also critical for
As embryonic kidney development progresses, nephron development. Mouse genetic studies
stromal cells are present in both the peripheral show that Foxd1 and Pod1 are necessary for nor-
renal cortex and the medulla surrounding devel- mal nephron patterning (in addition to ureteric
oping collecting ducts. At this time, the cortical morphogenesis) [54, 153]. Loss of Foxd1 in
stroma expresses Foxd1, Aldehyde dehydroge- mice leads to premature differentiation of stromal
nase 1 family, member A2 (Raldh2), Retinoic cells, which inhibits Bmp7-mediated nephron
acid receptor α (Rarα), and Rarβ2, while the progenitor differentiation [174]. Two recent
medullary stroma expresses Fgf7, Pod1, and studies have shown how complete ablation of
Bmp4. Many of these stromally expressed genes renal cortical stroma with diphtheria toxin leads
have been shown to be critical for nephrogenesis to abnormally thickened nephron progenitor
and ureteric branching morphogenesis by virtue caps and a decreased ability of progenitors to
of mouse knockout studies [54, 55, 57, 58, 153, differentiate [64, 175]. Mechanistically, it
169]. At birth, many of the developmental stromal appears that loss of the protocadherin Fat4 in the
cells have undergone apoptosis and are replaced stroma perturbs the activity of the transcription
by nephron segments such as loops of Henle factors, Yap and Taz, which in turn disrupts
[170]. Many stromal derivatives do survive giving Wnt9b signaling and nephron differentiation
rise to fibroblasts, lymphocyte-like cells, glomer- [64]. Thus, in addition to providing a “frame-
ular mesangial cells, renin-expressing cells, vas- work” for the rest of the developing kidney, the
cular smooth muscle cells, pericytes, and a renal stroma actively signals to other renal line-
subpopulation of peritubular endothelial cells ages and differentiates into cells that populate the
[168, 170, 171]. mature kidney.
As noted, signaling from the renal stroma is
critical for ureteric morphogenesis. Three genes/
pathways expressed within the stroma, retinoic
acid, Foxd1, and Pod1, modulate ureteric Vascular Development of the Kidney
branching by regulating expression of Ret, a
receptor tyrosine kinase expressed in ureteric The adult kidney receives approximately 25 % of
tips and required for ureteric development (see the cardiac output. Furthermore, the adult kidney
below). Vitamin A is converted to its active has a high complex vascular network with differ-
form, retinoic acid, by the enzyme Raldh2 in the ent functions and therefore different specialized
renal stroma. Moreover, blockade of retinoic acid endothelia depending on location [176]. Specifi-
signaling in mice, by deletion of Raldh2 or by cally, three major types of endothelial cells are
combined deletion of the retinoic acid receptors, present within the kidney, including fenestrated
Rarα and Rarβ2, leads to hypoplastic kidneys (in glomerular capillaries), fenestrated with dia-
with a reduction in the number of ureteric phragms (in peritubular capillaries and ascending
branches; the ureteric branching defects are linked vasa recta), and continuous capillaries
to the downregulation of Ret expression in mutant (in descending vasa recta) (Fig. 4). Not surpris-
embryos, which in the case of the retinoic acid ingly, these various endothelial cell types have
receptor mutants can be rescued by forced heterogeneous expression profiles and often
re-expression of Ret in the ureteric tissues [55, appear to have different developmental origins
56, 172]. Foxd1 (expressed in cortical stroma [21, 177, 178].
1 Embryonic Development of the Kidney 15

Fig. 4 Electron microscopy demonstrating the varied endothelium with diaphragms (c, h, i, arrow). (d, h, i)
renal endothelium. (a, e) Glomerular capillaries contain Descending vasa recta (DVR) possess endothelium that is
fenestrated endothelium without diaphragms (e, arrows) non-fenestrated, thick, and continuous (d, h, i). RBC red
and share a basement membrane (*) with podocyte foot blood cell, EC endothelial cell. Panels (a–d) scanning
processes (large arrowhead) that are separated by slit electron micrographs. Panels (e–i) transmission electron
diaphragms (small arrowhead). (b, f, g) Peritubular capil- micrographs (Reproduced with kind permission from
laries have fenestrated endothelial cells that are covered Springer Science+Business Media: Stolz DB and Sims-
with diaphragms (f, g, arrows) and have a thick basement Lucas S. Unwrapping the origins and roles of the renal
membrane (*) separating them from the tubular cells. endothelium, Pediatr Nephrol. 2015;30(6):865–72,
(c, h, i) Ascending vasa recta (AVR) also have fenestrated Figure 2)

Angiogenesis Versus Vasculogenesis progenitors (marked by Flk1/Vegfr2) form primi-


tive vascular networks, particularly within the
Blood vessels can form by angiogenesis, in which renal stroma, that subsequently join with and are
new vessels sprout from existing vessels, or by pruned by the angiogenic vessels [182]. The
vasculogenesis, in which de novo vessels form vasculogenic endothelial cell progenitors within
from endothelial progenitors (Fig. 5). Extensive the kidney appear to arise from the
linage tracing experiments and transplantation Osr1-expressing intermediate mesoderm, as is
studies have shown that both of likely occur in the case with the rest of the metanephric
renal vascular formation [176, 179–181]. kidney [45]. Finally, specification of the
The early renal artery and efferent arterioles endothelium (whether angiogenic or
appear to be primary sites from which new vasculogenic in origin), including arterial,
angiogenic vessels sprout within the developing venous, capillary, or lymphatic fates, is driven
kidney. Simultaneously, renal endothelial by growth factor signaling pathways and
16 C. Bates et al.

Fig. 5 Schematic diagram of vascular formation in the diagrams depicting how a combination of angiogenesis
developing mouse kidney. Top panels. Angiogenic ves- and vasculogenesis likely leads to vessel formation in the
sels (red) grow out from the major branches of the renal kidney. E10.5–12.5 = embryonic days 10.5–12.5
artery and track with the branching ureteric epithelium (Reproduced with kind permission from Springer Science
(orange). Middle panels. Vasculogenic vessels form from +Business Media: Stolz DB and Sims-Lucas
progenitor cells (yellow, middle panel) within the meta- S. Unwrapping the origins and roles of the renal endothe-
nephric mesenchyme (blue) and form a primitive vascular lium, Pediatr Nephrol. 2015;30(6):865–72, Figure 1)
plexus (yellow, right panel). Bottom panels. Schematic

transcription factors, including Vegf, Ephrin, peritubular capillaries has been less well defined.
Notch, and Sox [183]. Recent studies, however, have found that
peritubular capillaries arise from a combination
of resident endothelial progenitors as well as
Origins of the Peritubular Capillary invading angiogenic vessels [182, 184,
Endothelia 185]. One intriguing study found that Foxd1-pos-
itive renal cortical stroma cells give rise to a subset
While the formation of glomerular capillaries has of the peritubular endothelia but not the glomeru-
been extensively studied (see above), the origin of lar endothelia [184].
1 Embryonic Development of the Kidney 17

Molecular Control of Renal Vascular The Notch signaling pathway appears to regu-
Development late renal angiogenic vessel outgrowth [199].
Notch receptors induce sprouting by stimulating
A key signaling pathway mediating renal vascular the expression of Vegfr2 in vascular tip cells.
development is the VEGF pathway. VEGF Simultaneously Notch inhibits Vegfr2 signaling
ligands are expressed early in the metanephric in adjacent vascular stalk cells, causing them to
mesenchyme and later in the developing glomer- remain dormant. Thus Notch regulates the pattern
ular podocytes, distal tubules, and collecting of branching in angiogenic vessels.
ducts and at low levels in the proximal tubules
[68, 69]. Developing endothelial cells, including
those that arise from existing vessels and those Collecting System Development
forming de novo, express VEGF receptors; thus,
VEGF signaling appears to drive both angiogen- Ureteric bud formation begins in the 5th week of
esis and vasculogenesis within the kidney. Inter- gestation in humans and at embryonic day 10.5 in
estingly, Vegfr2 is present on the apical surface of mice. As noted previously, signals from the meta-
ureteric epithelium, which likely accounts for nephric mesenchyme cause the ureteric bud to
the stimulatory role of Vegf on ureteric growth form from the mesonephric duct and then invade
[132, 186]. the mesenchyme. Overall, collecting duct system
Hypoxia-inducible factors (HIFs), a family of development includes (i) ureteric bud outgrowth,
transcriptions factors, are likely master regulators (ii) branching of the ureteric bud, and (iii) pattern-
of angiogenesis and vasculogenesis within the ing of the collecting duct system, all of which is
developing kidney [187]. These molecules are discussed in more detail below.
activated during periods of low oxygenation, as
occurs during embryogenesis, and are
downregulated postnatally. The HIF genes are Ureteric Bud Induction and Outgrowth
largely located in the nephrogenic zone, including
podocytes, developing collecting ducts, and Failure of ureteric bud outgrowth results in renal
developing endothelial cells [187, 188]. HIF pro- aplasia, which can occur unilaterally or bilaterally
teins induce expression of VEGF ligands, Vegfr1, [200]. The GDNF–RET signaling pathway is cru-
and Vegfr2 during kidney development by directly cial for bud outgrowth. The receptor tyrosine
binding to hypoxia-responsive elements on those kinase RET and its coreceptor GFRα1 are
genes [189–192]. expressed in the mesonephric duct, the initial ure-
Angiopoietin (Ang) growth factors that bind to teric bud, and later in the branching ureteric tips,
Tie receptors also appear to have critical roles in while its ligand, GDNF, is present in the meta-
renal vascular development and are at least in part nephric mesenchyme (Fig. 6) [201–203]. Targeted
regulated by HIF and VEGF signaling [193, deletion of Gdnf, Ret, or Gfrα1 in mice generally
194]. Ang1, which is expressed in the metaneph- results in bilateral renal aplasia due to a lack of
ric mesenchyme, maturing nephron tubules, and ureteric bud outgrowth [202, 204–209]. Heterozy-
podocytes, signals through Tie2, which is gous mutations of RET have also been identified
expressed on endothelial cells [195]. Conditional in humans with bilateral renal aplasia, and a rare
deletion of Ang1 or Tie2 in mice leads to glomer- RET polymorphism has been reported in individ-
ular capillary defects including endothelial cells uals with nonsyndromic vesicoureteral reflux
that do not attach to the basement membrane [196, [210, 211]. Moreover, the renal aplastic pheno-
197]. Ang2, expressed in vascular smooth muscle type is not fully penetrant in a subset of Gdnf /
cells and pericytes, binds to Tie1 that is expressed or Ret/ mutant mice [202, 204], suggesting that
by endothelial cells. Genetic deletion of Ang2 other molecular pathways play a role in ureteric
leads to upregulation of Tie2 signaling and signif- bud outgrowth. Some examples of other pathways
icant defects in renal peritubular capillaries [198]. include signaling through integrins such as
18 C. Bates et al.

Fig. 6 Schematic diagram of the molecular control of bud induction to a single site from the mesonephric duct.
ureteric bud induction. GDNF is secreted from the meta- Sprouty1 (in the mesonephric duct) and Bmp4 (in tailbud-
nephric mesenchyme and binds to its receptor, Ret (and its derived mesenchyme around the mesonephric duct)
coreceptor GFRα1) on the mesonephric duct, to induce repress GDRF–Ret signaling and thus restrict ureteric
ureteric bud formation. Slit2/Robo2 and FoxC1 inhibit bud induction to its proper site
the domain of GDNF expression and thus limit ureteric

α8 integrin [84] and enzymes involved in proteo- At least three genes, Foxc1, Slit2, and Robo2,
glycan synthesis such as heparan sulfate are thought to be crucial in restricting Gdnf to the
2-sulfotransferase (Hs2st) [212]. posterior intermediate mesoderm (Fig. 6). Homo-
Many studies have focused on the molecular zygous mutant mice for all three genes develop
mechanisms that regulate GDNF–RET expression ectopic ureteric buds, multiple ureters,
and/or signaling. Prior to kidney development, hydroureter, and anterior expansion of Gdnf
Ret is expressed throughout the mesonephric expression [216, 217]. Foxc1 encodes a transcrip-
duct, and Gdnf is present throughout the interme- tion factor co-expressed with Gdnf in the meta-
diate mesoderm adjacent to the mesonephric duct nephric mesenchyme [216]. In the central nervous
[50, 201]. At the time of ureteric bud induction, system, the secreted protein Slit2 functions as a
Gdnf expression becomes restricted to the poste- chemorepellent during migration of axons that
rior intermediate mesoderm next to the site of express its receptor Robo2 [218, 219]. In the
ureteric bud outgrowth; once the ureteric bud has developing kidney, Slit2 is expressed in the meso-
invaded the mesenchyme, Ret expression nephric duct, and Robo2 is detected in the meta-
becomes restricted to ureteric bud tips [50]. In nephric mesenchyme [220]. ROBO2 missense
vitro studies with Gdnf-soaked agarose beads mutations in humans have been identified in fam-
show that the entire length of the mesonephric ilies with vesicoureteral reflux and/or duplex
duct is competent to respond to Gdnf by initiating kidneys [221].
ectopic ureteric bud formation [201, 213]. More- Two other genes, Sprouty1 (Spry1) and Bmp4,
over, mice that ectopically express Gdnf or Ret act in a negative feedback loop with GDNF–RET
in vivo develop renal malformations such as signaling (Fig. 6). Loss of Spry1, which is nor-
duplex kidney and hydronephrosis [214, mally expressed in the mesonephric duct, results
215]. Together, these data show that GDNF–RET in ectopic ureteric bud induction, multiple ureters,
signaling is highly spatially regulated for a single multiplex kidneys, and hydroureter [222,
ureteric bud to form in the correct location from 223]. Spry1 mutant embryonic kidneys have
the mesonephric duct. increased expression of Gdnf and GDNF–RET
1 Embryonic Development of the Kidney 19

target genes and have increased sensitivity to remodeling of central (medullary) segments
GDNF-induced ureteric induction in organ cul- [2]. At this stage, four to seven new nephrons
ture. Bmp4 is expressed in the tailbud-derived are induced around each tip of a terminal
mesenchyme (different than renal mesenchyme) collecting duct branch [2, 43].
immediately next to the mesonephric duct and Localized cell proliferation contributes to ini-
ureteric bud [169, 224]. Mice heterozygous for tial ureteric bud outgrowth from the mesonephric
Bmp4 have ectopic or duplicated ureteric buds, duct, formation and growth of ampullae, and elon-
resulting in hypodysplastic kidneys, hydroureter- gation of ureteric branches [61, 108, 234,
onephrosis, and ureteral duplications [225, 235]. Cell survival is also critical for normal
226]. In vitro, Bmp4 has been shown to block renal branching morphogenesis; defects in cell
the ability of Gdnf to induce ureteric bud out- survival are associated with renal cystic dysplasia
growth from the mesonephric duct [85, and urinary tract dilatation. Moreover, targeted
227]. BMP4 mutations have also been described deletion of bcl2 [236] and AP-2 [237], genes
in humans with renal tract malformations [228]. critical for cell survival, results in increased apo-
The downstream effects of GDNF–RET sig- ptosis and collecting duct cysts in mice. In addi-
naling, namely, ureteric bud proliferation, sur- tion, experimental models of fetal and neonatal
vival, and ureteric outgrowth and branching, are urinary tract obstruction lead to apoptosis in
mediated by the transcription factors, Etv4 and dilated collecting ducts [238, 239].
Etv5. Combined deletion of Etv4 and Etv5 causes Several signaling pathways are necessary for
bilateral renal aplasia in mice [229]. Etv4 and Etv5 branching morphogenesis. In addition to its role in
drive expression of several critical genes in the ureteric bud induction, GDNF–RET signaling has
ureteric bud tip, including Wnt11, Cxcr4, Mmp14, been shown to be critical for ureteric branching
Myb, and Met [229]. Furthermore, genetic dele- in vivo and in vitro [213, 215]. As noted, Wnt11,
tion studies in mice have shown that Wnt11 is expressed in ureteric tips, is necessary for
necessary for normal Gdnf expression in the meta- maintaining normal Gdnf expression; conversely,
nephric mesenchyme [230]. Wnt11 expression is reduced when Gdnf signaling
is absent. Furthermore, Wnt11 mutant mice
have defective ureteric branching morphogenesis
Renal Branching Morphogenesis and thus develop renal hypoplasia
[230, 240]. Finally, conditional targeting of
After growing into the metanephric mesenchyme, β-catenin, a key mediator of canonical Wnt sig-
the ureteric bud bifurcates into a T-shaped struc- naling, in the ureteric lineage results in aberrant
ture. The ureteric bud then continues to branch, branching, loss of ureteric bud tip gene expres-
ultimately generating about 15 generations of sion, and premature expression of differentiated
branches, with the earliest branches remodeling collecting duct genes [241, 242].
to form the calyces and renal pelvis [231]. The Fibroblast growth factor signaling is also crit-
process of ureteric branching includes (1) expan- ical for ureteric branching. In vitro studies have
sion of the ureteric bud at its leading tip (termed shown that exogenous Fgf ligands differentially
the ampulla), (2) division of the ampulla to form modulate ureteric bud growth and proliferation
new branches, and (3) elongation of newly formed [243]. FGF10 preferentially stimulates prolifera-
branches [232, 233]. In humans, during the first tion at ureteric bud tips, whereas FGF7 increases
9 generations of branching, ureteric bud tips cell proliferation throughout the developing
induce formation of new nephrons from the sur- collecting ducts [243]. In vivo, global deletion of
rounding cap mesenchyme at about a 1:1 ratio Fgf7 or Fgf10 in mice results in ureteric branching
[2]. Ureteric bud branching is completed by the defects and hypoplastic kidneys [58, 244]. Condi-
20th–22nd week of human gestation, and subse- tional targeting studies in mice have revealed that
quent collecting duct maturation occurs by elon- Fgfr2 is likely the key Fgf receptor mediating
gation of peripheral (cortical) segments and effects on ureteric branching [245–247]. Loss of
20 C. Bates et al.

Fgfr2 in the ureteric bud results in hypoplastic differentiating glomeruli, nephron tubules, and
ureteric ampullae with reduced proliferation and collecting ducts [250]. In contrast, the developing
increased apoptosis, ultimately leading to a sig- renal medulla expands longitudinally, perpendic-
nificant reduction in ureteric branching and hypo- ular to the axis of cortical growth, due to elonga-
plastic kidneys. In addition, an interesting study tion of outer medullary collecting ducts
revealed that while combined loss of Sprouty1 and [250]. Stromal cells may be a source of stimula-
Gdnf in mice largely rescues ureteric defects com- tory cues for the medullary growth [250]; studies
pared to when either locus is deleted alone, addi- have shown that mice lacking the stromal tran-
tional loss of Fgf10 in the combined mutants led scription factors Foxd1 and Pod1 have abnormal
to complete loss of ureteric branching and renal medullary collecting duct patterning [54, 66,
aplasia [248]; thus, FGF signals appear to be able 251]. Finally, apoptosis appears to participate in
to largely substitute for GDNF in promoting ure- remodeling the branched medullary ureteric tis-
teric morphogenesis in the absence of Sprouty1. sues into elongated tubules, as programmed cell
Finally, a combination of in vitro and in vivo death normally occurs prominently in developing
experiments revealed that Fgf signaling acts in a medullary ureteric epithelia that become the
coordinated fashion with Wnt11 and Gdnf to reg- papilla, calyces, and renal pelvis [108].
ulate ureteric morphogenesis, in concert with Multiple genes have been implicated in the
Sprouty genes [249]. differentiation of cortical and medullary
collecting ducts, including those that encode for
soluble growth factors (Fgf7, Fgf10, Bmp4,
Patterning of the Medullary Bmp5, and Wnt7b), proteoglycans (Gpc3), cell
and Cortical Collecting Ducts cycle regulatory proteins (p57KIP2), and compo-
nents of the renin–angiotensin axis (angiotensin
From the 22nd–34th week of human gestation [2] and angiotensin type 1 and 2 receptors). Fgf7
and embryonic day 15 birth in mice [43], the mutant mice have marked papillary underdevel-
cortical (peripheral) and medullary (central) opment, while Fgf10 null kidneys exhibit medul-
regions of the kidney become established. The lary dysplasia with fewer loops of Henle and
relatively compact, circumferential renal cortex medullary collecting ducts, increased medullary
comprises approximately 70 % of the mature kid- stroma, and enlargement of the renal calyx [58,
ney volume [250]. The renal medulla develops 244]. The ability of FGF ligands to bind properly
modified a cone shape and occupies the remainder to their receptors requires interactions with cell
of the mature kidney volume [250]. The apex of surface proteoglycans, including glypicans
the medullary cone consists of collecting ducts [252]. Glypican-3 (GPC3) is required for normal
converging in the inner medulla and is termed medullary patterning in humans and mice [253,
the papilla. Ultimately, medullary collecting 254]. Moreover, the medullary dysplasia observed
ducts become morphologically distinct from cor- in Gpc3-deficient mice appears to result from
tical collecting ducts. Medullary collecting ducts unrestrained proliferation and overgrowth of the
become elongated and linear and remain relatively ureteric bud and collecting ducts, followed by
unbranched in a region devoid of glomeruli. In aberrant apoptosis [253, 254]. The Gpc3/ med-
contrast, collecting ducts in the renal cortex ullary defects appear to be driven by altered
remain branched and induce nephrogenic cap responses of mutant collecting duct cells to
mesenchyme to form nephrons throughout growth factors including Fgfs [254–256]. Finally,
nephrogenesis. mice lacking the cell cycle protein p57KIP2 dem-
These morphological differences are likely due onstrate medullary dysplasia, with fewer inner
in part to distinct axes of growth in the developing medullary collecting ducts [257]. Together, these
renal cortex and medulla. The renal cortex grows studies reveal the importance of balanced cell
circumferentially, which preserves the organiza- proliferation and apoptosis in medullary
tion of the peripheral tissues, including collecting duct patterning.
1 Embryonic Development of the Kidney 21

Proper elongation and growth of medullary or ureterovesical junction stenoses, renal dyspla-
collecting ducts also appears to rely on oriented sia or hypoplasia, multicystic dysplastic kidney,
cell divisions. Studies have shown that canonical and renal aplasia [107].
Wnt signaling in collecting ducts via Wnt7b leads
to proper oriented cell division and survival [258,
259]. Furthermore, α3β1 integrin and the receptor Lower Urinary Tract Development
tyrosine kinase c-Met act in concert to regulate
Wnt7b expression and signaling in medullary Anatomic and Functional Development
collecting ducts [259].
Finally, angiotensin (Agt) and angiotensin Concurrent with metanephric kidney formation,
receptors (Agtrs) appear critical for the develop- the embryonic ureter and bladder develop the
ment of the renal calyces, pelvis, and ureter. Mice former functioning to propel urine into the latter
lacking Agt or Agtr1 genes demonstrate progres- which stores urine until an appropriate time to
sive widening of the calyx and atrophy of the expel it via the urethra. Similar to the kidney, the
papillae and underlying medulla [260, ureter and bladder undergo maturation largely due
261]. These defects appear to be caused by to reciprocal interactions between an epithelium
decreased proliferation of the smooth muscle (i.e., urothelium) and surrounding mesenchyme
cells that line the renal pelvis. Loss of Agtr2 that forms the lamina propria, muscle, and adven-
causes a range of renal anomalies secondary to titia (Fig. 7). For a recent detailed review on
ureteric mispatterning, including vesicoureteral anatomic and molecular control of lower urinary
reflux, duplex kidney, renal ectopia, ureteropelvic tract development, see [262].

Fig. 7 H&E-stained
sections from E15.5 and
P1 mouse ureters and
bladders. E15.5 ureters and
bladders have early
urothelium (u), an inner
layer of mesenchyme
(future lamina propria,
arrows), and an outer layer
of condensing mesenchyme
(future muscle, m). P1
ureters and bladders have a
more stratified urothelium
(u) and well-developed
lamina propria (arrows) and
outer muscle (m) layers.
The adventitial layer of
fibroblasts that surrounds
the muscle in both tissues is
not labeled. Ureters =100
magnification; bladders
=40 magnification
22 C. Bates et al.

Ureter development begins simultaneously bladder. The ability of the ureter to undergo peri-
with metanephric kidney development around staltic waves of contraction followed by relaxa-
the 5th week of gestation in humans and at tion appears to be intrinsic and not dependent on
E10.5 in the mouse, when the ureteric bud arises urine flow; cultured explants of E13.5 mouse ure-
from the mesonephric duct [262]. Thus the ters attached to kidneys begin to undergo sponta-
embryonic origin of the ureteral urothelium is neous peristaltic contractions within a few days,
the intermediate mesoderm, the same as the meta- as do isolated and cultured E15.5 ureters
nephric kidney. By E11.5 in the mouse, the ure- [268]. Elegant studies recently identified a popu-
teric bud has been segmented into a distal portion lation of pacemaker cells at the junction of the
that will develop into the ureter and a proximal renal pelvis and the kidney that express
end that has invaded the metanephric mesen- hyperpolarization-activated cation-3 (Hcn3)
chyme to eventually branch and form the channels (a family of channels that are also pre-
collecting ducts and renal pelvis (see above). sent in cardiac pacemakers) [269]. Loss of Hcn3
The mesenchyme surrounding the early develop- activity in mice leads to abnormal coordination
ing ureter consists largely of tailbud-derived mes- and frequency of ureter contractions [269]. Fol-
enchyme that appears to be crucial for directing lowing this study, another described a population
the distal ureteric bud toward a ureter fate of secondary pacemakers that are located in the
[263]. Between E10.5 and E13.5, the nascent muscle of the mouse proximal ureter (starting at
ureter transitions from attaching to the nephric the ureteropelvic junction) and that have morpho-
duct to emptying directly into the early bladder logical and molecular features similar to intestinal
(see below). By E13.5, a thin outer ring of ureteral pacemakers including expression of c-kit
mesenchyme condenses and expresses alpha [268]. Thus, like the cardiac conduction system,
smooth muscle actin (αSMA) mRNA, the first the ureter has primary and secondary pacemakers
marker of differentiation toward a smooth muscle that act to drive urinary propulsion in a coordi-
fate [264]. αSMA protein expression is not noted nated fashion.
until E14.5 in the proximal portion of the ureter The embryonic bladder initially forms around
(nearest the kidney) and then throughout the entire the 5th gestational week in humans and at
length of ureter by E16.5; thus, ureter muscle E11.5–12.5 in the mouse [262, 270]. Unlike the
development progresses in a rostral to caudal ureter, bladder urothelium is derived from the
direction [265]. Concurrent with development of endodermal urogenital sinus, formed from the
the mesenchymal layers, the ureteral urothelium ventral region of the cloaca. At E11.5–12.5, the
gradually matures from a simple epithelium to a urogenital sinus further subdivides into an ante-
stratified epithelium consisting of at least three rior portion, which will become the bladder, and a
cell types: basal, intermediate, and superficial/ posterior portion that forms the urethra and por-
umbrella cells. Each of these cell types has distinct tions of the female vagina. The mesenchyme that
structural features and molecular markers [266]; surrounds the bladder urothelium is largely
moreover, recent data strongly suggests that thought to be from splanchnic mesoderm,
urothelial basal cells, arising from the original although fate-mapping studies reveal that tailbud
ureteric epithelium, serve as the progenitors for mesenchyme also contributes to bladder mesen-
other ureteral urothelial cell types [266]. A unique chyme (similar to the ureter) [263]. By E13.5, the
feature of urothelium (compared with other epi- bladder is recognized as a distinct structure that is
thelia) is the apical expression of urothelial attached directly to the ureters. As is the case with
plaques, consisting of uroplakins, which likely ureters, αSMA mRNA expression in the develop-
have several functions, including providing a per- ing bladder muscle precedes protein expression;
meability barrier and acting as a binding site for mRNA expression appears as early as E11.5 in
uropathogenic E. coli [266]. mice [264], while protein expression begins at
An important function of the ureter is to con- E13.5 [270]. Unlike the ureter (and many other
tinuously propel urine from the renal pelvis to the organs with smooth muscle such as the intestine)
1 Embryonic Development of the Kidney 23

that has a thin ring of mesenchyme that begins to While the connection of the ureters with the
differentiate into muscle, the entire outer half of bladder is critical, how this occurs has been the
the bladder mesenchyme condenses simulta- subject of some debate. What is clear is that
neously and strongly expresses αSMA by E15.5 between E12.5 and E13.5, the common nephric
[264, 271]. Similar to the ureter, the bladder duct (caudal portion of the mesonephric duct
urothelium matures from a simple epithelium to between the ureter base and future bladder)
a stratified epithelium, consisting of cell types moves adjacent to the bladder, allowing for the
similar to the ureter, most of which also express ureters and rostral nephric ducts (future male
uroplakins and urothelial plaques [266]. While gonadal excretory ducts) to separate and empty
bladder urothelial basal cells were originally directly into the bladder. The triangular portion of
thought to be the progenitor cell for the other the bladder demarcated by the entry points of the
urothelial cell types, recent careful lineage tracing ureters and the bladder neck (where the remaining
studies revealed the presence of a previously embryonic nephric ducts empty) is known as the
unidentified transient population of cells, known trigone. Historically the common nephric duct
as “P” cells that serve as the progenitors in was thought to become incorporated into the blad-
embryos [272]; moreover, there are dynamic der to form the trigone. However, elegant fate-
changes in relative composition of bladder mapping studies reveal that the common nephric
urothelial cell types throughout development duct undergoes complete apoptosis starting at
(Fig. 8). E12.5 in the mouse and that the urothelium of
the trigone originates entirely from the bladder
urothelium [273]. Moreover, a separate study
P-cell Foxa2+ Upk+ P63+ Shh+ Krt5–
showed that the muscle present in the trigone
l-cell Foxa2– Upk+ P63+ Shh+ Krt5– arises mostly from the bladder with only a few
S-cell Foxa2– Upk+ P63– Shh– Krt5– fibers emanating from ureteral muscle
[274]. Thus, the trigone originates mostly from
B-cell Foxa2– Upk– P63+ Shh+ Krt5+
bladder tissues and not the mesonephric duct.
100%

Molecular Control of Ureter


75% and Bladder Development

50% Ureter
The pathways critical for early ureteric bud for-
25% mation were already covered in the section on
kidney development above. Thus, an overview
0% of the molecular control of ureter development
E11 E13 E14 E16 E18 Adult will be presented in this section (Fig. 9). Once
the ureteric bud has formed, Bmp4 is secreted by
Fig. 8 Graph demonstrating the proportion of differ-
ent murine bladder urothelial cell types during devel- tailbud-derived mesenchyme surrounding the dis-
opment. “P” cells, an early transient progenitor population tal ureteric bud, driving the epithelium toward a
appear first, followed dynamic changes in the proportion of urothelial fate; moreover, ectopic Bmp4 expres-
their derivatives [intermediate (I ), superficial (S), and basal
sion around proximal portions of the ureteric bud
(B) cells] through adulthood. Immunohistochemical
markers of each cell type are listed (Foxa2 forkhead box directs it to become urothelium instead of
A2, Upk uroplakins, P63 tumor protein P63, Shh sonic collecting duct epithelium [263]. The transcrip-
hedgehog, Krt5 keratin 5) (Reproduced with kind permis- tion factor Tbx18 is also critical for normal
sion from Elsevier. Gandhi, D, et al. Retinoid signaling in
urothelial development and is expressed through-
progenitors controls specification and regeneration of the
urothelium. Developmental Cell, volume 26, pp 469–482, out the mesenchyme investing the distal ureter.
2013, adapted from Figure 2) The genetic ablation of Tbx18 in mice leads to
24 C. Bates et al.

Ptch

Hcn3
Smo
Shh
Muscle Gli3R

Adventitia UPKs Fzd Kit


β-catenin

Adventitia
Wnt
Tbx18
Ptch

s
ell
lc
Bmp4

lia
he
Shh Smo
s
ell Bmp4

ot
c Smad
Ur
al receptor
m lls
ro ce Tshz3
St le
Lamina propria c
Dlg1 us SMC genes
m

tia
(stromal cells) Urothelium th

nti
oo
Sm

ve
Ad
Agtr1

Cnb1 AngII

Fig. 9 Genetic pathways regulating ureter develop- proteins) and Tshz3 regulate smooth muscle morphogene-
ment. The cell layers of the developing ureter include the sis. Wnt ligands in urothelium bind to Fzd receptors and
urothelium (green) and mesenchymal lamina propria/stro- stabilize β-catenin to stimulate smooth muscle develop-
mal cells (pink), smooth muscle (yellow), and adventitia ment and repress adventitial expansion. Angiotensin
(blue). Tbx18, expressed in mesenchyme, is critical for 2 (AngII) binds to type 1 receptors (Agtr1) that along
smooth muscle and urothelial development. Uroplakins with calcineurin b1 subunits also pattern smooth muscle.
(UPKs) are not only markers of urothelium but critical Dlg1 is critical for lamina propria/stromal cell morphogen-
for urothelial morphogenesis. Sonic hedgehog (Shh) is esis (Reproduced with kind permission from Wiley Peri-
secreted by urothelium, binding to patched (Ptch) receptors odicals, Inc. Rasouly, HM and Lu W. Lower urinary tract
to regulate morphogenesis of smooth muscle and Hcn3 and development and disease. Wiley Interdisciplinary
c-Kit expressing pacemakers [via interactions with Reviews: Systems Biology and Medicine, volume 5, pp
Smoothened (Smo) and Gli3 repressor (Gli3R)]. Other 307–342, adapted from Figure 3)
Shh targets including Bmp4 (acting through Smad

severe mispatterning of the mesenchyme and then Signaling from the ureteral urothelium has also
secondary defects in urothelial development been shown to be critical for mesenchymal pat-
[277]. Finally, not only are uroplakins markers terning. The growth factor, Sonic hedgehog (Shh),
of urothelial maturation, but they are also critical is secreted by the urothelium and binds to its
for normal urothelial morphogenesis. Loss of receptor, Patched 1, in surrounding mesenchyme.
either uroplakin II or uroplakin IIIa leads to severe Conditional ablation of Shh in developing ureteric
urothelial plaque defects, hypoplastic superficial epithelium leads to loss of mesenchymal prolifer-
cells, urothelial leakiness, hydronephrosis, and ation and smooth muscle differentiation
vesicoureteral reflux [276, 277]. Moreover, muta- [265]. Furthermore, Shh signaling from
tions in UPKIIIa in humans have been associated urothelium has also been shown to be critical for
with severe renal dysplasia and reflux [279], the formation of Hcn3 and c-kit expressing pace-
whereas the role of UPKII in humans is still makers within the ureteral muscle, via the hedge-
unclear [279]. hog signaling mediators Smoothened and Gli13
1 Embryonic Development of the Kidney 25

repressor [268]. In addition, downstream targets Bladder


of Shh signaling have been identified as necessary
for ureteral muscle development. One target, Compared with the ureter, much less is known
Bmp4, is essential for normal ureteral about the molecular control of bladder develop-
muscle investment around the ureter, particularly ment. Some of the pathways critical for formation
at the ureterovesical junction (in addition to of the urogenital sinus (future bladder) have been
having its aforementioned role in urothelial dif- identified. Bidirectional signaling between
ferentiation) [280]. The transcription factor, ephrin-B2 and EphB2, a ligand and its receptor
Teashirt 3 (Tshz3), downstream of both Shh and tyrosine kinase, is critical for septation of the
Bmp4, is critical for proximal ureteral cloaca into the ventral urogenital sinus and the
smooth muscle differentiation and for normal dorsal anorectal canal [287]. Sonic hedgehog sig-
ureteral peristaltic function; deletion of Tshz3 in naling is also critical for the formation of the
mice leads to severe muscle patterning defects and urogenital sinus. In mice, the loss of Shh or com-
congenital hydronephrosis [281]. A role for SHH pound mutations in the downstream hedgehog
in human urinary tract development was mediators, Gli2 and Gli3, leads to failure of cloa-
confirmed in patients with Pallister–Hall syn- cal septation [288]. During later stages of bladder
drome, who have mutations in GLI3 and urinary formation, urothelial cell stratification is depen-
tract anomalies such as hydroureter and dent on retinoid signaling emanating from the
hydronephrosis [268, 282]. Finally, Wnt lamina propria surrounding the urothelium; forced
ligands are also secreted by the urothelium, bind expression of a dominant negative retinoic acid
to frizzled receptors in the mesenchyme, and sig- receptor in developing mouse urothelium leads to
nal primarily by stabilization of β-catenin a loss of S cells, I cells, and uroplakins [272].
(Ctnnb1). Genetic ablation of Ctnnb1 leads to There are also some limited data on genetic
failure of mesenchymal proliferation and differ- pathways critical for bladder mesenchyme devel-
entiation into smooth muscle cells, with concur- opment. As is true in the ureter, several studies
rent expansion of the outer adventitial fibroblast have shown that Shh signaling from the bladder
layer [283]. urothelium is necessary for bladder mesenchyme
Other genes within the ureteral mesenchyme morphogenesis; loss of Shh in mice leads to a
have been shown to be critical for normal mesen- complete loss of smooth muscle formation
chymal development. As alluded to, loss of the [289–292]. Another mouse line, termed the
transcription factor Tbx18 leads to decreased megabladder mouse (mgb/), is a random trans-
proliferation and failure of smooth muscle differ- gene insertional mutant that lacks outer mesen-
entiation and ultimately severe hydrouretero- chymal condensation, has very limited αSMA
nephrosis [275]. Similarly, conditional ablation expression, and ultimately develops a massive
of the calcineurin b1 subunit in developing bladder with no functional detrusor [271]. While
ureteral mesenchyme leads to reduced smooth the gene disrupted in mgb/ mice is still
muscle proliferation and hydronephrosis unknown, Shh signaling appears perturbed, and
[284]. Furthermore, genetic ablation of the angio- the bladders lack expression of myocardin, a tran-
tensin type 1 receptor (which is expressed in ure- scription factor critical for smooth muscle
teral mesenchyme) leads to smooth muscle development [293].
hypoplasia, lack of peristalsis, and ultimately
hydronephrosis [285]. Finally, discs large
homolog 1 (Dlg1) is the only gene identified to Ureter–Bladder Anastomosis
date that is necessary for lamina propria/stromal
cell development; genetic deletion of Dlg1 in There are some data on genetic pathways neces-
mice led to an absence of the entire ureteral lamina sary for the proper connection between the ureter
propria layer and disorganized muscle and bladder, i.e., apoptosis of the common nephric
development [286]. duct starting at E12.5 in mice. Retinoid signaling
26 C. Bates et al.

from the bladder has been shown to be critical for 8. Piscione TD, Rosenblum ND. The molecular
this process. Genetic ablation of retinaldehyde control of renal branching morphogenesis: current
knowledge and emerging insights. Differentiation.
dehydrogenase-2, an enzyme expressed in the 2002;70(6):227–46.
urogenital sinus and necessary for retinoic acid 9. Dressler GR. The cellular basis of kidney develop-
synthesis, led to persistence of the common neph- ment. Annu Rev Cell Dev Biol. 2006;22:509–29.
ric duct with ureters ending blindly in the meso- 10. Costantini F. Renal branching morphogenesis: con-
cepts, questions, and recent advances. Differentiation.
nephric duct [273]. In addition, Ret has also been 2006;74(7):402–21.
shown to be essential for ureter–bladder anasto- 11. Shah MM, Sampogna RV, Sakurai H, Bush KT,
mosis. Mice with a point mutation in tyrosine Nigam SK. Branching morphogenesis and kidney
1015, the phospholipase Cγ binding site on Ret disease. Development. 2004;131(7):1449–62.
12. Yu J, McMahon AP, Valerius MT. Recent genetic
(RetY1015), have a persistent common nephric studies of mouse kidney development. Curr Opin
duct due to enhanced proliferation and decreased Genet Dev. 2004;14(5):550–7.
apoptosis [294]. 13. Little MH, McMahon AP. Mammalian kidney devel-
Lower urinary tract defects contribute substan- opment: principles, progress, and projections. Cold
Spring Harb Perspect Biol [Research Support, N.I.
tially to chronic kidney disease in children, neces- H., Extramural Review]. 2012;4(5):1–18.
sitating a better understanding about the genes 14. Costantini F, Kopan R. Patterning a complex organ:
regulating lower urinary tract morphogenesis. branching morphogenesis and nephron segmentation
While our understanding of the molecular control in kidney development. Dev Cell [Research Support,
N.I.H., Extramural Review]. 2010;18(5):698–712.
of ureter and bladder development trails that of the 15. Hwang DY, Dworschak GC, Kohl S, Saisawat P,
kidney, more studies are emerging on how the Vivante A, Hilger AC, et al. Mutations in 12 known
ureter and bladder form. dominant disease-causing genes clarify many con-
genital anomalies of the kidney and urinary tract.
Kidney Int. 2014;85(6):1429–33.
16. Chen S, Bellew C, Yao X, Stefkova J, Dipp S,
Saifudeen Z, et al. Histone deacetylase (HDAC)
References activity is critical for embryonic kidney gene
expression, growth, and differentiation. J Biol
1. Osathanondh V, Potter EL. Development of human Chem [Research Support, N.I.H., Extramural
kidney as shown by microdissection. Arch Path. Research Support, Non-U.S. Gov’t]. 2011;286(37):
1966;82:391–402. 32775–89.
2. Potter EL. Normal and abnormal development of the 17. Chen S, El-Dahr SS. Histone deacetylases in kidney
kidney. Chicago: Year Book Medical Publishers; development: implications for disease and therapy.
1972. Pediatr Nephrol [Review]. 2013;28(5):689–98.
3. Hinchliffe SA, Sargent PH, Howard CV, Chan YF, 18. McLaughlin N, Wang F, Saifudeen Z, El-Dahr SS. In
van Velzen D. Human intrauterine renal growth situ histone landscape of nephrogenesis. Epigenetics
expressed in absolute number of glomeruli assessed [Research Support, N.I.H., Extramural]. 2014;9
by the disector method and Cavalieri principle. Lab (2):222–35.
Invest. 1991;64(6):777–84. 19. Ho J, Kreidberg JA. The long and short of
4. Hughson M, Farris 3rd AB, Douglas-Denton R, Hoy microRNAs in the kidney. J Am Soc Nephrol.
WE, Bertram JF. Glomerular number and size in 2012;23(3):400–4.
autopsy kidneys: the relationship to birth weight. 20. Ho J, Ng KH, Rosen S, Dostal A, Gregory RI,
Kidney Int. 2003;63(6):2113–22. Kreidberg JA. Podocyte-specific loss of functional
5. Rodriguez MM, Gomez AH, Abitbol CL, Chandar JJ, microRNAs leads to rapid glomerular and tubular
Duara S, Zilleruelo GE. Histomorphometric analysis injury. J Am Soc Nephrol. 2008;19(11):2069–75.
of postnatal glomerulogenesis in extremely preterm 21. Marrone AK, Stolz DB, Bastacky SI, Kostka D,
infants. Pediatr Dev Pathol. 2004;7(1):17–25. Bodnar AJ, Ho J. MicroRNA-17~92 is required for
6. Keller G, Zimmer G, Mall G, Ritz E, Amann nephrogenesis and renal function. J Am Soc Nephrol.
K. Nephron number in patients with primary hyper- 2014;25(7):1440–52.
tension. N Engl J Med. 2003;348(2):101–8. 22. Ho J, Pandey P, Schatton T, Sims-Lucas S, Khalid M,
7. Hoy WE, Hughson MD, Singh GR, Douglas-Denton- Frank MH, et al. The pro-apoptotic protein Bim is a
R, Bertram JF. Reduced nephron number and microRNA target in kidney progenitors. J Am Soc
glomerulomegaly in Australian Aborigines: a group Nephrol [Research Support, N.I.H., Extramural
at high risk for renal disease and hypertension. Kid- Research Support, Non-U.S. Gov’t]. 2011;22(6):
ney Int. 2006;70(1):104–10. 1053–63.
1 Embryonic Development of the Kidney 27

23. Lelievre-Pegorier M, Vilar J, Ferrier ML, Moreau E, 36. Welham SJ, Riley PR, Wade A, Hubank M, Woolf
Freund N, Gilbert T, et al. Mild vitamin A deficiency AS. Maternal diet programs embryonic kidney gene
leads to inborn nephron deficit in the rat. Kidney Int. expression. Physiol Genomics [Research Support,
1998;54(5):1455–62. Non-U.S. Gov’t]. 2005;22(1):48–56.
24. Goodyer P, Kurpad A, Rekha S, Muthayya S, 37. Tse HK, Leung MB, Woolf AS, Menke AL, Hastie
Dwarkanath P, Iyengar A, et al. Effects of maternal ND, Gosling JA, et al. Implication of Wt1 in the
vitamin A status on kidney development: a pilot pathogenesis of nephrogenic failure in a mouse
study. Pediatr Nephrol [Multicenter Study Research model of retinoic acid-induced caudal regression syn-
Support, Non-U.S. Gov’t]. 2007;22(2):209–14. drome. Am J Pathol [Research Support, Non-U.S.
25. Harding SD, Armit C, Armstrong J, Brennan J, Gov’t]. 2005;166(5):1295–307.
Cheng Y, Haggarty B, et al. The GUDMAP 38. Sanker S, Cirio MC, Vollmer LL, Goldberg ND,
database–an online resource for genitourinary McDermott LA, Hukriede NA, et al. Development
research. Development. 2011;138(13):2845–53. of high-content assays for kidney progenitor cell
26. McMahon AP, Aronow BJ, Davidson DR, Davies JA, expansion in transgenic zebrafish. J Biomol Screen
Gaido KW, Grimmond S, et al. GUDMAP: the geni- [Research Support, N.I.H., Extramural]. 2013;18
tourinary developmental molecular anatomy project. (10):1193–202.
J Am Soc Nephrol. 2008;19(4):667–71. 39. Liu L, Fan XD. CRISPR-Cas system: a powerful tool
27. Diez-Roux G, Banfi S, Sultan M, Geffers L, Anand S, for genome engineering. Plant Mol Biol. 2014;85
Rozado D, et al. A high-resolution anatomical atlas of (3):209–18.
the transcriptome in the mouse embryo. PLoS Biol 40. Drummond IA, Majumdar A, Hentschel H, Elger M,
[Research Support, Non-U.S. Gov’t]. 2011;9(1): Solnica-Krezel L, Schier AF, et al. Early development
e1000582. of the zebrafish pronephros and analysis of mutations
28. Avner ED, Ellis D, Temple T, Jaffe R. Metanephric affecting pronephric function. Development.
development in serum-free organ culture. 1998;125:4655–67.
In Vitro Cell Dev Biol. 1982;18:675–82. PMID: 41. Vize PD, Seufert DW, Carroll TJ, Wallingford
7129481. JB. Model systems for the study of kidney develop-
29. Sariola H, Saarma M, Sainio K, Arumae U, Palgi J, ment: use of the pronephros in the analysis of organ
Vaahtokari A, et al. Dependence of kidney morpho- induction and patterning. Dev Biol.
genesis on the expression of nerve growth factor 1997;188:189–204.
receptor. Science [Research Support, Non-U.S. 42. Staack A, Donjacour AA, Brody J, Cunha GR, Carroll
Gov’t]. 1991;254(5031):571–3. P. Mouse urogenital development: a practical
30. Woolf AS, Kolatsi-Joannou M, Hardman P, approach. Differentiation. 2003;71(7):402–13.
Andermarcher E, Moorby C, Fine LG, et al. Roles 43. Saxen L. Organogenesis of the kidney. Cambridge:
of hepatocyte growth factor/scatter factor and the met Cambridge University Press; 1987.
receptor in the early development of the metanephros. 44. James RG, Kamei CN, Wang Q, Jiang R, Schultheiss
J Cell Biol [Research Support, Non-U.S. Gov’t]. TM. Odd-skipped related 1 is required for develop-
1995;128(1–2):171–84. ment of the metanephric kidney and regulates forma-
31. Sims-Lucas S. Analysis of 3D branching pattern: tion and differentiation of kidney precursor cells.
hematoxylin and eosin method. Methods Mol Biol. Development. 2006;133(15):2995–3004.
2012;886:73–86. 45. Mugford JW, Sipila P, McMahon JA, McMahon
32. Sims-Lucas S, Argyropoulos C, Kish K, McHugh K, AP. Osr1 expression demarcates a multi-potent pop-
Bertram JF, Quigley R, et al. Three-dimensional ulation of intermediate mesoderm that undergoes pro-
imaging reveals ureteric and mesenchymal defects gressive restriction to an Osr1-dependent nephron
in Fgfr2-mutant kidneys. J Am Soc Nephrol. progenitor compartment within the mammalian kid-
2009;20(12):2525–33. ney. Dev Biol [Research Support, N.I.H., Extramural
33. Short KM, Combes AN, Lefevre J, Ju AL, Georgas Research Support, Non-U.S. Gov’t]. 2008;324(1):
KM, Lamberton T, et al. Global quantification of 88–98.
tissue dynamics in the developing mouse kidney. 46. Dressler GR, Deutsch U, Chowdhury K, Nornes HO,
Dev Cell [Research Support, Non-U.S. Gov’t]. Gruss P. Pax-2, a new murine paired-box-containing
2014;29(2):188–202. gene and its expression in the developing excretory
34. Tarantal AF, Han VK, Cochrum KC, Mok A, system. Development. 1990;109:787–95.
daSilva M, Matsell DG. Fetal rhesus monkey model 47. Bouchard M, Souabni A, Mandler M, Neubuser A,
of obstructive renal dysplasia. Kidney Int [Research Busslinger M. Nephric lineage specification by Pax2
Support, U.S. Gov’t, P.H.S.]. 2001;59(2):446–56. and Pax8. Genes Dev [Research Support, Non-U.S.
35. Yang SP, Woolf AS, Quinn F, Winyard Gov’t]. 2002;16(22):2958–70.
PJ. Deregulation of renal transforming growth 48. Fujii T, Pichel JG, Taira M, Toyama R, Dawid IB,
factor-beta1 after experimental short-term ureteric Westphal H. Expression patterns of the murine LIM
obstruction in fetal sheep. Am J Pathol [Research class homeobox gene lim1 in the developing brain
Support, Non-U.S. Gov’t]. 2001;159(1):109–17. and excretory system. Dev Dyn. 1994;1:73–83.
28 C. Bates et al.

49. Grote D, Souabni A, Busslinger M, Bouchard M. Pax 63. Barasch J, Yang J, Ware CB, Taga T, Yoshida K,
2/8-regulated Gata 3 expression is necessary for mor- Erdjument-Bromage H, et al. Mesenchymal to epithe-
phogenesis and guidance of the nephric duct in the lial conversion in rat metanephros is induced by LIF.
developing kidney. Development. 2006;133(1): Cell. 1999;99(4):377–86.
53–61. 64. Das A, Tanigawa S, Karner CM, Xin M, Lum L,
50. Pachnis V, Mankoo B, Costantini F. Expression of the Chen C, et al. Stromal-epithelial crosstalk regulates
c-ret proto-oncogene during mouse embryogenesis. kidney progenitor cell differentiation. Nat Cell Biol.
Development. 1993;119:1005–17. 2013;15(9):1035–44.
51. Erickson RA. Inductive interactions in the develop- 65. Yang J, Blum A, Novak T, Levinson R, Lai E,
ment of the mouse metanephros. J Exp Zool. Barasch J. An epithelial precursor is regulated by the
1968;169(1):33–42. ureteric bud and by the renal stroma. Dev Biol.
52. Grobstein C. Morphogenetic interaction between 2002;246(2):296–310.
embryonic mouse tissues separated by a membrane 66. Cui S, Schwartz L, Quaggin SE. Pod1 is required in
filter. Nature. 1953;172:869–71. stromal cells for glomerulogenesis. Dev Dyn.
53. Grobstein C. Inductive interaction in the development 2003;226(3):512–22.
of the mouse metanephros. J Exp Zool. 67. Clark AT, Bertram JF. Molecular regulation of neph-
1955;130:319–40. ron endowment. Am J Physiol. 1999;276(4 Pt 2):
54. Hatini V, Huh SO, Herzlinger D, Soares VC, Lai F485–97.
E. Essential role of stromal mesenchyme in kidney 68. Schrijvers BF, Flyvbjerg A, De Vriese AS. The role
morphogenesis revealed by targeted disruption of of vascular endothelial growth factor (VEGF) in
Winged Helix transcription factor BF-2. Genes Dev. renal pathophysiology. Kidney Int. 2004;65
1996;10:1467–78. (6):2003–17.
55. Mendelsohn C, Batourina E, Fung S, Gilbert T, Dodd 69. Simon M, Rockl W, Hornig C, Grone EF, Theis H,
J. Stromal cells mediate retinoid-dependent functions Weich HA, et al. Receptors of vascular endothelial
essential for renal development. Development. growth factor/vascular permeability factor (VEGF/
1999;126:1139–48. VPF) in fetal and adult human kidney: localization
56. Batourina E, Gim S, Bello N, Shy M, Clagett- and [125I]VEGF binding sites. J Am Soc Nephrol.
Dame M, Srinivas S, et al. Vitamin A controls epithe- 1998;9(6):1032–44.
lial/mesenchymal interactions through Ret expres- 70. Evan AP, Gattone 2nd VH, Schwartz
sion. Nat Genet. 2001;27:74–8. GJ. Development of solute transport in rabbit proxi-
57. Levinson RS, Batourina E, Choi C, mal tubule. II. Morphologic segmentation. Am J
Vorontchikhina M, Kitajewski J, Mendelsohn Physiol. 1983;245(3):F391–407.
CL. Foxd1-dependent signals control cellularity in 71. Fetterman GH, Shuplock NA, Philipp FJ, Gregg
the renal capsule, a structure required for normal HS. The growth and maturation of human glomeruli
renal development. Development [Research Support, and proximal convolutions from term to adulthood:
U.S. Gov’t, P.H.S.]. 2005;132(3):529–39. studies by microdissection. Pediatrics.
58. Qiao J, Uzzo R, Obara-Ishihara T, Degenstein L, 1965;35:601–19.
Fuchs E, Herzlinger D. FGF-7 modulates ureteric 72. Neiss WF. Histogenesis of the loop of Henle in the rat
bud growth and nephron number in the developing kidney. Anat Embryol. 1982;164(3):315–30.
kidney. Development. 1999;126:547–54. 73. Nakai S, Sugitani Y, Sato H, Ito S, Miura Y,
59. Gao X, Chen X, Taglienti M, Rumballe B, Little MH, Ogawa M, et al. Crucial roles of Brn1 in distal tubule
Kreidberg JA. Angioblast-mesenchyme induction of formation and function in mouse kidney. Develop-
early kidney development is mediated by Wt1 and ment. 2003;130(19):4751–9.
Vegfa. Development. 2005;132(24):5437–49. 74. Neiss WF, Klehn KL. The postnatal development of
60. Tufro-McReddie A, Norwood VF, Aylor KW, Botkin the rat kidney, with special reference to the chemodif-
SJ, Carey RM, Gomez RA. Oxygen regulates vascu- ferentiation of the proximal tubule. Histochemistry.
lar endothelial growth factor-mediated 1981;73(2):251–68.
vasculogenesis and tubulogenesis. Dev Biol. 75. Nishinakamura R, Matsumoto Y, Nakao K,
1997;183(2):139–49. Nakamura K, Sato A, Copeland NG, et al. Murine
61. Meyer TN, Schwesinger C, Bush KT, Stuart RO, homolog of SALL1 is essential for ureteric bud inva-
Rose DW, Shah MM, et al. Spatiotemporal regulation sion in kidney development. Development.
of morphogenetic molecules during in vitro branching 2001;128:3105–15.
of the isolated ureteric bud: toward a model of 76. Xu PX, Zheng W, Huang L, Maire P, Laclef C, Silvius
branching through budding in the developing kidney. D. Six1 is required for the early organogenesis of
Dev Biol. 2004;275(1):44–67. mammalian kidney. Development. 2003;130(14):
62. Barasch J, Qiao J, McWilliams G, Chen D, Oliver JA, 3085–94.
Herzlinger D. Ureteric bud cells secrete multiple fac- 77. Kalatzis V, Sahly I, El-Amraoui A, Petit C. Eya1
tors, including bFGF, which rescue renal progenitors expression in the developing ear and kidney: towards
from apoptosis. Am J Physiol. 1997;273:F757–67. the understanding of the pathogenesis of Branchio-
1 Embryonic Development of the Kidney 29

Oto-Renal (BOR) syndrome. Dev Dyn. 92. Davies JA, Garrod DR. Induction of early stages of
1998;213:486–99. kidney tubule differentiation by lithium ions. Dev
78. Xu P-X, Adams J, Peters H, Brown MC, Heaney S, Biol. 1995;167(1):50–60.
Maas R. Eya1-deficient mice lack ears and kidneys 93. Hedgepeth CM, Conrad LJ, Zhang J, Huang HC, Lee
and show abnormal apoptosis of organ primordia. Nat VM, Klein PS. Activation of the Wnt signaling path-
Genet. 1999;23:113–7. way: a molecular mechanism for lithium action. Dev
79. Hellmich HL, Kos L, Cho ES, Mahon KA, Zimmer Biol. 1997;185(1):82–91.
A. Embryonic expression of glial cell-line derived 94. Klein PS, Melton DA. A molecular mechanism for the
neurotrophic factor (GDNF) suggests multiple devel- effect of lithium on development. Proc Natl Acad Sci
opmental roles in neural differentiation and epithelial- U S A. 1996;93(16):8455–9.
mesenchymal interactions. Mech Dev. 95. Carroll TJ, Park JS, Hayashi S, Majumdar A,
1996;54:95–105. McMahon AP. Wnt9b plays a central role in the
80. Kreidberg JA, Sariola H, Loring JM, Maeda M, regulation of mesenchymal to epithelial transitions
Pelletier J, Housman D, et al. WT-1 is required for underlying organogenesis of the mammalian urogen-
early kidney development. Cell. 1993;74:679–91. ital system. Dev Cell. 2005;9(2):283–92.
81. Boyle S, Shioda T, Perantoni AO, de Caestecker 96. Karner CM, Das A, Ma Z, Self M, Chen C, Lum L,
M. Cited1 and Cited2 are differentially expressed in et al. Canonical Wnt9b signaling balances progenitor
the developing kidney but are not required for cell expansion and differentiation during kidney
nephrogenesis. Dev Dyn [Research Support, N.I.H., development. Development [Research Support, N.I.
Extramural]. 2007;236(8):2321–30. H., Extramural Research Support, Non-U.S. Gov’t].
82. Self M, Lagutin OV, Bowling B, Hendrix J, Cai Y, 2011;138(7):1247–57.
Dressler GR, et al. Six2 is required for suppression of 97. Brown AC, Adams D, de Caestecker M, Yang X,
nephrogenesis and progenitor renewal in the develop- Friesel R, Oxburgh L. FGF/EGF signaling regulates
ing kidney. EMBO J. 2006;25(21):5214–28. the renewal of early nephron progenitors during
83. Cho EA, Patterson LT, Brookhiser WT, Mah S, embryonic development. Development [Research
Kintner C, Dressler GR. Differential expression and Support, American Recovery and Reinvestment Act
function of cadherin-6 during renal epithelium devel- Research Support, N.I.H., Extramural Research Sup-
opment. Development. 1998;125(5):803–12. port, Non-U.S. Gov’t]. 2011;138(23):5099–112.
84. M€uller U, Wang D, Denda S, Meneses JJ, Pedersen 98. Barak H, Huh SH, Chen S, Jeanpierre C,
RA, Reichardt LF. Integrin α8β1 is critically impor- Martinovic J, Parisot M, et al. FGF9 and FGF20
tant for epithelial-mesenchymal interactions during maintain the stemness of nephron progenitors in
kidney morphogenesis. Cell. 1997;88:603–13. mice and man. Dev Cell. 2012;22(6):1191–207.
85. Brophy PD, Ostrom L, Lang KM, Dressler 99. Hains D, Sims-Lucas S, Kish K, Saha M, McHugh K,
GR. Regulation of ureteric bud outgrowth by Pax2- Bates CM. Role of fibroblast growth factor receptor
dependent activation of the glial derived neurotrophic 2 in kidney mesenchyme. Pediatr Res [Research Sup-
factor gene. Development. 2001;128:4747–56. port, N.I.H., Extramural]. 2008;64(6):592–8.
86. Kobayashi A, Kwan KM, Carroll TJ, McMahon AP, 100. Poladia DP, Kish K, Kutay B, Hains D, Kegg H,
Mendelsohn CL, Behringer RR. Distinct and sequen- Zhao H, et al. Role of fibroblast growth factor recep-
tial tissue-specific activities of the LIM-class homeo- tors 1 and 2 in the metanephric mesenchyme. Dev
box gene Lim1 for tubular morphogenesis during Biol [Research Support, N.I.H., Extramural].
kidney development. Development [Research Sup- 2006;291(2):325–39.
port, N.I.H., Extramural Research Support, 101. Sims-Lucas S, Cusack B, Baust J, Eswarakumar VP,
U.S. Gov’t, P.H.S.]. 2005;132(12):2809–23. Masatoshi H, Takeuchi A, et al. Fgfr1 and the IIIc
87. Rothenpieler UW, Dressler GR. Pax-2 is required for isoform of Fgfr2 play critical roles in the metanephric
mesenchyme-to-epithelium conversion during kidney mesenchyme mediating early inductive events in kid-
development. Development. 1993;119:711–20. ney development. Dev Dyn [Research Support, N.I.
88. Torres M, Gomez-Pardo E, Dressler GR, Gruss H., Extramural]. 2011;240(1):240–9.
P. Pax-2 controls multiple steps of urogenital devel- 102. Bard JB. Growth and death in the developing mam-
opment. Development. 1995;121:4057–65. malian kidney: signals, receptors and conversations.
89. Tsang TE, Shawlot W, Kinder SJ, Kobayashi A, Bioessays. 2002;24(1):72–82.
Kwan KM, Schughart K, et al. Lim1 activity is 103. Dudley AT, Godin RE, Robertson EJ. Interaction
required for intermediate mesoderm between FGF and BMP signaling pathways regulates
differentiation in the mouse embryo. Dev Biol. development of metanephric mesenchyme. Genes
2000;223(1):77–90. Dev. 1999;13:1601–13.
90. Shawlot W, Behringer RR. Requirement for Lim1 in 104. Koseki C, Herzlinger D, Al-Awqati Q. Apoptosis in
head-organizer function. Nature. 1995;374:425–30. metanephric development. J Cell Biol. 1992;119(5):
91. Hilliard SA, Yao X, El-Dahr SS. Mdm2 is required for 1327–33.
maintenance of the nephrogenic niche. Dev Biol. 105. Plisov SY, Yoshino K, Dove LF, Higinbotham KG,
2014;387(1):1–14. Rubin JS, Perantoni AO. TGF beta 2, LIF and FGF2
30 C. Bates et al.

cooperate to induce nephrogenesis. Development. 119. Perantoni AO, Timofeeva O, Naillat F, Richman C,
2001;128(7):1045–57. Pajni-Underwood S, Wilson C, et al. Inactivation of
106. Araki T, Saruta T, Okano H, Miura M. Caspase activ- FGF8 in early mesoderm reveals an essential role in
ity is required for nephrogenesis in the developing kidney development. Development. 2005;132(17):
mouse metanephros. Exp Cell Res. 1999;248(2): 3859–71.
423–9. 120. Gerber SD, Steinberg F, Beyeler M, Villiger PM,
107. Nishimura H, Yerkes E, Hohenfellner K, Miyazaki Y, Trueb B. The murine Fgfrl1 receptor is essential for
Ma J, Hunley TE, et al. Role of the angiotensin type the development of the metanephric kidney. Dev Biol.
2 receptor gene in congenital anomalies of the kidney 2009;335(1):106–19.
and urinary tract, CAKUT, of mice and men. Mol 121. Majumdar A, Lun K, Brand M, Drummond
Cell. 1999;3:1–10. IA. Zebrafish no isthmus reveals a role for pax2.1 in
108. Coles HSR, Burne JF, Raff MC. Large-scale normal tubule differentiation and patterning events in the
cell death in the developing rat kidney and its reduc- pronephric primordia. Development. 2000;127(10):
tion by epidermal growth factor. Development. 2089–98.
1993;117:777–84. 122. Wallingford JB, Carroll TJ, Vize PD. Precocious
109. Winyard PJD, Nauta J, Lirenman DS, Hardman P, expression of the Wilms’ tumor gene xWT1 inhibits
Sams VR, Risdon RA, et al. Deregulation of cell embryonic kidney development in Xenopus laevis.
survival in cystic and dysplastic renal development. Dev Biol. 1998;202(1):103–12.
Kidney Int. 1996;49:135–46. 123. Ryan G, Steele-Perkins V, Morris JF, Rauscher 3rd FJ,
110. Marrone AK, Stolz DB, Bastacky SI, Kostka D, Dressler GR. Repression of Pax-2 by WT1 during
Bodnar AJ, Ho J. MicroRNA-17~92 is required for normal kidney development. Development.
nephrogenesis and renal function. J Am Soc Nephrol. 1995;121(3):867–75.
2014;25(7):1440–52. 124. Pelletier J, Schalling M, Buckler AJ, Rogers A, Haber
111. Karavanov AA, Karavanova I, Perantoni A, Dawid DA, Housman D. Expression of the Wilms’ tumor
IB. Expression pattern of the rat Lim-1 homeobox gene WT1 in the murine urogenital system. Genes
gene suggests a dual role during kidney development. Dev. 1991;5:1345–56.
Int J Dev Biol. 1998;42:61–6. 125. Dressler GR, Wilkinson JE, Rothenpieler UW,
112. Stewart CL, Kaspar P, Brunet LJ, Bhatt H, Gadi I, Patterson LT, Silliams-Simons L, Westphal
Kontgen F, et al. Blastocyst implantation depends on H. Deregulation of Pax-2 expression in transgenic
maternal expression of leukaemia inhibitory factor. mice generates severe kidney abnormalities. Nature.
Nature. 1992;359(6390):76–9. 1993;362:65–7.
113. Sanford LP, Ormsby I, Gittenberger-de Groot AC, 126. Dressler GR, Douglass EC. Pax-2 is a DNA-binding
Sariola H, Friedman R, Boivin GP, et al. TGFβ2 protein expressed in embryonic kidney and
knockout mice have multiple developmental defects Wilms tumor. Proc Natl Acad Sci U S A. 1992;89:
that are non-overlapping with other TGFβ knockout 1179–83.
phenotypes. Development. 1997;124:2659–70. 127. Cheng HT, Miner JH, Lin M, Tansey MG, Roth K,
114. McPherron AC, Lawler AM, Lee SJ. Regulation of Kopan R. Gamma-secretase activity is dispensable for
anterior/posterior patterning of the axial skeleton mesenchyme-to-epithelium transition but required for
by growth/differentiation factor 11. Nat Genet. podocyte and proximal tubule formation in develop-
1999;22(3):260–4. ing mouse kidney. Development. 2003;130(20):
115. Herzlinger D, Qiao J, Cohen D, Ramakrishna N, 5031–42.
Brown AMC. Induction of kidney epithelial morpho- 128. Cheng HT, Kim M, Valerius MT, Surendran K,
genesis by cells expressing wnt-1. Dev Biol. Schuster-Gossler K, Gossler A, et al. Notch2, but
1994;166:815–8. not Notch1, is required for proximal fate acquisition
116. Kispert A, Vainio S, McMahon AP. Wnt-4 is a mes- in the mammalian nephron. Development. 2007;134
enchymal signal for epithelial transformation of meta- (4):801–11.
nephric mesenchyme in the developing kidney. 129. Wang P, Pereira FA, Beasley D, Zheng H. Presenilins
Development. 1998;125:4225–34. are required for the formation of comma- and
117. Yoshino K, Rubin JS, Higinbotham KG, Uren A, S-shaped bodies during nephrogenesis. Development.
Anest V, Plisov SY, et al. Secreted Frizzled-related 2003;130(20):5019–29.
proteins can regulate metanephric development. 130. Boyle SC, Kim M, Valerius MT, McMahon AP,
Mech Dev. 2001;102(1–2):45–55. Kopan R. Notch pathway activation can replace the
118. Grieshammer U, Cebrian C, Ilagan R, Meyers E, requirement for Wnt4 and Wnt9b in mesenchymal-to-
Herzlinger D, Martin GR. FGF8 is required for cell epithelial transition of nephron stem cells. Develop-
survival at distinct stages of nephrogenesis and for ment [Research Support, N.I.H., Extramural].
regulation of gene expression in nascent nephrons. 2011;138(19):4245–54.
Development [Research Support, N.I.H., Extramural 131. Kreidberg JA. Podocyte differentiation and
Research Support, Non-U.S. Gov’t Research Support, glomerulogenesis. J Am Soc Nephrol. 2003;14(3):
U.S. Gov’t, P.H.S.]. 2005;132(17):3847–57. 806–14.
1 Embryonic Development of the Kidney 31

132. Robert B, St John PL, Hyink DP, Abrahamson WT1 results in the characteristic urogenital abnormal-
DR. Evidence that embryonic kidney cells expressing ities of Denys-Drash syndrome. Proc Natl Acad Sci U
flk-1 are intrinsic, vasculogenic angioblasts. Am J S A. 1999;96(6):2931–6.
Physiol. 1996;271(3 Pt 2):F744–53. 147. Hammes A, Guo JK, Lutsch G, Leheste JR,
133. Hyink DP, Tucker DC, St John PL, Landrock D, Ziegler U, et al. Two splice variants of
Leardkamolkarn V, Accavitti MA, Abrass CK, the Wilms’ tumor 1 gene have distinct functions dur-
et al. Endogenous origin of glomerular endothelial ing sex determination and nephron formation. Cell.
and mesangial cells in grafts of embryonic kidneys. 2001;106(3):319–29.
Am J Physiol. 1996;270(5 Pt 2):F886–99. 148. Guo JK, Menke AL, Gubler MC, Clarke AR,
134. Ricono JM, Xu YC, Arar M, Jin DC, Barnes JL, Harrison D, Hammes A, et al. WT1 is a key regulator
Abboud HE. Morphological insights into the of podocyte function: reduced expression levels cause
origin of glomerular endothelial and mesangial crescentic glomerulonephritis and mesangial sclero-
cells and their precursors. J Histochem Cytochem. sis. Hum Mol Genet. 2002;11(6):651–9.
2003;51(2):141–50. 149. Yang Y, Jeanpierre C, Dressler GR, Lacoste M,
135. Sariola H, Ekblom P, Lehtonen E, Saxen Niaudet P, Gubler MC. WT1 and PAX-2 podocyte
L. Differentiation and vascularization of the meta- expression in Denys-Drash syndrome and isolated
nephric kidney grafted on the chorioallantoic mem- diffuse mesangial sclerosis. Am J Pathol. 1999;154
brane. Dev Biol. 1983;96(2):427–35. (1):181–92.
136. Nagata M, Nakayama K, Terada Y, Hoshi S, 150. Barbaux S, Niaudet P, Gubler M-C, Gr€ unfeld J-P,
Watanabe T. Cell cycle regulation and differentiation Jaubert F, Kuttenn F, et al. Donor splice-site mutations
in the human podocyte lineage. Am J Pathol. in WT1 are responsible for Frasier syndrome. Nat
1998;153(5):1511–20. Genet. 1997;17:467–70.
137. Garrod DR, Fleming S. Early expression of desmo- 151. Klamt B, Koziell A, Poulat F, Wieacker P, Scambler P,
somal components during kidney tubule morphogen- Berta P, et al. Frasier syndrome is caused by defective
esis in human and murine embryos. Development. alternative splicing of WT1 leading to an altered ratio
1990;108(2):313–21. of WT1+/KTS splice isoforms. Hum Mol Genet.
138. Pavenstadt H, Kriz W, Kretzler M. Cell biology of the 1998;7:709–14.
glomerular podocyte. Physiol Rev. 2003;83(1): 152. Coppes MJ, Liefers GJ, Higuchi M, Zinn AB, Balfe
253–307. JW, Williams BR. Inherited WT1 mutation in Denys-
139. Miner JH, Sanes JR. Collagen IValpha 3, alpha 4, and Drash syndrome. Cancer Res. 1992;52(21):6125–8.
alpha 5 chains in rodent basal laminae: sequence, 153. Quaggin SE, Schwartz L, Cui S, Igarashi P,
distribution, association with laminins, and develop- Deimling J, Post M, et al. The basic-helix-loop-helix
mental switches. J Cell Biol. 1994;127(3):879–91. protein pod1 is critically important for kidney and
140. Miner JH, Li C. Defective glomerulogenesis in the lung organogenesis. Development.
absence of laminin alpha5 demonstrates a develop- 1999;126:5771–83.
mental role for the kidney glomerular basement mem- 154. Sadl V, Jin F, Yu J, Cui S, Holmyard D, Quaggin S,
brane. Dev Biol. 2000;217(2):278–89. et al. The mouse Kreisler (Krml1/MafB) segmenta-
141. Miner JH, Sanes JR. Molecular and functional defects tion gene is required for differentiation of glomerular
in kidneys of mice lacking collagen alpha 3(IV): visceral epithelial cells. Dev Biol. 2002;249
implications for Alport syndrome. J Cell Biol. (1):16–29.
199;135(5):1403–13. 155. Miner JH, Morello R, Andrews KL, Li C, Antignac C,
142. Noakes PG, Miner JH, Gautam M, Cunningham JM, Shaw AS, et al. Transcriptional induction of slit
Sanes JR, Merlie JP. The renal glomerulus of mice diaphragm genes by Lmx1b is required in
lacking s-laminin/laminin ß2: nephrosis despite podocyte differentiation. J Clin Invest. 2002;109(8):
molecular compensation by laminin ß1. Nature 1065–72.
Genet. 1995;10:400–6. 156. Dreyer SD, Zhou G, Baldini A, Winterpacht A,
143. Ekblom P. Formation of basement membranes in Zabel B, Cole W, et al. Mutations in LMX1B cause
embryonic kidney: an immunohistological study. abnormal skeletal patterning and renal dysplasia in
J Cell Biol. 1981;91:1–10. nail patella syndrome. Nat Genet. 1998;19:47–50.
144. Sariola H, Timpl R, von der Mark K, Mayne R, Fitch 157. Lemley KV. Kidney disease in nail-patella syndrome.
JM, Linsenmayer TF, et al. Dual origin of glomerular Pediatr Nephrol [Research Support, Non-U.S. Gov’t
basement membrane. Dev Biol. 1984;101:86–96. Review]. 2009;24(12):2345–54.
145. Gao F, Maiti S, Sun G, Ordonez NG, Udtha M, Deng 158. Harvey SJ, Jarad G, Cunningham J, Goldberg S,
JM, et al. The Wt1+/R394W mouse displays Schermer B, Harfe BD, et al. Podocyte-specific dele-
glomerulosclerosis and early-onset renal failure char- tion of dicer alters cytoskeletal dynamics and causes
acteristic of human Denys-Drash syndrome. Mol Cell glomerular disease. J Am Soc Nephrol. 2008;19(11):
Biol. 2004;24(22):9899–910. 2150–8.
146. Patek CE, Little MH, Fleming S, Miles C, Charlieu JP, 159. Shi S, Yu L, Chiu C, Sun Y, Chen J, Khitrov G,
Clarke AR, et al. A zinc finger truncation of murine et al. Podocyte-selective deletion of dicer induces
32 C. Bates et al.

proteinuria and glomerulosclerosis. J Am Soc 174. Fetting JL, Guay JA, Karolak MJ, Iozzo RV, Adams
Nephrol. 2008;19(11):2159–69. DC, Maridas DE, et al. FOXD1 promotes nephron
160. Kitamoto Y, Tokunaga H, Tomita K. Vascular endo- progenitor differentiation by repressing decorin in
thelial growth factor is an essential molecule for the embryonic kidney. Development. 2014;141(1):
mouse kidney development: glomerulogenesis and 17–27.
nephrogenesis. J Clin Invest. 1997;99(10):2351–7. 175. Hum S, Rymer C, Schaefer C, Bushnell D, Sims-
161. Tufro A, Norwood VF, Carey RM, Gomez Lucas S. Ablation of the renal stroma defines its
RA. Vascular endothelial growth factor induces critical role in nephron progenitor and vasculature
nephrogenesis and vasculogenesis. J Am Soc patterning. PLoS One. 2014;9(2):e88400.
Nephrol. 1999;10(10):2125–34. 176. Stolz DB, Sims-Lucas S. Unwrapping the origins and
162. Woolf AS, Yuan HT. Angiopoietin growth factors and roles of the renal endothelium. Pediatr Nephrol. 2014.
Tie receptor tyrosine kinases in renal vascular devel- 177. Yu J, Valerius MT, Duah M, Staser K, Hansard JK,
opment. Pediatr Nephrol. 2001;16(2):177–84. Guo JJ. Identification of molecular compartments and
163. Lindahl P, Hellström M, Kalén M, Karlsson L, genetic circuitry in the developing mammalian kid-
Pekny M, Pekna M, et al. Paracrine PDGF-B/PDGF- ney. Development. 2012;139(10):1863–73.
Rß signaling controls mesangial cell development in 178. Brunskill EW, Potter SS. Gene expression programs
kidney glomeruli. Development. 1998;125:3313–22. of mouse endothelial cells in kidney development and
164. Leveen P, Pekny M, Gebre-Medhin S, Swolin B, disease. PLoS One. 2010;5(8):e12034.
Larsson E, Betsholtz C. Mice deficient for PDGF B 179. Abrahamson DR, Robert B, Hyink DP, St John PL,
show renal, cardiovascular, and hematological abnor- Daniel TO. Origins and formation of microvascula-
malities. Genes Dev. 1994;8:1875–87. ture in the developing kidney. Kidney Int Suppl
165. Soriano P. Abnormal kidney development and hema- [Research Support, Non-U.S. Gov’t Research Sup-
tological disorders in PDGF ß-receptor mutant mice. port, U.S. Gov’t, P.H.S. Review]. 1998;67:S7–11.
Genes Dev. 1994;8:1888–96. 180. Robert B, St John PL, Abrahamson DR. Direct visu-
166. McCright B, Gao X, Shen L, Lozier J, Lan Y, alization of renal vascular morphogenesis in Flk1
Maguire M, et al. Defects in development of the heterozygous mutant mice. Am J Physiol [Research
kidney, heart and eye vasculature in mice homozy- Support, U.S. Gov’t, P.H.S.]. 1998;275(1 Pt 2):
gous for a hypomorphic Notch2 mutation. Develop- F164–72.
ment. 2001;128:491–502. 181. Sequeira Lopez ML, Gomez RA. Development of
167. Li W, Hartwig S, Rosenblum ND. Developmental the renal arterioles. J Am Soc Nephrol. 2011;22(12):
origins and functions of stromal cells in the normal 2156–65.
and diseased mammalian kidney. Dev Dyn. 2014;243 182. Lancrin C, Sroczynska P, Serrano AG, Gandillet A,
(7):853–863. Ferreras C, Kouskoff V, et al. Blood cell generation
168. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, from the hemangioblast. J Mol Med [Research Sup-
Nowlin BT, Bonventre JV, et al. Fate tracing reveals port, Non-U.S. Gov’t Review]. 2010;88(2):167–72.
the pericyte and not epithelial origin of 183. Kume T. Specification of arterial, venous, and lym-
myofibroblasts in kidney fibrosis. Am J Pathol phatic endothelial cells during embryonic develop-
[Research Support, N.I.H., Extramural Research ment. Histol Histopathol. 2010;25(5):637–46.
Support, U.S. Gov’t, Non-P.H.S.]. 2010;176(1): 184. Sims-Lucas S, Schaefer C, Bushnell D, Ho J,
85–97. Logar A, Prochownik E, et al. Endothelial progenitors
169. Miyazaki Y, Oshima Y, Fogo A, Hogan BLM, Ichi- exist within the kidney and lung mesenchyme. PLoS
kawa I. Bone morphogenetic protein 4 regulates the One. 2013;8(6):e65993.
budding site and elongation of the mouse ureter. 185. Schmidt-Ott KM, Chen X, Paragas N, Levinson RS,
J Clin Invest. 2000;105:863–73. Mendelsohn CL, Barasch J. c-kit delineates a distinct
170. Cullen-McEwen LA, Caruana G, Bertram JF. The domain of progenitors in the developing kidney. Dev
where, what and why of the developing renal stroma. Biol [Research Support, N.I.H., Extramural Research
Nephron Exp Nephrol [Review]. 2005;99(1):e1–8. Support, Non-U.S. Gov’t]. 2006;299(1):238–49.
171. Lemley KV, Kriz W. Anatomy of the renal 186. Marlier A, Schmidt-Ott KM, Gallagher AR,
interstitium. Kidney Int. 1991;39(3):370–81. Barasch J, Karihaloo A. Vegf as an epithelial cell
172. Rosselot C, Spraggon L, Chia I, Batourina E, morphogen modulates branching morphogenesis of
Riccio P, Lu B, et al. Non-cell-autonomous retinoid embryonic kidney by directly acting on the ureteric
signaling is crucial for renal development. Develop- bud. Mech Dev [Research Support, Non-U.S. Gov’t].
ment [Research Support, N.I.H., Extramural Research 2009;126(3–4):91–8.
Support, Non-U.S. Gov’t]. 2010;137(2):283–92. 187. Freeburg PB, Robert B, St John PL, Abrahamson
173. Quaggin SE, Vanden Heuvel GB, Igarashi P. Pod-1, a DR. Podocyte expression of hypoxia-inducible factor
mesoderm-specific basic-helix-loop-helix protein (HIF)-1 and HIF-2 during glomerular development.
expressed in mesenchymal and glomerular epithelial J Am Soc Nephrol. 2003;14(4):927–38.
cells in the developing kidney. Mech Dev. 188. Steenhard BM, Freeburg PB, Isom K, Stroganova L,
1998;71:37–48. Borza DB, Hudson BG, et al. Kidney development
1 Embryonic Development of the Kidney 33

and gene expression in the HIF2alpha knockout 202. Schuchardt A, D'Agati V, Larsson-Blomberg L,
mouse. Dev Dyn. 2007;236(4):1115–25. Costantini F, Pachnis V. Defects in the kidney and
189. Kappel A, Ronicke V, Damert A, Flamme I, Risau W, enteric nervous system of mice lacking the tyrosine
Breier G. Identification of vascular endothelial kinase receptor Ret. Nature. 1994;367:380–3.
growth factor (VEGF) receptor-2 (Flk-1) promoter/ 203. Enomoto H, Araki T, Jackman A, Heuckeroth RO,
enhancer sequences sufficient for angioblast and Snider WD, Johnson EMJ, et al. GFRα 1-deficient
endothelial cell-specific transcription in transgenic mice have deficits in the enteric nervous system and
mice. Blood. 1999;93(12):4284–92. kidneys. Neuron. 1998;21:317–24.
190. Gerber HP, Condorelli F, Park J, Ferrara 204. Pichel JG, Shen L, Sheng HZ, Granholm A-C,
N. Differential transcriptional regulation of the two Drago J, Grinberg A, et al. Defects in enteric inner-
vascular endothelial growth factor receptor genes. vation and kidney development in mice lacking
Flt-1, but not Flk-1/KDR, is up-regulated by hypoxia. GDNF. Nature. 1996;382:73–6.
J Biol Chem. 1997;272(38):23659–67. 205. Sanchez MP, Silos-Santiago I, Frisen J, He B, Lira
191. Levy AP, Levy NS, Wegner S, Goldberg SA, Barbacid M. Renal agenesis and the absence of
MA. Transcriptional regulation of the rat vascular enteric neurons in mice lacking GDNF. Nature.
endothelial growth factor gene by hypoxia. J Biol 1996;382:70–3.
Chem. 1995;270(22):13333–40. 206. Schuchardt A, D'Agati V, Pachnis V, Costantini
192. Liu Y, Cox SR, Morita T, Kourembanas S. Hypoxia F. Renal agenesis and hypodysplasia in ret-k mutant
regulates vascular endothelial growth factor gene mice result from defects in ureteric bud development.
expression in endothelial cells. Identification of a 5’ Development. 1996;122:1919–29.
enhancer. Circ Res. 1995;77(3):638–43. 207. Cacalano G, Farinas I, Wang LC, Hagler K, Forgie A,
193. Woolf AS, Gnudi L, Long DA. Roles of angiopoietins Moore M, et al. GFRalpha1 is an essential receptor
in kidney development and disease. J Am Soc component for GDNF in the developing nervous sys-
Nephrol. 2009;20(2):239–44. tem and kidney. Neuron. 1998;21:53–62.
194. Simon MP, Tournaire R, Pouyssegur J. The 208. Jain S, Encinas M, Johnson EM, Jr., Milbrandt
angiopoietin-2 gene of endothelial cells is J. Critical and distinct roles for key RET tyrosine
up-regulated in hypoxia by a HIF binding site docking sites in renal development. Genes Dev
located in its first intron and by the central factors [Research Support, N.I.H., Extramural Research Sup-
GATA-2 and Ets-1. J Cell Physiol. 2008;217(3): port, Non-U.S. Gov’t]. 2006;20(3):321–33.
809–18. 209. Jain S, Knoten A, Hoshi M, Wang H, Vohra B,
195. Kolatsi-Joannou M, Li XZ, Suda T, Yuan HT, Woolf Heuckeroth RO, et al. Organotypic specificity of key
AS. Expression and potential role of angiopoietins RET adaptor-docking sites in the pathogenesis of
and Tie-2 in early development of the mouse meta- neurocristopathies and renal malformations in mice.
nephros. Dev Dyn. 2001;222(1):120–6. J Clin Invest [Research Support, N.I.H., Extramural
196. Jeansson M, Gawlik A, Anderson G, Li C, Research Support, Non-U.S. Gov’t]. 2010;120(3):
Kerjaschki D, Henkelman M, et al. Angiopoietin-1 778–90.
is essential in mouse vasculature during development 210. Skinner MA, Safford SD, Reeves JG, Jackson ME,
and in response to injury. J Clin Invest. Freemerman AJ. Renal aplasia in humans is
2011;121(6):2278–89. associated with RET mutations. Am J Hum Genet.
197. Tachibana K, Jones N, Dumont DJ, Puri MC, Bern- 2008;82(2):344–51.
stein A. Selective role of a distinct tyrosine residue on 211. Yang Y, Houle AM, Letendre J, Richter A. RET
Tie2 in heart development and early hematopoiesis. Gly691Ser mutation is associated with primary
Mol Cell Biol. 2005;25(11):4693–702. vesicoureteral reflux in the French-Canadian popula-
198. Pitera JE, Woolf AS, Gale NW, Yancopoulos GD, tion from Quebec. Hum Mutat [Research Support,
Yuan HT. Dysmorphogenesis of kidney cortical Non-U.S. Gov’t]. 2008;29(5):695–702.
peritubular capillaries in angiopoietin-2-deficient 212. Bullock SL, Fletcher JM, Beddington RSP, Wilson
mice. Am J Pathol. 2004;165(6):1895–906. VA. Renal agenesis in mice homozygous for a
199. Tung JJ, Tattersall IW, Kitajewski J. Tips, stalks, gene trap mutation in the gene encoding heparan
tubes: notch-mediated cell fate determination and sulfate 2-sulfotransferase. Genes Dev.
mechanisms of tubulogenesis during angiogenesis. 1998;12:1894–906.
Cold Spring Harb Perspect Med. 2012;2(2):a006601. 213. Pepicelli CV, Kispert A, Rowitch D, McMahon
200. Piscione TD, Rosenblum ND. The malformed kid- AP. GDNF induces branching and increased cell pro-
ney: disruption of glomerular and tubular develop- liferation in the ureter of the mouse. Dev Biol.
ment. Clin Genet. 1999;56(5):343–58. 1997;192:193–8.
201. Sainio K, Suvanto P, Davies J, Wartiovaara J, 214. Srinivas S, Wu Z, Chen C-M, D’Agati V, Costantini
Wartiovaara K, Saarma M, et al. Glial-cell-line- F. Dominant effects of RET receptor misexpression
derived neurotrophic factor is required for bud initia- and ligand-independent RET signaling on
tion from ureteric epithelium. Development. ureteric bud development. Development.
1997;124:4077–87. 1999;126:1375–86.
34 C. Bates et al.

215. Shakya R, Jho EH, Kotka P, Wu Z, Kholodilov N, Soc Nephrol [Research Support, Non-U.S. Gov’t].
Burke R, et al. The role of GDNF in patterning the 2008;19(5):891–903.
excretory system. Dev Biol. 2005;283(1):70–84. 229. Lu BC, Cebrian C, Chi X, Kuure S, Kuo R, Bates CM,
216. Kume T, Deng K, Hogan BL. Murine forkhead/ et al. Etv4 and Etv5 are required downstream of
winged helix genes Foxc1 (Mf1) and Foxc2 (Mfh1) GDNF and Ret for kidney branching morphogenesis.
are required for the early organogenesis of the kidney Nat Genet [Research Support, N.I.H., Extramural
and urinary tract. Development. 2000;127:1387–95. Research Support, Non-U.S. Gov’t]. 2009;41
217. Grieshammer U, Le M, Plump AS, Wang F, Tessier- (12):1295–302.
Lavigne M, Martin GR. SLIT2-mediated ROBO2 230. Majumdar A, Vainio S, Kispert A, McMahon J,
signaling restricts kidney induction to a single site. McMahon AP. Wnt11 and Ret/Gdnf pathways coop-
Dev Cell. 2004;6(5):709–17. erate in regulating ureteric branching during meta-
218. Tessier-Lavigne M, Goodman CS. The molecular nephric kidney development. Development.
biology of axon guidance. Science. 2003;130(14):3175–85.
1996;274:1123–33. 231. Al-Awqati Q, Goldberg MR. Architectural patterns in
219. Brose K, Bland KS, Wang KH, Arnott D, Henzel W, branching morphogenesis in the kidney. Kidney Int.
Goodman CS, et al. Slit proteins bind Robo receptors 1998;54:1832–42.
and have an evolutionarily conserved role in repulsive 232. Watanabe T, Costantini F. Real-time analysis of ure-
axon guidance. Cell. 1999;96:795–806. teric bud branching morphogenesis in vitro. Dev Biol.
220. Piper M, Georgas K, Yamada T, Little M. Expression 2004;271(1):98–108.
of the vertebrate Slit gene family and their putative 233. Lin Y, Zhang S, Tuukkanen J, Peltoketo H,
receptors, the Robo genes, in the developing murine Pihlajaniemi T, Vainio S. Patterning parameters asso-
kidney. Mech Dev. 2000;94:213–7. ciated with the branching of the ureteric bud regulated
221. Lu W, van Eerde AM, Fan X, Quintero-Rivera F, by epithelial-mesenchymal interactions. Int J Dev
Kulkarni S, Ferguson H, et al. Disruption of ROBO2 Biol. 2003;47(1):3–13.
is associated with urinary tract anomalies and confers 234. Fisher CE, Michael L, Barnett MW, Davies JA. Erk
risk of vesicoureteral reflux. Am J Hum Genet [Com- MAP kinase regulates branching morphogenesis in
parative Study]. 2007;80(4):616–32. the developing mouse kidney. Development.
222. Basson MA, Watson-Johnson J, Shakya R, 2001;128(21):4329–38.
Akbulut S, Hyink D, Costantini FD, et al. Branching 235. Michael L, Davies JA. Pattern and regulation of cell
morphogenesis of the ureteric epithelium during kid- proliferation during murine ureteric bud develop-
ney development is coordinated by the opposing ment. J Anat. 2004;204(4):241–55.
functions of GDNF and Sprouty1. Dev Biol. 236. Sorenson CM, Rogers SA, Korsmeyer SJ,
2006;299(2):466–77. Hammerman MR. Fulminant metanephric apoptosis
223. Basson MA, Akbulut S, Watson-Johnson J, Simon R, and abnormal kidney development in bcl-2-deficient
Carroll TJ, Shakya R, et al. Sprouty1 is a critical mice. Am J Physiol. 1995;268:F73–81.
regulator of GDNF/RET-mediated kidney induction. 237. Moser M, Pscherer A, Roth C, Becker J, M€ ucher G,
Dev Cell. 2005;8(2):229–39. Zerres K, et al. Enhanced apoptotic cell death of
224. Dudley AT, Robertson EJ. Overlapping expression renal epithelial cells in mice lacking
domains of bone morphogenetic protein family mem- transcription factor AP-2ß. Genes Dev.
bers potentially account for limited tissue defects in 1997;11:1938–48.
BMP7 deficient embryos. Dev Dyn. 238. Chevalier RL. Growth factors and apoptosis in neo-
1997;208:349–62. natal ureteral obstruction. J Am Soc Nephrol.
225. Pope IV JC, Brock III JW, Adams MC, Stephens FD, 1996;7:1098–105.
Ichikawa I. How they begin and how they end: classis 239. Tarantal AF, Han VK, Cochrum KC, Mok A,
and new theories for the development and deteriora- daSilva M, Matsell DG. Fetal rhesus monkey model
tion of congenital anomalies of the kidney and urinary of obstructive renal dysplasia. Kidney Int.
tract, CAKUT. J Am Soc Nephrol. 1999;10:2018–28. 2001;59:446–56.
226. Ichikawa I, Kuwayama F, Pope JCT, Stephens FD, 240. Kispert A, Vainio S, Shen L, Rowitch DH, McMahon
Miyazaki Y. Paradigm shift from classic anatomic AP. Proteoglycans are required for maintenance of
theories to contemporary cell biological views of Wnt-11 expression in the ureter tips. Development.
CAKUT. Kidney Int. 2002;61(3):889–98. 1996;122:3627–37.
227. Bush KT, Sakurai H, Steer DL, Leonard MO, 241. Bridgewater D, Cox B, Cain J, Lau A, Athaide V,
Sampogna RV, Meyer TN, et al. TGF-beta superfam- Gill PS, et al. Canonical WNT/beta-catenin signaling
ily members modulate growth, branching, shaping, is required for ureteric branching. Dev Biol.
and patterning of the ureteric bud. Dev Biol. 2008;317(1):83–94.
2004;266(2):285–98. 242. Marose TD, Merkel CE, McMahon AP, Carroll
228. Weber S, Taylor JC, Winyard P, Baker KF, Sullivan- TJ. Beta-catenin is necessary to keep cells of ureteric
Brown J, Schild R, et al. SIX2 and BMP4 mutations bud/Wolffian duct epithelium in a precursor state. Dev
associate with anomalous kidney development. J Am Biol. 2008;314(1):112–26.
1 Embryonic Development of the Kidney 35

243. Qiao J, Bush KT, Steer DL, Stuart RO, Sakurai H, 257. Zhang P, Liégeois NJ, Wong C, Finegold M, Thomp-
Wachsman W, et al. Multiple fibroblast growth factors son JC, Silverman A, et al. Altered cell differentiation
support growth of the ureteric bud but have different and proliferation in mice lacking p57KIP2 indicates a
effects on branching morphogenesis. Mech Dev. role in Beckwith-Wiedemann syndrome. Nature.
2001;109(2):123–35. 1997;387:151–8.
244. Ohuchi H, Hori Y, Yamasaki M, Harada H, Sekine K, 258. Yu J, Carroll TJ, Rajagopal J, Kobayashi A, Ren Q,
Kato S, et al. FGF10 acts as a major ligand for FGF McMahon AP. A Wnt7b-dependent pathway regu-
receptor 2 IIIb in mouse multi-organ development. lates the orientation of epithelial cell division and
Biochem Biophys Res Commun. 2000;277:643–9. establishes the cortico-medullary axis of the mamma-
245. Poladia DP, Kish K, Kutay B, Hains D, Kegg H, lian kidney. Development [Research Support, N.I.H.,
Zhao H, et al. Role of fibroblast growth factor recep- Extramural Research Support, Non-U.S. Gov’t].
tors 1 and 2 in the metanephric mesenchyme. Dev 2009;136(1):161–71.
Biol. 2006;291(2):325–39. 259. Liu Y, Chattopadhyay N, Qin S, Szekeres C,
246. Sims-Lucas S, Cusack B, Eswarakumar VP, Zhang J, Vasylyeva T, Mahoney ZX, et al. Coordinate integrin
Wang F, Bates CM. Independent roles of Fgfr2 and and c-Met signaling regulate Wnt gene expression
Frs2alpha in ureteric epithelium. Development. during epithelial morphogenesis. Development
2011;138(7):1275–80. [Research Support, N.I.H., Extramural Research Sup-
247. Zhao H, Kegg H, Grady S, Truong HT, Robinson ML, port, Non-U.S. Gov’t]. 2009;136(5):843–53.
Baum M, et al. Role of fibroblast growth factor 260. Niimura F, Labostky PA, Kakuchi J, Okubo S,
receptors 1 and 2 in the ureteric bud. Dev Biol. Yoshida H, Oikawa T, et al. Gene targeting in mice
2004;276(2):403–15. reveals a requirement for angiotensin in the develop-
248. Michos O, Cebrian C, Hyink D, Grieshammer U, ment and maintenance of kidney morphology and
Williams L, D'Agati V, et al. Kidney development in growth factor regulation. J Clin Invest.
the absence of Gdnf and Spry1 requires Fgf10. PLoS 1995;96:2947–54.
Genet. 2010;6(1):e1000809. 261. Miyazaki Y, Tsuchida S, Nishimura H, Pope IV JC,
249. Chi L, Zhang S, Lin Y, Prunskaite-Hyyrylainen R, Harris RC, McKanna JM, et al. Angiotensin induces
Vuolteenaho R, Itaranta P, et al. Sprouty proteins the urinary peristaltic machinery during the perinatal
regulate ureteric branching by coordinating reciprocal period. J Clin Invest. 1998;102:1489–97.
epithelial Wnt11, mesenchymal Gdnf and stromal 262. Rasouly HM, Lu W. Lower urinary tract development
Fgf7 signalling during kidney development. Devel- and disease. Wiley Interdiscip Rev Syst Biol Med.
opment. 2004;131(14):3345–56. 2013;5(3):307–42.
250. Cebrian C, Borodo K, Charles N, Herzlinger 263. Brenner-Anantharam A, Cebrian C, Guillaume R,
DA. Morphometric index of the developing murine Hurtado R, Sun TT, Herzlinger D. Tailbud-derived
kidney. Dev Dyn. 2004;231(3):601–8. mesenchyme promotes urinary tract segmentation
251. Bard J. A new role for the stromal cells in kidney via BMP4 signaling. Development. 2007;134(10):
development. Bioessays. 1996;18(9):705–7. 1967–75.
252. Bonneh-Barkay D, Shlissel M, Berman B, Shaoul E, 264. McHugh KM. Molecular analysis of smooth muscle
Admon A, Vlodavsky I, et al. Identification of development in the mouse. Dev Dyn. 1995;204(3):
glypican as a dual modulator of the biological activity 278–90.
of fibroblast growth factors. J Biol Chem. 265. Yu J, Carroll TJ, McMahon AP. Sonic hedgehog
1997;272:12415–21. regulates proliferation and differentiation of mesen-
253. Cano-Gauci DF, Song H, Yang H, McKerlie C, chymal cells in the mouse metanephric kidney. Devel-
Choo B, Shi W, et al. Glypican-3-deficient opment [Research Support, Non-U.S. Gov’t Research
mice exhibit developmental overgrowth and Support, U.S. Gov’t, P.H.S.]. 2002;129(22):5301–12.
some of the renal abnormalities typical of Simpson- 266. Wu XR, Kong XP, Pellicer A, Kreibich G, Sun
Golabi-Behmel syndrome. J Cell Biol. TT. Uroplakins in urothelial biology, function, and
1999;146:255–64. disease. Kidney Int. 2009;75(11):1153–65.
254. Grisaru S, Cano-Gauci D, Tee J, Filmus J, Rosenblum 267. Weiss RM, Guo S, Shan A, Shi H, Romano RA,
ND. Glypican-3 modulates BMP- and FGF-mediated Sinha S, et al. Brg1 determines urothelial cell fate
effects during renal branching morphogenesis. Dev during ureter development. J Am Soc Nephrol.
Biol. 2001;231:31–46. 2013;24(4):618–26.
255. Jackson SM, Nakato H, Sugiura M, Jannuzi A, 268. Cain JE, Islam E, Haxho F, Blake J, Rosenblum
Oakes R, Kaluza V, et al. Dally, a drosophila glypican, ND. GLI3 repressor controls functional development
controls cellular responses to the TGF-ß-related mor- of the mouse ureter. J Clin Invest [Research Support,
phogen Dpp. Development. 1997;124:4113–20. Non-U.S. Gov’t]. 2011;121(3):1199–206.
256. Tsuda M, Kamimura K, Nakato H, Archer M, 269. Hurtado R, Bub G, Herzlinger D. The pelvis-kidney
Staatz W, Fox B, et al. The cell-surface proteoglycan junction contains HCN3, a hyperpolarization-
dally regulates wingless signalling in Drosophila. activated cation channel that triggers ureter peristal-
Nature. 1999;400:276–80. sis. Kidney Int. 2010;77(6):500–8.
36 C. Bates et al.

270. Price KL, Woolf AS, Long DA. Unraveling the dominant Pallister-Hall syndrome. Nat Genet.
genetic landscape of bladder development in mice. 1997;15(3):266–8.
J Urol [Research Support, Non-U.S. Gov’t]. 283. Trowe MO, Airik R, Weiss AC, Farin HF, Foik AB,
2009;181(5):2366–74. Bettenhausen E, et al. Canonical Wnt signaling regu-
271. Singh S, Robinson M, Nahi F, Coley B, Robinson lates smooth muscle precursor development in the
ML, Bates CM, et al. Identification of a unique trans- mouse ureter. Development. 2012;139:2009–3108.
genic mouse line that develops megabladder, obstruc- 284. Chang CP, McDill BW, Neilson JR, Joist HE, Epstein
tive uropathy, and renal dysfunction. J Am Soc JA, Crabtree GR, et al. Calcineurin is required in
Nephrol. 2007;18(2):461–71. urinary tract mesenchyme for the development of
272. Gandhi D, Molotkov A, Batourina E, Schneider K, the pyeloureteral peristaltic machinery. J Clin Invest.
Dan H, Reiley M, et al. Retinoid signaling in pro- 2004;113(7):1051–8.
genitors controls specification and regeneration of the 285. Miyazaki Y, Tsuchida S, Nishimura H, Pope JCt,
urothelium. Dev Cell [Research Support, N.I.H., Harris RC, McKanna JM, et al. Angiotensin induces
Extramural Research Support, Non-U.S. Gov’t]. the urinary peristaltic machinery during the perinatal
2013;26(5):469–82. period. J Clin Invest [Research Support, Non-U.S.
273. Batourina E, Tsai S, Lambert S, Sprenkle P, Viana R, Gov’t Research Support, U.S. Gov’t,
Dutta S, et al. Apoptosis induced by vitamin A sig- P.H.S.]. 1998;102(8):1489–97.
naling is crucial for connecting the ureters to the 286. Mahoney ZX, Sammut B, Xavier RJ, Cunningham J,
bladder. Nat Genet. 2005;37(10):1082–9. Go G, Brim KL, et al. Discs-large homolog 1 regulates
274. Viana R, Batourina E, Huang H, Dressler GR, smooth muscle orientation in the mouse ureter. Proc
Kobayashi A, Behringer RR, et al. The development Natl Acad Sci U S A. 2006;103(52):19872–7.
of the bladder trigone, the center of the anti-reflux 287. Dravis C, Yokoyama N, Chumley MJ, Cowan CA,
mechanism. Development. 2007;134(20):3763–9. Silvany RE, Shay J, et al. Bidirectional signaling
275. Airik R, Bussen M, Singh MK, Petry M, Kispert mediated by ephrin-B2 and EphB2 controls urorectal
A. Tbx18 regulates the development of the ureteral development. Dev Biol. 2004;271(2):272–90.
mesenchyme. J Clin Invest. 2006;116(3):663–74. 288. Mo R, Kim JH, Zhang J, Chiang C, Hui CC, Kim
276. Kong XT, Deng FM, Hu P, Liang FX, Zhou G, Auer- PC. Anorectal malformations caused by defects in
bach AB, et al. Roles of uroplakins in plaque forma- sonic hedgehog signaling. Am J Pathol. 2001;
tion, umbrella cell enlargement, and urinary tract 159(2):765–74.
diseases. J Cell Biol. 2004;167(6):1195–204. 289. Baskin L, DiSandro M, Li Y, Li W, Hayward S,
277. Hu P, Deng FM, Liang FX, Hu CM, Auerbach AB, Cunha G. Mesenchymal-epithelial interactions in
Shapiro E, et al. Ablation of uroplakin III gene results bladder smooth muscle development: effects of the
in small urothelial plaques, urothelial leakage, local tissue environment. J Urol. 2001;165(4):
and vesicoureteral reflux. J Cell Biol. 2000;151(5): 1283–8.
961–72. 290. DiSandro MJ, Li Y, Baskin LS, Hayward S, Cunha
278. Jenkins D, Bitner-Glindzicz M, Malcolm S, Hu CC, G. Mesenchymal-epithelial interactions in bladder
Allison J, Winyard PJ, et al. De novo Uroplakin IIIa smooth muscle development: epithelial specificity.
heterozygous mutations cause human renal J Urol. 1998;160(3 Pt 2):1040–6; discussion 79.
adysplasia leading to severe kidney failure. J Am 291. Cao M, Tasian G, Wang MH, Liu B, Cunha G, Baskin
Soc Nephrol. 2005;16(7):2141–9. L. Urothelium-derived Sonic hedgehog promotes
279. Jenkins D, Bitner-Glindzicz M, Malcolm S, Allison J, mesenchymal proliferation and induces bladder
de Bruyn R, Flanagan S, et al. Mutation analyses smooth muscle differentiation. Differentiation
of Uroplakin II in children with renal tract [Research Support, N.I.H., Extramural]. 2010;
malformations. Nephrol Dial Transplant. 2006; 79(4–5):244–50.
21(12):3415–21. 292. Cheng W, Yeung CK, Ng YK, Zhang JR, Hui CC,
280. Wang GJ, Brenner-Anantharam A, Vaughan ED, Kim PC. Sonic Hedgehog mediator Gli2 regulates
Herzlinger D. Antagonism of BMP4 signaling bladder mesenchymal patterning. J Urol [Research
disrupts smooth muscle investment of the ureter Support, Non-U.S. Gov’t]. 2008;180(4):1543–50.
and ureteropelvic junction. J Urol. 2009;181(1): 293. DeSouza KR, Saha M, Carpenter AR, Scott M,
401–7. McHugh KM. Analysis of the Sonic Hedgehog sig-
281. Caubit X, Lye CM, Martin E, Core N, Long DA, naling pathway in normal and abnormal bladder
Vola C, et al. Teashirt 3 is necessary for ureteral development. PLoS One [Research Support, N.I.H.,
smooth muscle differentiation downstream of SHH Extramural]. 2013;8(1):e53675.
and BMP4. Development [Research Support, 294. Hoshi M, Batourina E, Mendelsohn C, Jain S. Novel
Non-U.S. Gov’t]. 2008;135(19):3301–10. mechanisms of early upper and lower urinary tract
282. Kang S, Graham Jr JM, Olney AH, Biesecker patterning regulated by RetY1015 docking tyrosine in
LG. GLI3 frameshift mutations cause autosomal mice. Development. 2012;139(13):2405–15.
Development of Glomerular Circulation
and Function 2
Alda Tufro and Ashima Gulati

Contents Introduction
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
From the Malpighian corpuscle description and
Development of the Kidney Vasculature . . . . . . . . . . . 38
Vasculogenesis and Angiogenesis . . . . . . . . . . . . . . . . . . . . 38
Bowman’s sketch to defining its ultrastructure and
molecular function, the ways we look at the kid-
The Glomerular Filtration Barrier . . . . . . . . . . . . . . . . . 41
ney glomerulus have evolved tremendously. The
Components of the GFB . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
Glomerular Filtration Barrier (GFB) Selective first systematic exploration of the body with a
Permeability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46 microscope led to the identification of “Malpi-
Glomerular Hemodynamics and Assessment of ghian corpuscles” as “glands” within the kidney
Renal Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46 [1]. Two centuries later, a more sophisticated
Renal Blood Flow: Basic Concepts . . . . . . . . . . . . . . . . . . . 47 microscope enabled Sir William Bowman to iden-
Perinatal Considerations for Renal Blood Flow tify glomerular capillary tufts in animal and
and GFR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
The Concept of Clearance . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50 human kidneys and demonstrate a relationship
between the capillary tuft and the renal tubule
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
[2]. Since then, the understanding of the human
glomerulus as a specialized structure uniquely
adapted for renal filtration at the proximal part of
the nephron has considerably advanced.
The human definitive kidney is a highly
complex organ system with an average number
of ~1 million functional units called nephrons.
Nephrogenesis involves the development of the
glomerulus (glomerulogenesis) and the renal
tubule (tubulogenesis) from mesenchymal pro-
genitors residing in the metanephric mesenchyme
and ureteric bud. The specification, maintenance,
and commitment of nephron progenitors and the
regulatory processes that transform nephron pro-
genitors into a functional nephron are
detailed elsewhere ([3]; see ▶ Chaps. 1, “Embry-
A. Tufro (*) • A. Gulati
onic Development of the Kidney,” and ▶ 18,
Department of Pediatrics, Nephrology Section, Yale
School of Medicine, New Haven, CT, USA “Translational Research Methods: Renal Stem
e-mail: alda.tufro@yale.edu Cells” in this text). The current chapter aims to
# Springer-Verlag Berlin Heidelberg 2016 37
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_2
38 A. Tufro and A. Gulati

combine established knowledge and new findings delivery to all nephron segments and for fluid and
pertaining to the structural and functional aspects solute reabsorption from them. As a result of fluid
of glomerular development, describe intricacies of removal by glomerular ultrafiltration, oncotic
the glomerular filtration barrier at the molecular pressure in postglomerular capillaries increases
level, and provide insight into future research above hydrostatic pressure. Thus, peritubular cap-
areas. illaries are poised for fluid and solute
Three attributes make the human glomerulus a reabsorption.
fascinating focus of research and clinical signifi-
cance: first is the synchrony among
vasculogenesis, angiogenesis, and epithelial and Vasculogenesis and Angiogenesis
stromal differentiation; second, the glomerular
unique ultrastructure provides controlled regula- The formation of the kidney vasculature com-
tion of filtration while acting as a barrier; and prises in situ differentiation of endothelial cells
third, the glomerulus is the major controller of from hemangioblasts present in the metanephric
renal hemodynamics and provides functional mesenchyme and capillary assembly, a process
advantages for effective filtration. Each of these called vasculogenesis, and angiogenesis wherein
attributes and their relevance for clinical use will capillary growth occurs by the sprouting and/or
be discussed in the following sections. splitting of existing capillaries within or surround-
ing the developing kidney [5–9]. In vivo,
vasculogenesis and angiogenesis might occur
Development of the Kidney sequentially or simultaneously [10].
Vasculature
Vasculogenesis and Angiogenesis
The kidney vasculature develops in a patterned Contribute to the Kidney Vasculature
fashion, allowing structural and functional devel- The origin of kidney endothelial cells and the
opment of the glomerular circulation that eventu- mechanisms involved in kidney vascularization
ally handles 20 % of the cardiac output for the have been extensively examined in experimental
purpose of clearing metabolic waste products and models and debated for quite some time. Genetic
toxins and maintaining fluid and electrolyte bal- fate mapping and the identification of molecular
ance [4]. The complex architecture of the renal guidance cues from angiogenic factors have
vasculature is essential for the organ function and largely resolved these controversies. The hypoth-
comprises an arterial tree and three capillary beds. esis of extrarenal angiogenic origin of kidney
Single renal arteries branch and direct over 90 % endothelial cells is supported by elegant interspe-
of the renal blood flow to glomeruli in the renal cies transplantation experiments and by the
cortex. The glomerular capillary bed length absence of vascular development in embryonic
amplifies the area for filtration, allowing the kidneys cultured ex vivo [11–13]. As shown in
highest fluid outflow rate in the body. Glomerular other embryonic vascular beds [14], Sariola and
capillaries are flanked by high-resistance afferent colleagues showed evidence of angiogenesis in
and efferent arterioles, which regulate the rate of undifferentiated embryonic mouse kidney rudi-
blood flow through the capillary bed and thereby ments after the transplantation onto avian chorio-
glomerular filtration. Two postglomerular capil- allantoic membrane [15, 16]. Indeed, grafts were
lary networks emerge in series from efferent arte- well vascularized, and both the glomerular and the
rioles. Efferent arterioles from superficial vessel endothelium expressed an avian nuclear
glomeruli give rise to cortical peritubular capil- marker, suggesting that the glomerular endothe-
laries, while those from the juxtamedullary glo- lium is derived from extrinsic (avian) vasculature
meruli give rise to vasa recta. Close alignment of rather than by the differentiation of endothelial
the postglomerular microvasculature with cortical cells native to the metanephric mesenchyme
and medullary renal tubules is critical for oxygen [15–17]. Abrahamson et al. demonstrated that
2 Development of Glomerular Circulation and Function 39

embryonic kidneys grafted under the renal cap- nephrogenic cortex of wild-type mice develop
sule of newborn mice were vascularized by host transgene-expressing glomerular capillary loops
endothelium, whereas adult hosts failed to [31]. In contrast, glomerular Tie-1/LacZ + vessels
vascularize the grafts [18], suggesting that the do not develop in rudiments placed in organ cul-
angiogenic activity of the host is crucial to this ture on 20 % O2 [31]. This capability of Flk1+
process. angioblasts to give rise to a primitive vasculature
The hypothesis of the endogenous, has subsequently been confirmed in vitro and
vasculogenic origin of glomerular endothelial in vivo explant assays [18, 19, 32, 33]. These
cells is supported by the identification of native observations imply that endothelial progenitors
endothelial precursors in the metanephric mesen- present at the onset of mouse nephrogenesis dif-
chyme, by the exposure of avascular metanephric ferentiate and undergo morphogenesis to become
kidneys to hypoxia or angiogenic factors, and by glomerular capillaries when experimental condi-
grafting experiments [18–21]. These findings tions resemble those found in the metanephros
were facilitated by the landmark discovery of in vivo, such as moderate hypoxia. Together, a
vascular endothelial growth factor (VEGF) recep- large body of experimental data demonstrates that
tors Flk1 and Flt1, as indispensable for endothe- both vasculogenesis and angiogenesis contribute
lial differentiation and vascular assembly and thus to the formation of kidney vasculature under the
genetic markers of endothelial precursors influence of a variety of growth factors and guid-
[22–24]. Flk1+ and Flt1+ angioblasts were ance proteins.
detected within the avascular metanephric mesen- Blood vessels are comprised of endothelial
chyme [19–21], demonstrating that endothelial cells and associated vascular mural cells including
progenitors in situ enable vasculogenesis in the pericytes and vascular smooth muscle cells. While
developing kidney [25–28]. Flt-1 and Flk-1 are the data outlined above provide evidence for the
expressed in isolated cells before any morpho- presence of endothelial progenitors in the meta-
logic evidence of vascular development in the nephric blastema as well as ingrowing endothelial
metanephric blastema [25]. Within 24 h in culture, cells, genetic fate mapping experiments demon-
Flk-1-expressing cells align to form cord-like strate that Foxd1+ cells present at the periphery of
structures, followed by the acquisition of lumen the metanephric blastema give rise to most vascu-
and typical endothelial cell phenotype in the fol- lar mural cells, thus contributing to the overall
lowing 2 days. As renal vascularization proceeds, vascular structural development [34].
Flt-1 and Flk-1 are expressed in contiguous endo-
thelial cells [25]. Exposure of avascular kidney Angiogenic Factor Signaling Is Critical
rudiments to hypoxia similar to that occurring in for Patterning the Kidney Vasculature
embryonic tissues leads Flk1+ angioblasts present The kidney endothelium undergoes specification
within the metanephric blastema to form primitive into distinct histological types in arteries, veins,
vascular networks via the upregulation of VEGF capillaries, and lymphatics, including continuous
[29]. Similarly, exposure to exogenous VEGF endothelium or endothelium fenestrated with dia-
enables avascular embryonic kidneys to develop phragms. This specification is adapted to the tis-
capillaries [25, 30]. Genetically tagged LacZ- sue permeability characteristics and is determined
Flk1+ embryonic mouse kidneys grafted into the by local microenvironmental cues, irrespectively
anterior chamber of the rat eye develop glomeruli of the endothelial cell lineage [35]. For example,
vascularized by graft rather than host endothelial the grafting of embryonic vessels with fenestrated
cells [31]. Remarkably, the glomerular basement endothelium into the brain resulted in tight con-
membrane and mesangial matrix were noted to be tinuous endothelium typical of the blood-brain
exclusively of graft origin, implying the self- barrier [36]. Though the progenitor lineage
sufficient capability of the metanephric mesen- might contribute to endothelial cell heterogeneity
chyme to form the glomerulus [19]. Furthermore, within the kidney, endothelium specification by
Tie-1/LacZ metanephros transplanted into the local environmental cues involves dynamic cross
40 A. Tufro and A. Gulati

talk between various signaling pathways that development [25, 26]. Moreover, tightly regulated
communicate by way of growth factors [37]. Sol- VEGF-A is required to establish and maintain a
uble angiogenic factors and guidance proteins normal podocyte phenotype, i.e., foot processes
including VEGF-A, basic fibroblast growth factor linked by slit diaphragms, as revealed by
(bFGF), platelet-derived growth factor (PDGF), both loss- and gain-of-function mouse models
and semaphorin 3A play a key role in kidney [45, 52–54]. VEGF-A signals in autocrine and
vascularization. paracrine fashion in podocytes, renal tubules,
VEGF-A is a pleiotropic glycoprotein origi- and endothelial cells promoting the survival,
nally described as a vascular permeability and proliferation, and migration in vitro and in vivo
endothelial growth factor [38–40]. VEGF is a [44, 45, 55–62]. VEGF-A receptors are most
direct-acting specific endothelial cell mitogen abundant in endothelial cells, but they are
that stimulates angiogenesis and regulates embry- expressed by multiple cells, including podocytes
onic vessel development in a gene dosage- and tubular cells [44, 57, 59, 60, 62–64].
dependent manner [41]. VEGF-A is required for Two VEGF-A tyrosine kinase receptors,
endothelial cell differentiation, survival, prolifer- VEGFR-1 [previously known as fms-like tyrosine
ation, and migration, as well as for vascular kinase-1 (Flt-1)] and VEGFR-2 [formerly murine
assembly, maintenance, and remodeling; thus it fetal liver kinase 1 (flk-1)/human kinase insert
is critical for physiological angiogenesis [42, 43]. domain receptor (KDR)], and two co-receptors,
During kidney development, VEGF-A neuropilin-1 and neuropilin-2 (NRP1 and NRP2),
expressed in the metanephric mesenchyme acts have been described. VEGF-A signals are trans-
as a chemoattractant for endothelial cells and duced through VEGFR2 and NRP1 and NRP2
directs their migration toward developing neph- amplify VEGFR2 signals, while VEGFR1 func-
rons [30]. Glomerular development starts when tions mostly as a decoy [65]. The importance of
nephrons are at the S-shaped body stage, at VEGF-A signaling for embryonic vascular devel-
which time podocytes differentiate and express opment is illustrated by gene deletion experi-
VEGF-A, leading endothelial progenitors to ments. Both heterozygous and homozygous
migrate into and differentiate in the adjacent “vas- VEGF-A knockout mice die during embryogene-
cular cleft.” Podocytes synthesize three VEGF-A sis due to major vascular defects [66, 67]. Flk-1-
isoforms (VEGF121-165-189) by alternative splicing deficient mice die in utero because of an early
[44]. VEGF-A isoforms with differences in size, defect in the development of hematopoietic cells
membrane, and extracellular matrix-binding prop- and endothelial cells [22]. Flt-1-deficient mice die
erties (VEGF121 and the most abundant VEGF165 later in utero, due to the disorganization of the
are secreted) enable gradient formation and endo- early embryonic vasculature [23, 68].
thelial cell chemoattraction [30]. The glomerular Hypoxia leads to the stabilization of HIF-1α, a
endothelium forms a single capillary loop transcriptional activator of hypoxia-inducible
initially that later forms the mature glomerular genes, including VEGF-A, erythropoietin, and
tuft. VEGF-A is indispensable for normal devel- other angiogenic factors expressed in the kidney,
opment of glomerular capillaries [45] and to such as angiopoietin-1, angiopoietin-2, PDGFBB,
acquire their fenestrated phenotype [46–48]. The and FGFβ [10, 69]. Although it is likely that
principal source of VEGF-A in the renal glomer- several signaling pathways contribute to the
ulus is the podocyte [49]. Continuous expression angiogenic response to hypoxia in kidney devel-
of VEGF-A in podocytes and tubular cells and opment resulting in glomerular vascularization,
of Flk1 on the adjacent endothelia induces and undoubtedly VEGF-A plays a critical role
maintains the fenestrated endothelia and also reg- [10]. Accordingly, the deletion of podocyte
ulates vascular permeability [26, 48, 50, 51]. VEGF-A reduces the glomerular endothelial
Podocytes and tubular cells express VEGF-A cells [45]. In contrast, the ablation of semaphorin
throughout life, unlike other tissues that cease 3A, a guidance protein that functions as a negative
expressing VEGF-A at the completion of regulator of endothelial cell survival and
2 Development of Glomerular Circulation and Function 41

migration, which is also secreted by podocytes peritubular capillaries [77]. Moreover, vasa recta
[70, 71], results in glomerular capillary hyperpla- postnatal development is controlled by angioten-
sia [72]. Conversely, excess podocyte semaphorin sin II and mediated by increased tubular VEGF
3A leads to glomerular endothelial apoptosis and secretion [82]. Notably, the deletion of Foxd1
a low number of glomerular endothelial cells progenitors, a transcription factor expressed in
[72]. Together, these studies indicate that glomer- the renal stroma, disrupts peritubular capillary
ular capillary development depends on pro- and development, recruitment of vascular mural
antiangiogenic signaling pathways shared with cells, and nephron patterning [4]. This suggests
other vascular beds, while it is clear that that regulators of the local environment, as
podocytes play a key role controlling discussed above in regard to hypoxia, directly or
glomerulogenesis. indirectly influence epithelial-endothelial-mural
Other hypoxia-regulated angiogenic factors cell cross talk and ultimately the patterning of
control the relationship between endothelial and the renal vasculature.
mesangial cells during glomerular development,
as revealed by targeted deletion experiments. The
ablation of PDGF B, or its receptor PDGFR beta, The Glomerular Filtration Barrier
expressed by endothelial and mesangial cells,
respectively, and involved in vessel maturation, The renal glomerulus controls the filtration of
results in glomeruli with aneurysm-like glomeru- water and solutes while at the same time acting
lar capillaries and lacking mesangial cells as a barrier retaining vital molecules such as
[73, 74]. Similarly, the targeted ablation of plasma proteins. The glomerular filter functions
CXCL12, its receptor CXCR4 or RBP-J, a trans- as a semipermeable, macromolecular sieve capa-
ducer of notch signaling, and angiopoietin1 ble of excluding molecules larger than serum
results in single-loop or ballooned glomerular albumin (MM 66,400) [83, 84]. The glomerular fil-
capillaries associated with mesangial cell deficit tration barrier (GFB) separating the vasculature
in some mutants [75–79]. The studies summa- from the urinary space consists of a three-layered
rized here illustrate the critical role of hypoxia- in-series structural arrangement of highly special-
regulated angiogenic factors and their signaling in ized cells that interact with one another (Fig. 1).
the differentiation, proliferation, migration, and The components of the GFB include three layers:
mutual regulation among glomerular endothe- the fenestrated endothelial cells lined by a
lium, mesangial cells, and podocytes. glycocalyx, the glomerular basement membrane
The development of postglomerular capillary (GBM), and the slit diaphragm, which links the
beds is thought to be driven and regulated by a neighboring podocyte foot processes.
similar array of angiogenic factors secreted by
renal tubules and endothelial or mural cell pre-
cursors, including VEGF-A, angiopoietins, stro- Components of the GFB
mal cell-derived factor-1, erythropoietin, and
angiotensin II. These factors are involved in the Table 1 and see also ▶ Chaps. 1, “Embryonic
remarkable alignment of postglomerular capil- Development of the Kidney,” and ▶ 18, “Transla-
laries with the renal tubules. The deletion of tional Research Methods: Renal Stem Cells” of
VEGF-A from developing renal tubules results this text.
in a hypoplastic medulla with fewer peritubular
capillaries [79, 80]. Angiopoietin-2 regulates The Podocyte
peritubular capillary architecture by promoting Podocytes, the glomerular visceral epithelial cells
vascular mural cell differentiation in the presence that reside within the Bowman’s space and bathe
of VEGF [81]. SDF-1 signaling from vascular in the glomerular filtrate, are a specialized and
mural cell progenitors to its receptor in endothelial anatomically unique feature of the glomerulus.
cells regulates the size and distribution of The podocyte has microtubule-based cellular
42 A. Tufro and A. Gulati

Fig. 1 Ultrastructure of a
typical glomerular capillary
loop: podocyte foot
processes ( fp), slit-
diaphragm (thin arrow),
glomerular basement
membrane (GBM), capillary
(cap) endothelial cell (EC)
with fenestrations (thick
arrows)

extensions known as primary processes and the filtration of large molecules such as ferritin
actin-based interdigitating secondary processes (MM ~500,000) is hindered at the GBM, whereas
known as foot processes, which rest on the GBM smaller proteins such as horseradish peroxidase
[85, 86]. Mature podocyte foot processes are (MM 40,000) are filtered by the GBM but retained
connected by modified adherens junctions called by the SD [91–93]. In addition to its structural
slit diaphragms (SD). During glomerulogenesis, function as a filtration barrier and cell-cell junc-
columnar podocytes are joined by tight junctions, tion, the SD is a signaling protein complex that
which migrate toward the basal side and acquire dynamically determines podocyte cell behavior.
SD features as differentiation proceeds. The major Extracellular domains of nephrin, neph1,
molecular components of the glomerular filtration P-cadherin, and FAT1 contribute to the SD protein
barrier are described in Table 1. complex, which includes multiple scaffolding,
The ultrastructure of the SD has been conven- receptors, and actin-binding proteins, through
tionally studied using transmission electron direct and indirect protein interactions. The list
microscopy (TEM) (Fig. 1), EM tomography, of SD complex members is ever growing, and
and scanning EM [87–90]. Rodewald and their roles are documented in gene ablation exper-
Karnovsky described the SD as “zipper structure iments, as well as human mutations leading to
arrangement” with regular pores with a mean proteinuria and glomerular disease ([94–111],
width of ~40A, which would restrict the passage Table 1).
of most serum proteins [87]. Tryggvason
et al. [89] provided new insight into the three- The Glomerular Basement Membrane
dimensional molecular structure of the podocyte The GBM is the extracellular gel-like matrix com-
slit diaphragm by identifying the extracellular ponent that lies between podocytes and endothe-
domain of nephrin as the backbone of the SD lial cells, composed of four major
and irregular pores with similar average width. macromolecules: laminin, type IV collagen,
Recent examination of the slit diaphragm using nidogen, and heparan sulfate proteoglycan [112]
scanning EM challenges the view of an ordered (see Table 1). In the mature GBM, laminin is a
“zipper-like” SD structure and suggests a trimer consisting of α5, β2, and γ1 laminin chains
heteroporous structure with ellipsoidal pores of (α5β2γ1 or 521), and collagen IV is a trimer
~120 A radius measured by digital morphometry consisting of α3, α4, and α5 chains [113]. In con-
[90]. Tracer studies followed by TEM show that trast, laminin α1β2γ1 and α1 and α2 collagen IV
2 Development of Glomerular Circulation and Function 43

Table 1 Known molecular components of the glomerular filtration barrier (Refs. [94–113])
Protein/
localization Gene Function Characteristic References
Slit diaphragm
Nephrin NPHS1 Finnish form of congenital Transmembrane protein [94–100,
nephrotic syndrome localized to SD 103, 104]
Infantile nephrotic syndrome
Autosomal recessive
inheritance
Neph-family Neph1 (Kirrel) Phenotype of the mice lacking Neph1–3 are components of [101, 102]
proteins Neph2 Neph1 resembles that of the SD
(Neph1–3) (Kirrel3) nephrin-deficient mice
Neph3
(Kirrel2)
Podocin NPHS2 Autosomal recessive steroid- Transmembrane protein [105, 106]
resistant nephrotic syndrome interacts with nephrin and
(infantile and childhood) neph1
CD2-associated CD2AP Autosomal recessive infantile Multidomain scaffolding [107–109]
protein steroid-resistant nephrotic protein at the SD
syndrome
Phospholipase C PLCE1 Autosomal recessive infantile Component of SD [110]
epsilon 1 and early childhood steroid-
resistant nephrotic syndrome;
major gene of DMS
Transient TRPC6 Autosomal dominant Component of SD [111]
receptor Juvenile and adult onset FSGS
potential cation
channel
subfamily C
member 6
FAT FAT Component of SD, co-localizes Adhesion molecule, large [195]
with ZO-1 protocadherin at SD contains
a larger extracellular domain
than traditional cadherins
Zonula ZO-1 Most commonly associated Present in cytoplasmic [195]
occludens-1 with tight junctions, sometimes component of SD
associated with adherens
junctions
Podocyte
WT-1 WT-1 First identified as a tumor Zinc finger transcription [193]
suppressor gene for Wilms factor and RNA-binding
tumor protein
Expression restricted to the
podocyte
Denys-Drash syndrome –
diffuse mesangial sclerosis
within the glomeruli
Frasier syndrome –
glomerulosclerosis
(continued)
44 A. Tufro and A. Gulati

Table 1 (continued)
Protein/
localization Gene Function Characteristic References
LMX1B LMX1B Regulates podocyte-specific LIM-homeodomain [193]
gene expression; KO mice transcription factor
show reduced expression of
foot processes
Unique role in renal
development and in the
patterning of the skeletal
system; mutation causes
nail-patella syndrome
Pod1/epicardin/ Pod1 KO mice glomerular Basic helix-loop-helix [193]
capsulin development arrests at single transcription factor
capillary loop stage
Integrins Alpha3beta1 integrin Adhesion protein in the [194]
Major integrin expressed by GBM
podocytes
Important for podocyte
differentiation
Receptor for some isoforms of
laminin
Major laminin-binding integrin
Alpha1beta1
Alpha2beta 2 are collagen IV
receptors
Podocalyxin Podxl Maintains podocyte cell Sulfated cell surface [195]
separation sialomucin-charged protein
GLEPP1 GLEPP1 Deficient mice are susceptible Cell surface protein [195]
to hypertension Receptor tyrosine
phosphatase
Synaptopodin Synaptopodin Foot process assembly Actin-associated [195]
cytoskeletal protein
Alpha-actinin 4 ACTN4 Familial FSGS has been Actin-binding protein [113]
described
VEGF-A VEGF Endothelial cell mitogen for Glycoprotein [25, 26]
endothelial cell differentiation,
survival, proliferation, and
migration
Angiopoietin-1 Angiopoietin-1 Remodeling and maturation of 70 kDa glycoprotein [31]
capillaries
GBM
Laminin LAMB2 Juvenile isoforms LM-111 and Large (~800 kd) heterotrimer [194]
(humans) LM-511 replaced by mature of alpha, beta, and gamma
isoform LM-521 in mature glycoprotein chains
glomeruli
No known human mutations in
LAMA1/B1/C1/A5
Mutations in human LAMB2
gene known (Pierson
syndrome)
(continued)
2 Development of Glomerular Circulation and Function 45

Table 1 (continued)
Protein/
localization Gene Function Characteristic References
Type-IV COL4A3, A4, Alpha-1,2 in early nephron, Triple helical heterotrimer of [194]
collagen A5 shift to 3,4,5 subunits in mature collagen IV alpha chains
GBM
Mutations in human COL4A3,
A4 (autosomal recessive), A5
(most common, X-linked)
genes – Alport syndrome
Nidogen One of the integral basement Two isoforms A and B, each [112, 113]
membrane protein consisting of a single
Function not yet determined polypeptide chain
Agrin (most Negatively charged GBM Heparan sulfate [112, 113]
abundant) and components originally thought proteoglycans with
perlecan to contribute to the charge negatively charged sulfated
selective barrier (however, glycosaminoglycan side
current evidence challenges chains
this REF)

are expressed in the developing GBM [114]. Spe- The Glomerular Endothelial Cell Layer
cific basement membrane protein isoforms are and Its Contributions to the Filtration
crucial for glomerular development, morphology, Barrier
and function, as mutations in laminin β2 The endothelium functions as a barrier, regulates
(LAMB2) or collagen IV (Alport) alter the GBM vasomotor tone, and controls tissue inflammation
structure and lead to progressive glomerular dis- and thrombosis. Glomerular endothelial cells
ease. The mechanisms for transitions from imma- form a fenestrated capillary bed and play a
ture to adult-type isoforms of the GBM proteins role in the filtration function [121]. Endothelial
are not completely understood, but these substitu- fenestrations are transcellular holes that allow
tions are important for the structural and func- the plasma flowing through the capillaries to
tional integrity of the GFB [113, 115–117]. It reach the GBM, even though fenestrations
has long been accepted that the net negative are plugged by a glycocalyx-like material
charge of the GBM is a crucial component of the consisting of sulfated proteoglycans and
glomerular capillary wall’s filtration barrier to glycoproteins that impart barrier properties
plasma albumin, which is also negatively charged [51, 55–57]. The glomerular endothelial
and should therefore be repelled by the GBM surface layer has a thickness similar to that of
[91]. However, the concept of charge selectivity the GBM consisting of two elements, the
has recently been called into question. Removal of glycocalyx and the cell coat [122]. It has been
these negatively charged proteoglycans has no proposed that the endothelial glycocalyx might
effect on the glomerular filtration barrier perme- contribute significantly to the GFB
ability to either albumin or to a negatively charged permselectivity [122–127]. The glycocalyx com-
tracer [118]. It has been suggested that GBM ponents are covalently bound to the endothelial
charge plays a minor role in imparting the glomer- cell membrane and are attached to the glycocalyx
ular filter with charge selectivity and argued that by charge-charge interactions [128, 129]. Damage
the GBM functions as a gel where macromole- to the glycocalyx and endothelial cell coat causes
cules traffic by diffusion depending on their proteinuria in the absence of detectable damage to
molecular size [119] or their size and anionic the GBM or the podocytes, but the evidence
charge [120]. remains indirect [130, 131].
46 A. Tufro and A. Gulati

As discussed above, the assembly and integrity compatible for crossing the barrier. In general,
of endothelial cells are regulated by angiogenic there has been a discrepancy between these calcu-
factor signaling. VEGF-A is necessary for the lations and the in vivo descriptions of molecular
survival of endothelial cells, to establish and sieving, suggesting that other mechanisms might
maintain the integrity of endothelial fenestrae, as be involved [145, 146]. Kedem and Katchalsky
demonstrated by knockdown and loss of function have proposed flux equations for defining the
in vivo experiments, and endothelial cell damage physical properties of the GFB and require no
leading to TMA in humans treated with VEGF-A specification of its substructure [147]. Charge
receptor blockers [35, 46, 47, 61]. Angiopoietin-1 selectivity has been an age-old phenomenon
produced by mural cells supports endothelial cell assumed to operate at the level of the GBM due
survival and decreases vascular permeability by to the presence of negatively charged proteogly-
inhibiting VEGF-induced eNOS activation cans, which were thought to repel the anionic
[132]. Angiopoietin-2 is stored in Weibel-Palade proteins. However, present-day emphasis is on
bodies and secreted by activated endothelial cells. the endothelial layer glycocalyx property confer-
Angiopoietin-2 and angiopoietin-1 compete for ring charge selectivity to the GFB. Smithies pro-
signaling via the Tie2 receptor; in the presence posed that the size selectivity of the glomerulus
of VEGF-A, angiopoietin-2 leads to angiogenesis, resides solely in the GBM, which functions as a
whereas in the absence of VEGF-A, it causes concentrated gel and macromolecules permeate
endothelial cell apoptosis [133]. Davis et al. mostly by diffusion and also by fluid flow (con-
showed that podocyte-specific overexpression of vection). Hence, this gel permeation/ diffusion
angiopoietin-2 leads to apoptosis of glomerular model suggests that lower filtration rate causing
endothelial cells, without affecting the podocytes lower water flow and thus a higher concentration
[134]. In addition, mesangial cells affect the glo- of proteins in the proximal tubular fluid and the
merular endothelial cell properties and promote tubular mechanisms of reabsorption saturate ear-
glomerular endothelial cell survival by lier to increase proteinuria [119]. Data from chil-
inactivating TGFβ [78, 135–138]. dren with nephrotic syndrome fit this hypothesis
[148], provided the degree of podocyte efface-
ment is also taken into account, suggesting that
Glomerular Filtration Barrier (GFB) the latter influences the size selectivity of the GFB
Selective Permeability beyond limitations of GFR. Recently, Moeller and
colleagues proposed an electrokinetic model,
The glomerular filtration barrier has size-selective whereby a local electric field “streaming poten-
properties and differentially handles molecules of tial” is established across the glomerular filter that
varying size. Small molecules up to the size of prevents plasma proteins from entering or cross-
inulin filter freely, whereas large molecules such ing the filter [120]. This hypothesis fits data from
as plasma proteins are held back. The sieving several previous models but awaits further exper-
coefficient for a particular molecule is the ratio imental confirmation.
of its concentration in the glomerular filtrate rela-
tive to the plasma concentration. The GFB pore
theory assumes that the capillary wall consists of Glomerular Hemodynamics
cylindrical pores of two or various sizes allowing and Assessment of Renal Function
size-selective passage of molecules through them
[139–143]. The fiber matrix theory, an extension The molecular basis of glomerular architecture
of the pore theory, posits that the GFB consists of discussed previously argues that the glomerular
pores filled with a fiber matrix [144]. Various structure provides functional advantages leading
mathematical models have been proposed and to controlled glomerular filtration. Mammalian
applied to the porous substructure of the barrier glomerular capillary pressure averages
to predict macromolecular sizes that will be 50 mmHg, which is approximately 50 % of the
2 Development of Glomerular Circulation and Function 47

mean systemic arterial pressure, with waveform capillary pressure. Thus changes in arteriolar
characteristics similar to the central aorta resistance lead to changes in glomerular plasma
[149]. The glomerular capillaries form a high- flow and glomerular capillary pressure, which can
pressure system with about 4 times the pressure influence GFR. For example, during sympathetic
of systemic capillaries and confer a functional stimulation or increased angiotensin II, both affer-
advantage due to higher blood flow rates enabling ent and efferent resistance increase; thus RPF
higher clearance as they filter large volumes of decreases, but GFR remains constant due to the
plasma (~180 l/day). opposite effect of afferent and efferent resistance
on GFR. This intrinsic autoregulation occurring
via neurohumoral mechanisms is mainly a conse-
Renal Blood Flow: Basic Concepts quence of local adjustment of RVR secondary to
changes in efferent arteriolar tone. The efferent
Renal blood flow is approximately 1 L/min (20 % arteriole is normally in a state of only partial
of cardiac output) Fig. 2. Renal plasma flow is the constriction, and any increase or decrease in
portion of the renal blood flow available for ultra- blood flow is accompanied by a reciprocal change
filtration: RPF = (1-Hct)*RBF, thus given a nor- in glomerular filtration pressure, with the result
mal Hct of 40 %, RPF is approximately that the filtration rate remains relatively
600 ml/min. Renal blood flow (RBF) is deter- unchanged [150–152]. Though the efferent arteri-
mined systemically by the renal perfusion pres- ole seems to take precedence over the afferent as a
sure that depends on the systemic arterial blood controller of renal hemodynamics, the contribu-
pressure (SBP) and locally by the renal vascular tions of pre-and postglomerular vasculature vary
resistance (RVR), so RBF/SBP/RVR. owing to specific circumstances. The efferent arte-
The unique architecture of the renal vascula- riole seems to be the major regulator of renal
ture enables to control the RVR at two sites, the blood flow during conditions of stress or after
afferent and efferent arterioles, where significant the administration of angiotensin-converting
pressure drops occur, leading to relatively high enzyme inhibitors or receptor blockers
glomerular capillary pressure and low peritubular [150–152]. However, the large increase in RPF
to the remnant kidney after a nephrectomy leads to
a dramatic decrease in afferent arteriole resistance,
20 Fraction of cardiac output perfusing the kidneys driving a rapid increase in GFR. Generally,
%
increased glomerular plasma flow leads to
increased GFR. The reduction in hematocrit in
euvolemic patients increases RPFs and is a mech-
15
anism contributing to hyperfiltration injury in ane-
mic states [153]. Under normal conditions, the
glomerular filtrate is a steady fraction of the
10 RPF, the so-called filtration fraction
(FF) [149]. FF = GFR/RPF. Because normal
GFR is 125 ml/min and RPF 600 ml/min, normal
FF is 0.2. FF is higher at low RPF and when
5
efferent arteriole resistance is increased.

Age, days
0 Perinatal Considerations for Renal
15 20 30 40 50 60 Blood Flow and GFR
Fig. 2 Renal blood flow as a percentage of cardiac output,
plotted versus age, in growing rats between 17 and 60 days The fetal human kidney main function from ges-
of age (From Ref. [196]) tation week 16 onwards is to produce urine to
48 A. Tufro and A. Gulati

maintain normal amniotic fluid volume, as the lower renal perfusion, incomplete nephrogenesis,
fetal fluid-electrolyte homeostasis and solute or reduced nephron number associated with pre-
clearance are achieved through the placenta and maturity Figs. 4 and 5.
maternal kidney function. Though nephrogenesis The most elaborate quantitative data on post-
is complete by 34–36 weeks of gestation, the natal RPF in neonates and small children has been
efficiency of the renal excretory system akin to provided from the measurement of renal extrac-
most other organ systems is age dependent even in tion ratios of PAH. PAH clearance in neonates
postnatal life related to structural maturation increases from about 65 % to attain 90 % of
[154]. It is of utmost importance to the clinician adult value by 5 months of age. RPF increased
to be aware of the functional capacity of the new- from about 140 ml/min/1.73 m2 at 8 days of age in
born kidney and the extreme states of vulnerabil- a full-term infant to 580 ml/min/1.73 m2 at
ity posed by prematurity and at the same time 5 months of age. Comparatively, the RPF in nor-
understand the expected sequence of physiologic mal adults is 625 ml/min [159].
adaptations of kidney function that shall occur In preterm neonates, the RPF (PAH clearance)
with time during a successful transition to is presumably lower, though not measured. The
extrauterine life. renal hemodynamic alterations between fetal and
The proportion of cardiac output flowing adult life are mainly accompanied by a gradual
through the kidney increases with gestational decrease in RVR and consequent increments in
and postnatal age; the fetal kidney is receiving RBF [160]. See comment in PubMed Commons
only 2–3 % of the cardiac output in animals and below. Inulin clearance of premature and term
humans alike [155–157]. The low fetal renal neonates remains low despite the correction for
blood flow, which does not attain adult levels body size [161]. This has been attributed to factors
until 2 years of age, forms the basis for reduced related to the low permeability of the glomerular
capacity for glomerular filtration in the filtration barrier, small available surface area for
newborn period and early childhood [158] filtration, low systemic arterial pressure, and rela-
Fig. 3. The normally low GFR in neonates is tively high resistance of the afferent glomerular
further decreased in premature infants due to arteriole [154]. Measurements of filtration pres-
sure in guinea pigs suggest a 2.5-fold increase
2
within the first 2 postnatal months. This in con-
160 GFR, ml/ min/ 1.73m junction with maturational changes in permeabil-
150 ity of the filtration barrier and increase in available
surface area for filtration could account for a
20-fold increase in GFR [162]. GFR increases
during postnatal development and maturation in
various animal species [163, 164]. Barnett
100
et al. showed that GFR in preterm infants does
not increase postnatally as rapidly as in full-term
infants [165]. The overall developmental pattern
of postnatal GFR changes in the humans suggests
50 nonlinear increments in GFR that are much more
pronounced with postnatal age beyond 34 weeks
of gestation, coinciding with the time when
nephrogenesis is completed, as compared to
much slower GFR increases at lower gestational
Age, months
0 age (Fig. 6) [166]. However, this has been
0 5 10 15
questioned by recent estimates of GFR [167].
Fig. 3 Glomerular filtration rate (GFR) during the first Overall, neonates constitute a high-risk group
year of life (From Ref. [197]) requiring thoughtful fluid management and choice
2 Development of Glomerular Circulation and Function 49

Fig. 4 Creatinine CREATININE CLEARANCE


clearance measured within (ml/min/1.73 M2)
24–40 h of birth in 30-week
premature to 40-week r = 0.643
full-term infants p = < .001
(From Ref. [198]) 50

40

30

20

10

26 26 30 32 34 36 38 40
GESTATION (Weeks)

1.8
1− 30 days 31− 94 days
1.6 n = 52 n = 29
PLASMA CREATININE (mg/dl)

r = −0.575 r = 0.006
1.4 p < 0.001 PNS
1.2

1.0

0.8

0.6

0.4

0.2

10 20 30 40 50 60 70 80 90 100
AGE (Days)

Fig. 5 Plasma creatinine values during the first 3 months of life in low-birth-weight infants (less than 2,000 g). (From
Ref. [199])

and dosage of exogenous agents, aiming at renal hemodynamics and knowledge of glomeru-
primum non nocere. lar physiology. The classical concept of glomeru-
lar filtration stems from Ludwig’s filtration theory
GFR: Basic Concepts that states that water and solutes move across the
Our understanding of the GFR has followed a glomerular filtration barrier due to a hydrostatic
distinct timeline with advances in evaluation of pressure difference that favors filtration [168].
50 A. Tufro and A. Gulati

creatinine clearance (ml / min)


5

28 30 32 34 36 38 40
gestational age (wks)

Fig. 6 Increase in creatinine clearance with GA (gesta- life are represented by a mass plot of data with GA, in
tional + postnatal ages). Infants studied at birth were which studies from one infant at different ages are indi-
grouped for GA and are represented by mean values  cated by the same symbol (y = 0.406  11.548, r = 0.68,
1 SD connected by the solid line (y = 0.170  4.95, r = P <0.001) (Source: Ref. [166])
0.51, P <0.001). Infants studied during later extrauterine

This perhaps basic and widely accepted theory plus the capsular pressure. The equilibrium
was for some time refuted, and emphasis was oncotic pressure can be calculated as Po is the
laid on tubular secretion as a primary mechanism initial oncotic pressure. FF averages ~20 % and
for excretion [169]. However, observations of the taking Po as 24 mmHg [172], at filtration equilib-
direct passage of the dye indigo carmine from the rium, the plasma proteins will have been concen-
blood into the Bowman’s space [170] supported trated 1.25 times, and Po raised to 30 mmHg. The
the original filtration theory, also supported by filtration equilibrium will in principle be
Starling [171]. maintained so long as the glomerular circulation
The movement of water and solutes via ultra- is active and Bowman’s capsule contains free
filtration and diffusion during glomerular filtra- fluid.
tion requires that the hydrostatic pressure within
the glomerular capillaries (PG) exceeds the sum of
the oncotic pressure (Po) plus the capsular pres- The Concept of Clearance
sure (Pc), that is, the pressure existing within the
Bowman’s capsule and approximately equal to Renal clearance of a substance X is the volume of
interstitial pressure. Along the capillary, as hydro- plasma “cleared” of substance X by the kidneys in
static pressure decreases and plasma oncotic pres- a given time period (Cx; ml/min). The term “clear-
sure increases, at some point, the net ultrafiltration ance” entails a mathematical description of the
pressure dissipates and a filtration equilibrium is renal handling of various substances under phys-
achieved. This hydrostatic null point occurs in the iological and pathophysiological conditions and
proximal portion of the efferent arteriole so as to thus is used to measure kidney functional proper-
promote maximal GFR and maximal reabsorption ties such as GFR and the RBF [173, 174]. Clear-
in the peritubular capillaries [149, 152]. Since the ance is expressed as a rate (ml/min) and corrected
mean glomerular pressure is ~ 50–60 % of the (normalized) by body weight, kidney weight, or
mean systemic arterial pressure (~90 mmHg), an estimate of surface area. These corrections are
PG averages ~45 mmHg. At filtration equilib- indispensable in pediatrics, given the broad range
rium, this value should equal the oncotic pressure of normal patients size. Renal clearance of any
2 Development of Glomerular Circulation and Function 51

given substance results from glomerular filtration constant rate regardless of amount that is in solu-
and any combination of the following processes: tion within the plasma.
tubular secretion, tubular reabsorption, and The all-time gold standard for GFR determina-
intrarenal metabolism. tion is the inulin clearance method [151]. Inulin is
Cx = (Ux * V)/Px; Ux and Px are the concen- a small, inert polysaccharide, is a fructose poly-
trations of X in urine and plasma (mg/ml), respec- mer containing 32 fructose molecules, has a
tively, and V (ml/min) is the urine flow. Thus, Cx molecular weight of 5.7 KDa, and fulfills all the
is expressed as ml/min. aforementioned criteria. Conventionally, GFR has
been measured following a constant intravenous
Measurement of RBF infusion of inulin with periodic blood and urine
A plasma substance that is fully cleared on a sampling. This method is currently not used in
single passage through the kidneys can be used clinical practice but has value as a research tool
as a marker of renal plasma flow (RPF). Para- [175, 177]. Multiple variations of inulin clearance
aminohippuric acid (PAH), an organic acid not have been developed. A bolus injection of inulin
normally present in the body, is given intrave- and serial measurement of its plasma concentra-
nously to achieve a low concentration (<12 tion enables accurate calculation of GFR and is
mg/dl), and the clearance of PAH accurately esti- feasible [178, 179] but not frequently used. Inulin
mates RPF [175, 176]. Although conventionally a availability for human use and a somewhat cum-
constant infusion of PAH is infused and periodic bersome assay are major limiting factors for its
multiple plasma and urine samples are obtained, a clinical use. Inulin still remains as the gold stan-
single injection technique using 131I-labeled PAH dard validation tool against which all other
is more practical. Indirect assessment of RBF by methods can be tested and compared.
Doppler ultrasound is more often used in clinical Radioactive (3H)-labeled inulin is commonly
practice. Similarly, Doppler RBF probes are used experimentally in rodents and the only avail-
placed on the renal artery to assess pressure and able method for direct measurement of single-
flow in experimental studies, in addition to the nephron GFR (SNGFR) in micropuncture studies.
measurement of intraglomerular pressure by A surrogate for radiolabeled inulin is 1251-
micropuncture of superficial glomeruli [149]. iothalamate, which can be used in humans
because of minimal radiation exposure, has been
Measurement of GFR shown to have comparable accuracy
The most frequent clinical use of measuring clear- [180–182]. However, logistics of radioactive iso-
ance is in determining the GFR. While renal clear- topes used in medicine are making these methods
ance takes into account the cumulative commonly out of reach. The methods based on
performance of all nephrons, techniques for mea- renal clearance of exogenous markers such as
51
surement of single-kidney and single-nephron Cr-EDTA, 99Tc-DTPA, 1251-iothalamate, and
GFR have been employed in experimental set- iohexol are accurate but cumbersome with less
tings. In order for a substance to accurately mea- practical utility for clinical use. These methods
sure GFR, it must meet a series of criteria, as involve multiple periodic blood samples (4) as
defined by Homer Smith. These include the fol- the plasma clearance is calculated by the injected
lowing: (i) the substance in question is freely total dose divided by the total area under the curve
filtered by the kidneys; (ii) it is neither secreted (AUC) obtained by a number of blood samples
nor reabsorbed by the tubules; (iii) it is physiolog- after injection of the tracer. The simplified version
ically inert and nontoxic; (iv) it is neither cleared using less or a single blood sample is less accurate
nor metabolized in extrarenal sites; (v) it is neither but more feasible. However, clinical imprecision
synthesized, stored, nor metabolized within the especially for low GFRs further limits its utility.
kidney; (vi) it is not bound to proteins and the The tracer molecules are further inaccurate in
plasma level of the substance must remain reason- edema states when they get distributed into the
ably constant; and (vii) it should be cleared at a extracellular volume.
52 A. Tufro and A. Gulati

Fig. 7 Analysis of 100


log-transformed height/Scr 90
and iGFR, showing that 80
65.0 % of the variation in 70
log(iGFR) can be explained
60
by log(height/Scr).
Regression line and 50
nonparametric spline

iGFR
depicted by dashed curve 40
are superimposed (Source:
Fig. 1 of Ref. [184])
30

20
R-square = 65.0%
iGFR= 41.2 [ht/Scr]0.775

1/2 2/3 1 3/2 2


height [meters] / Scr

Creatinine as Endogenous Marker of GFR enzymatic or by mass spectrometry, and standard-


Methods utilizing endogenous markers such as ized values are approximately 25 % lower than
serum creatinine are the most commonly used in previously used colorimetric assays.
clinical practice. Creatinine is a small (113 Da) Pediatric GFR prediction models based on
molecule resulting from creatine and phosphocre- serum creatinine and height are reasonably accu-
atine metabolism, produced and excreted in the rate and widely used in clinical practice. The most
urine at a steady rate (20  5 mg/kg/day) propor- used and recommended is the Schwartz equation
tional to muscle mass. Serum creatinine is not for the calculation of estimated GFR, which nor-
bound to proteins and is freely filtered and also malizes the calculated GFR to a standard adult
secreted actively by the proximal tubule. Tubular surface area and has been updated taking into
secretion of creatinine amounts to ~16 % in nor- account the enzymatic creatinine assay [184]
mal adults, but increases up to 92 % at low GFRs; Fig. 7.
therefore the overestimation of GFR as assessed Creatinine clearance (ml/min/1.73 m2) = k 
by creatinine clearance is considerable in patients height (cm)/plasma creatinine (mg/dl); where k is
with GFR <40 ml/min [183]. Another limitation a constant = 0.413.
of creatinine as a marker of GFR is its dependence
on muscle mass, which renders it not meaningful Cystatin C and Beta-Trace Protein
in patients with decreased muscle mass, such as as Endogenous Markers of GFR
those with chronic malnutrition, malignancy, Plasma cystatin C, an endogenous small-
liver, heart, or neurologic disease. This limitation molecular-weight protein secreted by most tis-
is also relevant to healthy children, as muscle sues, freely filtered, and almost completely
mass varies drastically with growth and develop- degraded in the tubules, has been used as a marker
ment, as well as gender postpuberty. The measure- of GFR and validated in large studies including
ment of creatinine clearance entails timed urine children and by meta-analysis [185, 186]. It is
collection, which is cumbersome and potentially generally considered as accurate as serum creati-
impossible in small children as outpatients. Serum nine to estimate GFR and probably more accurate
creatinine is useful for population studies and for when GFR is below 75 ml/min (when the percent-
the longitudinal follow-up of kidney function in age of tubular secretion of creatinine increases)
individual patients deemed to have normal muscle [167, 185–187]. The main current drawback for
mass. Current serum creatinine assays are cystatin C use is the lack of standardization of
2 Development of Glomerular Circulation and Function 53

reference values using two different assays avail- 7. Espinoza-Valdez A, Femat R, Ordaz-Salazar FC. A
able [167] and cost. Beta-trace protein is an model for renal arterial branching based on graph
theory. Math Biosci. 2010;2010(225):36–43.
endogenous circulating low-molecular-weight 8. Tomake RJ. Assembly of the vasculature and its reg-
protein independent of size, gender, or muscle ulation. Berlin: Birkhäuser; 2001.
mass and is considered a potential surrogate 9. Sequeria López ML, Gomez RA. Desarrollo de la
GFR marker promising in pregnant women and vasculatura renal. Medicina. 2000;60:694
(In Spanish) // Sequeira Lopez ML, Gomez
neonates [188]. RA. Development of the renal arterioles. JASN.
2011;22:2156–65.
Newer Methods for GFR Measurement 10. Tufro A, Tufro-McReddie A, Norwood VF, Aylor
More recently, a fluorescence-based assay using KW, Botkin SJ, Carey RM, Gomez RA. Oxygen reg-
ulates vascular endothelial growth factor-mediated
fluorescein isothiocyanate-inulin (FITC-inulin) vasculogenesis and tubulogenesis. Dev Biol.
has been developed [189]. The rapid development 1997;183:139–49.
of fluorescent microscopy has led to some addi- 11. Potter EL. Development of the human glomerulus.
tional novel approaches to determining GFR Arch Pathol. 1965;80:241–55.
12. Grobstein C. Inductive interaction in the development
in vivo. Yu et al. used two-photon excitation of the mouse metanephros. J Exp Zool.
microscopy in rats with superficial glomeruli to 1955;130:319–40.
directly visualize GFR at near real-time speeds 13. Bernstein J, Cheng F, Roska J. Glomerular differenti-
[190]. The potential for such real-time GFR meth- ation in metanephric culture. Lab Invest.
1981;45:183–90.
odology is provocative, as it is portable to the 14. Poole TJ, Coffin JD. Vasculogenesis and angiogene-
bedside and does not depend on numerous sis: two distinct morphogenetic mechanisms establish
assumptions seen in some other methodologies. embryonic vascular pattern. J Exp Zool. 1989;251(2):
The two-compartment system controls for itself 224–31.
15. Sariola H, Ekblom P, Lehtonen E, Saxén
and does not require taking multiple blood sam- L. Differentiation and vascularization of the meta-
ples. It is highly likely that the use of fluorescence nephric kidney grafted on the chorioallantoic mem-
to accurately determine GFR in humans will ulti- brane. Dev Biol. 1983;96:427–35.
mately be accomplished noninvasively using sen- 16. Sariola H, Saarma M, Sainio K, Arumäe U, Palgi J,
Vaahtokari A, Thesleff I, Karavanov A. Dependence
sors that do not need to penetrate the skin or enter of kidney morphogenesis on the expression of nerve
the blood stream [191, 192]. growth factor receptor. Science. 1991;254
(5031):571–3.
17. Ekblom P, Sariola H, Karkinen-Jaaskelainen M,
Saxen L. The origin of the glomerular endothelium.
Cell Differ. 1982;11:35–9.
References 18. Abrahamson DR, Robert B, Hyink DP, St John PL,
Daniel TO. Origins and formation of microvascula-
1. Malpighi M. De viscerum structura exercitatio ture in the developing kidney. Kidney Int Suppl.
anatomica (la-cobi Montij, Bononiae); 1666. 1998;67:S7–11.
2. Bowman W. On the structure and use of the Malpi- 19. Hyink DP, Tucker DC, St John PL,
ghian bodies of the kidney, with observations on the Leardkamolkarn V, Accavitti MA, Abrass CK,
circulation through that gland. Philos Trans R Soc Abrahamson DR. Endogenous origin of glomerular
Lond B Biol Sci. 1842;132:57–80. endothelial and mesangial cells in grafts of embryonic
3. O’Brien LL, McMahon AP. Induction and patterning kidneys. Am J Physiol. 1996;270:F886–99.
of the metanephric nephron. Semin Cell Dev Biol. 20. Robert B, St John PL, Hyink DP, Abrahamson
2014. http://dx.doi.org/10.1016/j.semcdb.2014.08. DR. Evidence that embryonic kidney cells expressing
014. flk-1 are intrinsic, vasculogenic angioblasts. Am J
4. Herzlinger D, Hurtado R. Patterning the renal vascu- Physiol. 1996;271:F744–53.
lar bed. Semin Cell Dev Biol. 2014. http://dx.doi.org/ 21. Robert B, St John PL, Abrahamson DR. Direct
10.1016/j.semcdb.2014.08.002. visualization of renal vascular morphogenesis in
5. Noden DW. Embryonic origins and assembly of Flk1 heterozygous mutant mice. Am J Physiol.
blood vessels. Am Rev Respir Dis. 1989;140(4): 1998;275:F164–72.
1097–103. 22. Shalaby F, Rossant J, Yamaguchi TP. Failure of
6. Wilting JR, Christ B. Embryonic angiogenesis: a blood-island formation and vasculogenesis in Flk-1-
review. Naturwissenschaften. 1996;83:153–64. deficient mice. Nature. 1995;376:62–6.
54 A. Tufro and A. Gulati

23. Fong G-H, Rossant J, Gertsenstein M, Breitman secreted angiogenic mitogen. Science.
ML. Role of the Flt-1 receptor tyrosine kinase in 1989;246:1306–9. PubMed: 2479986.
regulating the assembly of vascular endothelium. 39. Keck PJ, Hauser SD, Krivi G, Sanzo K, Warren T,
Nature. 1995;376:66–70. Feder J, et al. Vascular permeability factor, an endo-
24. Yamaguchi TP, Dumont DJ, Conlon RA, Breitman thelial cell mitogen related to PDGF. Science.
ML. Rossant J: flk-1, an flt-related receptor tyrosine 1989;246:1309–12. PubMed: 2479987.
kinase, is an early marker for endothelial precursors. 40. Senger DR. Vascular endothelial growth factor: much
Development. 1993;118:489–98. more than an angiogenesis factor. Mol Biol Cell.
25. Tufro A, Norwood VF, Carey RM, Gomez 2010;21:377–9. PubMed: 20124007.
RA. Vascular endothelial growth factor induces 41. Ferrara N, Houck K, Jakeman L, Leung
nephrogenesis, and vasculogenesis. J Am Soc DW. Molecular and biological properties of the vas-
Nephrol. 1999;10:2125–34. cular endothelial growth factor family of proteins.
26. Simon M, Grone HJ, Johren O. Expression of vascu- Endocr Rev. 1992;13:18–32.
lar endothelial growth factor and its receptors in 42. Coultas L, Chawengsaksophak K, Rossant
human renal ontogenesis and adult kidney. Am J J. Endothelial cells and VEGF in vascular develop-
Physiol. 1995;268:F240–50. ment. Nature. 2005;438:937–45. PubMed: 16355211.
27. Flamme I, von Reutern M, Drexler HCA, Syed-Ali S, 43. Tischer E, Mitchell R, Haertman T, et al. The human
Risau W. Overexpression of vascular endothelial gene for vascular endothelial growth factor. Multiple
growth factor in the avian embryo induces hypervas- protein forms are encoded through alternative exon
cularization and increased vascular permeability splicing. J Biol Chem. 1991;266:11947–54.
without alterations of embryonic pattern formation. 44. Guan F, Villegas G, Teichman J, Mundel P, Tufro
Dev Biol. 1995;171:399–414. A. Autocrine VEGF-A system in podocytes regulates
28. Sims-Lucas S, Schaefer C, Bushnell D, Ho J, podocin and its interaction with CD2AP. Am J
Logar A, et al. Endothelial progenitors exist within Physiol Renal Physiol. 2006;291:F422–8. PubMed:
the kidney and lung mesenchyme. PLoS One. 16597608.
2013;8(6):e65993. 45. Eremina V, Sood M, Haigh J, Nagy A, Lajoie G,
29. Tufro-McReddie A, Norwood VF, Aylor KW, Botkin Ferrara N, et al. Glomerular-specific alterations of
SJ, Carey RM, Gomez RA. Oxygen regulates vascu- VEGF-A expression lead to distinct congenital and
lar endothelial growth factor-mediated acquired renal diseases. J Clin Invest.
vasculogenesis and tubulogenesis. Dev Biol. 2003;111:707–16. PubMed: 12618525.
1997;183:139–49. 46. Roberts WG, Palade GE. Neovasculature induced by
30. Tufro A. VEGF spatially directs angiogenesis during vascular endothelial growth factor is fenestrated. Can-
metanephric development in vitro. Dev Biol. cer Res. 1997;57:765–72.
2000;227:558–66. 47. Kamba T, et al. VEGF-dependent plasticity of fenes-
31. Loughna S, Landels EC, Woolf AS. Growth factor trated capillaries in the normal adult microvascula-
control of developing kidney endothelial cells. Exp ture. Am J Physiol. 2006;290:H560–76.
Nephrol. 1996;4:112–8. 48. Esser S, Wolburg K, Wolburg H, Breier G,
32. Woolf AS, Loughna S. Origin of glomerular Kurzchalia T, Risau W. Vascular endothelial growth
capillaries: is the verdict in? Exp Nephrol. factor induces endothelial fenestration in vitro. J Cell
1998;6:17–21. Biol. 1998;140:947–59.
33. Hyink DP, Abrahamson DR. Origin of the glomerular 49. Kretzler M, Schroppel B, Merkle M, et al. Detection
vasculature in the developing kidney. Semin Nephrol. of multiple vascular endothelial growth factor splice
1995;15:300–14. isoforms in single glomerular podocytes. Kidney Int
34. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Suppl. 1998;67:S159–61.
Nowlin BT, Bonventre JV, et al. Fate tracing reveals 50. Breier G, Albrecht U, Sterrer S, Risau W. Expression
the pericyte and not epithelial origin of of vascular endothelial growth factor during embry-
myofibroblasts in kidney fibrosis. Am J Pathol. onic angiogenesis and endothelial cell differentiation.
2010;176:85–97. Development. 1992;114(2):521–32.
35. Risau W. Differentiation of endothelium. FASEB 51. Brown LF, Berse B, Tognazzi K, Manseau EJ, Van de
J. 1995;9:926–33. Water L, Senger DR, Dvorak HF, Rosen
36. Risau W, Hallmann R, Albrecht U, Henke-Fahle S. Vascular permeability factor mRNA and protein
S. Brain induces the expression of an early cell sur- expression in human kidney. Kidney Int. 1992;42
face marker for blood–brain barrier-specific endothe- (6):1457–61.
lium. EMBO J. 1986;5:3179–83. 52. Eremina V, Cui S, Gerber H, Ferrara N, Haigh J,
37. Schell C, Wanner N, Huber TB. Glomerular Nagy A, et al. Vascular endothelial growth factor a
development–shaping the multi-cellular filtration signaling in the podocyte-endothelial compartment is
unit. Se. Cell Dev Biol. 2014;36:39–49. required for mesangial cell migration and survival. J
38. Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Am Soc Nephrol. 2006;17:724–35. PubMed:
Ferrara N. Vascular endothelial growth factor is a 16436493.
2 Development of Glomerular Circulation and Function 55

53. Veron D, Reidy K, Villegas G, Kopp J, Thomas D, 66. Carmeliet P, et al. Abnormal blood vessel develop-
Tufro A. Induction of podocyte VEGF-A ment and lethality in embryos lacking a single VEGF
overexpression in adult mice causes glomerular dis- allele. Nature. 1996;380:435–9.
ease. Kidney Int. 2010;77:989–99. PubMed: 67. Ferrara N, Carver-Moore K, Chen H. Heterozygous
20375978. embryonic lethality induced by targeted inactivation
54. Veron D, Reidy K, Marlier A, Villegas G, of the VEGF gene. Nature. 1996;380:439–42.
Kashgarian M, Tufro A. Induction of podocyte 68. Breier G, Risau W. The role of vascular endothelial
VEGF164 overexpression at different stages of devel- growth factor in blood vessel formation. Trends Cell
opment causes congenital nephrosis or steroid- Biol. 1996;6:454.
resistant nephrotic syndrome. Am J Pathol. 69. Carmeliet P. Mechanisms of angiogenesis and
2010;177:2225–33. PubMed: 20829436. arteriogenesis. Nat Med. 2000;6:389–95.
55. Jin J, Sison K, Li C, et al. Soluble FLT1 binds lipid 70. Eichmann A, Makinen T, Alitalo K. Neural guidance
micro-domains in podocytes to control cell morphol- molecules regulate vascular remodeling and vessel
ogy and glomerular barrier function. Cell. navigation. Genes Dev. 2005;19:1013–21.
2012;151:384–99. 71. Gelfand MV, Hong S, Gu C. Guidance from above:
56. Tufro A, Veron D. VEGF and podocytes in common cues direct distinct signaling outcomes in
diabetic nephropathy. Semin Nephrol. 2012;32: vascular and neural patterning. Trends Cell Biol.
385–93. 2009;19:99–110.
57. Bertuccio C, Veron D, Aggarwal PK, et al. Vascular 72. Reidy KJ, Villegas G, Teichman J, Veron D, Shen W,
endothelial growth factor receptor 2 direct interaction Jimenez J, et al. Semaphorin 3a regulates endothelial
with nephrin links VEGF-A signals to actin in kidney cell number and podocyte differentiation during
podocytes. J Biol Chem. 2011;286:39933–44. glomerular development. Development. 2009;136:
58. Lee S, Chen TT, Barber CL, Jordan MC, Murdock J, 3979–89.
Desai S, Ferrara N, Nagy A, Roos KP, Iruela- 73. Lindahl P, Hellstrom M, Kalen M, Karlsson L,
Arispe ML. Autocrine VEGF signaling is required Pekny M, Pekna M, et al. Paracrine PDGF-B/PDGF-
for vascular homeostasis. Cell. 2007;130(4): R beta signaling controls mesangial cell development
691–703. in kidney glomeruli. Development.
59. Villegas G, Lange-Sperandio GB, Tufro A. Autocrine 1998;125:3313–22.
and paracrine functions of vascular endothelial 74. Lindahl P, Johansson BR, Leveen P, Betsholtz
growth factor (VEGF) in renal tubular epithelial C. Pericyte loss and microaneurysm formation in
cells. Kidney Int. 2005;67:449–57. PubMed: PDGF-B-deficient mice. Science. 1997;277:242–5.
15673292. 75. Boyle SC, Liu Z, Kopan R. Notch signaling is
60. Kanellis J, Fraser S, Katerelos M, Power required for the formation of mesangial cells from a
DA. Vascular endothelial growth factor is a survival stromal mesenchyme precursor during kidney devel-
factor for renal tubular epithelial cells. Am J Physiol opment. Development. 2014;141:346–54.
Renal Physiol. 2000;278:F905–15. PubMed: 76. Lin EE, Sequeira-Lopez ML, Gomez RA. RBP-J in
10836978. FOXD1+ renal stromal progenitors is crucial for the
61. Eremina V, Jefferson JA, Kowalewska J, Hochster H, proper development and assembly of the kidney vas-
Haas M, Weisstuch J, et al. VEGF inhibition and renal culature and glomerular mesangial cells. Am J
thrombotic microangiopathy. N Engl J Med. Physiol Renal Physiol. 2014;306:F249–58.
2008;358:1129–36. PubMed: 18337603. 77. Takabatake Y, Sugiyama T, Kohara H, Matsusaka T,
62. Veron D, Villegas G, Aggarwal P, Moeckel G, Kurihara H, Koni PA, et al. The CXCL12 (SDF-1)/
Kashgarian M, Tufro A. Acute podocyte VEGF-A CXCR4 axis is essential for the development of
knockdown disrupts αVβ3 integrin signaling in the renal vasculature. J Am Soc Nephrol.
glomerulus. J Am Soc Nephrol. 2011;22:9A. 2009;20:1714–23.
PubMed: 21164024. 78. Jeansson M, Gawlik A, Anderson G, Li C,
63. M€uller-Deile J, Worthmann K, Saleem M, Tossidou I, Kerjaschki D, Henkelman M, et al. Angiopoietin-1
Haller H, Schiffer M. The balance of autocrine is essential in mouse vasculature during development
VEGF-A and VEGF-C determines podocyte survival. and in response to injury. J Clin Invest.
Am J Physiol Renal Physiol. 2009;297:F1656–67. 2011;121:2278–89.
PubMed: 19828679. 79. Dimke H, Sparks MA, Thomson BR, Frische S,
64. Ku CH, White KE, Dei Cas A, Hayward A, Coffman TM, Quaggin SE. Tubulovascular Cross-
Webster Z, Bilous R, et al. Inducible overexpression Talk by vascular endothelial growth factor a main-
of sFlt-1 in podocytes ameliorates glomerulopathy in tains peritubular microvasculature in kidney. J Am
diabetic mice. Diabetes. 2008;57:2824–33. PubMed: Soc Nephrol. 2015;26(5):1027–38.
18647955. 80. Tufro A. Tubular vascular endothelial growth factor-
65. Weis SM, Cheresh DA. Pathophysiological conse- A, erythropoietin, and medullary vessels: a trio linked
quences of VEGF-induced vascular permeability. by hypoxia. J Am Soc Nephrol. 2014;pii:
Nature. 2005;437:497–504. PubMed: 16177780. ASN.2014101004.
56 A. Tufro and A. Gulati

81. Pitera JE, Woolf AS, Gale NW, Yancopoulos GD, 96. Helmstädter M, L€ uthy K, Gödel M, Simons M,
Yuan HT. Dysmorphogenesis of kidney cortical Ashish ND, Rensing SA, Fischbach KF, Huber
peritubular capillaries in angiopoietin-2-deficient TB. Functional study of mammalian Neph proteins
mice. Am J Pathol. 2004;165:1895–906. in Drosophila melanogaster. PLoS One. 2012;7:
82. Madsen K, Marcussen N, Pedersen M, e40300.
Kjaersgaard G, Facemire C, Coffman TM, 97. Huber TB, Benzing T. The slit diaphragm: a signaling
et al. Angiotensin II promotes development of the plat- form to regulate podocyte function. Curr Opin
renal microcirculation through AT1 receptors. J Am Nephrol Hypertens. 2005;14:211–6.
Soc Nephrol. 2010;21:448–59. 98. Neumann-Haefelin E, Kramer-Zucker A, Slanchev K,
83. Pappenheimer JR. Über die Permeabilität der Hartleben B, Noutsou F, Martin K, Wanner N,
Glomerulummembranen in der Niere. Klir Ritter A, Gödel M, Pagel P, Fu X, M€ uller A,
Wochenschr. 1955;33:362. Baumeister R, Walz G, Huber TB. A model organism
84. Landis EM, Pappenheimer JR. Exchange of sub- approach: defining the role of Neph proteins as regu-
stances through the capillary walls. Handb Physiol. lators of neuron and kidney morphogenesis. Hum
1963;2(2):961. Mol Genet. 2010;19:2347–59.
85. Dressler GR. The cellular basis of kidney develop- 99. Rantanen M, Palmén T, Pätäri A, Ahola H,
ment. Annu Rev Cell Dev Biol. 2006;22:509–29. Lehtonen S, Aström E, Floss T, Vauti F, Wurst W,
86. Mundel P, Kriz W. Structure and function of Ruiz P, Kerjaschki D, Holthöfer H. Nephrin TRAP
podocytes: an update. Anat Embryol (Berl). mice lack slit diaphragms and show fibrotic glomeruli
1995;192:385–97. and cystic tubular lesions. J Am Soc Nephrol.
87. Rodewald R, Karnovsky MJ. Porous substructure of 2002;13:1586–94.
the glomerular slit diaphragm in the rat and mouse. 100. Ruotsalainen V, Ljungberg P, Wartiovaara J,
J Cell Biol. 1974;60:423–33. et al. Nephrin is specifically located at the slit dia-
88. Hora K, Ohno S, Ogushi H, Furukawa T, Furuta phragm of glomerular podocytes. Proc Natl Acad Sci
S. Three-dimensional study of glomerular slit dia- U S A. 1999;96:7962–7. PubMed: 10393930.
phragm by the quick-freezing and deep-etching rep- 101. Sellin L, Huber TB, Gerke P, Quack I, Pavenstädt H,
lica method. Eur J Cell Biol. 1990;53:402–6. Walz G. NEPH1 defines a novel family of podocin
89. Wartiovaara J, Ofverstedt LG, Khoshnoodi J, interacting proteins. FASEB J. 2003;17:115–7.
Zhang J, Makela E, Sandin S, Ruotsalainen V, 102. Donoviel DB, Freed DD, Vogel H, et al. Proteinuria
Cheng RH, Jalanko H, Skoglund U, Tryggvason and perinatal lethality in mice lacking NEPH1, a
K. Nephrin strands contribute to a porous slit dia- novel protein with homology to NEPHRIN. Mol
phragm scaffold as revealed by electron tomography. Cell Biol. 2001;21:4829–36.
J Clin Invest. 2004;114:1475–83. 103. Doné SC, Takemoto M, He L, et al. Nephrin is
90. Gagliardini E, Conti S, Benigni A, Remuzzi G, involved in podocyte maturation but not survival dur-
Remuzzi A. Imaging of the porous ultrastructure of ing glomerular development. Kidney Int.
the glomerular epithelial filtration slit. J Am Soc 2008;73:697–704.
Nephrol. 2012;21:2081–9. 104. Simons M, Huber TB. It’s not all about nephrin.
91. Farquhar MG, Wissig SL, Palade GE. Glomerular Kidney Int. 2008;73:671–3.
permeability. I. Ferritin transfer across the normal 105. Boute N, Gribouval O, Roselli S, Benessy F, Lee H,
glomerular capillary wall. J Exp Med. 1961;113:47. Fuchshuber A, Dahan K, Gubler MC, Niaudet P,
92. Farquhar MG, Palade GE. Glomerular permeability. Antignac C. NPHS2, encoding the glomerular protein
II. Ferritin transfer across the glomerular capillary podocin, is mutated in autosomal recessive steroid-
wall in nephrotic rats. J Exp Med. 1961;114:699. resistant nephrotic syndrome. Nat Genet.
93. Venkatachalam MA, Karnovsky MJ, Cotran 2000;24:349–54.
RS. Glomerular permeability. Ultrastructural studies 106. Miner JH. Focusing on the glomerular slit diaphragm:
in experimental nephrosis using horseradish peroxi- podocin enters the picture. Am J Pathol.
dase as a tracer. J Exp Med. 1969;130:381–99. 2002;160:3–5.
94. Kestilä M, Lenkkeri U, Männikkö M, Lamerdin J, 107. Asanuma K, Campbell KN, Kim K, Faul C, Mundel
McCready P, Putaala H, Ruotsalainen V, Morita T, P. Nuclear relocation of the nephrin and CD2AP-
Nissinen M, Herva R, Kashtan CE, Peltonen L, binding protein dendrin promotes apoptosis of
Holmberg C, Olsen A, Tryggvason K. Positionally podocytes. Proc Natl Acad Sci U S A. 2007;104:
cloned gene for a novel glomerular 10134–9.
protein–nephrin–is mutated in congenital nephrotic 108. Shih NY, Li J, Karpitskii V, Nguyen A, Dustin ML,
syndrome. Mol Cell. 1998;1:575–82. Kanagawa O, Miner JH, Shaw AS. Congenital
95. Wanner N, Noutsou F, Baumeister R, Walz G, Huber nephrotic syndrome in mice lacking CD2-associated
TB, Neumann-Haefelin E. Functional and spatial protein. Science. 1999;286:312–5.
analysis of C. elegans SYG-1 and SYG-2, orthologs 109. Li C, Ruotsalainen V, Tryggvason K, et al. CD2AP is
of the Neph/nephrin cell adhesion module directing expressed with nephrin in developing podocytes and
selective synaptogenesis. PLoS One. 2011;6:e23598. is found widely in mature kidney and elsewhere. Am J
2 Development of Glomerular Circulation and Function 57

Physiol Renal Physiol. 2000;279:F785–92. PubMed: function. Proc Natl Acad Sci U S A. 2003;100(7):
10997929. 4108–13.
110. Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, 120. Moeller MJ, Tenten V. Renal albumin filtration: alter-
Seelow D, N€urnberg G, Garg P, Verma R, Chaib H, native models to the standard physical barriers. Nat
Hoskins BE, Ashraf S, Becker C, Hennies HC, Rev Nephrol. 2013;9(5):266–77.
Goyal M, Wharram BL, Schachter AD, 121. Satchell S. The role of the glomerular endothelium in
Mudumana S, Drummond I, Kerjaschki D, albumin handling. Nat Rev Nephrol. 2013;9(12):
Waldherr R, Dietrich A, Ozaltin F, Bakkaloglu A, 717–25.
Cleper R, Basel-Vanagaite L, Pohl M, Griebel M, 122. Haraldsson B, Nyström J, Deen WM. Properties of
Tsygin AN, Soylu A, M€ uller D, Sorli CS, Bunney the glomerular barrier and mechanisms of proteinuria.
TD, Katan M, Liu J, Attanasio M, O’toole JF, Physiol Rev. 2008;88:451–87.
Hasselbacher K, Mucha B, Otto EA, Airik R, 123. Harvey SJ, Jarad G, Cunningham J, Rops AL, van der
Kispert A, Kelley GG, Smrcka AV, Gudermann T, Vlag J, Berden JH, Moeller MJ, Holzman LB, Bur-
Holzman LB, N€urnberg P, Hildebrandt F. Positional gess RW, Miner JH. Disruption of glomerular base-
cloning uncovers mutations in PLCE1 responsible for ment membrane charge through podocyte-specific
a nephrotic syndrome variant that may be reversible. mutation of agrin does not alter glomerular
Nat Genet. 2006;38:1397–405. permselectivity. Am J Pathol. 2007;171:139–52.
111. Reiser J, Polu KR, Möller CC, Kenlan P, Altintas 124. Goldberg S, Harvey SJ, Cunningham J,
MM, Wei C, Faul C, Herbert S, Villegas I, Avila- Tryggvason K, Miner JH. Glomerular filtration is
Casado C, McGee M, Sugimoto H, Brown D, normal in the absence of both agrin and perlecan-
Kalluri R, Mundel P, Smith PL, Clapham DE, Pollak heparan sulfate from the glomerular basement mem-
MR. TRPC6 is a glomerular slit diaphragm- associ- brane. Nephrol Dial Transplant. 2009;24:2044–51.
ated channel required for normal renal function. Nat 125. van den Hoven MJ, Wijnhoven TJ, Li JP, Zcharia E,
Genet. 2005;37:739–44. Dijkman HB, Wismans RG, Rops AL, Lensen JF, van
112. Timpl R. Structure and biological activity of basement den Heuvel LP, van Kuppevelt TH, Vlodavsky I,
membrane proteins. Eur J Biochem. 1989;180: Berden JH, van der Vlag J. Reduction of anionic
487–502. sites in the glomerular basement membrane by
113. Miner JH. Developmental biology of glomerular heparanase does not lead to proteinuria. Kidney Int.
basement membrane components. Curr Opin Nephrol 2008;73:278–87.
Hypertens. 1998;7:13–9. 126. Axelsson J, Sverrisson K, Rippe A, Fissell W, Rippe
114. St John PL, Wang R, Yin Y, Miner JH, Robert B, B. Reduced diffusion of charge modified, conforma-
Abrahamson DR. Glomerular laminin isoform transi- tionally intact anionic Ficoll relative to neutral Ficoll
tions: errors in metanephric culture are corrected by across the rat glomerular filtration barrier in vivo. Am
grafting. Am J Physiol Renal Physiol. 2001;280(4): J Physiol Renal Physiol. 2011;301:F708–12.
F695–705. 127. Hausmann R, Kuppe C, Egger H, Schweda F,
115. Miner JH, Patton BL, Lentz SI, Gilbert DJ, Snider Knecht V, Elger M, Menzel S, Somers D, Braun G,
WD, Jenkins NA, Copeland NG, Sanes JR. The lam- Fuss A, Uhlig S, Kriz W, Tanner G, Floege J, Moeller
inin alpha chains: expression, developmental transi- MJ. Electrical forces determine glomerular perme-
tions, and chromosomal locations of alpha1-5, ability. J Am Soc Nephrol. 2010;21:2053–8.
identification of heterotrimeric laminins 8-11, and 128. Ryan GB, Karnovsky MJ. Distribution of endogenous
cloning of a novel alpha3 isoform. J Cell Biol. albumin in the rat glomerulus: role of hemodynamic
1997;137:685–701. factors in glomerular barrier function. Kidney Int.
116. Miner JH, Sanes JR. Collagen IV α3, α4, and α5 1976;9:36–45.
chains in rodent basal laminae: sequence, distribution, 129. Bevan HS, Slater SC, Clarke H, et al. Acute laminar
association with laminins, and developmental shear stress reversibly & increases human glomerular
switches. J Cell Biol. 1994;127:879–91. endothelial cell permeability via activation of endo-
117. Poschl E, Schlotzer-Schrehardt U, Brachvogel B, thelial nitric oxide synthase. Am J Physiol Renal
Saito K, Ninomiya Y, Mayer U. Collagen IV is Physiol. 2011;301:F733–42.
essential for basement membrane stability but 130. Friden V, Oveland E, Tenstad O, et al. The glomerular
dispensable for initiation of its assembly during endothelial cell coat is essential for glomerular filtra-
early development. Development. 2004;131: tion. Kidney Int. 2011;79:1322–30.
1619–28. 131. Haraldsson B, Nyström J. The glomerular endothe-
118. Bohrer MP, Baylis C, Humes HD, Glassock RJ, Rob- lium: new insights on function and structure. Curr
ertson CR, Brenner BM. Permselectivity of the glo- Opin Nephrol Hypertens. 2012;21(3):258–63.
merular capillary wall. Facilitated filtration of 132. Oubaha M, Gratton JP. Phosphorylation of endothe-
circulating polycations. J Clin Invest. 1978;61:72–8. lial nitric oxide synthase by atypical PKC zeta con-
PubMed: 618914. tributes to angiopoietin-1-dependent inhibition of
119. Smithies O. Why the kidney glomerulus does not VEGF-induced endothelial permeability in vitro.
clog: a gel permeation/diffusion hypothesis of renal Blood. 2009;114(15):3343–51.
58 A. Tufro and A. Gulati

133. Eklund L, Saharinen P. Angiopoietin signaling in the 148. Bohman SO, Jaremko G, Bohlin AB, Berg U.
vasculature. Exp Cell Res. 2013;319(9):1271–80. Foot process fusion and glomerular filtration rate in
134. Davis B, Dei Cas A, Long DA, White KE, minimal change nephrotic syndrome. Kidney Int.
Hayward A, Ku CH, Woolf AS, Bilous R, Viberti G, 1984;25(4):696–700.
Gnudi L. Podocyte-specific expression of 149. Brenner BM, Troy JL, Daugharty TM. The dynamics
angiopoietin-2 causes proteinuria and apoptosis of of glomerular ultrafiltration in the rat. J Clin Invest.
glomerular endothelia. J Am Soc Nephrol. 2007; 1971;50:1776–80.
18(8):2320–9. 150. Chasis H, Ranges HA, Goldring W, Smith HW. The
135. Khan S, Lakhe-Reddy S, McCarty JH, control of renal blood flow and glomerular filtration in
et al. Mesangial cell integrin alphavbeta8 provides normal man. J Clin Invest. 1938;17:683.
glomerular endothelial cell cytoprotection by seques- 151. Smith HW, Goldring W, Chasis H. The measurement
tering TGF-beta and regulating PECAM-1. Am J of the tubular excretory mass, effective blood flow
Pathol. 2011;178:609–20. and filtration rate in the normal human kidney. J Clin
136. Clement LC, Avila-Casado C, Mace C, Invest. 1938;17:263.
et al. Podocyte-secreted angiopoietin-like-4 mediates 152. Smith HW, Chasis H, Goldring W, Ranges
proteinuria in glucocorticoid-sensitive nephrotic syn- HA. Glomerular dynamics in the normal human kid-
drome. Nat Med. 2011;17:117–22. ney. J Clin Invest. 1940;19:751–64.
137. Huang RL, Teo Z, Chong HC, et al. ANGPTL4 mod- 153. Lafferty HM, Anderson S, Brenner BM. Anemia: a
ulates vascular junction & integrity by integrin sig- potent modulator of renal hemodynamics in models of
naling and disruption of intercellular VE-cadherin progressive renal disease. Am J Kidney Dis. 1991;17
and claudin-5 clusters. Blood. 2011;118:3990–4002. (5 Suppl 1):2–7.
138. El-Banawy HS, Gaber EW, Maharem DA, Matrawy 154. Larsson L, Aperia A, Elinder G. Structural and func-
KA. Angiopoietin-2, & endothelial dysfunction and tional development of the nephron. Acta Paediatr
renal involvement in patients with systemic lupus Scand Suppl. 1983;305:56–60.
erythematosus. J Nephrol. 2011. doi:10.5301/ 155. Paton JB, Fisher DE, Peterson EN, DeLannoy CW,
jn.5000030. The effect of Angiopoeitin-2 and endo- Behrman RE. Cardiac output and organ blood
thelial dysfunction. flows in the baboon fetus. Biol Neonate. 1973;22
139. Pappenheimer JR. Passage of molecules through cap- (1):50–7.
illary walls. Physiol Rev. 1953;33:387–423. 156. Rudolph AM, Heymann MA. Circulatory changes
140. Blouch K, Deen WM, Fauvel JP, Bialek J, Derby G, during growth in the fetal lamb. Circ Res. 1970;26
Myers BD. Molecular configuration and glomerular (3):289–99.
size selectivity in healthy and nephrotic humans. Am 157. Rudolph AM, Heymann MA, Teramo KAW,
J Physiol. 1997;273:F430–7. et al. Studies on the circulation of the previable
141. Deen WM, Bridges CR, Brenner BM, Myers human fetus. Pediatr Res. 1971;5:452.
BD. Heteroporous model of glomerular size selectiv- 158. Rubin MI, Bruck E, Rapoport M. Maturation of renal
ity: application to normal and nephrotic humans. Am function in childhood; clearance studies. J Clin Invest.
J Physiol. 1985;249:F374–89. 1949;28(5 Pt 2):1144–62.
142. Öberg CM, Rippe B. A distributed two-pore model: 159. Calcagno PL, Rubin MI. Renal extraction of para-
theoretical implications and practical application to aminohippurate in infants and children. J Clin Invest.
the glomerular sieving of Ficoll. Am J Physiol Renal 1963;42:1632–9.
Physiol. 2014;306(8):F844–54. 160. Gruskin AB, Edelmann Jr CM, Yuan S. Maturational
143. Deen WM, Bohrer MP, Brenner BM. Macromolecule changes in renal blood flow in piglets. Pediatr Res.
transport across glomerular capillaries: application of 1970;4(1):7–13.
pore theory. Kidney Int. 1979;16(3):353–65. 161. Barnett HL, Hare K, McNamara H, Hare
144. Katz MA, Schaeffer Jr RC, Gratrix M, Mucha D, R. Measurement of glomerular filtration rate in pre-
Carbajal J. The glomerular barrier fits a two-pore- mature infants. J Clin Invest. 1948;27(6):691–9.
and-fiber-matrix model: derivation and physiologic 162. Spitzer A. Edelman CH Jr. Maturational changes in
test. Microvasc Res. 1999;57(3):227–43. pressure gradients for glomerular filtration. Am J
145. Drummond MC, Kristal B, Myers BD, Deen Physiol. 1971;221:1431–1435.
WM. Structural basis for reduced glomerular filtration 163. Horster M, Valtin H. Postnatal development of renal
capacity in nephrotic humans. J Clin Invest. function: micropuncture and clearance studies in the
1994;94:1187–95. dog. J Clin Invest. 1971;50(4):779–95.
146. Drummond MC, Deen WM. Structural determinants 164. Friis C. Postnatal development of renal function in
of glomerular hydraulic permeability. Am J Physiol piglets: glomerular filtration rate, clearance of PAH
Renal Fluid Electrolyte Physiol. 1994;266:F1–12. and PAH extraction. Biol Neonate. 1979;35(3–4):
147. Kedem O, Katchalsky A. Thermodynamic analysis of 180–7.
the permeability of biological membranes to 165. Barnett HL, Hare WK, et al. Influence of postnatal age
non-electrolytes. Biochim Biophys Acta. on kidney function of premature infants. Proc Soc
1958;27:229. Exp Biol Med. 1948;69(1):55–7.
2 Development of Glomerular Circulation and Function 59

166. Arant Jr BS. Developmental patterns of renal func- 183. Shemesh O, Golbetz H, Kriss JP, Myers
tional maturation compared in the human neonate. BD. Limitations of creatinine as a filtration marker in
J Pediatr. 1978;92(5):705–12. glomerulopathic patients. Kidney Int. 1985;28:830–8.
167. Abitbol CL, Seeherunvong W, Galarza MG, 184. Schwartz GJ, Munoz A, Schneider MF, et al. New
Katsoufis C, Francoeur D, Defreitas M, Edwards- equations to estimate GFR in children with CKD.
Richards A, Master Sankar Raj V, Chandar J, Duara S, J Am Soc Nephrol. 2009;20:629–37.
Yasin S, Zilleruelo G. Neonatal kidney size and function 185. Dharnidharka VR, Kwon C, Stevens G. Serum
in preterm infants: what is a true estimate of glomerular cystatin C is superior to serum creatinine as a marker
filtration rate? J Pediatr. 2014;164(5):1026–31. of kidney function: a meta-analysis. Am J Kidney
168. Carl Ludwig original reference: Ludwig CFW. Dis. 2002;40(2):221–6.
Beitraege zur Lehre vom Mechanismus der 186. Roos JF, Doust J, Tett SE, Kirkpatrick
Harnsekretion. Marburg: N.G. Elwert; 1843. CM. Diagnostic accuracy of cystatin C compared to
169. Heidenhain RP. Absonderungsvorgaenge. Sechster serum creatinine for the estimation of renal dysfunc-
Abschnitt. Die Harnabsonderung (Viertes Capitel. tion in adults and children–a meta-analysis. Clin
Die Absonderung der festen Harnbestandteile). In: Biochem. 2007;40(5–6):383–91.
Leipzig HL, editor. Handbuch d Physiol Fuenfter 187. Filler G, Yasin A, Medeiros M. Methods of assessing
Teil. Germany: Vogel; 1883. p. 341–43. renal function. Pediatr Nephrol. 2014;29(2):183–92.
170. Wearn JT, Richards AN. From: observations on the 188. Filler G, Kusserow C, Lopes L, Kobrzyński M. Beta-trace
composition of glomerular urine, with particular ref- protein as a marker of GFR–history, indications, and
erence to the problem of reabsorption in the renal future research. Clin Biochem. 2014;47(13–14):1188–94.
tubules. Am J Physiol. 1924;71:209–27. 189. Lorenz JN, Gruenstein E. A simple, nonradioactive
171. Starling EH. The glomerular functions of the kidney. method for evaluating single-nephron filtration rate using
J Physiol Lond. 1899;24:317–30. FITC-inulin. Am J Physiol. 1999;276(1 Pt 2):F172–7.
172. Wies CH, Peters JP. The osmotic pressure of proteins 190. Yu W, Sandoval RM, Molitoris BA. Quantitative
in whole serum. J Clin Invest. 1937;16:93. intravital microscopy using a generalized polarity
173. Levinsky NG, Berliner RW. Changes in composition concept for kidney studies. Am J Physiol Cell
of the ureter and bladder at low urine flow. Am J Physiol. 2005;289:C1197–208.
Physiol. 1959;196:549–53. 191. Wang E, Meier DJ, Sandoval RM, Von Hendy-
174. Levinsky NG, Lieberthal W. Clearance techniques. Willson VE, Presser BM, Bunch RM, Alloosh M,
In: Windhager E. editor. Handbook of physiology. Sturek MS, Schwartz GJ, Molitoris BA. A portable
Renal physiology. New York: Oxford University fiberoptic ratiometric fluorescence analyzer provides
Press; 1992, sect. 8, pp. 227–47. rapid point-of-care determination of glomerular filtra-
175. Shannon JA, Smith HW. The excretion of inulin, tion rate in large animals. Kidney Int. 2012;81:112–7.
xylose and urea by normal and phlorinized man. 192. Yu W, Sandoval RM, Molitoris BA. Rapid determi-
J Clin Invest. 1935;112:405–13. nation of renal filtration fraction using an optical
176. Heiskanen T, Weber T, Grasbeck R. Determination of ratiometric imaging approach. Am J Physiol Renal
I131 hippuric acid renal clearances using single- Physiol. 2007;292:F1837–80.
injection techniques. Scand J Clin Lab Invest. 193. Quaggin SE. Transcriptional regulation of podocyte
1968;21:211–5. specification and differentiation. Microsc Res Tech.
177. Smith HW. The kidney- structure and function in health 2002;57(4):208–11.
and disease. New York: Oxford University Press; 1951. 194. Abrahamson DR. Role of the podocyte (and glomer-
178. Earle Jr DP, Berliner RW. A simplified clinical procedure ular endothelium) in building the GBM. Semin
for measurement of glomerular filtration rate and renal Nephrol. 2012;32(4):342–9.
plasma flow. Proc Soc Exp Biol Med. 1946;62(2):262–4. 195. Patrakka J, Tryggvason K. Molecular make-up of the
179. Orlando R, Floreani M, Padrini R, Palatini glomerular filtration barrier. Biochem Biophys Res
P. Determination of inulin clearance by bolus intrave- Commun. 2010;396(1):164–9.
nous injection in healthy subjects and ascitic patients: 196. Aperia A, Herin P. Development of glomerular perfu-
equivalence of systemic and renal clearances as glo- sion rate and nephron filtration rate in rats 17 to
merular filtration markers. Br J Clin Pharmacol. 20 days old. Am J Physiol. 1975;228:1319.
1998;46(6):605–9. 197. Aperia A, Broberger O, Thodenius K,
180. Brismar J, Jacobsson BF, Jorulf H. Miscellaneous et al. Development of renal control of salt and fluid
adverse effects of low-versus high-osmolality contrast homeostasis during the first year of life. Acta Paediatr
media: a study revised. Radiology. 1991;179(1):19–22. Scand. 1975;64:393.
181. Berglund F. Renal clearance of inulin, polyfructosan- 198. Chevalier RL. Developmental renal physiology of
S and a polyethy-lene glycol (PE6 1000) in the rat. the low birth weight preterm newborn. J Urol.
Acta Physiol Scand. 1965;64:238–44. 1996;156(2 Pt 2):714–9.
182. Bing J, Effersoe P. Comparative tests of the 199. Stonestreet BS, Oh W. Plasma creatinine levels in
thiosulphate and creatinine clearances in rabbits and low-birth-weight infants during the first three months
cats. Acta Physiol Scand. 1948;15:231–6. of life. Pediatrics. 1978;61:788.
Renal Tubular Development
3
Michel Baum

Contents Developmental Features of Proximal


Tubule Water Transport . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
Organization of the Nephron . . . . . . . . . . . . . . . . . . . . . . . 62
Developmental Transport in the Thick
Principals of Membrane Transport . . . . . . . . . . . . . . . . 64 Ascending Limb . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
Maturation of Na+/K+-ATPase Along Developmental Transport in the Distal
the Nephron . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 65 Convoluted Tubule . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
Proximal Tubule Transport . . . . . . . . . . . . . . . . . . . . . . . . . 66 Development of Urinary Concentration
Developmental Glucose Transport . . . . . . . . . . . . . . . . . 67 and Dilution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80

Transport Amino Acid: Developmental Developmental Aspects of Distal Tubule


Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69 Acidification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82

Development of Organic Acid Transport . . . . . . . . . . 70 Developmental Features of Cortical Collecting


Tubule Sodium Transport . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
Development of Phosphate Transport . . . . . . . . . . . . . . 70
Developmental Aspects of Tubular Potassium
Development of Proximal Tubular Transport . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Acidification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
Development of Proximal Tubule NaCl
Transport . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75

M. Baum (*)
Departments of Pediatrics and Internal Medicine,
University of Texas Southwestern Medical Center at
Dallas, Dallas, TX, USA
e-mail: Michel.Baum@UTSouthwestern.edu

# Springer-Verlag Berlin Heidelberg 2016 61


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_76
62 M. Baum

Organization of the Nephron a constant composition of the extracellular fluid


volume, the mammalian kidney has evolved into a
The nephron is faced with the enormous task of highly specialized organ with thousands of units
maintaining a constant composition and volume called nephrons. Each human kidney has approx-
of the extracellular fluid. The amount of electro- imately one million nephrons. Each nephron is a
lytes and water ingested and absorbed must be tube consisting of epithelial cells and is divided
eliminated, and the waste products from metabo- into 12 specialized segments as shown in Fig. 1.
lism must also be excreted. This challenge is all The epithelial cells allow for the vectorial trans-
the more complex as our dietary intake is quite port of solutes. The proximal tubule is responsible
variable from day to day. Despite this variable for the bulk reclamation of solutes and water and
intake, there is virtually no change in the volume for the secretion of organic cations and anions.
or composition of the extracellular fluid volume Approximately two-thirds of the glomerular fil-
from day to day. trate is reabsorbed by the proximal tubule in an
There are two possible ways that our kidney isotonic fashion. Virtually all of the organic sol-
could balance ingestion and excretion. The kidney utes, as well as the majority of bicarbonate, phos-
could be a secretory organ where all the excess phate, and chloride, are reabsorbed in this
solutes and water ingested would be excreted by segment. The proximal tubule is divided into S1,
tubular secretion. This would be very inefficient S2, and S3 segments based on the rates of transport
and require an enormous amount of energy. In of some solutes and morphological changes that
addition, in times of a disturbance in the extracel- occur down the proximal tubule. The proximal
lular fluid volume such as a high salt intake or tubule transitions into the thin descending limb
volume loss from diarrhea, the regulatory systems which can be of variable length and then makes a
necessary to maintain a constant extracellular hairpin turn into the thin ascending limb which
fluid volume and composition while excreting aids in the generation of a concentrated urine. The
waste products would be very complex. On the length of the thin ascending and descending limb
other hand, the kidney could filter an enormous is variable among species with dessert rodents
quantity of extracellular fluid, which would be having very long thin limbs as they need to con-
very efficient in removing waste products, and serve water and excrete very concentrated urine.
reclaim the desired salt, organic solutes, and The length of the thin ascending and descending
water. With volume depletion, a greater fraction limbs increase as one goes from the superficial
of salt and water could be reabsorbed, and when cortex down to the medulla. The thin descending
one has a dietary indiscretion such as the salt limb expresses aquaporin 1, a water channel, on
intake from a pepperoni pizza, there could be a the apical and basolateral membranes making the
reduction in sodium reabsorption leading to an thin descending limb very permeable to water
increase in excretion to bring us back into balance. [1]. This segment does not actively transport sol-
The mammalian kidney actually uses both mech- utes and has a very low solute permeability. This
anisms to perform its job which is necessary for results in a concentrated fluid in the medulla with
our survival on land. The adult kidney filters a very high sodium chloride content in the luminal
~150 l of isotonic fluid a day and reclaims most fluid providing a passive driving force for sodium
of it, leaving the waste produces to be excreted in chloride diffusion in the thin ascending limb. The
about 1.5 l of urine. In addition, there are secretory thin ascending limb is impermeable to water but
processes for solutes such as organic anions and has a high permeability to NaCl [2]. The chloride
cations in the proximal tubule and secretory channel (CLC-K1) in the thin ascending limb is
mechanisms to excrete the excess acid generated developmentally regulated [3]. There is no
from metabolism in the distal nephron which aid expression in the fetus and in the first few days
in maintaining homeostasis. of life until the end of the first week in rats. There
To accomplish the remarkable task of reclama- is a correlation between CLC-K1 and urinary
tion of the necessary solutes and water to maintain osmolality suggesting an important role of this
3 Renal Tubular Development 63

DCT
7%
S1PCT
DCT
CT

PT
TAL
60%
30% CCT

S2PCT

CTAL

S3PCT OMCD
CCD
1-3%
MTAL

Thin DL IMCD
Thin AL

1%

Fig. 1 This cartoon depicts the nephron with its 12 initial cortical collecting tubule. The collecting duct is
segments. Shown in the figure are the nephron segments. made up of the cortical collecting tubule (CCT) and outer
S1, S2, and S3 PCT depict the three segments of the prox- medullary (OMCD) and inner medullary collecting duct
imal tubule. The loop of Henle consists of the thin (IMCD). Shown in yellow is the percentage of sodium
descending limb (thin DL) and thin ascending limb (thin reabsorbed by the proximal tubule (PT), thick ascending
AL) and the medullary (MTAL) and cortical thick ascend- limb (TAL), distal convoluted tubule (DCT), and cortical
ing limb (CTAL). The distal convoluted tubule is com- collecting duct (CCD). One percent of the filtered sodium
prised of the (DCT), connecting tubule (CT) and the is excreted

channel in generating a hypertonic medulla The thick ascending limb is the segment
[3]. Diffusion of NaCl causes a high interstitial responsible for ~30 % of sodium chloride trans-
osmolality. This loop structure along with the port and has a vital role in generating a concen-
thick ascending limb generates the countercurrent trated medulla. Apical sodium chloride absorption
multiplication system that results in a medullary is mediated by the bumetanide-sensitive
osmolality far greater than that of blood [4, 5]. The cotransporter. One of the unique features of this
importance of passive properties of the thin limbs segment is the fact that this segment is imperme-
in this countercurrent system is exemplified in able to water and thus the fluid leaving this seg-
mice which are deficient in aquaporin 1 [6]. The ment is hypotonic to blood. In addition, this
urine osmolality of aquaporin 1-deficient mice is segment has a very high paracellular permeability
greater than plasma but far less than control mice to cations and is responsible for much of calcium
that have normal aquaporin 1 expression [6]. In and magnesium reabsorption. The distal convo-
addition, aquaporin 1 knockout mice cannot luted tubule is responsible for ~5–10 % of NaCl
increase their urine osmolality in response to transport. NaCl transport in this segment is medi-
water deprivation unlike control mice. ated by the thiazide-sensitive cotransporter.
64 M. Baum

Active transcellular calcium and magnesium


transport also occurs in this segment.
The rest of the distal tubule is separated into the
connecting tubule and the cortical, outer, and Na+
Na+
inner medullary collecting tubule. These seg- 3HCO3−
ments are responsible for sodium absorption, H+
potassium secretion, final urinary acidification, Na+
ATP
and water absorption, the latter mediated by vaso-
pressin, which causes the insertion of aquaporin Glucose 3 Na+
OH−
2 water channels into the apical membrane. While 2 K+
the fraction of salt transport and renal acidification Cl− ADP+Pi
by the collecting tubule (or collecting duct) is a
small fraction of the capabilities of some of the
upstream nephron segments of that in other neph-
Cl−
ron segments, the collecting tubule is responsible
for the final modulation of the tubular fluid. Thus,
the final composition of urine and significant reg- Fig. 2 A proximal tubule renal cell which shows the Na+/
ulation of transport occur in these last segments. K+-ATPase on the basolateral membrane, an example of
primary active transport. Na+/K+-ATPase decreases the
intracellular sodium to about 10 mEq/l and increases the
intracellular potassium to approximately 140 mEq/l. The
Principals of Membrane Transport pump is electrogenic with potential of about 60 mV when
one compares the cell to the extracellular fluid. The sodium
The cells along the nephron are quite different in gradient provides the driving force for the apical Na+/H+
exchanger, and both the sodium gradient and the potential
the various nephron segments, which will be difference provide the driving force for the apical sodium
discussed in the subsequent sections. The cells in glucose transporter. The secretion of protons via the Na+/
each nephron segment are poised for vectorial H+ exchanger results in the driving force for the Cl/OH
transport. The apical and basolateral membranes exchanger which is an example of tertiary active transport.
Chloride is shown transversing the paracellular pathway.
are, by and large, a lipid bilayer which would be Bicarbonate is exiting the basolateral membrane via a
impermeable to water and solutes if there were not sodium bicarbonate cotransporter
specific proteins to facilitate transport across the
apical and basolateral membranes. In addition,
many transporters are regulated to adjust their example of primary active transport. This pump
rate of transport to meet the physiologic changes is vital to the generation of the low intracellular
in volume status or concentration of solutes in the sodium and high intracellular potassium concen-
extracellular milieux. tration as well as the negative intracellular poten-
The reabsorption of solutes along the nephron tial difference across the apical and basolateral
is characterized by active and passive transport membranes. Both the low intracellular sodium
processes. A typical cell is shown in Fig. 2. It and this potential difference can provide a
should be appreciated that if all active transport driving force for secondary active transport. For
was inhibited along the nephron, we would example, in Fig. 2, the reabsorption of glucose via
excrete urine with the composition and volume a sodium-dependent transporter utilizes both
of the glomerular ultrafiltrate. Passive the sodium gradient and the relative negative cell
transepithelial transport is, by and large, the result potential to bring glucose in the cell. The Na+/H+
of gradients generated by active transport. Most exchanger on this cell is electroneutral and
active transport is the result of the basolateral utilizes the sodium gradient to secrete protons
Na+/K+-ATPase. This transporter pumps three and reabsorb sodium. The sodium glucose and
sodiums out of the cell in exchange for two potas- the Na+/H+ exchanger are secondary active trans-
sium ions. The pump utilizes ATP and it is an port processes dependent on the basolateral
3 Renal Tubular Development 65

Na+/K+-ATPase. The secretion of protons will solute is entrained in fluid and hits a membrane or
cause the luminal pH to drop providing a favor- tight junction, it could pass through it and be
able driving force for the Cl/OH exchanger, an transported or bounce off and not be transported.
example of tertiary active transport. Thus, in sec- Direct measurements of solute drag in the proxi-
ondary and tertiary active transport, the trans- mal tubule of neonates and adults have shown that
porters do not directly utilize ATP directly; it contributes a negligible fraction to transport
however, inhibition of the ATP dependent [12–15].
Na+/K+-ATPase would bring these transport pro-
cesses to a halt.
In addition to active transport, a substantive Maturation of Na+/K+-ATPase Along
amount of passive transport occurs between cells the Nephron
across the tight junction. Active transport along
the nephron will generate ion and solute gradients The Na+/K+-ATPase is located on the basolateral
between the lumen and peritubular fluid. membrane of most tubules in the kidney [16]. It is
Depending on the permeability properties of the a heterodimer composed of an α- and β-subunit.
tight junction, passive absorption or secretion can There are four different α-subunits and three beta
occur. In the cell depicted in Fig. 2, there is pas- subunits on mammalian cells which have different
sive chloride transport across the paracellular functional properties [17]. There is also evidence
pathway. It has become apparent that the charac- for a small γ-subunit that is not required for
teristics of the tight junction vary along the neph- Na+/K+-ATPase activity [17, 18]. The γ-subunit
ron. The tight junction creates the primary binds to the α-subunit, stabilizes the enzyme, and
permeability barrier to diffusion of solutes across plays a regulatory role in enzyme activity [17, 18].
the paracellular pathway. Occludin and claudin The adult kidney expresses the α1 and β1 isoforms
proteins are localized to junctional fibrils and are of the Na+/K+-ATPase [19, 20]. The α-subunit is
transmembrane components of tight junctions the catalytic subunit and has the cation, ATP, and
[7–9]. These tight junction fibrils or strands are a ouabain binding sites [17]. The β-subunit is the
major factor determining the permeability proper- regulatory subunit and is essential for the function
ties of the paracellular pathway [7, 10, 11]. The of the enzyme [17, 21]. Several hormones that
claudin family of tight junction proteins now regulate sodium transport along the nephron act,
numbers 24. Occludin has a ubiquitous distribu- at least in part, by regulating the Na+/K+-ATPase
tion and is not responsible for the differential activity [17, 22–34]. Dopamine and atrial
permeability properties in the various nephron natriuretic factor inhibit, while alpha agonists
segments. The claudin isoforms present at the and angiotensin II stimulate, Na+/K+-ATPase
tight junction of various epithelia determine the activity [16]. Some hormones such as dopamine
resistance and the permeability properties of the have a direct effect to regulate the Na+/K+-ATPase
epithelia [7, 10, 11]. The distribution of claudin and are independent of an effect on transporters on
isoforms varies along the nephron and is respon- the apical membrane [22].
sible for the unique permeability properties of The Na+/K+-ATPase is responsible for lower-
each nephron segment. ing the intracellular sodium concentration and
The final form of passive transport is called establishing the negative cell potential difference.
solvent drag. Solvent drag has been postulated to The Na+-K+-ATPase provides the driving force
be responsible for a small fraction of transport in for sodium transport across the nephron. There is
the proximal tubule. The reabsorption of solutes a direct relationship between sodium transport
could result in water movement that could entrain and Na+/K+-ATPase activity factored per millime-
or carry solutes with it. For this to occur the solute ter of tubule along the nephron [35]. Indeed, an
would have to have a low reflection coefficient or increase in intracellular sodium concentration
high sieving coefficient (sieving coefficient = increases Na+/K+-ATPase activity [36]. Neonates
1refection coefficient). In other words when a have a lower renal Na+/K+-ATPase activity than
66 M. Baum

Fig. 3 Na+/K+-ATPase
activity is shown in nephron 25
segments of neonates and NEONATAL
adults. As is demonstrated,

Na-K-ATPase activity (mol Pi /kg dry weight/h)


there is a maturational MATURE
increase in Na+/K+-ATPase
20
activity in every nephron
segment [39]

15

10

10 8 7 7 9 10 1111 9 7 7 14
0
PCTSN PCTJM CTAL MTAL CCD MCD

do adults [25, 37–41]. As will be discussed, there


is a developmental increase in sodium transport in Proximal Tubule Transport
each nephron segment, which is paralleled by an
increase in Na+/K+-ATPase activity as shown in Proximal tubule transport is characterized by a
Fig. 3. phenomenon called threshold, which is depicted
The parallel maturational increase in sodium on Fig. 4. It is the threshold that keeps our serum
transport with Na+/K+-ATPase activity suggests bicarbonate at 25 mEq/l. If we were to ingest
that the maturational increase in apical sodium bicarbonate and try to raise our serum bicarbonate
transport may contribute to the postnatal increase level, we would have a bicarbonaturia and our
in Na+/K+-ATPase [35, 40]. In cell culture serum levels would return to 25 mEq/l as long as
studies an increase in intracellular sodium caused we were euvolemic. Our serum glucose is set by
a stimulation in Na+/K+-ATPase activity [42, 43] other factors well below the threshold level. As
as well as an increase in the α-subunit mRNA and shown in Fig. 4, if we increased the serum glucose
membrane pump density [44]. In addition to level, we would reabsorb more glucose until the
in vitro studies, there is evidence that a chronic load of glucose delivered to the proximal tubule
increase in Na+/H+ exchanger activity induced by exceeded its ability to reabsorb glucose and we
metabolic acidosis increased Na+/K+-ATPase would have glucosuria.
activity, an effect that was blocked by In the adult kidney there is a parallel change in
coadministration of the Na+/H+ exchange inhibi- proximal tubule transport with alterations in glo-
tor, amiloride [45]. Finally, there is a postnatal merular filtration rate. This phenomenon has been
increase in both serum thyroid hormone and designated glomerular tubular balance. If the rate
glucocorticoid levels with age [46–49]. of proximal tubular transport was fixed, an
Both glucocorticoids and thyroid hormone have increase in glomerular filtration rate would
been shown to increase Na+/K+-ATPase activity swamp the distal nephron with solutes and water,
[28, 38, 50–53]. and there would be a huge natriuresis and diuresis.
3 Renal Tubular Development 67

Transport Maximum
Reabsorbed Solute

Excreted Solute

Fig. 4 This figure depicts the concept of renal threshold. increases. At some point, the renal tubular absorption
As the delivered load increases to the tubule either by an reaches a maximum, called the threshold for that solute,
increase in the serum concentration or an increase in glo- and any further increase in the filtered load is excreted
merular filtration rate, the amount of solute absorbed

A similar phenomenon must occur during postna- transported by this segment, would increase over
tal development where there is a developmental twofold. However, this does not happen because
increase in glomerular filtration rate. If there was as the magnesium concentration rises above that
not a parallel increase in proximal tubule transport in the peritubular capillaries, magnesium is pas-
with the maturational increase in glomerular sively reabsorbed across the paracellular pathway
filtration rate, the neonate would die of dehydra- down the concentration gradient generated by the
tion when the glomerular filtration rate increased reabsorption of other solutes.
after birth.
While during postnatal development there is a
concomitant increase in tubular transport to Developmental Glucose Transport
accommodate or balance the increase in glomeru-
lar filtration rate [54–56], glomerular tubular bal- Glucose is reabsorbed solely by the proximal
ance is not present in the fetus [57]. Renal tubule. Physiologic studies have demonstrated
development is characterized by centrifugal mat- that the S1 proximal tubule reabsorbs glucose by
uration. The surface nephrons are relatively a high-capacity low-affinity transporter, while in
immature compared to the juxtamedullary neph- the late proximal tubule (S3), glucose transport is
rons. These immature nephrons with short proxi- via a low-capacity high-affinity transporter
mal tubules have glomerular tubular imbalance [59]. Similar axial heterogeneity of glucose trans-
[57]. This is clinically relevant as neonates born port kinetics was validated using cortical brush
before 34 weeks of gestation can have glucosuria border membrane vesicles to measure apical
and very premature neonates can have significant membrane transport and outer medullary brush
renal salt wasting [58]. border membrane vesicles which contain vesicles
The proximal tubule reabsorbs 60 % of the from the S3 segment [60]. The high-capacity
glomerular filtrate in an isoosmotic fashion. Due low-affinity sodium-dependent glucose trans-
to the fact that the proximal tubule has a relatively porter on the apical membrane designated is
high permeability to many ions, even solutes SGLT-2 [61]. SGLT-2 removes the bulk of the
which are not actively transported by this segment glucose from the glomerular ultrafiltrate. The
may get absorbed by diffusion across the low-capacity high-affinity transporter is desig-
paracellular pathway. For example, since the nated SGLT-1 [62, 63]. The glucose that is
proximal tubule reabsorbs over half of the glo- transported by the tubule exits across the
merular filtrate, the luminal concentration of a basolateral membrane by facilitative diffusion.
solute such as magnesium, which is not actively As shown in Fig. 5, SGLT-2 transports one
68 M. Baum

Fig. 5 Diagram of glucose Early Proximal Tubule


transport in the proximal
tubule. The early proximal
tubule has SGLT-2 on the
apical membrane which is a
high-capacity, low-affinity SGLT-2
ATP
transporter, while in the late
3 Na+
proximal tubule, the Na+
low-capacity high-affinity 2 K+
Glucose
transporter, SGLT1, is on ADP+Pi
the apical membrane.
Glucose exits the cell across Glucose
the basolateral membrane
by passive diffusion

Late Proximal Tubule

SGLT-1
ATP
2Na + 3 Na+

Glucose 2 K+
ADP+Pi
Glucose

sodium with one glucose molecule while SGLT-1 pathway. Whether sodium is recycled or chloride is
transports two sodiums with each glucose mole- reabsorbed is dependent on the relative sodium/
cule. A genetic mutation in SGLT-1 resulting in chloride permeability of the paracellular pathway.
loss of function causes glucose-galactose malab- Numerous studies using various techniques
sorption as this transporter is also present in the and animal species have shown that the fetus and
intestine [64–67]. Some patients with familial neonate transport glucose at a slower rate than the
glucosuria, a benign condition, have a mutation adult [57, 70–73]. These studies are of clinical
in SGLT-2 [68, 69]. This axial arrangement of relevance as premature neonates can have
glucose transporters, where there is a high- glucosuria [58, 74, 75]. Despite the fact that the
capacity, low-affinity transporter followed by a glomerular filtration rate of a premature neonate is
low-capacity, high-affinity glucose transporter, about 1 % of that of the adult and thus the filtered
results in reabsorption of virtually all of the fil- load delivered to the neonatal proximal tubule is
tered glucose. also about 1% of that of the adult, the filtered load
Sodium-dependent glucose reabsorption results exceeds the reabsorptive capacity for glucose
in a positive charge entering the proximal tubule transport in the premature neonate. Thus, there is
cell. This charge results in a lumen-negative a time during development where glomerular
transepithelial potential difference. This negative tubular balance is not present. Kinetic studies
potential provides a driving force for the absorption demonstrate that the Km for the glucose trans-
of an anion or the back diffusion of a cation porter is comparable between the fetus and adult;
(sodium) across the paracellular pathway. Thus however, the Vmax is much lower in the neonate
glucose transport can result in a net absorption of [76]. This indicates that the developmental
sodium chloride with sodium moving into the cell increase in transport is not due to a glucose trans-
with glucose and chloride across the paracellular porter isoform change but due to a maturational
3 Renal Tubular Development 69

increase in the number of glucose transporters on transported for each of these negatively charged
the apical membrane. amino acids [84, 85]. Brush border membrane
vesicle studies have shown that there are at least
two apical transporters for glutamate: one with a
Transport Amino Acid: Developmental high substrate affinity and one with a low affinity
Features [86]. The high-affinity transporter has been cloned
and designated SLC1A1 (EAAC1) [87]. SLC1A1
All amino acid transport occurs in the proximal is expressed on the apical membrane of the prox-
tubule. Unlike most cells which have amino acid imal tubule [88]. Eaac-1 knockout mice have a
transporters to provide substrates for protein syn- dicarboxylic aciduria proving the importance of
thesis, the proximal tubule mediates the vectorial this transporter in acidic amino acid transport
transport of amino acids from the glomerular fil- [89]. Recently, patients with SLC1A1 mutations
trate to the blood. The basic principal for amino have been found to have dicarboxylic aminoacid-
acid transport is similar to glucose. The uptake of uria and mental retardation likely due to the
amino acids is sodium dependent and electrogenic importance of SLC1A1 for glutamate transport
with the basolateral exit mediated by facilitated in the brain [90]. The basolateral transport of
passive diffusion. While there are 20 amino acids glutamate is via a sodium-dependent
that are utilized in the synthesis of proteins, there cotransporter indicating that the intracellular glu-
are not 20 different amino acid transporters on the tamate levels must be very high in the proximal
apical and basolateral membranes. Since some tubule to provide an adequate driving force for
amino acids are similar in structure and/or charge, sodium exit across the basolateral membrane
there is promiscuity among the classes of trans- [91]. In addition there is a sodium-independent
porters. We will briefly discuss the three major aspartate/glutamate transporter which is localized
classes of amino acid transporters. to the basolateral membrane designated
The neutral amino acids include leucine, AGT1 [92].
valine, isoleucine, methionine, phenylalanine, The lysine and arginine are basic amino acids
tyrosine, cysteine, glutamine, alanine, glycine, which utilize the same amino acid transporter as
serine, histidine, tryptophan, and proline. Trans- cystine. There are a number of basic amino acid
port of these amino acids is electrogenic with one transporters [93]. rBAT is a cystine/dibasic amino
sodium ion being transported with the amino acid acid transporter expressed along the proximal
across the apical membrane. B0AT1 (SLC6A19) tubule but predominantly in the S3 segment [94,
is expressed on the proximal tubule [77–79] and 95]. rBAT protein is undetectable in the fetal
transports all neutral amino acids, though there is kidney and is expressed at very low levels even
greater affinity for valine, leucine, isoleucine, and after weaning [96]. Mutations in SLC3A1, the
methionine [80]. Mutation of SLC6A19 causes gene encoding rBAT, result in type I cystinuria
Hartnup disease which is an autosomal recessive [97–100]. B0+AT is also a cystine transporter/
disorder of variable severity characterized by a dibasic amino acid transporter [101, 102]. Unlike
pellagra-like rash, cerebellar ataxia, and psycho- rBAT, B0+AT is predominantly expressed in the
logical and neurological disturbances [77, 79, early proximal convoluted tubule but it overlaps
81]. The symptoms of Hartnup disease are likely with the expression of rBAT [95, 103]. While both
the result of impaired intestinal and renal transport rBAT and B0+AT can function as cystine/dibasic
of tryptophan [82]. There are other neutral amino amino acid transporters, they likely function
acid transporters but their transport properties in vivo as a heterodimer [104].
have been less well characterized [81, 83]. Neonates have a generalized aminoaciduria
The acidic amino acids are aspartate and glu- which is more pronounced in premature neonates
tamate. They are transported across the apical [105–107]. While there have been numerous stud-
membrane of the proximal tubule in an electro- ies examining amino acid transport using a num-
genic fashion where two sodium ions are ber of species, most have utilized kidney slices or
70 M. Baum

suspensions of renal tubules. These studies are which has been localized to the apical membrane
complicated by the fact that one is looking simul- of the proximal tubule [119].
taneously at cellular uptake, often from a col- Para-aminohippurate is almost totally removed
lapsed tubule, metabolism of the amino acid, and from the blood with one pass through the kidney
basolateral exit. Examination of glycine transport and is used as a measure of renal blood flow.
in tubule suspensions and kidney slices of neo- Para-aminohippurate has been used to assess the
nates and adults have provided disparate results. maturation of renal blood flow in humans and to
Glycine uptake has been shown to be the same in determine the maturational changes in organic
neonates and adults in some studies [108, 109] anion transport. Studies in humans have shown
and lower in neonates than adults in others [110, that there is a maturational increase in para-
111]. The few studies looking at the brush border aminohippurate secretion with adult values being
membrane during development, a more direct way attained at about 2 years of age [120, 121]. Para-
of studying uptake transport across the apical aminohippurate secretion is less in premature than
membrane, have shown lower rates of transport term neonates [122].
in the neonate compared to the adult [112, 113]. There are a number of factors that could con-
tribute to the maturational increase in organic
anion secretion. Since organic anion secretion
requires an organic anion transporter (OAT), a
Development of Organic Acid sodium-dependent organic acid cotransporter,
Transport and the Na+/K+-ATPase to mediate intracellular
transport of the organic acid and an apical secre-
In addition to filtration and reclamation, the kid- tory mechanism to secrete the organic acid, a
ney has the ability to secrete some substances paucity in any of these transporters in neonates
through organic anion transport. There are five (compared to the adult) could be the rate-limiting
organic anion transporters (OATS) on the step. OAT1 and OAT2 have been shown to be
basolateral membrane of the proximal tubule present in the late gestation fetus, and mRNA
[114–118]. Different OATS have different sub- and protein expression increases during postnatal
strate specificities. Organic acids transported development [123, 124].
by OATS include prostaglandins, uric acid, One of the unique features of induction of
nonsteroidal anti-inflammatory drugs, β-lactam organic anion transport is that it can be induced
antibiotics, antiviral medications, para- prematurely during renal development by itself or
aminohippurate, probenecid, uric acid, another organic anion [125–128]. This is not true
bumetanide, salicylates, methotrexate, and many of adult animals where organic anions do not
others [115–117]. Many of these substances are cause a stimulation in transport [125]. In vitro
protein bound in the blood which limits their microperfusion studies have demonstrated that
excretion by glomerular filtration. The basolateral there was an intrinsic increase in the rate of trans-
uptake of OATS is an example of tertiary active port with postnatal age in rabbits and that
transport [115, 116]. OATS take up organic anions pretreatment with penicillin increased the rate of
into the cell predominantly in exchange for para-aminohippurate secretion in vitro [128]. The
α-ketoglutarate. The α-ketoglutarate that is mechanism of this induction of organic anion
exchanged for the organic acid enters the cell via transport by organic acids is unclear.
an α-ketoglutarate transporter. The energy for this
whole process is the basolateral Na+/K+-ATPase.
The organic acid transported inside the cell must Development of Phosphate Transport
exit across the apical membrane to enter the pri-
mordial urine. The mechanism for this is less well The adult ingests approximately 1–1.5 g of phos-
understood but includes members of the phorus a day and of that 80 % is absorbed. The
multidrug resistance protein family [115–117], adult must be in neutral phosphorus balance so
3 Renal Tubular Development 71

that the amount of phosphorus absorbed daily


must equal the amount excreted. The phosphorus
in the human body is predominantly in the form of
phosphate. At a pH of 7.4, there is a 4:1 ratio of 3Na+
HPO42/ H2PO41. The kidney maintains phos- IIa ATP
phate balance by the ability to regulate phosphate HPO42−
3 Na+
transport which predominantly occurs in the prox-
imal tubule. The main factors that regulate renal 2 K+
phosphate transport are dietary intake itself and a ADP+Pi
number of hormones including parathyroid hor- 2Na+
mone, FGF-23, and growth hormone. Phosphate IIc HPO42−
HPO42−
is essential for bone growth and 85 % of our
phosphate is in bones. In addition, phosphate is
involved in a myriad of enzymatic reactions and is
present in nucleotides, phospholipids, and pro-
teins. Unlike the adult, the neonate must be in
positive phosphate balance for growth. The
serum phosphate level is higher in the neonate Fig. 6 A proximal tubule cell reabsorbing phosphate is
shown. The top apical phosphate transporter NaPi-IIa is the
than the adult. This section will review phosphate predominant phosphate transporter in adult rodents. NaPi-
transport and then discuss developmental changes IIc is the predominant phosphate transporter on the apical
which occur in transport and its regulation which membrane of neonatal rodents. NaPi-IIa is electrogenic
allows the neonate to be in positive phosphate while NaPi-IIc is electroneutral. NaPi-IIc is the main phos-
phate transporter on the apical membrane of the proximal
balance. tubule in humans
The transporters involved in the regulation of
phosphate transport are shown in Fig. 6. The first
phosphate transporter cloned, designated NaPi-1, in neonates. However, early studies in human
did not have the characteristics previously identi- neonates found that the fraction of phosphate
fied in physiologic studies, and its function is still reabsorbed compared to the glomerular filtration
not clear [129, 130]. There are two sodium- rate was higher in neonates and infants than that in
dependent phosphate transporters on the apical adults [139–142]. In the first 24 h of life, human
membrane of the proximal tubule: one designated neonates reabsorb over 95 % of the filtered phos-
NaPi-IIa [131] and the other NaPi-IIc [132]. NaPi- phate [140]. This level drops to 90 % later in the
IIa is an electrogenic transporter that transports first week of life [139, 140]. This fractional
three sodium ions with one phosphate (HPO42), reabsorption of phosphate meets or exceeds the
while NaPi-IIc transports two sodium ions with reabsorptive capacity of adults and older children
every phosphate and is electroneutral [132, ingesting a normal phosphate diet [142].
133]. NaPi-IIa is regulated by PTH and dietary Animal studies demonstrated that young rats
phosphate intake [134–137]. Phosphate exits the had a greater rate of phosphate reabsorption than
proximal tubule by a transporter which has not yet adults [143]. This was seen in rats that received
been identified and characterized. NaPi-IIb is the parathyroidectomy indicating that an altered
phosphate transporter on the intestinal apical response to parathyroid hormone was not the fac-
membrane responsible for absorption of dietary tor that caused the disparity in renal phosphate
phosphate [138]. uptake [143]. Finally both young and adult rats
The serum phosphate levels are higher in neo- responded to a low-phosphate diet with an
nates than adults [139, 140]. Since the glomerular increase in the fractional reabsorption of phos-
filtration rate in the neonate is only a fraction that phate; the magnitude of phosphate absorption
of the adult, it is possible that this is the factor that was again higher in young animals than
is responsible for the relative hyperphosphatemia adults [143].
72 M. Baum

phosphate reabsorption has also been demon-


REABSORPTION RATE OF Pi / KW (mg/min/g KW)

strated in young rats compared to adult rats


Newborn
60 in vivo [145, 146].
Adult
Micropuncture studies of neonatal and adult
50 guinea pigs and rats have also demonstrated that
there is a higher intrinsic rate of phosphate trans-
40 port in young animals [147, 148]. Studies using
brush border membrane vesicles have demon-
30 strated that the maximal rate (Vmax) of phosphate
transport was severalfold higher in neonates than
adults, while there was no difference in the Km,
20
the phosphate concentration at half maximal
velocity [149]. A low-phosphate diet increased
10
the Vmax of the sodium phosphate transporter in
adult guinea pigs while a high-phosphate diet had
0
10 20 30 40 50 60 the opposite effect. There was no significant dif-
FILTETED LOAD OF Pi (mg/min) ference in Vmax in brush border membranes from
neonates gavaged with different phosphate-
Fig. 7 The rate of phosphate absorption by isolated per- containing diets [149]. As shown in Fig. 8, the
fused kidney preparation. As the filtered load increases,
there was an increase in the reabsorptive rate in both the
3-week-old rat has a maximal rate of phosphate
neonate and the adult kidney. At all filtered loads the rate of absorption that was higher than that of adult rats
phosphate reabsorption per gram of kidney weight was [150]. At all ages there was an augmentation in the
higher in the neonate (From Johnson and Spitzer [144], maximal capacity of phosphate reabsorption with
with permission)
a low-phosphate diet [150]. A greater effect of
phosphate deprivation on brush border membrane
While the above studies are consistent with an vesicle phosphate uptake and NaP-IIa protein
enhanced tubular reabsorptive capacity in neo- abundance was demonstrated in 4-week-old rats
nates, this could be due to a higher reabsorptive compared to older adult rats [148]. Finally, the
capacity of the neonatal tubule, a diminished driving force for phosphate entry across the apical
response to a phosphaturic factor, or the result of membrane may be greater in neonates. The intra-
an increased response to a substance which cellular phosphate concentration was almost 40 %
increases phosphate transport. To determine if lower in kidneys measured using NMR [151].
there was an inherent increase in tubular phos- Maturational studies examining the changes in
phate transport, Johnson and Spitzer examined NaPi-IIa expression have revealed that NaPi-IIa
phosphate absorption in neonatal and adult kid- mRNA and protein are not detected in developing
neys perfused in vitro [144]. As shown in Fig. 7, nephrons until there is a distinct brush border
neonatal kidneys had a higher phosphate membrane [152]. NaPi-IIa protein abundance
reabsorptive rate at any filtered load of phosphate was greater in brush border membranes from
which can only be due to an increased inherent 13-day-old rats compared to 22-day-old rats
rate of phosphate transport. In addition, they [152]. Others however have found that brush bor-
found that while addition of parathyroid hormone der membrane vesicle from suckling and adult rats
to the perfusate caused a phosphaturia in adult had a slower rate of phosphate uptake than wean-
kidneys, there was no increase in phosphate ling rats (21 days old) [153]. There was no change
excretion in neonatal kidneys. These studies in NaPi-IIa mRNA abundance, but NaPi-IIa pro-
directly demonstrate that neonates also have an tein abundance from brush border membrane ves-
attenuated effect of the primary factor regulating icles confirmed the transport findings that
phosphate transport, parathyroid hormone. A 21-day-old rats had the highest NaPi-IIa protein
blunted effect of parathyroid hormone on expression [153]. Studies comparing 28-day-old
3 Renal Tubular Development 73

14
* Diet
10 Low Pi
Normal Pi

Max RPl/GFR (umol/ml)


8 * High Pi

6
*
4 *
* *
2

0
IMMATURE YOUNG ADULT

Fig. 8 Age-dependent maximal phosphate reabsorption is stimulated phosphate absorption in all age groups. The
seen in 3–4-week-old rats (immature), 6–7-week-old rats maximal capacity for phosphate absorption was in the
(young), and 12–13-week-old rats (adult). All rats were immature groups which need phosphate for growth
parathyroidectomized. A low dietary phosphate intake (From Mulroney and Haramati [150], with permission)

rats to adult rats have also demonstrated greater hormone is not a significant regulator of phos-
brush border membrane NaPi-IIa protein abun- phate transport in the adult, this may not be the
dance in young rats than adults [148]. case in the growing animal. Administration of a
The high rates of phosphate transport in wean- growth hormone-releasing factor antagonist,
ling rodents suggested that there may be a which suppresses growth hormone secretion, has
developmentally regulated transporter with no effect on phosphate transport in adult rats but
greater expression at that time during develop- significantly reduces phosphate absorption in
ment [154]. Indeed, NaPi-IIc is a brush border young growing rats [158–160].
phosphate transporter with its highest expression There are a number of hormones that have been
at the time of weanling in the rat [132]. NaPi-IIc, shown to regulate phosphate transport including
like NaPi-IIa, is regulated by dietary phosphate insulin [161], fibroblast growth factor-23
uptake [132]. The relative importance of NaPi-IIa [162–164], frizzled-related protein 4 [165], and
and NaPi-IIc may be quite different in humans. Klotho [166, 167]. These hormones may have
Patients with hereditary hypophosphatemic rick- differential effects on phosphate transport in the
ets with hypercalciuria, a rare autosomal recessive neonate and adult, but this is yet to be determined.
characterized by hypophosphatemia secondary to
renal phosphate wasting and high vitamin D
levels, have a mutation, in the gene encoding Development of Proximal Tubular
NaPi-IIc. This suggests that NaPi-IIc may be the Acidification
predominant renal phosphate transporter in
humans and that NaPi-IIa cannot compensate for The proximal tubule reabsorbs 80 % of the filtered
the loss of NaPi-IIc [155]. bicarbonate. Luminal proton secretion is via the
Growth hormone increases phosphate trans- Na+/H+ exchanger and the H+-ATPase. In the
port via stimulation of IGF-1 in the proximal adult, one-third of proton secretion is via the
tubule [156]. Brush border membrane vesicle luminal H+-ATPase, and two-thirds is mediated
phosphate transport is increased in dogs that by the luminal Na+/H+ exchanger that is desig-
were administered growth hormone compared to nated NHE3 [168, 169]. The secreted proton
vehicle-treated controls [157]. While growth titrates the filtered HCO3 to generate H2CO3
74 M. Baum

a NHE8 Expression on BBMV b NHE3 Expression on BBMV

NHE8 NHE3

β-actin β-actin

1D 7D 14D 26D AD 1D 7D 14D 26D AD

1.2 1.2
*+
1.0 1.0

*
NHE-8/β-actin

NHE-3/β-actin
0.8 0.8
#
0.6 0.6

0.4 0.4

^ ^
0.2 0.2 # #

0.0 0.0
1D 7D 14D 26D AD 1D 7D 14D 26D AD

Fig. 9 Immunoblots of rat brush border membrane membrane. The expression of NHE3 is highest in the adult.
vesicles depict the changes in NHE8 (A) and NHE3 (B) The higher NHE3 protein abundance at 1 day of age is
protein abundance. As is seen there, there is higher expres- likely the result of the surge of glucocorticoids at the time
sion of NHE8 in the neonate than the adult brush border of birth (From Becker et al. [176], with permission)

which is converted to CO2 and H2O by luminal in the distal nephron which, by and large, is
carbonic anhydrase (carbonic anhydrase IV). CO2 responsible for the secretion of acid from metab-
diffuses into the cell and combines with H2O olism and new bone formation. Studies have
which is facilitated by intracellular carbonic shown that there is a maturational increase in
anhydrase (carbonic anhydrase II) to regenerate bicarbonate absorption during postnatal develop-
H2CO3. H2CO3 dissociates into a proton that can ment [73, 172], which accounts for the mutational
be secreted across the apical membrane and bicar- increase in the bicarbonate threshold.
bonate that exits the basolateral membrane via the The greatest developmental changes in proxi-
basolateral Na(HCO3)3 cotransporter. The sodium mal tubule acidification occur on the apical mem-
that enters the cell via the Na+/H+ exchanger exits brane. There is a fourfold increase in Na+/H+
the basolateral membrane by the Na+/K+-ATPase, exchanger activity during postnatal maturation
which provides the driving force for luminal pro- and an even greater increase in apical H+-ATPase
ton secretion by the Na+/H+ exchanger. There are activity [168, 173, 174]. Low levels of Na+/H+
maturational changes in most of these processes exchanger activity have been measured in the
that will be described below. fetus as well [175]. Despite the fact that there
Neonates have a lower serum bicarbonate con- was Na+/H+ exchanger activity on the apical
centration than adults, which is the result of a membrane of the neonatal rat at 1–2 weeks of
lower threshold for bicarbonate [170]. Premature age, as shown in Fig. 9, there was virtually no
neonates can have physiologic bicarbonate con- NHE3 on the brush border membrane [174, 176],
centrations as low as 15 mEq/l [171]. The lower the Na+/H+ exchanger on the apical membrane of
bicarbonate threshold is mediated in large part by the adult proximal tubule [177, 178].
the lower rate of proximal tubule acidification. NHE3 knockout mice have been shown to
The fine-tuning of renal acidification is mediated have substantive proximal tubule apical
3 Renal Tubular Development 75

membrane Na+/H+ exchanger activity [179]. The Na+/H+ activity and NHE3 protein abundance
apical Na+/H+ exchanger likely responsible for [46, 172, 188]. Both thyroid hormone and
this non-NHE3 Na+/H+ exchanger activity is glucocorticoids increase NHE3 activity by
NHE8, a recently discovered NHE isoform increasing transcription [189, 190]. Glucocorti-
([180, 181]). NHE8 has sodium-dependent proton coids have also recently been shown to increase
extrusion capabilities, which made it a potential the insertion of NHE3 into the apical membrane of
candidate for the developmental isoform proximal tubular cells by a posttranscriptional
[182]. As shown in Fig. 9, apical NHE8 was mechanism [191].
predominantly present in neonatal proximal Interestingly, neither prevention of the matura-
tubules at a time when NHE3 was almost tional increase in glucocorticoids or thyroid hor-
undetectable [176]. Thus there is a developmental mone alone can totally prevent the postnatal
Na+/H+ exchanger isoform. increase in Na+/H+ exchanger activity and NHE3
The Na(HCO3)3 symporter mediates bicarbon- mRNA and protein abundance [46, 172, 192,
ate exit in both the neonatal and adult proximal 193]. Thus, there appears to be some redundancy
tubule. While there is a maturational increase in in the postnatal maturational triggers for NHE3.
basolateral membrane Na(HCO3)3 symporter As demonstrated in Fig. 10, prevention of the
activity, it is relatively small compared to the maturational increase in both hormones results
magnitude of the change in Na+/H+ exchanger in the prevention of the postnatal increase in
activity on the apical membrane [183]. In addition NHE3 mRNA, protein abundance, and Na+/H+
to bicarbonate exit, the basolateral membrane Na exchanger activity [194].
(HCO3)3 symporter plays an important role in pH
regulation of the proximal tubule cell [183].
Carbonic anhydrase increases the rate of the Development of Proximal Tubule NaCl
interconversion of CO2 and H2O to H2CO3. Transport
Carbonic anhydrase II is located intracellularly
in proximal and distal tubule acidifying cells and The glomerulus generates an ultrafiltrate of
comprises ~95 % of total cellular carbonic plasma and delivers the fluid to the proximal
anhydrase activity. Carbonic anhydrase IV is on tubule. In the early proximal tubule, the
the apical and basolateral membrane of renal acid- transcellular reabsorption of sodium with glucose
ifying cells and comprises ~5 % of carbonic and amino acids results in a lumen-negative
anhydrase activity [184]. Both carbonic potential difference. This lumen-negative poten-
anhydrase II and IV increase during maturation tial provides a driving force for either chloride
of proximal and distal acidification, but neither is absorption or sodium secretion across the
likely a limiting factor causing the maturational paracellular pathway. Whether chloride is
increase in renal acidification in the proximal or absorbed or sodium is secreted is dependent on
distal tubule [185–187]. the relative permeabilities of chloride to sodium in
Since the developmental increase in renal acid- the proximal tubule. The relative chloride to
ification is due primarily to apical proton secre- sodium permeability is higher in the adult than
tion, many studies have examined the cause for the neonate, so passive chloride transport second-
the increase in Na+/H+ exchanger activity. There ary to the lumen-negative potential difference in
is a substantive increase in both thyroid hormone the early proximal tubule does not occur to a
and glucocorticoids during postnatal develop- significant degree in the neonate [14, 195].
ment, and both hormones increase in parallel The reabsorption of bicarbonate in preference
with the increase in proximal tubule Na+/H+ to chloride by the early proximal tubule causes the
exchanger activity [46–49]. Administration of luminal fluid in the late proximal tubule to have a
either glucocorticoids or thyroid hormone prior higher chloride and lower bicarbonate concentra-
to the maturational increase in either hormone tion than that of the peritubular fluid [196,
results in a precocious increase in exchanger 197]. The axial changes in luminal fluid
76 M. Baum

1000
* greater than 9 day (unpaired T-test)
+ less than 9 day (unpaired T-test)
**
** greater than 9 day, Adx and Adx-HypoT
800
**
JH (pmol/mm.min)

600

400 *

200
+

0
Adx- Adx-HypoT
9 Day Adx Sham
HypoT +Replacement

Fig. 10 Apical Na+/H+ exchanger activity in perfused 9-day-old rats had a lower rate of Na+/H+ exchanger activ-
tubules in 9-day-old rats and adults that were adrenalecto- ity than the sham adult. Neonatal adrenalectomy did not
mized as neonates, adults that were adrenalectomized and totally prevent the maturational increase in Na+/H+
hypothyroid since the neonatal period, and adults that were exchanger activity, but the maturational increase in
adrenalectomized and hypothyroid (adx-HypoT) since the Na+/H+ exchanger activity was abrogated in the adrenal-
neonatal period but given thyroid and glucocorticoid ectomized hypothyroid group (From Gupta et al. [194],
replacement before study and sham controls. The with permission)

composition are depicted in Fig. 11. The chloride driving force for active transcellular NaCl trans-
permeability is greater than that for bicarbonate so port is the basolateral Na+/K+-ATPase that has
that the concentration gradient is a driving force lower activity in the neonatal proximal tubule
for passive chloride diffusion across the [39, 40, 209]. There are a number of factors that
paracellular pathway. The diffusion of chloride regulate proximal tubule NaCl transport including
across the paracellular pathway results in a renal nerves, dopamine, and angiotensin II which
lumen-positive potential difference and a driving are shown in Table 1. The serum levels of most
force for the paracellular transport of sodium hormones that regulate sodium absorption are
resulting in net passive NaCl absorption. equal or higher in the neonate than the adult, but
In the adult proximal tubule, approximately in general there is a blunted response to the action
half of sodium chloride is active and transcellular of most regulatory hormones.
and half is passive and paracellular [193, 198, There are also changes in the properties of the
199]. Active chloride transport is mediated by paracellular pathway during postnatal develop-
parallel action of the Na+/H+ and Cl/base ment [14, 195, 210, 211]. Most importantly, the
exchangers on the apical membrane [193, permeability of the proximal tubule to chloride
200–202]. It is still somewhat unclear what the ions is less in the neonate than the adult [14,
nature of the base is as there is evidence for 195, 211]. The low permeability to chloride ions
chloride exchange for hydroxyl, formate, and oxa- results in almost no passive paracellular chloride
late ions [193, 203–207]. The rate of active transport in the neonate [14, 195]. As previously
transcellular chloride transport is lower in the noted, the permeability properties of an epithe-
neonate than the adult. This is due to the lower lium are determined by the expression of a family
rate of the apical Cl/base exchanger [193, 208] of proteins called claudins. The claudin proteins in
and the Na+/H+ exchanger [168, 173, 174]. The the tight junction change during the postnatal
3 Renal Tubular Development 77

2.0 Developmental Features of Proximal


Tubule Water Transport
1.8 Inulin

1.6 The proximal tubule reabsorbs most of the glo-


merular filtrate without a significant change in the
1.4 Cl− luminal osmolality. For this to occur, the proximal
tubule must be very permeable to water. Water
1.2
Na+ movement is predominantly through the cell and
TF
1.0 not across the paracellular pathway [213,
P Osm
214]. The constitutively water-permeable proxi-
0.8 mal tubule and thin descending limb have water
channels on the apical and basolateral membranes
0.6 HCO3−
[1]. The isoform of this water channel is aquaporin
0.4 1 [215–218]. Direct evidence of transcellular
Amino
acids water transport comes from aquaporin 1 knockout
0.2 mice which have a marked decrease in proximal
Glucose tubule sodium absorption [219]. There is a paucity
0
0 25 50 75 100 of water channels in the fetal kidney, and an
% Proximal tubule length increase in expression of aquaporin 1 does not
occur until birth [220, 221].
Fig. 11 The axial changes in proximal tubular transport Direct measurements of water permeability
are depicted on this figure. The early proximal tubule
preferentially reabsorbs glucose, amino acids, and bicar-
have demonstrated that the neonatal rabbit proxi-
bonate leaving the luminal chloride solution higher than mal tubule has a higher water permeability than
the blood in the peritubular capillaries. The early proximal that of the adult [222]. To determine the mecha-
tubule has a lumen-negative transepithelial potential dif- nism for the higher water permeability in neonatal
ference due to sodium-dependent glucose and amino acid
reabsorption. The higher luminal chloride concentration in
rabbit tubules, studies were performed examining
the late proximal tubule provides a driving force for pas- the water permeability of apical and basolateral
sive chloride absorption across the paracellular pathway membrane vesicles [223–225]. Despite the higher
causing a lumen-positive potential difference (From Rector transepithelial water permeability, the water per-
[197], with permission)
meability of the apical and basolateral membrane
development. Claudins 6, 9, and 13 are present in was lower in the neonate than the adult and there
the neonatal proximal tubule but not in the adult was less aquaporin 1 expression on both the apical
[210]. The claudin isoform responsible for the low and basolateral membranes of the proximal tubule
paracellular chloride permeability in the neonate of the neonate [224, 225]. The apparent paradox
and the factors that cause the claudin isoform between the higher water permeability in the neo-
changes during development have yet to be natal proximal tubule and the lower water perme-
determined. ability of the apical and basolateral membranes
Of the potential factors that cause the maturational was resolved with measurements of the contribu-
changes in paracellular chloride transport, only thy- tion of the intracellular compartment to water
roid hormone has been examined [212]. Administra- movement in the neonatal and adult proximal
tion of thyroid hormone prior to the normal tubule. The intracellular compartment was found
maturational increase results in an increase in chlo- to cause a large resistance to water flow, and
ride permeability. On the other hand, maintaining a neonatal proximal tubule intracellular compart-
hypothyroid state into adulthood prevents the matu- ment was less of a constraint to transcellular
rational increase in chloride permeability. It is as yet water movement than that of the adult [214].
to be determined if thyroid hormone is the factor that The postnatal increase in glucocorticoids is a
causes the proximal tubule claudin isoform changes likely factor in mediating the above noted matu-
during postnatal development. rational changes in water permeability and
78 M. Baum

Table 1 Regulation of Sodium Transport


Effect on urinary Effect of hormone on sodium in
sodium excretion in Neonatal serum level compared neonatal sodium excretion
Hormone adult to adult compared to adult References
Renin Level responds appropriately in [327, 330,
neonate 342, 343]

Aldosterone Increases distal [325–331]


sodium absorption

Atrial Increases sodium Probably [344] [345–348]


natriuretic excretion
peptide

Dopamine Decreases proximal [25,


tubule sodium 349–351]
absorption

PGE-2 Causes natriuresis Blunted synthesis in the [353, 354]


and diuresis medullary and cortical
collecting tubule [352]

aquaporin 1 expression. Administration of gluco- provide a driving force for luminal sodium entry.
corticoids to neonatal rabbits resulted in an A cartoon of a thick ascending limb cell is
increase in brush border membrane water perme- shown in Fig. 12. Sodium enters the cell via the
ability and aquaporin 1 expression [226]. Of inter- furosemide- or bumetanide-sensitive Na+/K+/
est, the developmental increase in thyroid 2Cl cotransporter [228, 229]. The Na+/K+/2Cl
hormone was not shown to be a factor mediating cotransporter is electroneutral, yet there is a
these postnatal maturational changes in water lumen-positive transepithelial potential difference
transport [227]. in the thick ascending limb [230]. The positive
luminal potential is generated by apical potassium
recycling via the potassium channel ROMK1
Developmental Transport in the Thick [231, 232]. With recycling of potassium back
Ascending Limb into the lumen, there is the net reabsorption of
1 sodium and 2 chloride ions which exit the cell
The thick ascending limb (TAL) is responsible for across the basolateral membrane. Sodium exits
the reabsorption of 30 % of filtered NaCl. The the cell via the Na+/K+-ATPase. Chloride predom-
thick ascending limb is impermeable to water. inantly exits the cell via a chloride channel desig-
The reabsorption of NaCl without water by the nated ClC-Kb [233–235]. A subunit of this
cortical and medullary TAL along with the distal chloride channel designated barttin is important
convoluted tubule generates a luminal fluid with in the function of ClC-Kb [236]. The lumen-
an osmolality of 50 mOsm/kg water. In the positive potential difference, generated by potas-
absence of ADH action on the collecting tubule, sium secretion into the lumen, generates a driving
this dilute urine will be excreted. The medullary force for the passive reabsorption of cations
interstitial hypertonicity is largely generated by including Mg2+ and Ca2+. The thick ascending
active NaCl reabsorption without water transport limb has a very high permeability to magnesium
in that tubular segment. and calcium ions which results in a substantial
The reabsorption of NaCl is due to secondary fraction of filtered calcium and magnesium being
active transport with the basolateral Na+/K+- reabsorbed passively across the paracellular path-
ATPase generating a low intracellular sodium to way in this segment [237–241]. The unique
3 Renal Tubular Development 79

the latter parts of the diluting segment. Human


neonates are able to dilute their urine to the same
level as an adult (50 mOsm/kg water). This is vital
Cl− since neonates ingest a hypotonic fluid, mother’s
Na+
2Cl−
milk. Sodium transport in the thick ascending
K+ ATP
limb has been directly examined in vitro and
3Na+ shown to be fivefold lower in the neonate than in
the adult [249].
2K+
The Na+/K+/2Cl cotransporter can first be
ADP+P
detected in the mid-late gestation rat thick ascend-
K+ ing limb and macula densa [250]. In the rat there is
K+
K+
a postnatal maturational increase in Na+/K+/2Cl
cotransporter, ROMK, Na+/K+-ATPase mRNA,
Cl− and protein abundance but no change in ClC-K
mRNA abundance [251]. Administration of dexa-
methasone before the normal maturational increase
at the time of weaning resulted in a premature
Fig. 12 The figure depicts a thick ascending limb cell. On increase in urinary concentrating ability and
the apical membrane is the Na+/K+/2Cl cotransporter that increase in Na+/K+/2Cl cotransporter, Na+/K+-
is sensitive to loop diuretics and the potassium channel ATPase mRNA, and protein abundance but no
designated ROMK that is depicted as the green arrow. change in ROMK protein abundance [251]. There
The basolateral membrane has a Na+/K+-ATPase; a KCl
cotransporter; a chloride channel designated ClC-Kb, with is also a postnatal maturational increase in thick
its accessory channel designated barttin in yellow; and a ascending limb Na+/K+-ATPase activity [39, 252,
potassium channel. A loss of function mutation in the 253]. The maturational increase in thick ascending
Na+/K+/2Cl cotransporter, ROMK, ClC-Kb, or barttin limb Na+/K+-ATPase activity could be accelerated
results in Bartter’s syndrome
precociously by glucocorticoids and prevented by
neonatal adrenalectomy [252, 253].
permeability properties of the thick ascending
limb are due to the expression of claudin
16 which is mutated in familial hypomagnesemia Developmental Transport in the Distal
with hypercalciuria and nephrocalcinosis where Convoluted Tubule
there is renal magnesium and calcium
wasting [242]. The distal convoluted tubule reabsorbs approxi-
Mutations of the Na+/K+/2Cl cotransporter mately 7 % of the filtered sodium. This segment is
[243], ROMK [244, 245], ClC-Kb [246], and water impermeable, and further reabsorption of
barttin [236, 247] all cause Bartter’s syndrome salt without water occurs in this segment resulting
(see this text, ▶ Chap. 38, “Renal Tubular in the nadir of luminal fluid osmolality. The trans-
Disorders of Electrolyte Regulation in Children”). porters responsible for NaCl transport in the distal
Barttin is also expressed in the ear where it is a convoluted tubule are shown in Fig. 13. Sodium
subunit of ClC-Ka and ClC-Kb. Mutations entry is via the thiazide-sensitive cotransporter
in barttin cause sensory neural hearing loss [254–257]. Mutations in the thiazide-sensitive
[236, 247]. cotransporter cause Gitelman’s syndrome
Micropuncture studies studying fluid from the [258–260] (see this text, ▶ Chap. 38, “Renal
early distal tubule showed that the osmolality of Tubular Disorders of Electrolyte Regulation in
the fluid was significantly lower in the adult than Children”). There is also evidence for parallel
the neonatal rat [248]. This is consistent with Na+/H+ and Cl/base exchange mediating sodium
lower rates of sodium transport in the thick entry in the rat [261]. The isoform of this Na+/H+
ascending limb, but this study did not examine exchanger is NHE2 [262]. Chloride exits the cell
80 M. Baum

dog [270]. This limited ability to excrete a sodium


load was not due to the lower glomerular filtration
rate in the neonate [270]. Since sodium transport
Cl− is less in the proximal tubule, thick ascending
limb, and collecting tubule, the nephron segment
K+ where there was avid neonatal sodium
reabsorption was unclear. It must be stated that it
Na+ was never considered that the volume of distribu-
ATP
3 Na+ tion of the saline infusion was greater in the neo-
Cl−
nate than the adult to account for these findings. A
2 K+ classical micropuncture study in developing rats
ADP+Pi
suggested that the sodium retention in the neonate
Cl−
may be the result of avid sodium reabsorption in
the distal convoluted tubule [269].
K+

Development of Urinary
Concentration and Dilution
Fig. 13 Distal convoluted cell showing the transporters
involved in active sodium reabsorption. The apical NaCl
The urine exiting the distal convoluted tubule and
transporter is the thiazide-sensitive cotransporter. entering the collecting duct has an osmolality of
Inactivating mutations in the thiazide-sensitive ~50 mOsm/kg water. Whether the urine will dilute
cotransporter lead to Gitelman’s syndrome or is maximally concentrated depends on the pres-
ence or absence of vasopressin (ADH). Neonates
via a basolateral chloride channel ClC-K [263] can dilute their urine to nearly the adult level of
and a KCl cotransporter KCNQ1 [264, 50 mOsm/kg water [273–275]. Thus the neonate
265]. There is also a basolateral potassium chan- that drinks mother’s milk can excrete free water
nel [266]. There is active transcellular calcium [274–276]. However, unlike the normal adult
and magnesium transport in the distal convoluted where it is almost impossible to drink enough
tubule, but there is no information about the water to cause hyponatremia (>30 l/day), neo-
developmental expression and regulation of nates have a rather limited ability to generate and
these transporters [267, 268]. excrete free water. Therefore improperly mixed
The distal convoluted tubule is very difficult to and dilute formula or administration of excessive
study in vitro and in vivo. No studies have been amounts of dilute fluid via NG or G tube can result
performed to date examining the relative abun- in hyponatremia [274].
dance of the thiazide-sensitive cotransporter in The maximum urine osmolality of the term
the neonate and adult. A study has been preformed human neonate is ~400–600 mOsm/kg water
that suggests that there is increased sodium [277–279]. The adult urine osmolality of 1,200
absorption in 24- compared to 40-day-old rats mOsm/kg water is achieved at ~1.5–2 years of age
[269, 270]. Before detailing this study, it is impor- [279]. There are many potential developmental
tant to know that studies have shown that the factors that could limit the ability of the neonate
neonate is less able to excrete a salt load compared to generate maximally concentrated urine. The
to the adult [270–272]. For example, administra- factors involved in urine concentration include:
tion of isotonic saline equal to 10 % of the dogs
weight to an adult dog resulted in excretion of 1. The ability to sense an increase in serum osmo-
50 % of the salt load over 8 h, but only 10 % lality or decrease in extracellular volume and
of the salt load was excreted in the 1–2-week-old secrete vasopressin.
3 Renal Tubular Development 81

2. There must be functional vasopressin receptors deprivation indicating that basolateral water
on the basolateral membrane of the movement is primarily via aquaporin 3 [291,
collecting duct. 292]. Vasopressin causes the intracellular vesicles
3. The collecting duct must be able to containing aquaporin 2 to fuse with the apical
generate cAMP. membrane resulting in the insertion of aquaporin
4. The loop of Henle must have generated a con- 2 into the apical membrane [290]. There is a
centrated interstitium. developmental increase in aquaporin 2 expression
5. The architecture of the medulla must not limit [286, 287, 293–295]. The maturational increase in
the concentrating ability. aquaporin 2 is accelerated by administration of
6. The collecting tubule must have aquaporins on synthetic glucocorticoids prior to the normal post-
the apical and basolateral membrane. natal increase in plasma glucocorticoids
7. There must not be extracellular or intracellular [295]. Neither aquaporin 3 nor aquaporin 4 are
mechanisms upregulated in the neonate which factors impairing urinary concentration in the neo-
limit urinary concentrating abilities. nate [286, 296].
The fetus and neonate respond to increases in
During prenatal and postnatal maturation, serum osmolality, stress, and hypovolemia with
there are anatomical changes that occur affecting appropriate increases in plasma vasopressin
urinary concentrating ability. There is an increase levels. Fetal sheep have an increase in plasma
in the medullary capillary density, a decrease in vasopressin and plasma osmolality with an infu-
medullary interstitial connective tissue, and an sion of hypertonic saline and increase in plasma
increased presence and length of the thin limbs, osmolality [297–299]. Plasma vasopressin also
and the tubules become more tightly packed with increases in fetal sheep in response to volume
cells in the loop decreasing in height with matu- depletion induced by hemorrhage or diuretics
ration [280, 281]. The length of the papilla [297, 300, 301]. Despite the fact that fetal sheep
increases linearly from day 10 to day 40 in the can increase serum vasopressin levels, infusion of
rat [282]. All these developmental anatomical vasopressin resulted in a blunted increase in urine
changes are necessary for the countercurrent mul- osmolality compared to adult sheep [302]. It
tiplication system to be maximally efficient. is difficult to directly relate these experimental
Accompanied by these anatomical changes are findings to humans. Comparison between a rela-
concomitant increases in the medullary sodium tively stressful vaginal delivery and a cesarean
and urea concentration [282, 283]. Administration section has consistently demonstrated higher
of a high-protein diet or urea to human neonates vasopressin levels in neonates born vaginally
results in an increased ability to concentrate urine [303–306]. However, there was no correlation
implying that the ability of urea to accumulate in with vasopressin levels and the degree of perinatal
the medulla can be limited by dietary intake [284, asphyxia in one study [305], while there was a
285]. Adrenalectomy in neonatal rats prevented correlation in another [303]. The balance of data
the maturational increase in urine osmolality suggests that the human fetus and neonate can
while administration of glucocorticoids prior to respond to appropriate stimuli with vasopressin
the maturational increase in serum glucocorti- secretion.
coids caused a premature increase in urinary con- There is a developmental increase in vasopres-
centrating ability [282]. sin receptors in the kidney; however, vasopressin
Principal cells in the collecting tubule and the receptor abundance does not appear to be a limit-
cells in the medullary collecting duct express ing factor in urinary concentration in the neonate
aquaporin 2 on the apical membrane and [307, 308]. Exogenous administration of vaso-
aquaporins 3 and 4 on the basolateral membrane pressin has been shown to increase the urine
[286–290]. Aquaporin 3 null mice have diabetes osmolality in fetal sheep showing that vasopressin
insipidus whereas aquaporin 4 null mice only action on the collecting tubule is a prenatal event
have a small concentrating defect after water [302]. Vasopressin acts in the collecting tubule by
82 M. Baum

increasing cAMP. There is some discrepancy content [318]. This is unusual for humans but not
about the extent of cAMP production during post- for some mammals which ingest an alkali diet
natal maturation in comparison to adults. The [319]. Both α- and β-intercalated cells have dif-
balance of data supports that conclusion; although ferences in the polarized localization of both the
vasopressin stimulates cAMP production in the Na+/K+-ATPase and the Cl/HCO3 as shown in
neonate, the amount of cAMP generated is less Fig. 14.
than that of the adult [307, 309–311]. The neonatal cortical collecting tubule has
The response of the collecting tubule to vaso- fewer and less differentiated intercalated cells
pressin has been examined in vitro than that of the adult segment [320, 321]. The
[312–315]. Neonatal CT water permeability did rate of both bicarbonate secretion and luminal
not increase in response to vasopressin to the same acidification is less in the neonate than in the
extent as that seen in the adult [312–315]. Studies adult [319, 321, 322]. The intercalated cells in
have shown that the phosphodiesterase activity is the inner medullary collecting duct were much
greater in the neonatal collecting tubule more differentiated in appearance and secreted
[313]. The water permeability of the neonatal protons at a rate comparable to the adult [319,
collecting tubule was identical to the adult in the 322]. The α-intercalated cell of the CT, as noted
presence of a phosphodiesterase inhibitor demon- above, has a luminal H+-K+ ATPase which causes
strating that the predominant factor limiting the proton secretion and potassium absorption from
action of vasopressin in the collecting tubule was the luminal fluid. It is activated under states of
the enhanced degradation of cAMP generated by metabolic acidosis and hypokalemia [323]. The
vasopressin in the neonatal collecting tubule H+-K+ ATPase can function at comparable rates in
[313]. Vasopressin increases prostaglandin pro- the neonatal cortical collecting tubule as that of
duction in the collecting tubule which attenuates the adult [324].
the vasopressin-mediated increase in cAMP pro-
duction [316, 317]. Vasopressin-mediated cAMP
production, while less in the neonatal collecting Developmental Features of Cortical
tubule, increases to adult levels in the presence of Collecting Tubule Sodium Transport
indomethacin, a prostaglandin synthesis inhibitor
[317]. Thus, prostaglandin production by the While relatively little sodium is reabsorbed by the
collecting tubule may attenuate the effect of vaso- cortical collecting tubule compared to the proxi-
pressin in the neonatal tubule to a greater extent mal nephron segments, it is the final nephron
than the adult tubule [317]. segment responsible for regulating sodium
absorption and thus is vitally important for the
regulation of sodium homeostasis. Sodium
Developmental Aspects of Distal absorption occurs in the principal cell of the
Tubule Acidification collecting tubule which is shown in Fig. 14.
Sodium transport is through the epithelial sodium
The distal nephron makes the final adjustment in channel designated ENaC which has three sub-
renal acidification. The distal nephron, under nor- units. The driving force for sodium entry across
mal circumstances, secretes the protons equal to ENaC is the low intracellular sodium concentra-
that generated from metabolism and in children tion and the potential difference across the luminal
the protons liberated from the formation of bone. membrane generated by the basolateral Na+/K+-
The cortical collecting tubule has two types of ATPase. The Na+/K+-ATPase undergoes a matu-
cells that are involved in renal acidification, as rational increase in this segment but is not the
shown in Fig. 14. The α-intercalated cell is limiting factor of the maturational increase in
responsible for proton secretion. The sodium absorption [39]. The abundance of
β-intercalated cell can secrete bicarbonate when ENaC on the apical membrane is by and large
the animal is alkalotic or eats a diet with an alkali determined by aldosterone in the adult, but
3 Renal Tubular Development 83

Fig. 14 Three cells of the


cortical collecting tubule
are demonstrated. The Principal Cell
principal cell is shown ATP
above with sodium entering
Na+
the cell down its 3 Na+
electrochemical gradient
via a channel designated
ENaC. This generates a ADP+Pi
lumen-negative K+
transepithelial potential K+
difference. Potassium is
secreted into the lumen
down its electrochemical Cl−
gradient via ROMK.
Chloride is moving through
the paracellular pathway
down the electrical gradient α-Intercalated Cell
generated by the lumen- ADP+Pi
negative potential
Cl−
difference. The cell below is
an alpha intercalated cell H+
which secretes protons into HCO3−
the cell lumen via a H+- ATP
ATPase. The bicarbonate Cl−
generated in this process is
secreted across the
basolateral membrane via a
Cl/HCO3 exchanger. The
third cell is a beta
intercalated cell which
secretes bicarbonate ions in
the face of metabolic β-Intercalated Cell Cl−
alkalosis. This cell is the
reverse of an alpha
ATP
intercalated cell, but the
HCO3−
Cl/HCO3 exchanger is a
H+
different isoform
Cl−
ADP+Pi

aldosterone has a blunted effect in the neonate there is a developmental increase in mRNA and
despite the fact that there are higher serum levels protein abundance of each during postnatal matu-
in the neonate and ample aldosterone receptors ration of this segment [333–337].
[325–331].
Sodium transport in the cortical collecting
tubule increases with postnatal development Developmental Aspects of Tubular
[331, 332]. This increase in sodium transport is Potassium Transport
paralleled by an increase in the apical expression
of ENaC, since ENaC expression is the limiting Neonates have higher serum potassium levels
factor in sodium absorption in this segment [333]. than adults. Potassium is the predominant intra-
ENaC is composed of α-, β-, and γ-subunits, and cellular cation, and neonates must be in positive
84 M. Baum

potassium balance for growth unlike adults which the CHIP28 water channel in rat kidney. Am J
excrete the quantity of potassium absorbed from Physiol. 1992;263:C1225–33.
2. Imai M, Kokko JP. Mechanism of sodium and chlo-
their diet in their urine [323]. Adult animals are ride transport in the thin ascending limb of Henle.
more readily able to excrete an exogenous potas- J Clin Invest. 1976;58:1054–60.
sium load than a neonate [338]. Approximately 3. Kobayashi K, Uchida S, Mizutani S, Sasaki S,
half of the filtered potassium is reabsorbed in the Marumo F. Developmental expression of CLC-K1 in
the postnatal rat kidney. Histochem Cell Biol.
proximal tubule of the adult and neonate by pas- 2001;116:49–56.
sive diffusion across the paracellular pathway 4. Kokko JP, Rector Jr FC. Countercurrent multiplica-
[339]. The loop of Henle reabsorbs 80 % of the tion system without active transport in inner medulla.
delivered potassium in the adult and only 45 % in Kidney Int. 1972;2:214–23.
5. Sands JM, Kokko JP. Countercurrent system. Kidney
the neonate [339]. Thus, the adult delivers approx- Int. 1990;38:695–9.
imately 10 % of the filtered potassium to the distal 6. Ma T, Yang B, Gillespie A, Carlson EJ, Epstein CJ,
nephron while the neonate delivers 25 %. The Verkman AS. Severely impaired urinary concentrat-
distal convoluted tubule, connecting tubule, and ing ability in transgenic mice lacking aquaporin-1
water channels. J Biol Chem. 1998;273:4296–9.
cortical collecting duct are the sites of potassium 7. Anderson JM, Van Itallie CM. Tight junctions and the
secretion and final modulation of urine potassium molecular basis for regulation of paracellular perme-
excretion. ability. Am J Physiol. 1995;269:G467–75.
Potassium secretion in a principal cell is 8. Mitic LL, Anderson JM. Molecular architecture of
tight junctions. Annu Rev Physiol. 1998;60:121–42.
depicted in Fig. 14. Sodium enters the principal 9. Mitic LL, Van Itallie CM, Anderson JM. Molecular
cell through the apical sodium channel down the physiology and pathophysiology of tight junctions
favorable electrochemical gradient generated by the I. Tight junction structure and function: lessons from
Na+/K+-ATPase on the basolateral membrane. This mutant animals and proteins. Am J Physiol
Gastrointest Liver Physiol. 2000;279:G250–4.
results in a lumen-negative potential difference and 10. Colegio OR, Van Itallie CM, McCrea HJ, Rahner C,
a favorable electrochemical gradient for potassium Anderson JM. Claudins create charge-selective chan-
secretion. While there is a maturational increase in nels in the paracellular pathway between epithelial
the sodium channel and the Na+/K+-ATPase, these cells. Am J Physiol Cell Physiol. 2002;283:C142–7.
11. Colegio OR, Itallie CV, Rahner C, Anderson
are not the limiting factors for potassium secretion. JM. Claudin extracellular domains determine
There are two potassium channels in the collecting paracellular charge selectivity and resistance but not
tubule: a channel designated ROMK is on the apical tight junction fibril architecture. Am J Physiol Cell
membrane of principal cells and a flow-dependent Physiol. 2003;284:C1346–54.
12. Corman B, Di Stefano A. Does water drag solutes
channel that is activated by stretch designated maxi- through kidney proximal tubule? Pflugers Arch.
K channel [323]. There is a maturational increase in 1983;397:35–41.
potassium secretion in cortical collecting tubules 13. Jacobson HR, Kokko JP, Seldin DW, Holmberg
perfused in vitro [332]. The secretion of potassium C. Lack of solvent drag of NaCl and NaHCO3 in
rabbit proximal tubules. Am J Physiol. 1982;243:
by principal cells is paralleled by the maturational F342–8.
increase in potassium channels (ROMK) on the 14. Quigley R, Baum M. Developmental changes in
apical membrane of the principal cell [337, rabbit proximal straight tubule paracellular
340]. There is also a developmental increase in the permeability. Am J Physiol Renal Physiol.
2002;283:F525–31.
maxi-K channel [341]. Potassium secretion is regu- 15. Quigley R, Flynn M, Baum M. Neonatal and adult
lated by aldosterone, and there is resistance to the rabbit renal brush border membrane vesicle solute
action of aldosterone despite higher serum levels in reflection coefficients. Biol Neonate.
the neonate [325–331]. 1999;76:106–13.
16. Aperia A. 2011 Homer Smith Award: to serve and
protect: classic and novel roles for Na+, K+  adeno-
sine triphosphatase. J Am Soc Nephrol.
References 2012;23:1283–90.
17. Blanco G, Mercer RW. Isozymes of the Na-K-
1. Sabolic I, Valenti G, Verbavatz J-M, Van Hoek AN, ATPase: heterogeneity in structure, diversity in func-
Verkman AS, Ausiello DA, Brown D. Localization of tion. Am J Physiol. 1998;275:F633–50.
3 Renal Tubular Development 85

18. Rivard CJ, Almeida NE, Berl T, Capasso JM. The 34. Takemoto F, Cohen HT, Satoh T, Katz AI. Dopamine
gamma subunit of Na/K-ATPase: an exceptional, inhibits Na/K-ATPase in single tubules and cultured
small transmembrane protein. Front Biosci. cells from distal nephron. Pflugers Arch.
2005;10:2604–10. 1992;421:302–6.
19. Farman N. Na, K-pump expression and distribution in 35. Garg LC, Knepper MA, Burg MB. Mineralocorticoid
the nephron. Miner Electrolyte Metab. 1996;22: effects on Na-K-ATPase in individual nephron seg-
272–8. ments. Am J Physiol. 1981;240:F536–44.
20. Orlowski J, Lingrel JB. Tissue-specific and develop- 36. Kaplan JH. Biochemistry of Na,K-ATPase. Annu Rev
mental regulation of rat Na, K-ATPase catalytic alpha Biochem. 2002;71:511–35.
isoform and beta subunit mRNAs. J Biol Chem. 37. Aperia A, Larrson L. Induced development of proxi-
1988;263:10436–42. mal tubular NaKATPase, basolateral cell membranes
21. McDonough AA, Geering K, Farley RA. The sodium and fluid reabsorption. Acta Physiol Scand.
pump needs its beta subunit. FASEB J. 1984;121:133–41.
1990;4:1598–605. 38. Aperia A, Larrson L, Zetterstrom R. Hormonal induc-
22. Aperia A, Bertorello A, Seri I. Dopamine causes tion of Na-K-ATPase in developing proximal tubular
inhibition of Na+-K+-ATPase activity in rat proximal cells. Am J Physiol. 1981;241:F356–60.
convoluted segments. Am J Physiol. 1987;252: 39. Schmidt U, Horster M. Na-K-activated ATPase: activ-
F39–45. ity maturation in rabbit nephron segments dissected
23. Bertorello AM, Katz AI. Short-term regulation of in vitro. Am J Physiol. 1977;233:F55–60.
renal Na-K-ATPase activity: physiological relevance 40. Schwartz GH, Evan AP. Development of solute trans-
and cellular mechanisms. Am J Physiol. 1993;265: port in rabbit proximal tubule. III. Na-K-ATPase
F743–55. activity. Am J Physiol. 1984;246:F845–52.
24. Ewart HS, Klip A. Hormonal regulation of the Na(+)- 41. Wang ZM, Yasui M, Celsi G. Glucocorticoids regu-
K(+)-ATPase: mechanisms underlying rapid and late the transcription of Na(+)-K(+)-ATPase genes in
sustained changes in pump activity. Am J Physiol. the infant rat kidney. Am J Physiol. 1994;267:
1995;269:C295–311. C450–5.
25. Fukuda Y, Bertorello A, Aperia A. Ontogeny of the 42. Harris RC, Seifter JL, Lechene C. Coupling of Na-H
regulation of Na+, K(+)-ATPase activity in the renal exchange and Na-K pump activity in cultured rat
proximal tubule cell. Pediatr Res. 1991;30:131–4. proximal tubule cells. Am J Physiol. 1986;251:
26. Katz AI, Satoh T, Takemoto F, Cohen HT. Novel C815–24.
pathways of Na-K-ATPase regulation in kidney 43. Larsson SH, Rane S, Fukuda Y, Aperia A, Lechene
cells. Contrib Nephrol. 1993;101:7–11. C. Changes in Na influx precede post-natal increase in
27. McDonough AA, Farley RA. Regulation of Na,K- Na, K-ATPase activity in rat renal proximal tubular
ATPase activity. Curr Opin Nephrol Hypertens. cells. Acta Physiol Scand. 1990;138:99–100.
1993;2:725–34. 44. Cramb G, Cutler CP, Lamb JF, McDevitt T, Ogden
28. Nakhoul F, Thompson CB, McDonough PH, Owler D, Voy C. The effects of monensin on the
AA. Developmental change in Na, K-ATPase abundance of mRNA(alpha) and of sodium pumps in
alpha1 and beta1 expression in normal and human cultured cells. Q J Exp Physiol.
hypothyroid rat renal cortex. Am J Nephrol. 1989;74:53–63.
2000;20:225–31. 45. Fukuda Y, Aperia A. Differentiation of Na+  K+ pump
29. Satoh T, Cohen HT, Katz AI. Intracellular signaling in in rat proximal tubule is modulated by Na+  H+
the regulation of renal Na-K-ATPase. I. Role of cyclic exchanger. Am J Physiol. 1988;255:F552–7.
AMP and phospholipase A2. J Clin Invest. 46. Baum M, Dwarakanath V, Alpern RJ, Moe
1992;89:1496–500. OW. Effects of thyroid hormone on the neonatal
30. Satoh T, Cohen HT, Katz AI. Regulation of renal Na- renal cortical Na+/H+ antiporter. Kidney Int.
K-ATPase by eicosanoids: central role of the cyto- 1998;53:1254–8.
chrome P450-monooxygenase pathway. Trans Assoc 47. Henning SJ. Plasma concentrations of total and free
Am Physicians. 1992;105:86–92. corticosterone during development in the rat. Am J
31. Satoh T, Cohen HT, Katz AI. Different mechanisms of Physiol. 1978;235:E451–6.
renal Na-K-ATPase regulation by protein kinases in 48. Henning SJ, Leeper LL, Dieu DN. Circulating
proximal and distal nephron. Am J Physiol. 1993;265: corticosterone in the infant rat: the mechanism of
F399–405. age and thyroxine effects. Pediatr Res.
32. Satoh T, Cohen HT, Katz AI. Intracellular signaling in 1986;20:87–92.
the regulation of renal Na-K-ATPase. II. Role of 49. Walker P, Dubois JD, Dussault JH. Free thyroid hor-
eicosanoids. J Clin Invest. 1993;91:409–15. mone concentrations during postnatal development in
33. Satoh T, Ominato M, Cohen HT, Katz AI. Role of the the rat. Pediatr Res. 1980;14:247–9.
phospholipase C-protein kinase C pathway in proxi- 50. Capasso G, Lin JT, De Santo NG, Kinne R. Short term
mal tubule Na-K-ATPase regulation. Trans Assoc Am effect of low doses of tri-iodothyronine on proximal
Physicians. 1993;106:196–200. tubular membrane Na-K-ATPase and potassium
86 M. Baum

permeability in thyroidectomized rats. Pflugers Arch. 67. Melin K, Meeuwisse GW. Glucose-galactose malab-
1985;403:90–6. sorption. A genetic study. Acta Paediatr Scand.
51. Celsi G, Nishi A, Akusjarvi G, Aperia A. Abundance 1969;188:19–24.
of Na(+)-K(+)-ATPase mRNA is regulated by gluco- 68. Calado J, Soto K, Clemente C, Correia P, Rueff
corticoid hormones in infant rat kidneys. Am J J. Novel compound heterozygous mutations in
Physiol. 1991;260:F192–7. SLC5A2 are responsible for autosomal recessive
52. Celsi G, Wang ZM, Akusjarvi G, Aperia A. Sensitive renal glucosuria. Hum Genet. 2004;114:314–16.
periods for glucocorticoids’ regulation of Na+, K(+)- 69. Magen D, Sprecher E, Zelikovic I, Skorecki K. A
ATPase mRNA in the developing lung and kidney. novel missense mutation in SLC5A2 encoding
Pediatr Res. 1993;33:5–9. SGLT2 underlies autosomal-recessive renal
53. Garg LC, Tisher CC. Effects of thyroid hormone on glucosuria and aminoaciduria. Kidney Int.
Na-K-adenosine triphosphatase activity along the rat 2005;67:34–41.
nephron. J Lab Clin Med. 1985;106:568–72. 70. Arant Jr BS, Edelmann Jr CM, Nash MA. The renal
54. Horster M, Valtin H. Postnatal development of renal reabsorption of glucose in the developing canine kid-
function: micropuncture and clearance studies in the ney: a study of glomerulotubular balance. Pediatr Res.
dog. J Clin Invest. 1971;50:779–95. 1974;8:638–46.
55. Kon V, Hughes ML, Ichikawa I. Physiologic basis for 71. Beck JC. Glucose and sodium transport in brush-
the maintenance of glomerulotubular balance in border membrane vesicles from fetal rabbit kidney.
young growing rats. Kidney Int. 1984;25:391–6. Ann N Y Acad Sci. 1985;456:457–9.
56. Spitzer A, Brandis M. Functional and morphologic 72. Foreman JW, Medow MS, Wald H, Ginkinger K,
maturation of the superficial nephrons. Relationship Segal S. Developmental aspects of sugar transport
to total kidney function. J Clin Invest. by isolated dog renal cortical tubules. Pediatr Res.
1974;53:279–87. 1984;18:719–23.
57. Merlet-Benichou C, Pegorier M, Muffat-Joly M, 73. Schwartz GH, Evan AP. Development of solute trans-
Augeron D. Functional and morphologic patterns of port in rabbit proximal tubule. I. HCO3 and glucose
renal maturation in the developing guinea pig. Am J absorption. Am J Physiol. 1983;245:F382–90.
Physiol. 1981;36:H1467–75. 74. Haworth JC, MacDonald MS. Reducing sugars in the
58. Arant Jr BS. Developmental patterns of renal func- urine and blood of premature babies. Arch Dis Child.
tional maturation compared in the human neonate. 2004;32:417–21.
J Pediatr. 1978;92:705–12. 75. Tuvad F, Vesterdal J. The maximal tubular transfer of
59. Barfuss DW, Schafer JA. Differences in active and glucose and para-aminohippurate in premature
passive glucose transport along the proximal nephron. infants. Acta Paediatr Scand. 1953;42:337–45.
Am J Physiol. 1981;241:F322–32. 76. Beck JC, Lipkowitz MS, Abramson
60. Turner RJ, Moran A. Heterogeneity of sodium- RG. Characterization of the fetal glucose transporter
dependent D-glucose transport sites along the proxi- in rabbit kidney: comparison with the adult brush
mal tubule: evidence from vesicle studies. Am J border electrogenic Na+-glucose symporter. J Clin
Physiol. 1982;242:F406–14. Invest. 1988;82:379–87.
61. Wells RG, Pajor AM, Kanai Y, Turk E, Wright EM, 77. Kleta R, Romeo E, Ristic Z, Ohura T, Stuart C, Arcos-
Hediger MA. Cloning of a human kidney cDNA with Burgos M, Dave MH, Wagner CA, Camargo SR,
similarity to the sodium-glucose cotransporter. Am J Inoue S, Matsuura N, Helip-Wooley A,
Physiol. 1992;263:F459–65. Bockenhauer D, Warth R, Bernardini I, Visser G,
62. Hediger MA, Coady MJ, Ikeda TS, Wright Eggermann T, Lee P, Chairoungdua A, Jutabha P,
EM. Expression cloning and cDNA sequencing of Babu E, Nilwarangkoon S, Anzai N, Kanai Y,
the Na+/glucose co-transporter. Nature. Verrey F, Gahl WA, Koizumi A. Mutations in
1987;330:379–81. SLC6A19, encoding B0AT1, cause Hartnup disorder.
63. Ikeda TS, Hwang ES, Coady MJ, Hirayama BA, Nat Genet. 2004;36:999–1002.
Hediger MA, Wright EM. Characterization of a Na+/ 78. Romeo E, Dave MH, Bacic D, Ristic Z, Camargo SM,
glucose cotransporter cloned from rabbit small intes- Loffing J, Wagner CA, Verrey F. Luminal kidney
tine. J Membr Biol. 1989;110:87–95. and intestine SLC6 amino acid transporters of
64. Elsas LJ. Glucose reabsorption in familial renal gly- B0AT-cluster and their tissue distribution in Mus
cosuria and glucose-galactose malabsorption. Birth musculus. Am J Physiol Renal Physiol. 2006;290:
Defects Orig Artic Ser. 1970;6:21–2. F376–83.
65. Elsas LJ, Hillman RE, Patterson JH, Rosenberg 79. Seow HF, Broer S, Broer A, Bailey CG, Potter SJ,
LE. Renal and intestinal hexose transport in familial Cavanaugh JA, Rasko JE. Hartnup disorder is caused
glucose-galactose malabsorption. J Clin Invest. by mutations in the gene encoding the neutral amino
1970;49:576–85. acid transporter SLC6A19. Nat Genet.
66. Meeuwisse GW, Melin K. Glucose-galactose malab- 2004;36:1003–7.
sorption. A clinical study of 6 cases. Acta Paediatr 80. Bohmer C, Broer A, Munzinger M, Kowalczuk S,
Scand. 1969;58 Suppl 188:3–18. Rasko JE, Lang F, Broer S. Characterization of
3 Renal Tubular Development 87

mouse amino acid transporter B0AT1 (slc6a19). 94. Bertran J, Werner A, Moore ML, Stange G,
Biochem J. 2005;389:745–51. Markovich D, Biber J, Testar X, Zorzano A,
81. Broer A, Cavanaugh JA, Rasko JE, Broer S. The Palacin M, Murer H. Expression cloning of a cDNA
molecular basis of neutral aminoacidurias. Pflugers from rabbit kidney cortex that induces a single trans-
Arch. 2006;451:511–17. port system for cystine and dibasic and neutral amino
82. Jonas AJ, Butler IJ. Circumvention of defective neu- acids. Proc Natl Acad Sci U S A. 1992;89:5601–5.
tral amino acid transport in Hartnup disease using 95. Fernandez E, Carrascal M, Rousaud F, Abian J,
tryptophan ethyl ester. J Clin Invest. 1989;84:200–4. Zorzano A, Palacin M, Chillaron J. rBAT-b(0,+)AT
83. Ristic Z, Camargo SM, Romeo E, Bodoy S, Bertran J, heterodimer is the main apical reabsorption system for
Palacin M, Makrides V, Furrer EM, Verrey F. cystine in the kidney. Am J Physiol Renal Physiol.
Neutral amino acid transport mediated by ortholog 2002;283:F540–8.
of imino acid transporter SIT1/SLC6A20 in opossum 96. Furriols M, Chillaron J, Mora C, Castello A,
kidney cells. Am J Physiol Renal Physiol. 2006;290: Bertran J, Camps M, Testar X, Vilaro S, Zorzano A,
F880–7. Palacin M. rBAT, related to L-cystine transport,
84. Kanai Y, Nussberger S, Romero MF, Boron WF, is localized to the microvilli of proximal
Hebert SC, Hediger MA. Electrogenic properties of straight tubules, and its expression is regulated in
the epithelial and neuronal high affinity glutamate kidney development. J Biol Chem.
transporter. J Biol Chem. 1995;270:16561–8. 1993;268:27060–8.
85. Samarzija I, Fromter E. Electrophysiological analysis 97. Calonge MJ, Gasparini P, Chillaron J, Chillon M,
of rat renal sugar and amino acid transport. IV. Basic Gallucci M, Rousaud F, Zelante L, Testar X,
amino acids. Pflugers Arch. 1982;393:210–14. Dallapiccola B, Di Silverio F. Cystinuria caused by
86. Weiss SD, McNamara PD, Pepe LM, Segal mutations in rBAT, a gene involved in the transport of
S. Glutamine and glutamic acid uptake by rat renal cystine. Nat Genet. 1994;6:420–5.
brushborder membrane vesicles. J Membr Biol. 98. Calonge MJ, Volpini V, Bisceglia L, Rousaud F,
1978;43:91–105. Sanctis L, Beccia E, Zelante L, Testar X, Zorzano A,
87. Kanai Y, Hediger MA. Primary structure and func- Estivill X, Gasparini P, Nunes V, Palacin M. Genetic
tional characterization of a high-affinity glutamate heterogeneity in cystinuria: the SLC3A1 gene is
transporter. Nature. 1992;360:467–71. linked to type I but not to type III cystinuria. Proc
88. Shayakul C, Kanai Y, Lee WS, Brown D, Rothstein Natl Acad Sci U S A. 1995;92:9667–71.
JD, Hediger MA. Localization of the high-affinity 99. Pras E, Raben N, Golomb E, Arber N,
glutamate transporter EAAC1 in rat kidney. Am J Aksentijevich I, Schapiro JM, Harel D, Katz G,
Physiol. 1997;273:F1023–9. Liberman U, Pras M, Kastner DL. Mutations in the
89. Peghini P, Janzen J, Stoffel W. Glutamate transporter SLC3A1 transporter gene in cystinuria. Am J Hum
EAAC-1-deficient mice develop dicarboxylic amino- Genet. 1995;56:1297–303.
aciduria and behavioral abnormalities but no 100. Pras E, Sood R, Raben N, Aksentijevich I, Chen X,
neurodegeneration. EMBO J. 1997;16:3822–32. Kastner DL. Genomic organization of SLC3A1, a
90. Bailey CG, Ryan RM, Thoeng AD, Ng C, King K, transporter gene mutated in cystinuria. Genomics.
Vanslambrouck JM, Auray-Blais C, Vandenberg RJ, 1996;36:163–7.
Broer S, Rasko JE. Loss-of-function mutations in the 101. Feliubadalo L, Font M, Purroy J, Rousaud F,
glutamate transporter SLC1A1 cause human dicar- Estivill X, Nunes V, Golomb E, Centola M,
boxylic aminoaciduria. J Clin Invest. Aksentijevich I, Kreiss Y, Goldman B, Pras M,
2011;121:446–53. Kastner DL, Pras E, Gasparini P, Bisceglia L,
91. Sacktor B, Rosenbloom IL, Liang CT, Cheng Beccia E, Gallucci M, de Sanctis L, Ponzone A,
L. Sodium gradient- and sodium plus potassium Rizzoni GF, Zelante L, Bassi MT, George Jr AL,
gradient-dependent L-glutamate uptake in renal Manzoni M, De Grandi A, Riboni M, Endsley JK,
basolateral membrane vesicles. J Membr Biol. Ballabio A, Borsani G, Reig N, Fernandez E,
1981;60:63–71. Estevez R, Pineda M, Torrents D, Camps M,
92. Matsuo H, Kanai Y, Kim JY, Chairoungdua A, Kim Lloberas J, Zorzano A, Palacin M. Non-type I cystin-
DK, Inatomi J, Shigeta Y, Ishimine H, uria caused by mutations in SLC7A9, encoding a
Chaekuntode S, Tachampa K, Choi HW, Babu E, subunit (bo,+AT) of rBAT. Nat Genet. 1999;23:52–7.
Fukuda J, Endou H. Identification of a novel Na+  102. Font MA, Feliubadalo L, Estivill X, Nunes V,
independent acidic amino acid transporter with struc- Golomb E, Kreiss Y, Pras E, Bisceglia L,
tural similarity to the member of a heterodimeric Bisceglia L, D’Adamo AP, Zelante L, Gasparini P,
amino acid transporter family associated with Bassi MT, George Jr AL, Manzoni M, Riboni M,
unknown heavy chains. J Biol Chem. Ballabio A, Borsani G, Reig N, Fernandez E,
2002;277:21017–26. Zorzano A, Bertran J, Palacin M. Functional analysis
93. Segal S, McNamara PD, Pepe LM. Transport interac- of mutations in SLC7A9, and genotype-phenotype
tion of cystine and dibasic amino acids in renal brush correlation in non-Type I cystinuria. Hum Mol
border vesicles. Science. 1977;197:169–71. Genet. 2001;10:305–16.
88 M. Baum

103. Mizoguchi K, Cha SH, Chairoungdua A, Kim DK, 118. Youngblood GL, Sweet DH. Identification and
Shigeta Y, Matsuo H, Fukushima J, Awa Y, functional assessment of the novel murine
Akakura K, Goya T, Ito H, Endou H, Kanai organic anion transporter Oat5 (Slc22a19) expressed
Y. Human cystinuria-related transporter: localization in kidney. Am J Physiol Renal Physiol. 2004;287:
and functional characterization. Kidney Int. F236–44.
2001;59:1821–33. 119. van Aubel RA, Smeets PH, Peters JG, Bindels RJ,
104. Pfeiffer R, Loffing J, Rossier G, Bauch C, Meier C, Russel FG. The MRP4/ABCC4 gene encodes a novel
Eggermann T, Loffing-Cueni D, Kuhn LC, Verrey apical organic anion transporter in human kidney
F. Luminal heterodimeric amino acid transporter proximal tubules: putative efflux pump for urinary
defective in cystinuria. Mol Biol Cell. cAMP and cGMP. J Am Soc Nephrol.
1999;10:4135–47. 2002;13:595–603.
105. Brodehl J, Gellissen K. Endogenous renal transport of 120. Calcagno PL, Rubin MI. Renal extraction of para-
free amino acids in infancy and childhood. Pediatrics. aminohippurate in infants and children. J Clin Invest.
1968;42:395–404. 1963;42:1632–9.
106. Dustin JP, Moore S, Bigwood EJ. Chromatographic 121. Rubin MI, Bruck E, Rapoport M. Maturation of renal
studies on the excretion of amino acids in early function in childhood: clearance studies. J Clin Invest.
infancy. Metabolism. 1955;4:75–9. 1949;28:1144–62.
107. Scriver CR, Davies E. Endogenous renal clearance 122. Fawer CL, Torrado A, Guignard JP. Maturation of
rates of free amino acids in pre-pubertal children. renal function in full-term and premature neonates.
(Employing an accelerated procedure for elution Helv Paediatr Acta. 1979;34:11–21.
chromatography of basic amino acids on ion 123. Nakajima N, Sekine T, Cha SH, Tojo A,
exchange resin). Pediatrics. 1965;36:592–8. Hosoyamada M, Kanai Y, Yan K, Awa S, Endou
108. Reynolds R, Roth KS, Hwang SM, Segal S. On the H. Developmental changes in multispecific organic
development of glycine transport systems by rat renal anion transporter 1 expression in the rat kidney. Kid-
cortex. Biochim Biophys Acta. 1978;511:274–84. ney Int. 2000;57:1608–16.
109. Roth KS, Hwang SM, London JW, Segal S. Ontogeny 124. Pavlova A, Sakurai H, Leclercq B, Beier DR, Yu AS,
of glycine transport in isolated rat renal tubules. Am J Nigam SK. Developmentally regulated expression of
Physiol. 1977;233:F241–6. organic ion transporters NKT (OAT1), OCT1, NLT
110. Baerlocher KE, Scriver CR, Mohyuddin F. The (OAT2), and Roct. Am J Physiol Renal Physiol.
ontogeny of amino acid transport in rat kidney. 2000;278:F635–43.
I. Effect on distribution ratios and intracellular metab- 125. Hirsch GH, Hook JB. Maturation of renal organic acid
olism of proline and glycine. Biochim Biophys Acta. transport: substrate stimulation by penicillin. Science.
1971;249:353–63. 1969;165:909–10.
111. Medow MS, Foreman JW, Bovee KC, Segal 126. Hirsch GH, Hook JB. Maturation of renal organic acid
S. Developmental changes of glycine transport in the transport: substrate stimulation by penicillin and
dog. Biochim Biophys Acta. 1982;693:85–92. p-aminohippurate (PAH). J Pharmacol Exp Ther.
112. Chesney RW, Gusowski N, Lippincitt S, Zelikovic 1970;171:103–8.
I. Renal adaptation to dietary amino acid alteration is 127. Hirsch GH, Hook JB. Stimulation of renal organic
expressed in immature renal brush border mem- acid transport and protein synthesis by penicillin. J
branes. Pediatr Nephrol. 1988;2:146–50. Pharmacol Exp Ther. 1970;174:152–8.
113. Medow MS, Roth KS, Goldmann DR, Ginkinger K, 128. Schwartz GJ, Goldsmith DI, Fine
Hsu BY, Segal S. Developmental aspects of proline LG. p-aminohippurate transport in the proximal
transport in rat renal brush border membranes. Proc straight tubule: development and substrate stimula-
Natl Acad Sci U S A. 1986;83:7561–4. tion. Pediatr Res. 1978;12:793–6.
114. Bahn A, Prawitt D, Buttler D, Reid G, Enklaar T, 129. Biber J, Caderas G, Stange G, Werner A, Murer
Wolff NA, Ebbinghaus C, Hillemann A, Schulten H. Effect of low-phosphate diet on sodium/phosphate
HJ, Gunawan B, Fuzesi L, Zabel B, Burckhardt cotransport mRNA and protein content and on oocyte
G. Genomic structure and in vivo expression of the expression of phosphate transport. Pediatr Nephrol.
human organic anion transporter 1 (hOAT1) gene. 1993;7:823–6.
Biochem Biophys Res Commun. 2000;275:623–30. 130. Werner A, Moore ML, Mantei N, Biber J, Semenza G,
115. Launay-Vacher V, Izzedine H, Karie S, Hulot JS, Murer H. Cloning and expression of cDNA for a
Baumelou A, Deray G. Renal tubular drug trans- Na/Pi cotransport system of kidney cortex. Proc Natl
porters. Nephron Physiol. 2006;103:97–106. Acad Sci U S A. 1991;88:9608–12.
116. Sekine T, Miyazaki H, Endou H. Molecular physiol- 131. Magagnin S, Werner A, Markovich D, Sorribas V,
ogy of renal organic anion transporters. Am J Physiol Stange G, Biber J, Murer H. Expression cloning of
Renal Physiol. 2006;290:F251–61. human and rat renal cortex Na/Pi cotransport. Proc
117. Wright SH, Dantzler WH. Molecular and cellular Natl Acad Sci U S A. 1993;90:5979–83.
physiology of renal organic cation and anion trans- 132. Segawa H, Kaneko I, Takahashi A, Kuwahata M,
port. Physiol Rev. 2004;84:987–1049. Ito M, Ohkido I, Tatsumi S, Miyamoto K.
3 Renal Tubular Development 89

Growth-related renal type II Na/Pi cotransporter. in the juvenile rat. Am J Physiol Regul Integr Comp
J Biol Chem. 2002;277:19665–72. Physiol. 2001;280:R1524–33.
133. Forster IC, Hernando N, Biber J, Murer H. Proximal 149. Neiberger RE, Barac-Nieto M, Spitzer A. Renal
tubular handling of phosphate: a molecular perspec- reabsorption of phosphate during development: trans-
tive. Kidney Int. 2006;70:1548–59. port kinetics in BBMV. Am J Physiol. 1989;257:
134. Kempson SA, Lotscher M, Kaissling B, Biber J, F268–74.
Murer H, Levi M. Parathyroid hormone action on 150. Mulroney SE, Haramati A. Renal adaptation to
phosphate transporter mRNA and protein in rat renal changes in dietary phosphate during development.
proximal tubules. Am J Physiol. 1995;268:F784–91. Am J Physiol. 1990;258:F1650–6.
135. Levi M, Arar M, Kaissling B, Murer H, Biber 151. Barac-Nieto M, Corey H, Liu SM, Spitzer A. Role of
J. Low-Pi diet increases the abundance of an apical intracellular phosphate in the regulation of renal phos-
protein in rat proximal-tubular S3 segments. Pflugers phate transport during development. Pediatr Nephrol.
Arch. 1994;426:5–11. 1993;7:819–22.
136. Levi M, Lotscher M, Sorribas V, Custer M, Arar M, 152. Traebert M, Lotscher M, Aschwanden R, Ritthaler T,
Kaissling B, Murer H, Biber J. Cellular mechanisms Biber J, Murer H, Kaissling B. Distribution of the
of acute and chronic adaptation of rat renal P sodium/phosphate transporter during postnatal ontog-
(i) transporter to alterations in dietary P(i). Am J eny of the rat kidney. J Am Soc Nephrol.
Physiol. 1994;267:F900–8. 1999;10:1407–15.
137. Takahashi F, Morita K, Katai K, Segawa H, 153. Taufiq S, Collins JF, Ghishan FK. Posttranscriptional
Fujioka A, Kouda T, Tatsumi S, Nii T, Taketani Y, mechanisms regulate ontogenic changes in rat renal
Haga H, Hisano S, Fukui Y, Miyamoto KI, Takeda sodium-phosphate transporter. Am J Physiol.
E. Effects of dietary Pi on the renal Na+  dependent 1997;272:R134–41.
Pi transporter NaPi-2 in thyroparathyroidectomized 154. Spitzer A, Barac-Nieto M. Ontogeny of renal phos-
rats. Biochem J. 1998;333(Pt 1):175–81. phate transport and the process of growth. Pediatr
138. Hilfiker H, Hattenhauer O, Traebert M, Forster I, Nephrol. 2001;16:763–71.
Murer H, Biber J. Characterization of a murine type 155. Bergwitz C, Roslin NM, Tieder M, Loredo-Osti JC,
II sodium-phosphate cotransporter expressed in mam- Bastepe M, Abu-Zahra H, Frappier D, Burkett K,
malian small intestine. Proc Natl Acad Sci U S A. Carpenter TO, Anderson D, Garabedian M,
1998;95:14564–9. Sermet I, Fujiwara TM, Morgan K, Tenenhouse HS,
139. Connelly JP, Crawford JD, Watson J. Studies of Juppner H. SLC34A3 mutations in patients with
neonatal hyperphosphatemia. Pediatrics. 1962;30: hereditary hypophosphatemic rickets with
425–32. hypercalciuria predict a key role for the sodium-
140. Hohenauer L, Rosenberg TF, Oh W. Calcium and phosphate cotransporter NaPi-IIc in maintaining
phosphorus homeostasis on the first day of life. Biol phosphate homeostasis. Am J Hum Genet.
Neonate. 1970;15:49–56. 2006;78:179–92.
141. Dean RFA, McCance RA. Phosphate clearance in 156. Quigley R, Baum M. Effects of growth hormone
infants and adults. J Physiol. 1948;107:182–6. and insulin-like growth factor I on rabbit proximal
142. Richmond JB, Kravitz H, Segar W, Kravitz H. Renal convoluted tubule transport. J Clin Invest.
clearance of endogenous phosphate in infants and 1991;88:368–74.
children. Proc Soc Exp Biol Med. 1951;77:83–7. 157. Hammerman MR, Karl IE, Hruska KA. Regulation of
143. Caverzasio J, Bonjour JP, Fleisch H. Tubular handling canine renal vesicle Pi transport by growth hormone
of Pi in young growing and adult rats. Am J Physiol. and parathyroid hormone. Biochim Biophys Acta.
1982;242:F705–10. 1980;603:322–35.
144. Johnson V, Spitzer A. Renal reabsorption of phos- 158. Haramati A, Mulroney SE, Lumpkin MD. Regulation
phate during development: whole-kidney events. of renal phosphate reabsorption during development:
Am J Physiol. 1986;251:F251–6. implications from a new model of growth hormone
145. Haramati A, Mulroney SE, Webster SK. Develop- deficiency. Pediatr Nephrol. 1990;4:387–91.
mental changes in the tubular capacity for phosphate 159. Mulroney SE, Lumpkin MD, Haramati A. Antagonist
reabsorption in the rat. Am J Physiol. 1988;255: to GH-releasing factor inhibits growth and renal Pi
F287–91. reabsorption in immature rats. Am J Physiol.
146. Webster SK, Haramati A. Developmental changes in 1989;257:F29–34.
the phosphaturic response to parathyroid hormone in 160. Woda CB, Halaihel N, Wilson PV, Haramati A,
the rat. Am J Physiol. 1985;249:F251–5. Levi M, Mulroney SE. Regulation of renal NaPi-2
147. Kaskel FJ, Kumar AM, Feld LG, Spitzer A. Renal expression and tubular phosphate reabsorption by
reabsorption of phosphate during development: tubu- growth hormone in the juvenile rat. Am J Physiol
lar events. Pediatr Nephrol. 1988;2:129–34. Renal Physiol. 2004;287:F117–23.
148. Woda C, Mulroney SE, Halaihel N, Sun L, Wilson 161. DeFronzo RA, Goldberg M, Agus ZS. The effects of
PV, Levi M, Haramati A. Renal tubular sites of glucose and insulin on renal electrolyte transport.
increased phosphate transport and NaPi-2 expression J Clin Invest. 1976;58:83–90.
90 M. Baum

162. Baum M, Schiavi S, Dwarakanath V, Quigley NHE8 in the rat proximal tubule. Am J Physiol
R. Effect of fibroblast growth factor-23 on phosphate Renal Physiol. 2007;293:F255–61.
transport in proximal tubules. Kidney Int. 177. Biemesderfer D, Pizzonia J, Abu-Alfa A, Exner M,
2005;68:1148–53. Reilly R, Igarashi P, Aronson PS. NHE3: a Na+/H+
163. Bowe AE, Finnegan R, Jan de Beur SM, Cho J, exchanger isoform of renal brush border. Am J
Levine MA, Kumar R, Schiavi SC. FGF-23 inhibits Physiol. 1993;265:F736–42.
renal tubular phosphate transport and is a PHEX 178. Wu MS, Biemesderfer D, Giebisch G, Aronson
substrate. Biochem Biophys Res Commun. PS. Role of NHE3 in mediating renal brush border
2001;284:977–81. Na+  H+ exchange. Adaptation to metabolic acido-
164. Shimada T, Muto T, Hasegawa H, Yamazaki Y, sis. J Biol Chem. 1996;271:32749–52.
Takeuchi Y, Fujita T, Fukumoto S, Yamashita 179. Choi JY, Shah M, Lee MG, Schultheis PJ, Shull GE,
T. FGF-23 is a novel regulator of mineral homeostasis Muallem S, Baum M. Novel amiloride-sensitive
with unique properties controlling, vitamin D metab- sodium-dependent proton secretion in the mouse
olism and phosphate reabsorption. J Bone Miner Res. proximal convoluted tubule. J Clin Invest.
2002;17:S425. 2000;105:1141–6.
165. Berndt T, Craig TA, Bowe AE, Vassiliadis J, 180. Goyal S, Vanden Heuvel G, Aronson PS. Renal
Reczek D, Finnegan R, de Beur SMJ, Schiavi SC, expression of novel Na+/H+ exchanger isoform
Kumar R. Secreted frizzled-related protein 4 is a NHE8. Am J Physiol Renal Physiol. 2003;284:
potent tumor-derived phosphaturic agent. J Clin F467–73.
Invest. 2003;112:785–94. 181. Goyal S, Mentone S, Aronson PS. Immunoloca-
166. Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, lization of NHE8 in rat kidney. Am J Physiol Renal
Nandi A, Rosenblatt KP, Baum MG, Schiavi S, Hu Physiol. 2005;288:F530–8.
MC, Moe OW, Kuro-o M. Regulation of fibroblast 182. Zhang J, Bobulescu IA, Goyal S, Aronson PS, Baum
growth factor-23 signaling by klotho. J Biol Chem. MG, Moe OW. Characterization of Na+/H+ exchanger
2006;281:6120–3. NHE8 in cultured renal epithelial cells. Am J Physiol
167. Prie D, Beck L, Urena P, Friedlander G. Recent find- Renal Physiol. 2007;293:F761–6.
ings in phosphate homeostasis. Curr Opin Nephrol 183. Baum M, Quigley R. Maturation of proximal tubular
Hypertens. 2005;14:318–24. acidification. Pediatr Nephrol. 1993;7:785–91.
168. Baum M. Developmental changes in rabbit 184. Schwartz GJ. Physiology and molecular biology of
juxtamedullary proximal convoluted tubule acidifica- renal carbonic anhydrase. J Nephrol. 2002;15 Suppl
tion. Pediatr Res. 1992;31:411–14. 5:S61–74.
169. Preisig PA, Ives HE, Cragoe Jr EJ, Alpern RJ, Rector 185. Winkler CA, Kittelberger AM, Watkins RH,
Jr FC. Role of the Na+/H+ antiporter in rat proximal Maniscalco WM, Schwartz GJ. Maturation of car-
tubule bicarbonate absorption. J Clin Invest. bonic anhydrase IV expression in rabbit kidney. Am
1987;80:970–8. J Physiol Renal Physiol. 2001;280:F895–903.
170. Edelmann CMJ, Soriano JR, Boichis H, Gruskin AB, 186. Karashima S, Hattori S, Ushijima T, Furuse A,
Acosta M. Renal bicarbonate reabsorption and hydro- Nakazato H, Matsuda I. Developmental changes in
gen ion excretion in normal infants. J Clin Invest. carbonic anhydrase II in the rat kidney. Pediatr
1967;46:1309–17. Nephrol. 1998;12:263–8.
171. Schwartz GJ, Haycock GB, Edelmann Jr CM, Spitzer 187. Schwartz GJ, Olson J, Kittelberger AM,
A. Late metabolic acidosis: a reassessment of the Matsumoto T, Waheed A, Sly WS. Postnatal devel-
definition. J Pediatr. 1979;95:102–7. opment of carbonic anhydrase IV expression in rabbit
172. Baum M, Quigley R. Prenatal glucocorticoids stimu- kidney. Am J Physiol. 1999;276:F510–20.
late neonatal juxtamedullary proximal convoluted 188. Baum M, Moe OW, Gentry DL, Alpern RJ. Effect of
tubule acidification. Am J Physiol. 1991;261: glucocorticoids on renal cortical NHE-3 and NHE-1
F746–52. mRNA. Am J Physiol. 1994;267:F437–42.
173. Baum M. Neonatal rabbit juxtamedullary proximal 189. Baum M, Amemiya M, Dwarakanath V, Alpern RJ,
convoluted tubule acidification. J Clin Invest. Moe OW. Glucocorticoids regulate NHE-3 transcrip-
1990;85:499–506. tion in OKP cells. Am J Physiol. 1996;270:F164–9.
174. Shah M, Gupta N, Dwarakanath V, Moe OW, Baum 190. Cano A, Baum M, Moe OW. Thyroid hormone stim-
M. Ontogeny of Na+/H+ antiporter activity in rat ulates the renal Na/H exchanger NHE3 by transcrip-
proximal convoluted tubules. Pediatr Res. tional activation. Am J Physiol. 1999;276:C102–8.
2000;48:206–10. 191. Bobulescu IA, Dwarakanath V, Zou L, Zhang J,
175. Beck JC, Lipkowitz MS, Abramson RG. Ontogeny of Baum M, Moe OW. Glucocorticoids acutely increase
Na/H antiporter activity in rabbit renal brush cell surface Na+/H+ exchanger-3 (NHE3) by activa-
border membrane vesicles. J Clin Invest. tion of NHE3 exocytosis. Am J Physiol Renal
1991;87:2067–76. Physiol. 2005;289:F685–91.
176. Becker AM, Zhang J, Goyal S, Dwarakanath V, 192. Baum M, Biemesderfer D, Gentry D, Aronson
Aronson PS, Moe OW, Baum M. Ontogeny of PS. Ontogeny of rabbit renal cortical NHE3 and
3 Renal Tubular Development 91

NHE1: effect of glucocorticoids. Am J Physiol. 209. Horster M. Expression of ontogeny in individual


1995;268:F815–20. nephron segments. Kidney Int. 1982;22:550–9.
193. Shah M, Quigley R, Baum M. Maturation of proximal 210. Abuazza G, Becker A, Williams SS, Chakravarty S,
straight tubule NaCl transport: role of thyroid hor- Truong HT, Lin F, Baum M. Claudins 6, 9, and 13 are
mone. Am J Physiol Renal Physiol. 2000;278: developmentally expressed renal tight junction pro-
F596–602. teins. Am J Physiol Renal Physiol. 2006;291:
194. Gupta N, Dwarakanath V, Baum M. Maturation of the F1132–41.
Na/H antiporter (NHE3) in the proximal tubule of the 211. Sheu JN, Baum M, Bajaj G, Quigley R. Maturation of
hypothroid adrenalectomized rat. Am J Physiol Renal rabbit proximal convoluted tubule chloride perme-
Physiol. 2004;287:F521–7. ability. Pediatr Res. 1996;39:308–12.
195. Baum M, Quigley R. Maturation of rat proximal 212. Baum M, Quigley R. Thyroid hormone modulates
tubule chloride permeability. Am J Physiol Regul rabbit proximal straight tubule paracellular perme-
Integr Comp Physiol. 2005;289:R1659–64. ability. Am J Physiol Renal Physiol. 2004;286:
196. Liu FY, Cogan MG. Axial heterogeneity in the rat F477–82.
proximal convoluted tubule. I. Bicarbonate, chloride, 213. Preisig PA, Berry CA. Evidence for transcellular
and water transport. Am J Physiol. 1984;247: osmotic water flow in rat proximal tubules. Am J
F816–21. Physiol. 1985;249:F124–31.
197. Rector Jr FC. Sodium, bicarbonate, and chloride 214. Quigley R, Baum M. Water transport in neonatal and
absorption by the proximal tubule. Am J Physiol. adult rabbit proximal tubules. Am J Physiol Renal
1983;244:F461–71. Physiol. 2002;283:F280–5.
198. Alpern RJ, Howlin KJ, Preisig PA. Active and passive 215. Nielsen S, Agre P. The aquaporin family of water
components of chloride transport in the rat proximal channels in kidney. Kidney Int. 1995;48:1057–68.
convoluted tubule. J Clin Invest. 1985;76:1360–6. 216. Nielsen S, Smith BL, Christensen EI, Knepper MA,
199. Baum M, Berry CA. Evidence for neutral Agre P. CHIP28 water channels are localized in con-
transcellular NaCl transport and neutral basolateral stitutively water-permeable segments of the nephron.
chloride exit in the rabbit convoluted tubule. J Clin J Cell Biol. 1993;120:371–83.
Invest. 1984;74:205–11. 217. Nielsen S, Frokiaer J, Marples D, Kwon TH, Agre P,
200. Aronson PS, Giebisch G. Mechanisms of chloride Knepper MA. Aquaporins in the kidney: from mole-
transport in the proximal tubule. Am J Physiol. cules to medicine. Physiol Rev. 2002;82:205–44.
1997;273:F179–92. 218. Preston GM, Agre P. Isolation of the cDNA for eryth-
201. Shah M, Quigley R, Baum M. Neonatal rabbit prox- rocyte integral membrane protein of 28 kilodaltons:
imal tubule basolateral membrane Na+/H+ antiporter member of an ancient channel family. Proc Natl Acad
and Cl-/base exchange. Am J Physiol. 1999;276: Sci U S A. 1991;88:11110–14.
R1792–7. 219. Schnermann J, Chou CL, Ma T, Traynor T, Knepper
202. Sheu JN, Quigley R, Baum M. Heterogeneity of chlo- MA, Verkman AS. Defective proximal tubular fluid
ride/base exchange in rabbit superficial and reabsorption in transgenic aquaporin-1 null mice.
juxtamedullary proximal convoluted tubules. Am J Proc Natl Acad Sci U S A. 1998;95:9660–4.
Physiol. 1995;268:F847–53. 220. Bondy C, Chin E, Smith BL, Preston GM, Agre
203. Aronson PS. 1994 Homer W. Smith Award. From P. Developmental gene expression and tissue distri-
flies to physiology–accidental findings along the bution of the CHIP28 water-channel protein. Proc
trail of renal NaCl transport. J Am Soc Nephrol. Natl Acad Sci U S A. 1993;90:4500–4.
1995;5:2001–13. 221. Smith B, Baumgarten R, Nielsen S, Raben D, Zeidel
204. Aronson PS. Role of ion exchangers in mediating ML, Agre P. Concurrent expression of erythroid and
NaCl transport in the proximal tubule. Kidney Int. renal aquaporin CHIP and appearance of water chan-
1996;49:1665–70. nel activity in perinatal rats. J Clin Invest.
205. Aronson PS. Ion exchangers mediating NaCl trans- 1993;92:2035–41.
port in the renal proximal tubule. Cell Biochem 222. Quigley R, Baum M. Developmental changes in rab-
Biophys. 2002;36:147–53. bit juxtamedullary proximal convoluted tubule water
206. Aronson PS, Kuo SM. Heterogeneity of anion permeability. Am J Physiol. 1996;271:F871–6.
exchangers mediating chloride transport in the prox- 223. Mulder J, Baum M, Quigley R. Diffusional water
imal tubule. Ann N Y Acad Sci. 1989;574:96–101. permeability (PDW) of adult and neonatal rabbit
207. Kurtz I, Nagami G, Yanagawa N, Li L, Emmons C, renal brush border membrane vesicles. J Membr
Lee I. Mechanism of apical and basolateral Na(+)- Biol. 2002;187:167–74.
independent Cl/base exchange in the rabbit superfi- 224. Quigley R, Harkins EW, Thomas PJ, Baum M.
cial proximal straight tubule. J Clin Invest. Maturational changes in rabbit renal brush border
1994;94:173–83. membrane vesicle osmotic water permeability.
208. Shah M, Quigley R, Baum M. Maturation of rabbit J Membr Biol. 1998;164:177–85.
proximal straight tubule chloride/base exchange. Am 225. Quigley R, Gupta N, Lisec A, Baum M. Maturational
J Physiol. 1998;274:F883–8. changes in rabbit renal basolateral membrane vesicle
92 M. Baum

osmotic water permeability. J Membr Biol. 241. Rocha AS, Magaldi JB, Kokko JP. Calcium and phos-
2000;174:53–8. phate transport in isolated segments of rabbit Henle’s
226. Mulder J, Haddad MN, Baum M, Quigley loop. J Clin Invest. 1977;59:975–83.
R. Glucocorticoids increase osmotic water permeabil- 242. Simon DB, Lu Y, Choate KA, Velazquez H, Al
ity (Pf) in neonatal proximal tubule brush border Sabban E, Praga M, Casari G, Bettinelli A,
membrane. Am J Physiol Regul Integr Comp Physiol. Colussi G, Rodriguez-Soriano J, McCredie D,
2005;188:R1417–21. Milford D, Sanjad S, Lifton RP. Paracellin-1, a renal
227. Mulder J, Haddad MN, Vernon K, Baum M, Quigley tight junction protein required for paracellular Mg2+
R. Hypothyroidism increases osmotic water perme- resorption. Science. 1999;285:103–6.
ability (Pf) in the developing renal brush border mem- 243. Simon DB, Karet FE, Hamdan JM, DiPietro A,
brane. Pediatr Res. 2003;53:1001–7. Sanjad SA, Lifton RP. Bartter’s syndrome,
228. Hebert SC, Gamba G. Molecular cloning and charac- hypokalaemic alkalosis with hypercalciuria, is caused
terization of the renal diuretic-sensitive electroneutral by mutations in the Na-K-2Cl cotransporter NKCC2.
sodium-(potassium)-chloride cotransporters. Clin Nat Genet. 1996;13:183–8.
Investig. 1994;72:692–4. 244. Lu M, Wang T, Yan Q, Yang X, Dong K, Knepper
229. Hebert SC, Gamba G, Kaplan M. The electroneutral MA, Wang W, Giebisch G, Shull GE, Hebert
Na(+)-(K+)-Cl- cotransport family. Kidney Int. SC. Absence of small conductance K+ channel
1996;49:1638–41. (SK) activity in apical membranes of thick
230. Greger R, Schlatter E. Presence of luminal K+, a ascending limb and cortical collecting duct in
prerequisite for active NaCl transport in the cortical ROMK (Bartter’s) knockout mice. J Biol Chem.
thick ascending limb of Henle’s loop of rabbit kidney. 2002;277:37881–7.
Pflugers Arch. 1981;392:92–4. 245. Simon DB, Karet FE, Rodriguez-Soriano J, Hamdan
231. Mennitt PA, Wade JB, Ecelbarger CA, Palmer LG, JH, DiPietro A, Trachtman H, Sanjad SA, Lifton
Frindt G. Localization of ROMK channels in the rat RP. Genetic heterogeneity of Bartter’s syndrome
kidney. J Am Soc Nephrol. 1997;8:1823–30. revealed by mutations in the K+ channel, ROMK.
232. Xu JZ, Hall AE, Peterson LN, Bienkowski MJ, Nat Genet. 1996;14:152–6.
Eessalu TE, Hebert SC. Localization of the ROMK 246. Simon DB, Bindra RS, Mansfield TA, Nelson-
protein on apical membranes of rat kidney nephron Williams C, Mendonca E, Stone R, Schurman S,
segments. Am J Physiol. 1997;273:F739–48. Nayir A, Alpay H, Bakkaloglu A, Rodriguez-
233. Adachi S, Uchida S, Ito H, Hata M, Hiroe M, Soriano J, Morales JM, Sanjad SA, Taylor CM,
Marumo F, Sasaki S. Two isoforms of a chloride Pilz D, Brem A, Trachtman H, Griswold W, Richard
channel predominantly expressed in thick ascending GA, John E, Lifton RP. Mutations in the chloride
limb of Henle's loop and collecting ducts of rat kid- channel gene, CLCNKB, cause Bartter’s syndrome
ney. J Biol Chem. 1994;269:17677–83. type III. Nat Genet. 1997;17:171–8.
234. Kieferle S, Fong P, Bens M, Vandewalle A, Jentsch 247. Birkenhager R, Otto E, Schurmann MJ, Vollmer M,
TJ. Two highly homologous members of the ClC Ruf EM, Maier-Lutz I, Beekmann F, Fekete A,
chloride channel family in both rat and human kidney. Omran H, Feldmann D, Milford DV, Jeck N,
Proc Natl Acad Sci U S A. 1994;91:6943–7. Konrad M, Landau D, Knoers NV, Antignac C,
235. Kobayashi K, Uchida S, Mizutani S, Sasaki S, Sudbrak R, Kispert A, Hildebrandt F. Mutation of
Marumo F. Intrarenal and cellular localization of BSND causes Bartter syndrome with sensorineural
CLC-K2 protein in the mouse kidney. J Am Soc deafness and kidney failure. Nat Genet.
Nephrol. 2001;12:1327–34. 2001;29:310–14.
236. Estevez R, Boettger T, Stein V, Birkenhager R, 248. Zink H, Horster M. Maturation of diluting capacity in
Otto E, Hildebrandt F, Jentsch TJ. Barttin is a Cl loop of Henle of rat superficial nephrons. Am J
channel beta-subunit crucial for renal Cl Physiol. 1977;233:F519–24.
reabsorption and inner ear K+ secretion. Nature. 249. Horster M. Loop of Henle functional differentiation:
2001;414:558–61. in vitro perfusion of the isolated thick ascending
237. Bourdeau JE, Burg MB. Voltage dependence of cal- segment. Pflugers Arch. 1978;378:15–24.
cium transport in the thick ascending limb of Henle’s 250. Lee HW, Kim WY, Song HK, Yang CW, Han KH,
loop. Am J Physiol. 1979;236:F357–64. Kwon HM, Kim J. Sequential expression of NKCC2,
238. Brunette MG, Vigneault N, Carriere S. Micropuncture TonEBP, aldose reductase, and urea transporter-A in
study of magnesium transport along the nephron in developing mouse kidney. Am J Physiol Renal
the young rat. Am J Physiol. 1974;227:891–6. Physiol. 2007;292:F269–77.
239. Brunette MG, Vigneault N, Carriere S. Micropuncture 251. Stubbe J, Madsen K, Nielsen FT, Skott O, Jensen
study of renal magnesium transport in magnesium- BL. Glucocorticoid impairs growth of kidney outer
loaded rats. Am J Physiol. 1975;229:1695–701. medulla and accelerates loop of Henle differentiation
240. Imai M. Calcium transport across the rabbit thick and urinary concentrating capacity in rat kidney
ascending limb of Henle’s loop perfused in vitro. development. Am J Physiol Renal Physiol.
Pflugers Arch. 1978;374:255–63. 2006;291:F812–22.
3 Renal Tubular Development 93

252. Rane S, Aperia A. Ontogeny of Na-K-ATPase activity KCNQ1 in normal mouse kidney. Am J Physiol
in thick ascending limb and of concentrating capacity. Renal Physiol. 2007;292:F456–66.
Am J Physiol. 1985;249:F723–8. 267. Dai LJ, Ritchie G, Kerstan D, Kang HS, Cole DE,
253. Djouadi F, Wijkhuisen A, Bastin J. Coordinate devel- Quamme GA. Magnesium transport in the renal distal
opment of oxidative enzymes and Na-K-ATPase in convoluted tubule. Physiol Rev. 2001;81:51–84.
thick ascending limb: role of corticosteroids. Am J 268. Mensenkamp AR, Hoenderop JG, Bindels RJ. Recent
Physiol. 1992;263:F237–42. advances in renal tubular calcium reabsorption. Curr
254. Ellison DH, Velazquez H, Wright FS. Thiazide- Opin Nephrol Hypertens. 2006;15:524–9.
sensitive sodium chloride cotransport in early distal 269. Aperia A, Elinder G. Distal tubular sodium
tubule. Am J Physiol. 1987;253:F546–54. reabsorption in the developing rat kidney. Am J
255. Obermuller N, Bernstein P, Velazquez H, Reilly R, Physiol. 1981;240:F487–91.
Moser D, Ellison DH, Bachmann S. Expression 270. Goldsmith DI, Drukker A, Blaufox MD, Edelmann Jr
of the thiazide-sensitive Na-Cl cotransporter in CM, Spitzer A. Hemodynamic and excretory
rat and human kidney. Am J Physiol. 1995;269: response of the neonatal canine kidney to acute vol-
F900–10. ume expansion. Am J Physiol. 1979;237:F392–7.
256. Shimizu T, Yoshitomi K, Nakamura M, Imai M. Site 271. Aperia A, Broberger O, Thodenius K, Zetterstrom
and mechanism of action of trichlormethiazide in R. Renal response to an oral sodium load in newborn
rabbit distal nephron segments perfused in vitro. J full-term infants. Acta Paediatr Scand.
Clin Invest. 1988;82:721–30. 1972;61:670–6.
257. Velazquez H, Wright FS. Effects of diuretic drugs on 272. Dean RF, McCance RA. The renal response of infants
Na, Cl, and K transport by rat renal distal tubule. Am J and adults to the administration of hypertonic solu-
Physiol. 1986;250:F1013–23. tions of sodium chloride and urea. J Physiol.
258. Simon DB, Lifton RP. The molecular basis of 2004;109:81–7.
inherited hypokalemic alkalosis: Bartter’s and 273. Aperia A, Herin P, Lundin S, Melin P, Zetterstrom
Gitelman’s syndromes. Am J Physiol. 1996;271: R. Regulation of renal water excretion in newborn
F961–6. full-term infants. Acta Paediatr Scand.
259. Simon DB, Lifton RP. Ion transporter mutations in 1984;73:717–21.
Gitelman’s and Bartter’s syndromes. Curr Opin 274. McCance RA, Naylor NJ, Widdowson EM. The
Nephrol Hypertens. 1998;7:43–7. response of infants to a large dose of water. Arch
260. Simon DB, Nelson-Williams C, Bia MJ, Ellison D, Dis Child. 1954;29:104–9.
Karet FE, Molina AM, Vaara I, Iwata F, Cushner HM, 275. Rodriguez-Soriano J, Vallo A, Oliveros R,
Koolen M, Gainza FJ, Gitleman HJ, Lifton Castillo G. Renal handling of sodium in premature
RP. Gitelman’s variant of Bartter’s syndrome, and full-term neonates: a study using clearance
inherited hypokalaemic alkalosis, is caused by muta- methods during water diuresis. Pediatr Res.
tions in the thiazide-sensitive Na-Cl cotransporter. 1983;17:1013–16.
Nat Genet. 1996;12:24–30. 276. Leake RD, Zakauddin S, Trygstad CW, Fu P, Oh
261. Wang T, Agulian SK, Giebisch G, Aronson W. The effects of large volume intravenous fluid
PS. Effects of formate and oxalate on chloride absorp- infusion on neonatal renal function. J Pediatr.
tion in rat distal tubule. Am J Physiol. 1993;264: 1976;89:968–72.
F730–6. 277. Hansen JOL, Smith CA. Effects of withholding fluid
262. Chambrey R, Warnock DG, Podevin RA, Bruneval P, in the immediate post-natal period. Pediatrics.
Mandet C, Belair MF, Bariety J, Paillard 1953;12:99–113.
M. Immunolocalization of the Na+/H+ exchanger iso- 278. Polacek E, Vocel J, Neugebaurova L, Sebkova M,
form NHE2 in rat kidney. Am J Physiol. 1998;275: Vechetova E. The osmotic concentrating ability in
F379–86. healthy infants and children. Arch Dis Child.
263. Lourdel S, Paulais M, Marvao P, Nissant A, Teulon 1965;40:291–5.
J. A chloride channel at the basolateral membrane of 279. Winberg J. Determination of renal concentrating
the distal-convoluted tubule: a candidate ClC-K chan- capacity in infants and children without renal disease.
nel. J Gen Physiol. 2003;121:287–300. Acta Paediatr Scand. 1959;48:318–28.
264. Liapis H, Nag M, Kaji DM. K-Cl cotransporter 280. Boss JM, Dlouha H, Kraus M, Krecek J. The structure
expression in the human kidney. Am J Physiol. of the kidney in relation to age and diet in white rats
1998;275:C1432–7. during the weaning period. J Physiol.
265. Velazquez H, Silva T. Cloning and localization of 1963;168:196–204.
KCC4 in rabbit kidney: expression in distal convo- 281. Trimble ME. Renal response to solute loading in
luted tubule. Am J Physiol Renal Physiol. 2003;285: infant rats: relationship to anatomical development.
F49–58. Am J Physiol. 1970;219:1089–97.
266. Zheng W, Verlander JW, Lynch IJ, Cash M, Shao J, 282. Rane S, Aperia A, Eneroth P, Lundin S. Development
Stow LR, Cain BD, Weiner ID, Wall SM, Wingo of urinary concentrating capacity in weaning rats.
CS. Cellular distribution of the potassium channel Pediatr Res. 1985;19:472–5.
94 M. Baum

283. Stanier MW. Development of intra-renal solute gra- vasopressin and renin secretion in the fetal lamb.
dients in foetal and post-natal life. Pflugers Arch. Pediatr Res. 1980;14:869–71.
1972;336:263–70. 299. Weitzman RE, Fisher DA, Robillard J, Erenberg A,
284. Edelmann Jr CM, Barnett HL, Stark H. Effect of urea Kennedy R, Smith F. Arginine vasopressin response
on concentration of urinary nonurea solute in prema- to an osmotic stimulus in the fetal sheep. Pediatr Res.
ture infants. J Appl Physiol. 1966;21:1021–5. 1978;12:35–8.
285. Edelmann Jr CM, Barnett HL, Troupkou V. Renal 300. DeVane GW, Naden RP, Porter JC, Rosenfeld
concentrating mechanisms in newborn infants. Effect CR. Mechanism of arginine vasopressin release in
of dietary protein and water content, role of urea, and the sheep fetus. Pediatr Res. 1982;16:504–7.
responsiveness to antidiuretic hormone. J Anat. 301. Robillard JE, Weitzman RE, Fisher DA, Smith Jr
1960;39:1062–9. FG. The dynamics of vasopressin release and blood
286. Baum MA, Ruddy MK, Hosselet CA, Harris HW. The volume regulation during fetal hemorrhage in the
perinatal expression of aquaporin-2 and aquaporin-3 lamb fetus. Pediatr Res. 1979;13:606–10.
in developing kidney. Pediatr Res. 1998;43:783–90. 302. Robillard JE, Weitzman RE. Developmental aspects
287. Bonilla-Felix M, Jiang W. Aquaporin-2 in the imma- of the fetal renal response to exogenous arginine
ture rat: expression, regulation, and trafficking. J Am vasopressin. Am J Physiol. 1980;238:F407–14.
Soc Nephrol. 1997;8:1502–9. 303. DeVane GW, Porter JC. An apparent stress-induced
288. Knepper MA, Inoue T. Regulation of aquaporin-2 release or arginine vasopressin by human neonates.
water channel trafficking by vasopressin. Curr Opin J Clin Endocrinol Metab. 1980;51:1412–16.
Cell Biol. 1997;9:560–4. 304. Hadeed AJ, Leake RD, Weitzman RE, Fisher
289. Terris J, Ecelbarger CA, Marples D, Knepper MA, DA. Possible mechanisms of high blood levels of
Nielsen S. Distribution of aquaporin-4 water channel vasopressin during the neonatal period. J Pediatr.
expression within rat kidney. Am J Physiol. 1979;94:805–8.
1995;269:F775–85. 305. Polin RA, Husain MK, James LS, Frantz AG. High
290. Ward DT, Hammond TG, Harris HW. Modulation of vasopressin concentrations in human umbilical cord
vasopressin-elicited water transport by trafficking of blood–lack of correlation with stress. J Perinat Med.
aquaporin2-containing vesicles. Annu Rev Physiol. 1977;5:114–19.
1999;61:683–97. 306. Rees L, Forsling ML, Brook CG. Vasopressin con-
291. Ma T, Song Y, Yang B, Gillespie A, Carlson EJ, centrations in the neonatal period. Clin Endocrinol
Epstein CJ, Verkman AS. Nephrogenic diabetes (Oxf). 1980;12:357–62.
insipidus in mice lacking aquaporin-3 water channels. 307. Rajerison RM, Butlen D, Jard S. Ontogenic develop-
Proc Natl Acad Sci U S A. 2000;97:4386–91. ment of antidiuretic hormone receptors in rat kidney:
292. Ma T, Yang B, Gillespie A, Carlson EJ, Epstein CJ, comparison of hormonal binding and adenylate
Verkman AS. Generation and phenotype of a trans- cyclase activation. Mol Cell Endocrinol.
genic knockout mouse lacking the mercurial- 1976;4:271–85.
insensitive water channel aquaporin-4. J Clin Invest. 308. Ostrowski NL, Young III WS, Knepper MA, Lolait
1997;100:957–62. SJ. Expression of vasopressin V1a and V2 receptor
293. Sabolic I, Katsura T, Verbavatz JM, Brown D. The messenger ribonucleic acid in the liver and kidney of
AQP2 water channel: effect of vasopressin treatment, embryonic, developing, and adult rats. Endocrinol-
microtubule disruption, and distribution in neonatal ogy. 1993;133:1849–59.
rats. J Membr Biol. 1995;143:165–75. 309. Gengler WR, Forte LR. Neonatal development of rat
294. Yamamoto T, Sasaki S, Fushimi K, Ishibashi K, kidney adenyl cyclase and phosphodiesterase.
Yaoita E, Kawasaki K, Fujinaka H, Marumo F, Kihara Biochim Biophys Acta. 1972;279:367–72.
I. Expression of AQP family in rat kidneys during 310. Joppich R, Kiemann U, Mayer G, Haberle D. Effect
development and maturation. Am J Physiol. of antidiuretic hormone upon urinary concentrating
1997;272:F198–204. ability and medullary c-AMP formation in neonatal
295. Yasui M, Marples D, Belusa R, Eklof AC, Celsi G, piglets. Pediatr Res. 1979;13:884–8.
Nielsen S, Aperia A. Development of urinary concen- 311. Schlondorff D, Weber H, Trizna W, Fine
trating capacity: role of aquaporin-2. Am J Physiol. LG. Vasopressin responsiveness of renal adenylate
1996;271:F461–8. cyclase in newborn rats and rabbits. Am J Physiol.
296. Kim YH, Earm JH, Ma T, Verkman AS, Knepper MA, 1978;234:F16–21.
Madsen KM, Kim J. Aquaporin-4 expression in adult 312. Horster MF, Zink H. Functional differentiation of the
and developing mouse and rat kidney. J Am Soc medullary collecting tubule: influence of vasopressin.
Nephrol. 2001;12:1795–804. Kidney Int. 1982;22:360–5.
297. Leake RD, Weitzman RE, Weinberg JA, Fisher 313. Quigley R, Chakravarty S, Baum M. Antidiuretic
DA. Control of vasopressin secretion in the newborn hormone resistance in the neonatal cortical collecting
lamb. Pediatr Res. 1979;13:257–60. tubule is mediated in part by elevated phosphodies-
298. Siegel SR, Leake RD, Weitzman RE, Fisher terase activity. Am J Physiol Renal Physiol.
DA. Effects of furosemide and acute salt loading on 2004;286:F317–22.
3 Renal Tubular Development 95

314. Siga E, Horster MF. Regulation of osmotic water electrolyte balance and the function of the renin-
permeability during differentiation of inner medullary angiotensin-aldosterone system in newborn infants.
collecting duct. Am J Physiol. 1991;260:F710–16. Biol Neonate. 1979;35:60–5.
315. Bonilla-Felix M, Vehaskari VM, Hamm LL. Water 331. Vehaskari VM. Ontogeny of cortical collecting duct
transport in the immature rabbit collecting duct. sodium transport. Am J Physiol. 1994;267:F49–54.
Pediatr Nephrol. 1999;13:103–7. 332. Satlin LM. Postnatal maturation of potassium trans-
316. Bonilla-Felix M. Development of water transport in port in rabbit cortical collecting duct. Am J Physiol.
the collecting duct. Am J Physiol Renal Physiol. 1994;266:F57–65.
2004;287:F1093–101. 333. Satlin LM, Palmer LG. Apical Na+ conductance in
317. Bonilla-Felix M, John-Phillip C. Prostaglandins maturing rabbit principal cell. Am J Physiol.
mediate the defect in AVP-stimulated cAMP genera- 1996;270:F391–7.
tion in immature collecting duct. Am J Physiol. 334. Huber SM, Braun GS, Horster MF. Expression of the
1994;267:F44–8. epithelial sodium channel (ENaC) during ontogenic
318. McKinney TD, Burg MB. Bicarbonate transport by differentiation of the renal cortical collecting duct
rabbit cortical collecting tubules. Effect of acid and epithelium. Pflugers Arch. 1999;437:491–7.
alkali loads in vivo on transport in vitro. J Clin Invest. 335. Vehaskari VM, Hempe JM, Manning J, Aviles DH,
1977;60:766–8. Carmichael MC. Developmental regulation of ENaC
319. Mehrgut FM, Satlin LM, Schwartz GJ. Maturation subunit mRNA levels in rat kidney. Am J Physiol.
of. Am J Physiol. 1990;259:F801–8. 1998;274:C1661–6.
320. Evan AP, Satlin LM, Gattone VH, Connors B, 336. Watanabe S, Matsushita K, McCray Jr PB, Stokes
Schwartz GJ. Postnatal maturation of rabbit renal JB. Developmental expression of the epithelial Na+
collecting duct. II. Morphological observations. Am channel in kidney and uroepithelia. Am J Physiol.
J Physiol. 1991;261:F91–107. 1999;276:F304–14.
321. Satlin LM, Matsumoto T, Schwartz GJ. Postnatal mat- 337. Zolotnitskaya A, Satlin LM. Developmental expres-
uration of rabbit renal collecting duct. III. Peanut sion of ROMK in rat kidney. Am J Physiol. 1999;276:
lectin-binding intercalated cells. Am J Physiol. F825–36.
1992;262:F199–208. 338. Lorenz JM, Kleinman LI, Disney TA. Renal response
322. Satlin LM, Schwartz GJ. Postnatal maturation of rab- of newborn dog to potassium loading. Am J Physiol.
bit renal collecting duct: intercalated cell function. 1986;251:F513–19.
Am J Physiol. 1987;253:F622–35. 339. Lelievre-Pegorier M, Merlet-Benichou D, Roinel N,
323. Gurkan S, Estilo GK, Wei Y, Satlin LM. Potassium DeRouffignac C. Developmental pattern of water and
transport in the maturing kidney. Pediatr Nephrol. electrolyte transport in rat superficial nephrons. Am J
2007;22:915–25. Physiol. 1983;245:F15–21.
324. Constantinescu A, Silver RB, Satlin LM. H-K- 340. Satlin LM, Palmer LG. Apical K+ conductance in
ATPase activity in PNA-binding intercalated cells of maturing rabbit principal cell. Am J Physiol.
newborn rabbit cortical collecting duct. Am J Physiol. 1997;272:F397–404.
1997;272:F167–77. 341. Woda CB, Bragin A, Kleyman TR, Satlin LM. Flow-
325. Aperia A, Broberger O, Herin P, Zetterstrom dependent K+ secretion in the cortical collecting duct
R. Sodium excretion in relation to sodium intake is mediated by a maxi-K channel. Am J Physiol Renal
and aldosterone excretion in newborn pre-term and Physiol. 2001;280:F786–93.
full-term infants. Acta Paediatr Scand. 342. Drukker A, Goldsmith DI, Spitzer A, Edelmann Jr
1979;68:813–17. CM, Blaufox MD. The renin angiotensin system in
326. Beitins IZ, Bayard F, Levitsky L, Ances IG, newborn dogs: developmental patterns and response
Kowarski A, Migeon CJ. Plasma aldosterone concen- to acute saline loading. Pediatr Res. 1980;14:304–7.
tration at delivery and during the newborn period. J 343. Godard C, Geering JM, Geering K, Vallotton
Clin Invest. 1972;51:386–94. MB. Plasma renin activity related to sodium balance,
327. Fiselier TJ, Lijnen P, Monnens L, van Munster P, renal function and urinary vasopressin in the newborn
Jansen M, Peer P. Levels of renin, angiotensin I and infant. Pediatr Res. 1979;13:742–5.
II, angiotensin-converting enzyme and aldosterone in 344. Ekblad H, Kero P, Vuolteenaho O, Arjamaa O,
infancy and childhood. Eur J Pediatr. 1983;141:3–7. Korvenranta H, Shaffer SG. Atrial natriuretic peptide
328. Siegel SR, Fisher DA, Oh W. Serum aldosterone in the preterm infant. Lack of correlation with natri-
concentrations related to sodium balance in the new- uresis and diuresis. Acta Paediatr. 1992;81:978–82.
born infant. Pediatrics. 1974;53:410–13. 345. Brace RA, Miner LK, Siderowf AD, Cheung
329. Stephenson G, Hammet M, Hadaway G, Funder CY. Fetal and adult urine flow and ANF responses
JW. Ontogeny of renal mineralocorticoid receptors to vascular volume expansion. Am J Physiol.
and urinary electrolyte responses in the rat. Am J 1988;255:R846–50.
Physiol. 1984;247:F665–71. 346. Robillard JE, Weiner C. Atrial natriuretic factor in the
330. Sulyok E, Nemeth M, Tenyi I, Csaba IF, Varga F, human fetus: effect of volume expansion. J Pediatr.
Gyory E, Thurzo V. Relationship between maturity, 1988;113:552–5.
96 M. Baum

347. Ross MG, Ervin MG, Lam RW, Castro L, Leake RD, 351. Li XX, Albrecht FE, Robillard JE, Eisner GM, Jose
Fisher DA. Plasma atrial natriuretic peptide response PA. Gbeta regulation of Na/H exchanger-3 activity in
to volume expansion in the ovine fetus. Am J Obstet rat renal proximal tubules during development. Am J
Gynecol. 1987;157:1292–7. Physiol Regul Integr Comp Physiol. 2000;278:
348. Wei YF, Rodi CP, Day ML, Wiegand RC, Needleman R931–6.
LD, Cole BR, Needleman P. Developmental changes 352. Schlondorff D, Satriano JA, Schwartz GJ. Synthesis
in the rat atriopeptin hormonal system. J Clin Invest. of prostaglandin E2 in different segments of isolated
1987;79:1325–9. collecting tubules from adult and neonatal rabbits.
349. Aperia AC. Intrarenal dopamine: a key signal in the Am J Physiol. 1985;248:F134–44.
interactive regulation of sodium metabolism. Annu 353. Matson JR, Stokes JB, Robillard JE. Effects of inhi-
Rev Physiol. 2000;62:621–47. bition of prostaglandin synthesis on fetal renal func-
350. Kaneko S, Albrecht F, Asico LD, Eisner GM, tion. Kidney Int. 1981;20:621–7.
Robillard JE, Jose PA. Ontogeny of DA1 receptor- 354. Osborn JL, Hook JB, Bailie MD. Effect of saralasin
mediated natriuresis in the rat: in vivo and in vitro and indomethacin on renal function in developing
correlations. Am J Physiol. 1992;263:R631–8. piglets. Am J Physiol. 1980;238:R438–42.
Clinical Perinatal Urology
4
David A. Diamond and Richard S. Lee

Contents Diagnosis
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
Diagnostic Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 98 In a large prospective Swedish study, the inci-
Rationale for Prenatal Intervention . . . . . . . . . . . . . . . 105
dence of prenatally detected renal anomalies was
0.28 % in which two-thirds (0.18 %) were
Clinical Experience with Intervention
hydronephrosis. A British study, in which 99 %
for Prenatal Hydronephrosis . . . . . . . . . . . . . . . . . . . . . . 106
of the pregnant population in Stoke-on-Trent were
Postnatal Management of Infants with Prenatally scanned at 28 weeks’ gestation, demonstrated
Diagnosed Urologic Renal Abnormality . . . . . . . . . . 108
Unilateral Hydronephrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . 108 hydronephrosis prenatally in 1.40 % of cases,
Bilateral Hydronephrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109 which was confirmed postnatally in 0.65 % [2].
Renal Agenesis, Renal Ectopia, and Unilateral Others have reported a 1–3% incidence of
Multicystic Dysplastic Kidney . . . . . . . . . . . . . . . . . . . . . . 109 ANH [1–5]. These authors defined prenatal
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 109 hydronephrosis as an anteroposterior (A-P) diam-
eter of the renal pelvis greater than 5 mm but noted
the lack of consensus on the definition of antenatal
hydronephrosis [6–8]. With the rapid improve-
ment of ultrasound technology, the incidence of
detection of renal anomalies may be changing.
In a more recent prospective cohort study
(1999–2003), a 0.76 % incidence of urinary tract
abnormalities was detected, which was increased
as compared to an earlier cohort from the same
institution (0.3 %, 1989–1993) [9]. Many varia-
tions in the definition and management of ANH
exist in the literature and clinical practice, includ-
ing method and frequency of in utero testing,
radiographic documentation, classification, or
postnatal management [10–16].
When an abnormality of the urinary tract is
D.A. Diamond • R.S. Lee (*) determined by maternal-fetal ultrasound, several
Department of Urology, Harvard Medical School, Boston
questions should be raised by the ultrasonogra-
Children’s Hospital, Boston, MA, USA
e-mail: david.diamond@childrens.harvard.edu; richard. pher and consulting urologist. Combinations of
lee@childrens.harvard.edu

# Springer-Verlag Berlin Heidelberg 2016 97


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_3
98 D.A. Diamond and R.S. Lee

Table 1 Major diagnostic findings in prenatal imaging


Finding Comment
Kidney Hydronephrosis Assess degree
Unilateral and May be different
bilateral degrees
Parenchymal Should be less than
echogenicity spleen or liver; if
increased suggests
congenital kidney
disease. If associated
with moderate to
severe renal
enlargements suggests
genetic polycystic
kidney disease (PKD)
Fig. 1 Ultrasound appearance of normal fetal kidney with
Duplication Often with dilation of echolucent medullary pyramids distinguishable from the
upper pole; may be more echogenic cortical parenchyma. The cortical paren-
lower pole dilation chyma should be of lower echogenicity than adjacent liver
Cysts Small cysts associated or spleen
with dysplasia.
Simple cyst of upper
pole suggests
duplication with Diagnostic Findings
ureterocele, or ectopic
ureter may suggest
genetic cystic disease Kidney
(PKD or phenocopy) There are critical elements to an antenatal US
Urinoma Perinephric or exam that may help identify urologic pathology.
subcapsular A constellation of aberrations may indicate
Ureter Dilation/ Obstruction or reflux
pathology, particularly when placed in context
tortuosity
Bladder Distended Variation with time
with other clinical findings. Specific details of
Wall thickness In relation to filling the US need to be reported to assist antenatal
status counseling, which include: number, location,
Intravesical Ureterocele size, duplication, renal parenchyma
cystic structure (echogenicity), pelvic dilation, calyceal dilation,
“Keyhole” Dilated posterior urothelial thickening, and cystic disease. Kidneys
pattern urethra; PUV
should be appropriate in size for gestational age
Not visible Exstrophy
and relatively symmetric [17]. Large differences
Amniotic Absence; Impaired urine output
fluid oligohydramnios in size may indicate contralateral compensatory
Polyhydramnios May be seen with growth. Absence of the kidney in the normal
mild-moderate location may represent ectopia or agenesis/dys-
hydronephrosis plasia. The normal kidney should be elliptical in
Gender Penis/scrotum/ Sex-associated shape and have distinctive internal echolucency,
testes conditions (e.g., PUV)
representative of normal medullary pyramids
Spine Meningocele Neural tube defect
(Fig. 1). The appearance of the medullary pyra-
PUV posterior urethral valves
mids should not be confused with renal calyceal
dilation. The echogenicity of the kidney should be
slightly less than the corresponding spleen or
specific findings direct the differential diagnosis liver. Abnormalities of echogenicity with or with-
and permit more accurate prognosis and tailoring out hydronephrosis may indicate congenital renal
of postnatal evaluation. The principal findings and disease (genetic, environmental, syndromic, or
their implications are listed in Table 1. due to intrauterine insult).
4 Clinical Perinatal Urology 99

indicate a pop-off mechanism, which may protect


the kidney, particularly in cases of lower urinary
tract obstruction [19].

Amniotic Fluid
Critical to the evaluation of the urinary tract is the
assessment of the amniotic fluid (AF) level and
changes during pregnancy. After 16 weeks, AF
production shifts from placental transudate to fetal
urine, and by 20–22 weeks the vast majority of AF
is fetal urine [20]. Consequently, reduced AF or
oligohydramnios identified after 18–20 weeks
gestation may be a result of urinary tract obstruc-
tion or poor renal development [21].
Fig. 2 Unilateral hydroureteronephrosis at 35 weeks.
Note the dilated ureter (U ) and bladder (B). Postnatal External Genitalia
imaging confirmed this to be a ureterovesical junction Proper identification of the external genitalia can
obstruction
also be very valuable in cases of gender-specific
diagnosis, such as PUV. In cases of virilization
Ureter, Bladder, and Urinoma (e.g., congenital adrenal hyperplasia), a clitoris
Findings of ureteral dilation, bladder filling and may appear as a small phallus; and therefore the
emptying, bladder wall thickness, intravesical presence of scrotal testes is critical for male gen-
cystic structures, dilation of the posterior urethra der assignment [22–24]. Megalourethra, or a
(keyhole sign), urinoma, amount of amniotic dilated, elongated penile urethra, may be an iso-
fluid, intra-abdominal/pelvic mass, and external lated anomaly or associated with prune belly syn-
genitalia should be noted. Hydroureter is best drome [25] (Fig. 5). A female fetus with bilateral
seen in cross section of a full bladder but is often renal obstruction and bladder outlet obstruction
difficult to detect (Fig. 2). Duplication and ureteral would suggest a cloacal anomaly [26–28].
ectopia may also make this a difficult diagnosis.
Although the bladder is sometimes difficult to Hydronephrosis
image well, visualization of the bladder can be Hydronephrosis, or dilation of the renal pelvis, is
very informative, as a full bladder implies renal the most common urologic abnormality found on
function. Inability to identify the bladder on repeat US. Numerous grading systems have been devel-
studies should raise the question of bladder oped; however, there is no consensus on the best
exstrophy. Increased bladder wall thickness may and most consistent method of reporting ANH
indicate outlet obstruction, and dilation of the (Fig. 6). Measurement of APD has been used
posterior urethra (keyhole sign) is strongly sug- widely; however, there have been no formal stud-
gestive of posterior urethral valves (PUV) (Fig. 6). ies to determine the inter- and intra-observer
In duplex systems, ureteroceles may present as an reproducibility of ANH measurement, and APD
intravesical cystic structure and a dilated fails to describe pelvic configuration, calyceal
upper pole. dilation, and the laterality of findings, which
Perirenal urinoma may indicate an obstructive should be included. APD can be affected by ges-
condition [18] (Figs. 3c and 4). Typically it will tational age, hydration status of the mother, blad-
have the appearance of an anechoic structure der hypertonicity, and degree of bladder
around the kidney or be in a subcapsular location. distention. Since the dimensions of the renal pel-
When identified, a urinoma or urinary ascites is vis may normally increase with gestational age,
often associated with cases of severe bladder most investigators have adjusted threshold APD
obstruction, or PUV, in which the urinoma may values for early and later gestational age.
100 D.A. Diamond and R.S. Lee

Fig. 3 Images of a fetus with posterior urethral valves. (a) arrow). (c) Further imaging revealed a perinephric
Depicts bilateral echogenic kidneys with hydrouretero- urinoma (white arrow) surrounding the hydronephrotic
nephrosis (arrow). (b) Demonstrates the associated thick- kidney (black arrow)
ened bladder wall (black arrow) and hydroureter (white

degrees of ANH have the potential to


progress [29].
Varying the minimal APD threshold can sig-
nificantly alter the specificity and sensitivity of
APD as a measure of ANH and postnatal pathol-
ogy. To date there is no consensus on the optimal
APD threshold for determining the need for post-
natal follow-up. An APD cutoff of 15 mm for
determining obstruction yielded a postnatal sensi-
tivity of 73 % and specificity of 82 % [30]. A late
gestational age APD cutoff of 10 mm would
detect approximately 23 % of abnormal kidneys,
Fig. 4 Appearance of a fetal perinephric urinoma flatten- whereas a cutoff of 7 mm detected 68 % [31]. One
ing the contour of the kidney large systematic review estimated that only
11.9 % of total pathology presented with late
Unfortunately, a simple threshold APD value gestational age APD less than 9 mm while 39 %
which separates normal from abnormal does not of total pathology was noted at APD levels less
exist, as even severe cases of ANH have the than 15 mm [16]. Nearly identical results have
potential to resolve without incident, while mild been demonstrated by other investigators [32].
4 Clinical Perinatal Urology 101

What appears certain is that lower cutoffs will be


more sensitive in detecting postnatal pathology
but will incur a higher false-positive rate.

Categorizing ANH by APD


There is near agreement that APD greater
than 15 mm represents severe or significant
hydronephrosis, and some would also agree that
a lower threshold of 4–5 mm is an appropriate
value for considering APD to be abnormal
[16, 32–36]. Taking the above-noted limitations
into consideration, we define ANH in the 2nd and
3rd trimester using APD thresholds for which
the best available evidence provides prognostic
information (Table 2).

Diagnostic Accuracy
Predicting accurate postnatal diagnosis and out-
come, regardless of the prenatal information, still
remains challenging. The importance of accurate
Fig. 5 Fetal ultrasound appearance of a male with a diagnosis is particularly critical in cases where
dilated and patulous urethra typical of a megalourethra.
fetal intervention is considered, such as posterior
This may be seen in prune belly syndrome as well as in
isolation. This child had marked vesicoureteral reflux and urethral valves (PUV). In other cases, the ability
bilateral hydroureteronephrosis to identify some degree of ANH is adequate to
permit a postnatal evaluation if deemed clinically
appropriate.
A recent systematic review of the ANH litera-
ture attempted to determine the risk of a patholog-
ical diagnosis for patients with varying severities
of ANH [16]. In this review of 1,308 patients with
any ANH and postnatal radiographic follow-up,
36 % had a postnatal pathological diagnosis. The
degree of ANH was defined by the anterior-
posterior diameter (APD) identified in a particular
trimester (Table 2) [16]. The overall risk for any
pathology increased with the degree of
hydronephrosis, except for vesicoureteral reflux
which remained consistent regardless of the
degree of ANH (Table 3) [16].
Although the risk of pathology with degrees
of ANH appears to be increased, accurately
determining the diagnosis remains difficult [16].
Earlier studies reported false-positive rates up to
22 %. The majority of false positives were
nonobstructive causes of hydronephrosis, such
as high-grade reflux, large, nonobstructed,
Fig. 6 Unilateral renal pelvis dilation with no ureteral extrarenal pelves, or transient hydronephrosis
dilation in 37-week-old fetus [6, 37]. Similarly, the diagnosis of vesicoureteral
102 D.A. Diamond and R.S. Lee

reflux is extremely challenging to make in utero, In another population-based series, the sensitivity
as evidenced by the fact that the risk for in detecting valves was as low as 23 % [6].
vesicoureteral reflux is the same regardless of the Increased renal echogenicity and decreased
degree of ANH [16]. amniotic fluid have been suggested to be indica-
As another example, the accurate diagnosis of tive of obstructive conditions [39]. Although
posterior urethral valves (PUV), in which inter- the hallmark signs of an in utero diagnosis of
vention might be considered, has proven difficult. posterior urethral valves have been described
In one series, the false-positive rate was as high as (oligohydramnios, dilated posterior urethra, thick-
58 %, but the criteria for diagnosing valves were ened bladder, and hydroureteronephrosis), there
quite liberal and perhaps inappropriate [38]. are very few studies that have prospectively
examined the clinical urologic implications of
Table 2 Classification of antenatal hydronephrosis these findings alone or in combination [16].
(ANH) by anterior-posterior diameter (APD)
APD Ureteropelvic Junction Obstruction
ANH 2nd trimester 3rd trimester The basic features of ureteropelvic junction
classification (mm) (mm) obstruction (UPJO) in the fetus include dilation
1. Mild 7 9 of the renal pelvis and collecting system with no
2. Mild/ <10 <15 evidence of ureteral dilation (Fig. 6). The thresh-
moderate old for recommending postnatal follow-up is
3. Moderate 7–10 9–15
largely arbitrary, and currently there are no long-
4. Moderate/ 7 9
term prospective studies to best determine the
severe
5. Severe 10 15
degree of postnatal evaluation. Increasing severity
of ANH increased the risk of identifying postnatal

Table 3 Risk of pathology by degree of antenatal hydronephrosis


Degree of antenatal hydronephrosis
Postnatal pathology Mild Mild-moderate Moderate Moderate-severe Severe Trend
[% (95 % CI)/a] (N = 587) (N = 213) (N = 235) (N = 179) (N = 94) P-valueb
Any pathology 11.9 (4.5, 39.0 (32.6, 45.1 (25.3, 72.1 (47.6, 88.0) 88.3 <0.001
28.0) 45.7) 66.6) (53.7,
98.0)
UPJ 4.9 (2.0, 13.6 (9.6, 18.9) 17.0 (7.6, 36.9 (17.9, 61.0) 54.3 <0.001
11.9) 33.9) (21.7,
83.6)
VUR 4.4 (1.5, 10.8 (7.3, 15.7) 14.0 (7.1, 12.3 (8.4, 17.7) 8.5 (4.7, 0.10
12.1) 25.9) 15.0)
PUV 0.2 (0.0, 0.9 (0.2, 3.7) 0.9 (0.2, 6.7 (2.5, 16.6) 5.3 (1.2, <0.001
1.4) 2.9) 21.0)
Ureteral obstruction 1.2 (0.2, 11.7 (8.1, 16.8) 9.8 (6.3, 10.6 (7.4, 15.0) 5.3 (1.4, 0.025
8.0) 14.9) 18.2)
Otherc 1.2 (0.3, 1.9 (0.7, 4.9) 3.4 (0.5, 5.6 (3.0, 10.2) 14.9 (3.6, 0.002
4.0) 19.4) 44.9)
a
Pointwise 95 % confidence intervals were estimated by logistic regression with robust standard errors based on
generalized estimating equations with a working independence correlation structure to adjust for clustering by study
for all degrees of antenatal hydronephrosis except mild to moderate. Because only one study had subjects with mild-
moderate antenatal hydronephrosis, the pointwise 95 % confidence intervals had to be estimated using logistic regression
with unadjusted standard errors
b
Testing for trend in risks with increasing degree of antenatal hydronephrosis using logistic regression with robust
standard errors based on generalized estimating equations with a working independence correlation structure
c
Includes prune belly syndrome, VATER syndrome, solitary kidney, renal mass, and unclassified
4 Clinical Perinatal Urology 103

Fig. 7 Two different fetal images (a) at 19 weeks and (b) at 26 weeks of a multicystic dysplastic kidney with multiple
noncommunicating cysts

UPJO [16]. In the case of significant unilateral indications for surgical intervention [44]. Mild
hydronephrosis, currently, there is little rationale degrees of renal pelvic dilatation may resolve in
for in utero intervention. In a few cases with utero. Mandell et al. noted this occurred in 23 % of
massive dilation, therapeutic aspiration has been cases, with 66 % remaining stable and 9 % wors-
recommended for dystocia. In the case of bilateral ening over the course of the pregnancy [42]. Sim-
UPJO, the efficacy of in utero intervention is ilarly, Lee et al. noted that 88.1 % of mild ANH
difficult to assess. were found to have no postnatal pathology
Attempts to correlate prenatal ultrasound [16]. Severe forms of UPJO may be associated
appearance with postnatal outcomes have been with urinary ascites or perinephric urinomas,
complicated by the long-standing controversy which may be a predictor of nonfunction of the
regarding postnatal evaluation and management kidney [19, 45].
of UPJO. Grignon et al. developed a system of
grading hydronephrosis secondary to UPJO based Cystic Kidneys
on the APD and degree of calyceal dilatation The distinction between severe unilateral
[40, 41]. Mandell et al. attempted to correlate the hydronephrosis and a multicystic dysplastic kid-
degree of APD relative to gestational age with ney may occasionally be unclear. The findings of
subsequent need for postnatal surgical interven- multiple noncommunicating cysts, minimal or
tion [42]. They found the “at-risk” diameter to be absent renal parenchyma, and the absence of a
greater than or equal to 5 mm at 15–20 weeks’ central large cyst are diagnostic of a multicystic
gestation, greater than or equal to 8 mm at 20–30 dysplastic kidney (MCDK) (Fig. 7). Real-time
weeks’ gestation, and greater than 1 cm at over examination of the kidneys to help determine the
30 weeks’ gestation. An alternative system pro- communication of the calyces is essential. MCDK
posed by Kleiner et al. defined hydronephrosis as may be present in any ectopic location but typi-
the ratio of APD to A-P diameter of the kidney as cally are in the normal position. In addition,
being greater than 0.5 cm [43]; caliectasis was MCDK can be present in a duplex kidney, typi-
later added as an additional indicator of significant cally the upper pole. If detected early in the preg-
hydronephrosis. Others have suggested that nancy, the MCDK will involute over a period of
regardless of the degree of ANH, moderate or time either prenatally or postnatally [45].
severe postnatal hydronephrosis with evidence Bilaterally enlarged echogenic kidneys, partic-
of decreased renal function should be the ularly if associated with hepatobiliary dilatation or
104 D.A. Diamond and R.S. Lee

Fig. 8 Bilateral markedly enlarged echogenic (“bright”) kidneys (a) with a small cystic lesion (b) in a fetus with
oligohydramnios, secondary to autosomal recessive polycystic kidney disease

oligohydramnios, suggests autosomal recessive along with varying degrees of renal pelvic and
polycystic kidney disease (Fig. 8). Typically this calyceal dilation (Fig. 2). The best way to detect
is identified prior to 20 weeks’ gestation, but later ureteral dilation is at the level of the bladder,
development has been reported [42, 46, 47]. A preferably in the transverse view. It is not uncom-
more challenging finding is normal-sized, dif- mon for the distal ureter to be more dilated as
fusely echogenic kidneys that are not associated compared proximally. More extreme cases may
with other urologic lesions [48, 49]. Estroff be confused with single system ectopic ureters,
et al. described 19 cases (14 bilateral), particularly in males. In general, the differentia-
including 10 with normal function who survived tion is made postnatally.
and 4 with autosomal recessive polycystic kid-
neys who died [50]. In a separate multicenter Duplication Anomalies
retrospective study of 93 fetuses with Duplication anomalies are often recognized on the
hyperechogenic kidneys and a later diagnosis of basis of upper pole hydroureteronephrosis, asso-
nephropathy of varying etiology, only 1/3 of the ciated with either an obstructing ureterocele
fetuses had renal cysts irrespective of their within the bladder or ectopic ureter inserting out-
diagnosis. Twenty-eight had ARPKD (only side of the bladder [52] (Fig. 9). The upper pole
3 with cysts) and 31 had ADPKD (9 with cysts). may appear as a cystic dysplastic unit without
Typically, those with ADPKD appeared to have hydronephrosis and a concomitant ureterocele.
moderately enlarged hyperechogenic kidneys This is often termed ureterocele disproportion
with increased corticomedullary differentiation. [53]. Occasionally, lower pole dilation is due to
Additionally, as opposed to renal cyst character- ureteral obstruction by a very large ureterocele of
istics, associated malformations were the most the upper pole ureter. In cases of a very large
helpful clue to identify a diagnosis [51]. See also ureterocele, the ureterocele may be mistaken for
▶ Chap. 36, “Childhood Polycystic Kidney the bladder. Of note, single system ureteroceles
Disease.” are more likely to occur in boys and have variable
degrees of hydronephrosis of the entire kidney.
Ureterovesical Junction Obstruction Lower pole hydronephrosis may also be present
Less common than UPJO, ureterovesical obstruc- as a result of vesicoureteral reflux or more rarely a
tion (UVJ) is characterized by ureteral dilation lower pole UPJO.
4 Clinical Perinatal Urology 105

Fig. 9 Fetal image of duplex kidney with marked upper pole hydronephrosis (arrow) in contrast to a normal lower pole
(a). Associated with this image is the finding of a ureterocele (arrow) within the bladder (b)

Vesicoureteral Reflux mandates prompt postnatal intervention, and in


A firm diagnosis of vesicoureteral reflux (VUR) some cases, prenatal intervention may be
based on prenatal ultrasound is extremely difficult warranted. Fetal sonographic findings of PUV
to make, although intermittent hydronephrosis or include bilateral hydroureteronephrosis, a thick-
hydroureter is suggestive of VUR. Vesicoureteral walled bladder with a dilated posterior urethra,
reflux may be present in as many as 38 % of and, in more severe cases, dysplastic renal paren-
children with prenatal hydronephrosis [54]. Reflux chymal changes with perinephric urinomas
occurred in 42 % of children in whom postnatal and urinary ascites (Fig. 3) [58]. When character-
imaging revealed persistent upper tract abnormali- istic sonographic findings are present, the differ-
ties and in 25 % of those with normal findings on ential diagnosis includes prune belly syndrome
postnatal ultrasound but having a history of prena- (with or without urethral atresia), massive
tal dilation. Tibballs and Debrun reported that in vesicoureteral reflux, and certain cloacal anoma-
patients with prenatal dilation and postnatally nor- lies (in genetic females) [59–61]. Prenatal diag-
mal renal units by ultrasound, 25 % had grade III–V nostic accuracy for PUV is far from perfect but is
reflux [55]. The incidence of high-grade reflux was probably better than what was previously reported
greater in males than in females as noted in previ- (40 %) [38].
ous studies. In two systematic reviews of the ANH
literature, a 10–15 % incidence of VUR was iden-
tified regardless of the degree of ANH [16, 56], Rationale for Prenatal Intervention
indicating that ANH is not indicative of VUR and
may not be the appropriate trigger for postnatal The scientific rationale for prenatal treatment of
evaluation. In a neonate with prenatally detected hydronephrosis is to maximize normal develop-
hydronephrosis, the importance of diagnosing ment of renal and pulmonary function. These two
vesicoureteral reflux remains controversial, as aspects of fetal development are closely linked
VUR diagnosed on postnatal evaluation for ANH because urine comprises 90 % of amniotic fluid
is associated with earlier resolution of VUR [57]. volume, and oligohydramnios during the third
trimester has been causally related to pulmonary
Posterior Urethral Valves hypoplasia. Before embarking on prenatal surgi-
Perhaps the most important diagnosis to be made cal intervention for obstructive uropathy, it is crit-
prenatally is that of posterior urethral valves ical to assess the risk-benefit ratio. The time of
(PUV) in the male fetus. PUV, at the very least, onset of oligohydramnios has been shown to be an
106 D.A. Diamond and R.S. Lee

Table 4 Prenatal assessment of renal functional prognosis


Good Poor
Amniotic fluid Normal to moderately decreased Moderate to severely decreased
Sonographic appearance of kidneys Normal to echogenic Echogenic to cystic
Fetal urine Glick Johnson Glick Johnson
et al. [64] et al. [65] et al. [64] et al. [65]
Sodium (mEq/L) <100 <100 >100 >100
Chloride (mEq/L) <90 – >90 –
Osmolarity (mOsm/L) <210 <200 >210 >200
Calcium (mg/dL) – <8 – >8
β2-Microglobulin (mg/L) – <4 – >4
Total protein (mg/dL) – <20 – >20
Output (mL/h) >2 – <2 –
Sequential improvement in urinary – X – –
values
X, only in this series was the criterion used

important determinant of outcome [62, 63]. In a predictor of postnatal renal outcome demon-
fetuses in which adequate amniotic fluid was strated that there was insufficient evidence to sup-
documented at up to 30 weeks’ gestation in asso- port its effectiveness to predict renal function
ciation with a urologic abnormality, pulmonary [68]. From 23 studies, they identified
outcomes were satisfactory, and postnatal clinical 572 women, from which the two most accurate
problems were related to renal disease. It seems fetal urine tests were urinary calcium and sodium.
inappropriate to recommend late urinary tract β2-microglobulin was found to be less
decompression from a pulmonary or renal basis. accurate [68].
It is unclear whether early delivery, to permit earlier Findings on antenatal ultrasound have also
postnatal urinary decompression, is beneficial. been examined to predict long-term outcome
The most widely accepted indicator of salvage- postnatal renal function. In a systematic review
able renal function is analysis of fetal urine. When of 13 articles encompassing 251 women,
the urinary sodium is less than 100 mg/dL and oligohydramnios and the appearance of the renal
urine osmolarity less than 200 mOsm/dL, renal cortex (increased echogenicity or cystic changes)
function appears to be salvageable with in utero at the diagnosis of LUTO were the best factors to
intervention (Table 4) [64]. Serial aspirations of predict poor renal function (defined as Cr > 1.2
fetal urine have been reported to yield more valu- mg/dl) [69]. In this particular study, gestational
able results [65]. Guez et al. reported ten fetuses age at diagnosis (< 24 weeks) was not predictive
who underwent multiple urine samplings and in of renal function, which may be more of a reflec-
whom severe obstruction reduced sodium and tion of the inherent variability of the available
calcium reabsorption [66]. They concluded that literature.
fetal urinary chemistries were reasonably predic-
tive of severe but not moderate postnatal renal
impairment. Other investigators have suggested Clinical Experience with Intervention
the use of fetal urinary β2-microglobulin as an for Prenatal Hydronephrosis
indicator of tubular damage. Using this parameter,
poor renal outcome has been predicted with a The ability to diagnose severe prenatal
specificity of 83 % and sensitivity of 80 % [67]. hydronephrosis and advances in fetal intervention
The accuracy of these predictors has been chal- helped develop prenatal surgery for obstructive
lenged. A 2007 systematic review of fetal urine as uropathy. In 1982, Harrison et al. described the
4 Clinical Perinatal Urology 107

initial report of fetal surgery in a 21-week-old Table 5 Prenatal intervention for hydronephrosis
fetus with bilateral hydroureteronephrosis sec- Indications (prerequisites) Contraindications
ondary to PUV [70]. After the 1986 report of the Presumed obstructive Unilateral hydronephrosis
International Fetal Surgery Registry in which out- hydronephrosis, persistent with an adequately
comes did not seem to justify risk, a de facto or progressive, bilateral or functioning contralateral
in solitary unit kidney
moratorium on in utero urinary tract shunting
Otherwise healthy fetus Chromosomal
evolved [71]. abnormalities or presence
With improved technology and renewed inter- of associated severe
est in fetal shunting, most cases have been referred anomalies
to a small number of highly specialized centers Oligohydramnios Bilateral hydronephrosis
without oligohydramnios
actively engaged in prenatal surgery. The initial
No overt renal dysplasia Severely dysplastic
method of decompression with open surgery has kidneys
largely been replaced by in utero shunt placement. Adequate renal functional Evidence or urethral
Current practice uses the Rodeck shunt which lies potential based on urinary atresia
flat against the abdomen to minimize shunt dis- indices (see text)
lodgement. Complications included shunt dis- Informed consent Presence of a normal twin
lodgement and, in the case of the double-J shunt,
bowel herniation [72]. Amnioinfusion may be
needed to improve visualization for shunt place-
ment; however, this may lead to excessive fetal prenatal obstructive uropathy (Table 5) [79]. Serial
movement. Occasionally the fetus needs to be bladder sampling over 3 days has been used to
paralyzed for accurate placement. Very large blad- help determine if the fetus is a viable candidate.
ders may cause the shunt to be placed too high in The serial nature of the procedure allows one to
the abdomen resulting in dislodgement from the see the response of the fetal kidneys to bladder
bladder after it decompresses. decompression [65]. The principal reason for con-
Some investigators have explored the use of sidering vesicoamniotic shunting is to prevent
fetoscopic methods for direct intervention to pro- early neonatal pulmonary insufficiency and
vide prolonged bladder drainage, whereas others death. The risks that one accepts with intervention
have attempted direct endoscopic valve ablation include induction of premature labor, perforation
[73–77]. The proposed advantage of fetoscopic of fetal bowel and bladder, and fetal and/or mother
intervention over vesicoamniotic shunting is to hemorrhage and infection.
improve drainage and to restore normal cycling More recently, the ability to influence renal
of the bladder. There are no studies to determine if outcome in male patients with PUV but without
this method of decompression is adequate in the oligohydramnios has been suggested as a possible
face of significant prenatal bladder dysfunction. indication for in utero intervention. The principal
Furthermore, fetoscopic intervention also intro- goal of intervention is not to prevent pulmonary
duces the additional potential for iatrogenic injury hypoplasia and deaths but to prevent or delay
to the urethra, bladder neck, or external urethral end-stage renal failure. Although some reports
sphincter. In a systematic review of the literature, have shown promise in the ability to distinguish
four papers totaling 63 patients identified that fetal those fetuses with likely early renal failure from
cystoscopy as compared to vesicoamniotic shunts those with later-onset failure, the specificity and
had no significant improvement in perinatal sur- accuracy of methods using a combination of ultra-
vival [78]. Overall, experience with fetoscopic/ sound and urinary chemistries (sodium, β2-
endoscopic valve ablation is currently at the case microglobulin, and calcium) has not been well
report and experimental level, and long-term out- defined [80–82]. In summary, precise identifica-
come data is unknown. tion of those situations in which intervention may
Harrison et al. have clearly outlined the indi- benefit the fetus with obstructive uropathy
cations and contraindications of intervention for remains unclear.
108 D.A. Diamond and R.S. Lee

To date, the reported long-term outcomes of renal function. To further improve outcomes,
antenatal intervention for severe obstructive more sensitive and specific markers to better iden-
uropathy (e.g., PUV, prune belly syndrome, ure- tify which fetus will benefit from in utero shunting
thral atresia) are mixed [83–93]. Variability in need to be defined.
patient selection and assessment of outcome
within these studies has limited the ability to
determine if prenatal intervention has altered the Postnatal Management of Infants
postnatal course. A large systematic review of the with Prenatally Diagnosed Urologic
prenatal intervention for obstructive uropathy Renal Abnormality
showed a statistically significant perinatal sur-
vival advantage with shunting [89]; however, A child with a prenatal diagnosis of a urologic
lack of randomization of patient selection in the renal abnormality such as ANH should be care-
trails reviewed may have biased the results. Of the fully evaluated and followed by a pediatri-
studies that have reported long-term outcomes of curologist from birth. The vast majority of these
in utero vesicoamniotic shunting, many of the children appear entirely healthy and, in the
children have renal insufficiency (57 %) and absence of prenatal ultrasound findings, would
growth impairment (86 %) [83, 85, 86]. Recently, not have any indications for regular urologic
Baird et al. reported on long-term follow-up (5.8 follow-up. Parental anxiety is common and
years) of patients who have survived in utero should be addressed directly with prenatal
shunting [83]. They noted acceptable renal func- counseling and education.
tion in 44 %, 22 % with mild impairment, and
33 % with renal failure. Prune belly patients had
the best renal outcome (57 %), followed by PUV Unilateral Hydronephrosis
(43 %) and then urethral atresia (25 %).
The European-based multicenter randomized The presence of unilateral dilation of the kidney
clinical trial (PLUTO Trial: Percutaneous warrants postnatal evaluation in a timely but
Shunting for Lower Urinary Tract Obstruction) nonurgent fashion (3–8 weeks of life) with an
was recently conducted [94]. The initial goal ultrasound [80]. The most common diagnoses
was to enroll 150 singleton pregnancies with associated with this finding are UPJO, VUR, and
ultrasound evidence of LUTO to evaluate the UVJO/megaureter. Early ultrasound is unlikely to
safety and effectiveness of vesicoamniotic shunt miss a significant abnormality. A normal postnatal
(VAS) as compared to conservative management. ultrasound indicates that obstructive uropathy is
The study was stopped early because of poor not present; however, it does not clarify if the
enrollment, as only 31 patients were enrolled child has VUR [55].
and randomized (16 VAS and 15 controls). In The decision to perform a voiding cystoure-
the VAS group, there were 12 live births and throgram (VCUG) or initiate prophylactic antibi-
4 postnatal deaths at 28 days and and 12 live births otics in the newborn period is unclear. Although
and 8 postnatal deaths in the control group. All some groups advocate postnatal VCUG in any
postnatal deaths were from pulmonary hypopla- child with a history of antenatal hydronephrosis,
sia. Although underpowered, the study demon- others have questioned the value of this approach
strated a nonsignificant increase in survival in [95]. Although various guidelines and recommen-
the VAS group. Consistent with the results of the dations have been proposed with regard to VCUG
systematic review of existing prenatal interven- usage, there are no definitive studies that have
tion data, there was minimal likelihood of surviv- rigorously studied the likelihood of VUR based
ing with long-term normal renal function. on consistent ultrasound findings or other clinical
Overall, it appears that in utero intervention for characteristics. The current trend in management
the appropriate patient may reduce the risk of of ANH minimizes the postnatal prophylactic
neonatal mortality and may potentially improve antibiotics and testing for VUR in cases of
4 Clinical Perinatal Urology 109

resolved ANH or mild to moderate cases of per- corrected promptly. For children with suspected
sistent postnatal ANH [96]. lower urinary tract obstruction (e.g., PUV),
In general, infants with severe ANH should be prompt bladder decompression and antibiotic pro-
placed on a prophylactic antibiotic (amoxicillin, phylaxis should be initiated prior to radiographic
10–15 mg/kg/day) and undergo a VCUG. Severe intervention.
ANH may possibly indicate a higher grade of
VUR and may worsen the clinical presentation
of a child with a febrile urinary tract infection Renal Agenesis, Renal Ectopia,
[97, 98]. As for mild ANH, a recent prospective and Unilateral Multicystic Dysplastic
study of 192 infants with ANH noted that the Kidney
majority of patients with mild ANH had no sig-
nificant events during infancy [34]. In another Infants born with solitary kidneys (renal agenesis),
study, female infants with a history of ANH and renal ectopia, or unilateral multicystic dysplasia
postnatal uropathy had a higher risk of febrile should be evaluated postnatally by US and
urinary tract infection [33]. Regardless, no appro- VCUG. Functional studies such as a dimercapto-
priate prospective studies with coordinated and succinic acid study (DMSA) are occasionally
comprehensive postnatal follow-up have exam- needed to confirm the diagnosis. The need for
ined this question in a rigorous fashion to provide further screening is controversial. It has been
consensus guidelines [16, 56]. reported that of infants with a solitary kidney,
At our institution, children with moderate or 30 % have VUR, 11 % UPJO, and 7 % UVJO
severe ANH are placed on prophylactic antibiotics [99, 100]. Similarly those with renal ectopia (sim-
at birth. One possible exception is the circumcised ple or crossed fused ectopia) may also be at risk for
male. Children with moderate or severe ANH VUR in the ectopic or contralateral kidney (30 %)
undergo renal bladder ultrasound and VCUG post- [101–103]. However, there are others that report a
natally. Diuretic renography is reserved for those very low incidence of associated urologic anoma-
with no VUR and persistent moderate or severe lies and do not recommend screening [104].
postnatal hydronephrosis on serial US. Infants As for MCDK, these lesions are primarily uni-
with mild ANH and mild or no postnatal lateral, isolated, and associated with a good prog-
hydronephrosis are observed and followed clini- nosis. If at birth the US findings are not conclusive
cally. Infants with a significant degree of antenatal of MCDK, a DMSA study can be used to confirm
or postnatal ureteral dilation undergo ultrasound, the diagnosis, as there should be no uptake. Patients
VCUG, and possibly diuretic renography (after with MCDK are often thought to be similar to those
3 months) if clinically indicated. born with a solitary kidney. Additionally, patients
with MCDK have a reported increased frequency of
VUR and UPJO in the contralateral normal kidney
Bilateral Hydronephrosis [105, 106], but utilization of routine screening by
VCUG remains controversial [107].
Infants with bilateral hydroureteronephrosis may
have PUV, bilateral VUR, bilateral UPJO or Acknowledgments The authors acknowledge the assis-
UVJO, or a combination of the above. For the tance of Carol E. Barnewolt, M.D., for providing many of
the fetal ultrasound images.
child with bilateral hydroureteronephrosis sug-
gestive of bladder outlet obstruction, an ultra-
sound and VCUG should be performed
promptly. In boys, PUV is the most important References
diagnosis to be ruled out. In girls, an obstructing
1. Gunn TR, Mora JD, Pease P. Antenatal diagnosis of
ectopic ureterocele would be the most likely cause urinary tract abnormalities by ultrasonography after
for bladder outlet obstruction. In the event that an 28 weeks’ gestation: incidence and outcome. Am J
obstructive lesion is discovered, it should be Obstet Gynecol. 1995;172(2 Pt 1):479–86.
110 D.A. Diamond and R.S. Lee

2. Livera LN, Brookfield DS, Egginton JA, Hawnaur 18. Yerkes EB, Cain MP, Padilla LM. In utero perinephric
JM. Antenatal ultrasonography to detect fetal renal urinoma and urinary ascites with posterior urethral
abnormalities: a prospective screening programme. valves: a paradoxical pop-off valve? J Urol.
BMJ. 1989;298(6685):1421–3. 2001;166(6):2387–8.
3. Sairam S, Al-Habib A, Sasson S, Thilaganathan 19. Adorisio O, Silveri M, Colajacomo M, Bassani F,
B. Natural history of fetal hydronephrosis diagnosed Rivosecchi M. The impact of perinatal urinoma for-
on mid-trimester ultrasound. Ultrasound Obstet mation on renal function: our experience and review
Gynecol. 2001;17(3):191–6. of the literature. J Paediatr Child Health. 2011;47
4. Shamshirsaz AA, Ravangard SF, Egan JF, Prabulos (4):217–22.
AM, Shamshirsaz AA, Ferrer FA, et al. Fetal 20. Takeuchi H, Koyanagi T, Yoshizato T, Takashima T,
hydronephrosis as a predictor of neonatal urologic Satoh S, Nakano H. Fetal urine production at different
outcomes. J Ultrasound Med Off J Am Inst Ultra- gestational ages: correlation to various compromised
sound Med. 2012;31(6):947–54. fetuses in utero. Early Hum Dev. 1994;40(1):1–11.
5. Helin I, Persson PH. Prenatal diagnosis of urinary 21. Stiller RJ, Pinto M, Heller C, Hobbins
tract abnormalities by ultrasound. Pediatrics. JC. Oligohydramnios associated with bilateral
1986;78(5):879–83. multicystic dysplastic kidneys: prenatal diagnosis at
6. Scott JE, Renwick M. Urological anomalies in the 15 weeks’ gestation. J Clin Ultrasound. 1988;16
Northern Region Fetal Abnormality Survey. Arch (6):436–9.
Dis Child. 1993;68(1 Spec No):22–6. 22. Benacerraf BR, Saltzman DH, Mandell
7. Scott JE, Renwick M. Screening for fetal urological J. Sonographic diagnosis of abnormal fetal genitalia.
abnormalities: how effective? BJU Int. 1999;84 J Ultrasound Med. 1989;8(11):613–7.
(6):693–700. 23. Bromley B, Mandell J, Gross G, Walzer TB, Bena-
8. Scott JE, Wright B, Wilson G, Pearson IA, Matthews cerraf BR. Masculinization of female fetuses with
JN, Rose PG. Measuring the fetal kidney with ultra- congenital adrenal hyperplasia may already be pre-
sonography. Br J Urol. 1995;76(6):769–74. sent at 18 weeks. Am J Obstet Gynecol. 1994;171
9. Mallik M, Watson AR. Antenatally detected urinary (1):264–5.
tract abnormalities: more detection but less action. 24. Mandell J, Bromley B, Peters CA, Benacerraf
Pediatr Nephrol. 2008;23(6):897–904. BR. Prenatal sonographic detection of genital
10. Benacerraf BR, Mandell J, Estroff JA, Harlow BL, malformations. J Urol. 1995;153(6):1994–6.
Frigoletto Jr FD. Fetal pyelectasis: a possible associ- 25. Dillon E, Rose PG, Scott JE. Case report: the antena-
ation with Down syndrome. Obstet Gynecol. 1990;76 tal ultrasound diagnosis of megalourethra. Clin
(1):58–60. Radiol. 1994;49(5):354–5.
11. Corteville JE, Dicke JM, Crane JP. Fetal 26. Cilento Jr BG, Benacerraf BR, Mandell J. Prenatal
pyelectasis and Down syndrome: is genetic amnio- diagnosis of cloacal malformation. Urology. 1994;43
centesis warranted? Obstet Gynecol. 1992;79 (3):386–8.
(5 (Pt 1)):770–2. 27. Taipale P, Heinonen K, Kainulainen S, Seuri R,
12. Adra AM, Mejides AA, Dennaoui MS, Beydoun Heinonen S. Cloacal anomaly simulating
SN. Fetal pyelectasis: is it always “physiologic”? megalocystis in the first trimester. J Clin Ultrasound.
Am J Obstet Gynecol. 1995;173(4):1263–6. 2004;32(8):419–22.
13. Thompson MO, Thilaganathan B. Effect of routine 28. Ohno Y, Koyama N, Tsuda M, Arii Y. Antenatal
screening for Down’s syndrome on the significance of ultrasonographic appearance of a cloacal anomaly.
isolated fetal hydronephrosis. Br J Obstet Gynaecol. Obstet Gynecol. 2000;95(6 Pt 2):1013–5.
1998;105(8):860–4. 29. Pates JA, Dashe JS. Prenatal diagnosis and manage-
14. Chudleigh PM, Chitty LS, Pembrey M, Campbell ment of hydronephrosis. Early Hum Dev. 2006;82
S. The association of aneuploidy and mild fetal (1):3–8.
pyelectasis in an unselected population: the results 30. Coplen DE, Austin PF, Yan Y, Blanco VM, Dicke
of a multicenter study. Ultrasound Obstet Gynecol. JM. The magnitude of fetal renal pelvic dilatation can
2001;17(3):197–202. identify obstructive postnatal hydronephrosis, and
15. Fernbach SK, Maizels M, Conway JJ. Ultrasound direct postnatal evaluation and management. J Urol.
grading of hydronephrosis: introduction to the system 2006;176(2):724–7; discussion 7.
used by the Society for Fetal Urology. Pediatr Radiol. 31. Ismaili K, Hall M, Donner C, Thomas D,
1993;23(6):478–80. Vermeylen D, Avni FE. Results of systematic screen-
16. Lee RS, Cendron M, Kinnamon DD, Nguyen ing for minor degrees of fetal renal pelvis dilatation in
HT. Antenatal hydronephrosis as a predictor of post- an unselected population. Am J Obstet Gynecol.
natal outcome: a meta-analysis. Pediatrics. 2006;118 2003;188(1):242–6.
(2):586–93. 32. Wollenberg A, Neuhaus TJ, Willi UV, Wisser
17. Chitty LS, Altman DG. Charts of fetal size: kidney J. Outcome of fetal renal pelvic dilatation diagnosed
and renal pelvis measurements. Prenat Diagn. during the third trimester. Ultrasound Obstet Gynecol.
2003;23(11):891–7. 2005;25(5):483–8.
4 Clinical Perinatal Urology 111

33. Coelho GM, Bouzada MC, Lemos GS, Pereira AK, 48. Tsatsaris V, Gagnadoux MF, Aubry MC, Gubler MC,
Lima BP, Oliveira EA. Risk factors for urinary tract Dumez Y, Dommergues M. Prenatal diagnosis of
infection in children with prenatal renal pelvic dilata- bilateral isolated fetal hyperechogenic kidneys. Is it
tion. J Urol. 2008;179(1):284–9. possible to predict long term outcome? BJOG.
34. Coelho GM, Bouzada MC, Pereira AK, Figueiredo 2002;109(12):1388–93.
BF, Leite MR, Oliveira DS, et al. Outcome of isolated 49. Mashiach R, Davidovits M, Eisenstein B, Kidron D,
antenatal hydronephrosis: a prospective cohort study. Kovo M, Shalev J, et al. Fetal hyperechogenic kidney
Pediatr Nephrol. 2007;22(10):1727–34. with normal amniotic fluid volume: a diagnostic
35. Ahmad G, Green P. Outcome of fetal pyelectasis dilemma. Prenat Diagn. 2005;25(7):553–8.
diagnosed antenatally. J Obstet Gynaecol. 2005;25 50. Carr MC, Benacerraf BR, Estroff JA, Mandell
(2):119–22. J. Prenatally diagnosed bilateral hyperechoic kidneys
36. Feldman DM, DeCambre M, Kong E, Borgida A, with normal amniotic fluid: postnatal outcome. J Urol.
Jamil M, McKenna P, et al. Evaluation and follow- 1995;153(2):442–4.
up of fetal hydronephrosis. J Ultrasound Med. 51. Chaumoitre K, Brun M, Cassart M, Maugey-Laulom-
2001;20(10):1065–9. B, Eurin D, Didier F, et al. Differential diagnosis of
37. Hobbins JC, Romero R, Grannum P, Berkowitz RL, fetal hyperechogenic cystic kidneys unrelated to renal
Cullen M, Mahoney M. Antenatal diagnosis of renal tract anomalies: A multicenter study. Ultrasound
anomalies with ultrasound. I. Obstructive uropathy. Obstet Gynecol Off J Int Soc Ultrasound Obstet
Am J Obstet Gynecol. 1984;148(7):868–77. Gynecol. 2006;28(7):911–7.
38. Abbott JF, Levine D, Wapner R. Posterior urethral 52. Vergani P, Ceruti P, Locatelli A, Mariani E,
valves: inaccuracy of prenatal diagnosis. Fetal Diagn Paterlini G, Zorloni C, et al. Accuracy of prenatal
Ther. 1998;13(3):179–83. ultrasonographic diagnosis of duplex renal system.
39. Kaefer M, Peters CA, Retik AB, Benacerraf J Ultrasound Med. 1999;18(7):463–7.
BB. Increased renal echogenicity: a sonographic 53. Share JC, Lebowitz RL. Ectopic ureterocele without
sign for differentiating between obstructive and ureteral and calyceal dilatation (ureterocele dispro-
nonobstructive etiologies of in utero bladder disten- portion): findings on urography and sonography.
sion. J Urol. 1997;158(3 Pt 2):1026–9. AJR Am J Roentgenol. 1989;152(3):567–71.
40. Grignon A, Filiatrault D, Homsy Y, Robitaille P, 54. Zerin JM, Ritchey ML, Chang AC. Incidental
Filion R, Boutin H, et al. Ureteropelvic junction ste- vesicoureteral reflux in neonates with antenatally
nosis: antenatal ultrasonographic diagnosis, postnatal detected hydronephrosis and other renal abnormali-
investigation, and follow-up. Radiology. 1986;160 ties. Radiology. 1993;187(1):157–60.
(3):649–51. 55. Tibballs JM, De Bruyn R. Primary vesicoureteric
41. Grignon A, Filion R, Filiatrault D, Robitaille P, reflux–how useful is postnatal ultrasound? Arch Dis
Homsy Y, Boutin H, et al. Urinary tract dilatation in Child. 1996;75(5):444–7.
utero: classification and clinical applications. Radiol- 56. van Eerde AM, Meutgeert MH, de Jong TP, Giltay
ogy. 1986;160(3):645–7. JC. Vesico-ureteral reflux in children with prenatally
42. Mandell J, Blyth BR, Peters CA, Retik AB, Estroff detected hydronephrosis: a systematic review. Ultra-
JA, Benacerraf BR. Structural genitourinary defects sound Obstet Gynecol. 2007;29(4):463–9.
detected in utero. Radiology. 1991;178(1):193–6. 57. Estrada Jr CR, Passerotti CC, Graham DA, Peters CA,
43. Kleiner B, Callen PW, Filly RA. Sonographic analy- Bauer SB, Diamond DA, et al. Nomograms for
sis of the fetus with ureteropelvic junction obstruc- predicting annual resolution rate of primary
tion. AJR Am J Roentgenol. 1987;148(2):359–63. vesicoureteral reflux: results from 2,462 children. J
44. Chertin B, Pollack A, Koulikov D, Rabinowitz R, Urol. 2009;182(4):1535–41.
Hain D, Hadas-Halpren I, et al. Conservative treat- 58. Peters CA. Lower urinary tract obstruction: clinical
ment of ureteropelvic junction obstruction in children and experimental aspects. Br J Urol. 1998;81 Suppl
with antenatal diagnosis of hydronephrosis: lessons 2:22–32.
learned after 16 years of follow-up. Eur Urol. 2006;49 59. Kaefer M, Barnewolt C, Retik AB, Peters CA.
(4):734–9. The sonographic diagnosis of infravesical obstruction
45. Mandell J, Paltiel HJ, Peters CA, Benacerraf in children: evaluation of bladder wall thickness
BR. Prenatal findings associated with a unilateral indexed to bladder filling. J Urol. 1997;157
nonfunctioning or absent kidney. J Urol. 1994;152 (3):989–91.
(1):176–8. 60. Oliveira EA, Diniz JS, Cabral AC, Pereira AK, Leite
46. Edwards OP, Baldinger S. Prenatal onset of autosomal HV, Colosimo EA, et al. Predictive factors of fetal
dominant polycystic kidney disease. Urology. urethral obstruction: a multivariate analysis. Fetal
1989;34(5):265–70. Diagn Ther. 2000;15(3):180–6.
47. Zerres K, Hansmann M, Mallmann R, Gembruch 61. Osborne NG, Bonilla-Musoles F, Machado LE,
U. Autosomal recessive polycystic kidney disease. Raga F, Bonilla Jr F, Ruiz F, et al. Fetal megacystis:
Problems of prenatal diagnosis. Prenat Diagn. differential diagnosis. J Ultrasound Med Off J Am
1988;8(3):215–29. Inst Ultrasound Med. 2011;30(6):833–41.
112 D.A. Diamond and R.S. Lee

62. Mahony BS, Callen PW, Filly RA. Fetal urethral 75. Ruano R, Duarte S, Bunduki V, Giron AM, Srougi M,
obstruction: US evaluation. Radiology. 1985;157 Zugaib M. Fetal cystoscopy for severe lower urinary
(1):221–4. tract obstruction–initial experience of a single center.
63. Mandell J, Peters CA, Estroff JA, Benacerraf Prenat Diagn. 2010;30(1):30–9.
BR. Late onset severe oligohydramnios associated 76. Ruano R, Yoshizaki CT, Giron AM, Srougi M,
with genitourinary abnormalities. J Urol. 1992;148 Zugaib M. Cystoscopic placement of transurethral
(2 Pt 2):515–8. stent in a fetus with urethral stenosis. Ultrasound
64. Glick PL, Harrison MR, Golbus MS, Adzick NS, Obstet Gynecol. 2014;44(2):238–40.
Filly RA, Callen PW, et al. Management of the 77. Clifton MS, Harrison MR, Ball R, Lee H. Fetoscopic
fetus with congenital hydronephrosis II: prognostic transuterine release of posterior urethral valves: a new
criteria and selection for treatment. J Pediatr Surg. technique. Fetal Diagn Ther. 2008;23(2):89–94.
1985;20(4):376–87. 78. Morris RK, Ruano R, Kilby MD. Effectiveness of
65. Johnson MP, Corsi P, Bradfield W, Hume RF, fetal cystoscopy as a diagnostic and therapeutic inter-
Smith C, Flake AW, et al. Sequential urinalysis vention for lower urinary tract obstruction: a system-
improves evaluation of fetal renal function in obstruc- atic review. Ultrasound Obstet Gynecol. 2011;37
tive uropathy. Am J Obstet Gynecol. 1995;173 (6):629–37.
(1):59–65. 79. Harrison MR, Golbus MS, Filly RA, Anderson RL,
66. Guez S, Assael BM, Melzi ML, Tassis B, Nicolini Flake AW, Rosen M, et al. Fetal hydronephrosis:
U. Shortcomings in predicting postnatal renal func- selection and surgical repair. J Pediatr Surg. 1987;22
tion using prenatal urine biochemistry in fetuses with (6):556–8.
congenital hydronephrosis. J Pediatr Surg. 1996;31 80. Clautice-Engle T, Anderson NG, Allan RB, Abbott
(10):1401–4. GD. Diagnosis of obstructive hydronephrosis in
67. Tassis BM, Trespidi L, Tirelli AS, Bocconi L, infants: comparison sonograms performed 6 days
Zoppini C, Nicolini U. In fetuses with isolated and 6 weeks after birth. AJR Am J Roentgenol.
hydronephrosis, urinary beta 2-microglobulin and 1995;164(4):963–7.
N-acetyl-beta-D-glucosaminidase (NAG) have a lim- 81. Muller F, Dommergues M, Mandelbrot L, Aubry MC,
ited role in the prediction of postnatal renal function. Nihoul-Fekete C, Dumez Y. Fetal urinary biochemis-
Prenat Diagn. 1996;16(12):1087–93. try predicts postnatal renal function in children with
68. Morris RK, Quinlan-Jones E, Kilby MD, Khan bilateral obstructive uropathies. Obstet Gynecol.
KS. Systematic review of accuracy of fetal urine 1993;82(5):813–20.
analysis to predict poor postnatal renal function in 82. Dommergues M, Muller F, Ngo S, Hohlfeld P, Oury
cases of congenital urinary tract obstruction. Prenat JF, Bidat L, et al. Fetal serum beta 2-microglobulin
Diagn. 2007;27(10):900–11. predicts postnatal renal function in bilateral
69. Morris RK, Malin GL, Khan KS, Kilby uropathies. Kidney Int. 2000;58(1):312–6.
MD. Antenatal ultrasound to predict postnatal renal 83. Biard JM, Johnson MP, Carr MC, Wilson RD,
function in congenital lower urinary tract obstruction: Hedrick HL, Pavlock C, et al. Long-term outcomes
systematic review of test accuracy. BJOG. 2009;116 in children treated by prenatal vesicoamniotic
(10):1290–9. shunting for lower urinary tract obstruction. Obstet
70. Harrison MR, Nakayama DK, Noall R, de Lorimier Gynecol. 2005;106(3):503–8.
AA. Correction of congenital hydronephrosis in utero 84. Crombleholme TM, Harrison MR, Golbus MS,
II. Decompression reverses the effects of obstruction Longaker MT, Langer JC, Callen PW, et al. Fetal
on the fetal lung and urinary tract. J Pediatr Surg. intervention in obstructive uropathy: prognostic indi-
1982;17(6):965–74. cators and efficacy of intervention. Am J Obstet
71. Manning FA, Harrison MR, Rodeck C. Catheter Gynecol. 1990;162(5):1239–44.
shunts for fetal hydronephrosis and hydrocephalus. 85. Freedman AL, Johnson MP, Smith CA, Gonzalez R,
Report of the International Fetal Surgery Registry. N Evans MI. Long-term outcome in children after ante-
Engl J Med. 1986;315(5):336–40. natal intervention for obstructive uropathies. Lancet.
72. Robichaux 3rd AG, Mandell J, Greene MF, Benacer- 1999;354(9176):374–7.
raf BR, Evans MI. Fetal abdominal wall defect: a new 86. Holmes N, Harrison MR, Baskin LS. Fetal surgery for
complication of vesicoamniotic shunting. Fetal Diagn posterior urethral valves: long-term postnatal out-
Ther. 1991;6(1–2):11–3. comes. Pediatrics. 2001;108(1):E7.
73. Quintero RA, Shukla AR, Homsy YL, Bukkapatnam 87. McLorie G, Farhat W, Khoury A, Geary D, Ryan
R. Successful in utero endoscopic ablation of poste- G. Outcome analysis of vesicoamniotic shunting in a
rior urethral valves: a new dimension in fetal urology. comprehensive population. J Urol. 2001;166
Urology. 2000;55(5):774. (3):1036–40.
74. Sago H, Hayashi S, Chiba T, Ueoka K, Matsuoka K, 88. Salam MA. Posterior urethral valve: outcome of ante-
Nakagawa A, et al. Endoscopic fetal urethrotomy for natal intervention. Int J Urol. 2006;13(10):1317–22.
anterior urethral valves: a preliminary report. Fetal 89. Clark TJ, Martin WL, Divakaran TG, Whittle MJ,
Diagn Ther. 2008;24(2):92–5. Kilby MD, Khan KS. Prenatal bladder drainage in
4 Clinical Perinatal Urology 113

the management of fetal lower urinary tract obstruc- 98. Song SH, Lee SB, Park YS, Kim KS. Is antibiotic
tion: a systematic review and meta-analysis. Obstet prophylaxis necessary in infants with obstructive
Gynecol. 2003;102(2):367–82. hydronephrosis? J Urol. 2007;177(3):1098–101; dis-
90. Johnson MP, Bukowski TP, Reitleman C, Isada NB, cussion 101.
Pryde PG, Evans MI. In utero surgical treatment of 99. Atiyeh B, Husmann D, Baum M. Contralateral renal
fetal obstructive uropathy: a new comprehensive abnormalities in patients with renal agenesis and
approach to identify appropriate candidates for noncystic renal dysplasia. Pediatrics. 1993;91(4):
vesicoamniotic shunt therapy. Am J Obstet Gynecol. 812–5.
1994;170(6):1770–6; discussion 6–9. 100. Cascio S, Paran S, Puri P. Associated urological
91. Coplen DE, Hare JY, Zderic SA, Canning DA, Snyder anomalies in children with unilateral renal agenesis.
3rd HM, Duckett JW. 10-year experience with prena- J Urol. 1999;162(3 Pt 2):1081–3.
tal intervention for hydronephrosis. J Urol. 1996;156 101. Arena F, Arena S, Paolata A, Campenni A,
(3):1142–5. Zuccarello B, Romeo G. Is a complete urological
92. Ethun CG, Zamora IJ, Roth DR, Kale A, Cisek L, evaluation necessary in all newborns with asymptom-
Belfort MA, et al. Outcomes of fetuses with lower atic renal ectopia? Int J Urol. 2007;14(6):491–5.
urinary tract obstruction treated with vesicoamniotic 102. Gleason PE, Kelalis PP, Husmann DA, Kramer
shunt: a single-institution experience. J Pediatr Surg. SA. Hydronephrosis in renal ectopia: incidence, eti-
2013;48(5):956–62. ology and significance. J Urol. 1994;151(6):1660–1.
93. Tonni G, Vito I, Ventura A, Grisolia G, De Felice 103. Guarino N, Tadini B, Camardi P, Silvestro L, Lace R,
C. Fetal lower urinary tract obstruction and its Bianchi M. The incidence of associated urological
management. Arch Gynecol Obstet. 2013;287 abnormalities in children with renal ectopia. J Urol.
(2):187–94. 2004;172(4 Pt 2):1757–9; discussion 9.
94. Morris RK, Malin GL, Quinlan-Jones E, Middleton 104. Calisti A, Perrotta ML, Oriolo L, Ingianna D, Miele
LJ, Hemming K, Burke D, et al. Percutaneous V. The risk of associated urological abnormalities in
vesicoamniotic shunting versus conservative manage- children with pre and postnatal occasional diagnosis
ment for fetal lower urinary tract obstruction of solitary, small or ectopic kidney: is a complete
(PLUTO): a randomised trial. Lancet. 2013;382 urological screening always necessary? World J
(9903):1496–506. Urol. 2008;26:281–4.
95. Yerkes EB, Adams MC, Pope 4th JC, Brock 3rd 105. Kaneko K, Suzuki Y, Fukuda Y, Yabuta K, Miyano
JW. Does every patient with prenatal hydronephrosis T. Abnormal contralateral kidney in unilateral
need voiding cystourethrography? J Urol. 1999;162 multicystic dysplastic kidney disease. Pediatr Radiol.
(3 Pt 2):1218–20. 1995;25(4):275–7.
96. Nguyen HT, Herndon CD, Cooper C, Gatti J, 106. Miller DC, Rumohr JA, Dunn RL, Bloom DA, Park
Kirsch A, Kokorowski P, et al. The Society for Fetal JM. What is the fate of the refluxing contralateral
Urology consensus statement on the evaluation and kidney in children with multicystic dysplastic kidney?
management of antenatal hydronephrosis. J Pediatr J Urol. 2004;172(4 Pt 2):1630–4.
Urol. 2010;6(3):212–31. 107. Ismaili K, Avni FE, Alexander M, Schulman C,
97. Grazioli S, Parvex P, Merlini L, Combescure C, Collier F, Hall M. Routine voiding cystoure-
Girardin E. Antenatal and postnatal ultrasound in the thrography is of no value in neonates with unilateral
evaluation of the risk of vesicoureteral reflux. Pediatr multicystic dysplastic kidney. J Pediatr. 2005;146(6):
Nephrol. 2010;25(9):1687–92. 759–63.
Renal Dysplasia/Hypoplasia
5
Paul Goodyer and Indra R. Gupta

Contents Overview
Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
Clinical Presentation of Renal Hypoplasia/ Congenital renal hypoplasia/dysplasia refers to
Dysplasia In Utero . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115 the end result of a variety of pathogenic mecha-
Renal Agenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 118 nisms that disturb the primary induction of meta-
Unilateral Renal Agenesis (URA) . . . . . . . . . . . . . . . . . . . 118 nephric progenitor cells (renal agenesis),
Bilateral Renal Agenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 120 dysregulation of nephron number (disorders of
Renal Hypoplasia/Dysplasia . . . . . . . . . . . . . . . . . . . . . . . 120 progenitor pool size or ureteric bud branching),
Renal Hypoplasia and Defects in Ureteric Bud aberrant progenitor cell differentiation (dysplastic
Branching . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
tissue elements), disturbance of the renal vascula-
Renal Hypoplasia and Dysregulation of the
Nephron Progenitor Pool . . . . . . . . . . . . . . . . . . . . . . . . . . . . 122 ture (causing tubular dysgenesis), organ structure
Renal Tubular Dysplasia with Disturbance of (multicystic/dysplastic kidney), and tissue migra-
Embryonic Kidney Perfusion . . . . . . . . . . . . . . . . . . . . . . . . 123 tion (horseshoe kidney, ectopic kidney). Many of
Renal Hypoplasia Associated with Lower Tract
these disturbances are linked to mutation of genes
Anomalies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124
Environmental Causes of Renal Hypoplasia/ in molecular cascades involved in the develop-
Dysplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 124 ment of other organs; thus, renal hypoplasia/dys-
Subtle Renal Hypoplasia . . . . . . . . . . . . . . . . . . . . . . . . . . . 125 plasia can be seen with a variety of non-renal
malformations. It has been useful to assemble
Multicystic Dysplastic Kidney (MCDK) . . . . . . . . . . 126
large heterogeneous pediatric cohorts under the
Renal Ectopia and Fusion Anomalies . . . . . . . . . . . . . 129 umbrella term of congenital anomalies of the kid-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130 ney and urinary tract (CAKUT) as a way to study
classification and identification of underlying
genes without bias [1]. In this chapter we consider
forms of CAKUT associated with reduced func-
tional renal mass at birth.
P. Goodyer (*)
Division of Pediatric Nephrology, Montreal Children’s
Hospital, McGill University, Montreal, QC, Canada Clinical Presentation of Renal
e-mail: paul.goodyer@muhc.mcgill.ca
Hypoplasia/Dysplasia In Utero
I.R. Gupta
Department of Pediatrics and Department of Human
With the advent of routine antenatal ultrasono-
Genetics, Division of Pediatric Nephrology, Montreal
Children’s Hospital, McGill University, Montréal, QC, graphic screening, renal size and shape can be
Canada evaluated prior to birth (Fig. 1), and pediatric
# Springer-Verlag Berlin Heidelberg 2016 115
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_4
116 P. Goodyer and I.R. Gupta

Table 1 Kidney size by gestational age. Sagittal kidney


length and renal volumes at various gestational ages; mean
(50th percentile) and lower limit of normal (5th percentile)
are shown. Adapted from Ref. [2]
Gestational Kidney Kidney
age (weeks) length (mm) volume (ml)
50th % 5th % 50th % 5th %
15 12.5 10 – –
20 20 16 2 1.5
25 26 23 4 2.5
30 33 28 7 4
35 38 34 12 7.5
40 44 38 17 11

175 ml) is generated primarily by fetal urine [3].


Thus, amniotic fluid volume may be reduced in
fetuses with reduced functional renal mass. How-
ever, regulation of amniotic fluid volume is com-
plex and difficult to estimate by ultrasonography.
By term, urine volume varies widely, from 400 to
Fig. 1 Magnetic resonance imaging showing normal fetal 1200 ml/day; about half is eliminated via the fetal
kidney (arrow) and amniotic fluid (asterisk)
gastrointestinal tract while the remainder of the
(relatively hypotonic) fluid is transferred directly
nephrologists are increasingly involved in the to the placental compartment by diffusion and
management of pregnancies with renal abnormal- bulk flow. Despite rapid renal growth in the third
ities. Optimal management is best accomplished trimester, amniotic fluid volume increases by only
by a team that also includes the obstetrician, radi- 35–70 ml/week during this period until the 38th
ologists, geneticists, and neonatologists. Pediatric week of gestation and then declines by
nephrologists should offer opinion regarding the 125 ml/week to an average of 800 ml by
implications of antenatal renal hypoplasia/dyspla- 40 weeks [3]. Since accurate measures of amniotic
sia and meet with the family before birth to coun- fluid volume cannot be made by ultrasonography,
sel them about the rapid, often difficult, clinical oligohydramnios is often tracked by serial mea-
decisions that may unfold in the early postnatal surements of amniotic fluid index (AFI). With the
period. transducer held in the sagittal plane and perpen-
Using transvaginal ultrasound, the urine is first dicular to the floor, the largest vertical diameter
detected in the bladder by about 9 weeks of ges- (cm) in each of four quadrants is measured and
tation; renal size and amniotic fluid volume can be summed [4]. Nomograms for AFI in healthy preg-
assessed by transabdominal ultrasonography from nancies from 16 to 42 weeks were reported by
15 to 16 weeks of gestation onward. The diagno- Moore et al. [5]; approximate values for mean and
sis of antenatal renal hypoplasia (less than 5th 2.5th percentile for AFI at various gestational ages
percentile) is usually made during the second are summarized in Table 2. Although
and third trimester using nomograms for renal oligohydramnios represents a clinically signifi-
length and volume [2]; approximate mean and cant consequence of bilateral renal hypoplasia, it
5th percentile at selected gestational ages are sum- should be noted that it can also be seen with
marized in Table 1. premature rupture of membranes, fetal growth
By 16 weeks, fetal skin has become restriction, and fetal hypoxia (thought to cause
keratinized, and the amniotic fluid (about fetal vasopressin release). Furthermore, a modest
5 Renal Dysplasia/Hypoplasia 117

Table 2 Amniotic fluid index at various gestational ages.


Amniotic fluid index (AFI) is calculated by antenatal ultra-
sonography as the sum of the largest vertical diameter
(cm) of amniotic fluid in each of the four quadrants in the
image. Mean (50th percentile) and lower limit (2.5th per-
centile) for various gestational ages are adapted from
Moore et al., Am J Ob Gyn (1990)
AFI(cm) 50th percentile 2.5th percentile
16 12.1 7.3
20 14.1 8.6
25 14.7 8.9
30 14.5 8.2
35 14.0 7.0
40 12.3 6.3

decline in AFI (from 14.7 to 12.3 cm) is normal


during the third trimester (Table 2).
When renal hypoplasia is extreme, inadequate Fig. 2 Magnetic resonance imaging showing case of Fra-
amniotic fluid production causes a characteristic ser syndrome with the absence of the right kidney, hypo-
plastic left kidney (arrow), and anhydramnios (asterisk)
pattern of facial compression and pulmonary
hypoplasia (Potter syndrome) that may compro-
mise postnatal viability (Fig. 2) [6, 7]. By (identifiable by mercaptoacetyltriglycerine imag-
compromising fetal respiratory movements that ing); (c) urine volume sufficient to permit minimal
optimize fetal lung development, severe long-term oral nutrition (100 kcal/kg/day); and
oligohydramnios may lead to pneumothorax in (d) family and institutional resources that can
the immediate postnatal period, and limited gas sustain the dialytic therapy that successfully
exchange may require intubation and intensive achieves growth and development prior to renal
respiratory support. Antenatal ultrasonographic transplantation. Although the immediate compli-
assessment of lung volume is difficult and, when cations of pulmonary hypoplasia dominate initial
suspected, is better examined by magnetic reso- care, some degree of pulmonary recovery can be
nance imaging. In fetuses with falling AFI, an expected, and most babies who survive the first
argument is often made for early induction of week do not succumb to respiratory failure unless
labor once reasonable lung maturity can be oliguria leads to marked fluid overload. Judicious
expected. On the other hand, if some amniotic fluid restriction and diuretic therapy are critical in
fluid is present in the third trimester and mechan- the first days of life to minimize edema that could
ical compromise of lung development is less compromise gas exchange and make dialysis
extreme, it may be preferable to have the preg- catheter technically difficult to insert. Toward the
nancy go to term. This allows maximal somatic end of the first week, the baby’s urine volume
and renal growth in the intrauterine environment, (on diuretics) should be estimated to inform fam-
making dialysis technically easier and reducing ily and medical team whether survival on dialysis
the extrauterine growth needed to achieve trans- is feasible.
plantable body size. When amniotic fluid is absent at an early stage,
In babies who survive the immediate postnatal families should be offered formal counseling
respiratory crisis, the decision to embark on about the anticipated postnatal viability of the
chronic peritoneal dialysis is usually based on: infant and to prepare them for the rapid series of
(a) whether the baby can achieve sufficient lung decisions which may arise regarding the extent of
recovery to be weaned off respiratory support; “heroic” interventions ranging from intubation, to
(b) evidence of some functional renal parenchyma resuscitation, to dialysis. Because of the risk of
118 P. Goodyer and I.R. Gupta

pneumothorax, fetuses with renal hypoplasia each ureteric bud branch tip induce adjacent neph-
and/or low AFI should be delivered in tertiary ron progenitor cells to transform into the proximal
care hospitals where neonatal intensive care is segments (glomerulus, proximal tubule, loop of
available, ideally with prior awareness of the neo- Henle, and distal convoluted tubule) of individual
natology, nephrology, and genetics teams. When a nephrons. Each emerging nephron fuses to the
monogenic cause of renal hypoplasia is suspected, collecting duct formed by its parental ureteric
families should be offered genetic counseling. bud branch. By 36 weeks of gestation, the ureteric
bud has undergone 22–23 successive branching
events yielding 900,000 nephrons linked to a
Renal Agenesis common outflow tract in each kidney. Renal
hypoplasia/dysplasia arises through disturbance
Unlike lower species such as the fishes, human of one or more of these embryonic processes.
nephron formation ends prior to birth, and babies
are born with a surfeit of nephrons (about
900,000/kidney) that are usually sufficient for Unilateral Renal Agenesis (URA)
life. However, human nephron number varies
widely (>10-fold) among normal newborns URA occurs in about 1 in 2000 births [13]
(210,000–2,700,000/kidney), and at the low end (Fig. 3). It is estimated that about 5 % of URA
of this spectrum are babies with clinically impor- can be attributed to a unilateral multicystic/dys-
tant renal hypoplasia [8]. In the 1980s, Brenner’s plastic kidneys which have involuted prior to
group postulated that suboptimal nephron endow- evaluation. The incidence of URA in males
ment induces compensatory glomerular (63 %) is slightly higher than in females, and the
hyperfiltration, leading to slow but inexorable left kidney is more often absent (63 %) than the
glomerulosclerosis in postnatal life [9–11]; they right [14]. Westland et al. reviewed 2684 URA
hypothesized that this increases the risk of essen- patients from 43 reports (pediatric and adult
tial hypertension and that reduced renal reserve cohorts) and found that about one third had vari-
accelerates progressive renal insufficiency in ous urinary tract anomalies, including
response to acquired renal injury later in life. vesicoureteral reflux (24 %), ureteropelvic junc-
Although the mechanisms that set final neph- tion obstruction (6 %), megaureter (7 %),
ron are not well understood, it is clear that the ureterocoele (1 %), and posterior urethral vales
kidneys, adrenals, and gonads are derived from (1 %) [13]. One third were associated with
progenitor cells in the posterior columns of inter-
mediate mesoderm which run along the
rostrocaudal axis of the embryo (see also
▶ Chap. 1, “Embryonic Development of the
Kidney” of this text). Key transcription factor
genes (e.g., WT1, CITED1) prime some of these
progenitors for nephrogenesis, while another sub-
set of cells express genes (e.g., GATA3 and
PAX2) transform them into nephric ducts on
each side of the embryo. At about 4–5 weeks of
gestation, ureteric buds emerge from the two cau-
dally migrating nephric ducts and grow outward
into the flanking column of intermediate meso-
derm. The left and right ureteric buds are induced
to arborize by glial-derived neurotrophic factor
(GDNF), and branching is further regulated by
other growth factors [12]. WNT signals from Fig. 3 DMSA scan showing agenesis of the left kidney
5 Renal Dysplasia/Hypoplasia 119

syndromic extrarenal anomalies. From analysis of progressed to end-stage renal failure. The risk of
an informative subset of the reports, it was esti- renal failure among URA babies was similar to
mated that mean GFR was 100 ml/min/1.73 m2 that in babies with bilateral hypoplasia/dysplasia.
and that the majority of patients with an isolated This presumably reflects the associated lower tract
solitary functioning kidney can be expected to anomalies or contralateral renal dysplasia.
have stable renal function for many decades. URA is frequently associated with ipsilateral
However, 21 % had microalbuminuria, 16 % had malformations of the reproductive tract. Among
hypertension, and 10 % had glomerular filtration girls, URA is seen in the Herlyn-Werner-
rate <60 ml/min/1.73 m2 [13]. It is unclear Wunderlich syndrome (HWWS), characterized
whether this renal dysfunction is the consequence by bifid uterus (uterus didelphys), bifid vagina
of congenital nephron deficit or instead reflects (obstructed hemi-vagina in 60 %), and ipsilateral
associated dysplasia in the remaining kidney. In a URA [19, 20]. Women may be fertile after surgi-
pediatric cohort of URA/MCDK patients from the cal repair of the uterus [21]. HWWS should be
Netherlands [15], hypertension was seen in 31 %, suspected when antenatal ultrasonography reveals
and microalbuminuria was present in 23 %, indi- URA in association with a fluid collection behind
cating that these issues are generally evident the uterus [22]. URA is rarely associated with
before the age of 18 years. Mayer-Rokitansky-Kuster-Hauser (MRKH) syn-
URA obviously reduces total nephron mass drome in which amenorrhea is linked to absence
and usually induces compensatory renal hypertro- of Mullerian ducts [23]. Among boys, URA can
phy in the remaining kidney during fetal life. In an be associated with cryptorchidism or ipsilateral
ultrasonographic study of a mixed URA/MCDK absence of the vas deferens [24]. However, most
cohort, length of the single functioning kidney males of unilateral absence of the vas deferens
was estimated to be 23 % longer than normal (0.5–1.0 %) have normal kidneys. The presence
from the 20th week of gestation onward [2]. Cho of URA does not exclude bilateral absence of the
et al. proposed that the ratio of anterior-posterior vas deferens, and this should be ruled out, since it
to transverse kidney dimensions discriminates is a cause of male infertility.
appropriately hypertrophied kidneys (1.0) from URA is also associated with a variety of other
the ratio in babies with two normal kidneys malformation syndromes, particularly VACTERL
(0.8) [16]. syndrome (1 per 10–40,000 births) involving a
The majority of children with unilateral kidney variable combination of vertebral anomalies,
malformations are expected to retain stable renal anal atresia, cardiac defects, tracheoesophageal
function for most of their adult lives. However, fistula with esophageal atresia, renal anomalies,
not all babies with URA display appropriate com- and limb defects [25]. About one quarter of
pensatory hypertrophy of the single remaining VACTERL patients have URA, while others
kidney. In a study of postnatal renal hypertrophy have vesicoureteral reflux (27 %) or a multicystic
among 125 babies with various forms of solitary dysplastic kidney (18 %) [26]. Other rare URA
functioning kidney, all but six had renal length associations include Barakat [27], Fraser [28],
(as a function of body length) or renal volume Kabuki makeup [29], and Manitoba-oculo-
(as a function of body surface area) greater than tricho-anal (MOTA) syndromes [30].
the 50th percentile for normal, and about one third Recent genetic screens in large CAKUT kin-
had kidney size greater than the 95th percentile for dreds have yielded information regarding specific
normal [17]. Importantly, the babies with renal genes associated with non-syndromic URA. In
size less than the 50th percentile developed renal these studies, URA has been associated with het-
insufficiency (<60 ml/min/1.73 m2) [17]. Recent erozygous (dominant) mutations of CDC5L [31]
observations by Sanna-Cherchi suggest that the and RET [31, 32] and with homozygous or com-
lifelong risk for URA may be underestimated pound heterozygous (recessive) mutations of
[18]. Of 71 patients with a solitary functioning FRAS1 and FREM2 [33]. Recessive mutations
kidney followed for 30 years, 21 eventually of FRAS1 [33] and TRAP1 [26] genes were
120 P. Goodyer and I.R. Gupta

identified in several unrelated children with URA mutations of integrin alpha 8 (ITGA8) [38]. BRA
and VACTERL anomalies; recessive mutations of was noted in a family with recessive mutations of
GATA3 were reported in a child Barakat syn- the FGF20 gene [39] in a large kindred with
drome (deafness, seizures, and hypoparathyroid- X-linked Kallmann syndrome due to a KAL-1
ism) and URA [27]. The genetic basis for most gene mutations [40] and is seen occasionally
cases of isolated and syndromic URA is not yet with heterozygous RET mutations [41].
known.
Parents of a child with URA commonly ask
about whether contact sports should be avoided to
prevent loss of the functioning kidney. While the Renal Hypoplasia/Dysplasia
decision is ultimately an individual one, it should
be recognized that loss of a kidney due to trauma Renal hypoplasia/dysplasia (RHD) (Fig. 4) may
is extremely rare and is far less likely than the risk be unilateral or bilateral and may occur in associ-
of lethal car accident en route or serious brain ation with minor urinary tract malformations (e.g.,
injury incurred during the contact sport. vesicoureteral reflux) or in association with more
complex syndromes. In one large CAKUT series,
renal hypoplasia/dysplasia accounted for about
Bilateral Renal Agenesis one third of the cohort [18]. In a prospective
antennal ultrasound screen of 4000 healthy Japa-
Bilateral renal agenesis (BRA) is rare – about nese pregnancies, Hiraoko et al. noted renal hypo-
10 per 100,000 births in a report on congenital plasia in about 1:400 births [42].
anomalies among 601,545 births in the United Among 112 children with unilateral (URHD)
Kingdom [34]. BRA uniformly leads to or bilateral (BRHD) renal hypoplasia/dysplasia
anhydramnios and the Potter sequence in which analyzed by Sanna-Cherchi et al., 13 % had pro-
mechanical compression causes wide-set eyes, teinuria and 2 % had hypertension at the time of
flattened nose, receding chin, and large low-set diagnosis [18]. After 8–15 years of follow-up, 9 of
ears lacking cartilage [7]. Newborns also have
severe lung hypoplasia and clubbed feet and are
generally nonviable. However, Bienstock
et al. recently reported successful use of weekly
amnioinfusion from 24 weeks of gestation onward
to avert lethal lung hypoplasia. The affected child
required minimal postnatal respiratory support
and underwent successful peritoneal dialysis [35].
In most cases, the cause of BRA is not known.
However, reports of absent renal arteries and ure-
ters in the second trimester suggest arrest of kid-
ney development at an early stage. Roodhooft
et al. examined 40 siblings of 41 index cases
with BRA and found that 10 % had URA,
suggesting a heritable cause [36]. Bankier
et al. confirmed in an Australian cohort that
URA was found in sibs of babies with isolated
BRA but not in sibs of BRA associated with
multiple malformations [37]. Humbert
et al. recently reported a consanguineous Roma
Gypsy family in which 3 couples had multiple
pregnancies with BRA associated with recessive Fig. 4 Hypoplastic/dysplastic kidney
5 Renal Dysplasia/Hypoplasia 121

Fig. 5 Peritubular fibrous


cuff in an area of renal
dysplasia

Fig. 6 Ectopic cartilage in


an area of renal dysplasia

the children had progressed to end-stage renal Renal Hypoplasia and Defects
disease. In RHD, the presence of ureters, some in Ureteric Bud Branching
functional renal cortex, and basic reniform archi-
tecture reflects the fact that early formation of Hwang et al. screened 120 CAKUT patients with
ureteric buds and priming of the renal progenitor renal hypoplasia/dysplasia and 90 with unilateral
population have occurred. However, functional renal agenesis [31]. Dysfunctional missense
nephrons may be interspersed with patchy dys- mutations in eight genes were associated with
plastic tissue such as expanded stromal cells reduced renal mass (with or without anomalies
around tubules (Fig. 5) or islands of cartilage of the lower urinary tract). In the same cohort,
amid the functional nephrons (Fig. 6). Renal next-generation sequencing of 12 candidates
hypoplasia necessarily means that nephron num- (drawn from mouse models of unilateral renal
ber is reduced, making it likely that there has been agenesis) identified biallelic mild missense muta-
dysregulation of the nephron progenitor pool, ure- tions of 6 Fraser-syndrome-related genes [33]; a
teric bud arborization, or both. renal hypoplasia phenotype was associated with
122 P. Goodyer and I.R. Gupta

four of the six genes. Interestingly, many of the A plausible explanation for the congenital neph-
mutant CAKUT genes linked to reduced renal ron deficit is that stochastic loss of cells might
mass in these two studies are implicated in the slow the extension of ureteric bud branches
regulation of ureteric bud branching (e.g., RET, required before the next round of branching
HNF1B, BMP7, FRAS1, etc). The unique pheno- events can occur.
type of ureteric bud tip cells is crucially dependent Recently, a PAX2 missense mutation (p.
on expression of RET (the GDNF receptor); the Gly189Arg) was identified in a family with dom-
transcription factor HNF-1B [43]; and the growth inant focal segmental glomerulosclerosis (FSGS),
factor, BMP7 [44]. Fraser syndrome genes encode presenting in adulthood without any ocular phe-
proteins belonging to a complex that is essential notype [52]. Screening of a familial FSGS cohort
for GDNF expression in the metanephric mesen- identified similar missense mutations in 4 % of
chyme [33]. Other reports identify patients with families [52]. Although the missense mutations
renal/hypodysplasia associated with mutations of found in these patients are predicted to be patho-
genes involved in fibroblast growth factor- genic, they presumably permit some residual
induced branching (e.g., FGF20 and PAX2 function, accounting for late onset of renal
DSTYK) [26]. disease [53]. The finding in this family supports
Autosomal dominant renal-coloboma syn- Brenner’s hypothesis that individuals born with
drome (RCS) is a form of primary renal hypopla- suboptimal nephron number exhibit compensa-
sia causing progressive renal insufficiency in tory hypertrophy and that glomerular
childhood associated with colobomas of the hyperfiltration early in life gradually leads to
optic nerve, reduced ureteric bud branching dur- focal glomerulosclerosis.
ing embryogenesis, and low nephron number at
birth [45]. RCS is caused by heterozygous null
mutations of PAX2, encoding a transcription fac- Renal Hypoplasia and Dysregulation
tor expressed both in the ureteric bud and in the of the Nephron Progenitor Pool
metanephric mesenchyme [46]. Retrospectively,
children with “oligomeganephronia” who lacked In the subset of CAKUT patients with renal
obvious ocular colobomas were found to have hypoplasia, Hwang et al. identified heterozygous
PAX2 mutations, underscoring the wide variabil- (autosomal dominant) mutations of several genes
ity in extrarenal manifestations [47]. Typically, implicated in sustaining the nephron progenitor
children who inherit a heterozygous null PAX2 cell pool [31]. These included dysfunctional
allele present with relatively small kidneys at mutations of EYA1, SIX1 which cause the auto-
birth, proteinuria in the first months, and progres- somal dominant branchio-oto-renal (BOR)
sive renal insufficiency during the first years of syndrome (OMIM 113650), pairing renal
life [48]. Renal biopsy shows that glomeruli are hypoplasia with preauricular pits or branchial
greatly enlarged suggesting compensatory glo- fistulae, and variable degrees of deafness [54].
merular hyperfiltration. Most patients develop An expanded list of features includes a mixed
end-stage renal disease by young adulthood. form of conductive/sensorineural hearing loss,
Vesicoureteral reflux is present in about half of preauricular pits, a “cup-shaped” deformity of ear
RCS patients; mild high-frequency hearing loss is pinnae, external auditory canal stenosis, branchial
less common [48]. fistulae, and renal anomalies including primary
The nephron deficit in mutant Pax2 mice renal hypoplasia, ureteropelvic junction obstruc-
has been attributed to a primary defect in ureteric tion, and vesicoureteral reflux. Renal hypoplasia
bud branching during embryogenesis [49, 50]. ranges from slightly reduced kidney size to bilat-
Pax21Neu mice were found to have increased eral renal agenesis with perinatal death [55] and
programmed cell death of ureteric bud cells; loss may affect one kidney more than the other. Only
of Pax2 reduces expression of the antiapoptotic 5–10 % of BOR patients develop end-stage renal
gene, Naip, in ureteric bud cells [50, 51]. disease [56]. The prevalence of BOR syndrome is
5 Renal Dysplasia/Hypoplasia 123

about 1:40,000 and affects 2 % of children with Table 3 Syndromic renal hypoplasia/dysplasia genes.
profound deafness [56]. The cochlea is hypoplastic Syndromic renal hypoplasia/dysplasia associated with het-
erozygous mutations of various genes
and maybe simplified; dilatation of the vestibular
duct may be evident by MRI [57]. Gene Syndrome OMIM
About 40 % of BOR syndrome patients have PAX2 Renal-coloboma syndrome 120330
heterozygous inactivating mutations of the tran- EYA1 Branchio-oto-renal syndrome 113650
SIX1 Branchio-oto-renal syndrome 113650
scription factor EYA1 gene (8q13) [58]. EYA1 is
SIX5 Branchio-oto-renal syndrome 610896
expressed in progenitor cells of the embryonic
SALL1 Townes-Brocks syndrome 107480
kidney and first branchial arch that gives rise to
HNF1β Renal cortical cysts and diabetes 137920
the auricles, auditory canal, and ossicles of the
KAL1 Kallmann Syndrome 308700
ear; EYA1 is also expressed in hair cells of the FRAS1 Fraser syndrome 219000
cochlea [59]. Thus, EYA1 mutations appear to FREM2 Bifid nose, anorectal, renal 608980
cause renal hypoplasia, outer ear deformities, GATA3 Hypoparathyroidism with 146255
and conductive hearing loss by compromising deafness
progenitor cell fate in the embryonic kidney and GLI3 Pallister-Hall syndrome 146510
ear. Between 5 % and 10 % of BOR syndrome, LRP4 Cenani-Lenz syndrome 212780
patients have mutations of genes belonging to the RET Hirschsprung and CAKUT 142623
SIX transcription factor family. Several families BMP4 Isolated RHD –
bear mutations of SIX1 [60], and nearly 5 % have REN Renal tubular dysgenesis 179820
mutations of SIX5 [61]. Although loss of SIX2 in AGT Renal tubular dysgenesis 106150
mutant mice reduces nephron number, Hwang AGTR1 Renal tubular dysgenesis 106165
ACE Renal tubular dysgenesis 106180
et al. identified only one patient with a SIX2
mutation in their CAKUT cohort, and this did
not involve significant renal hypoplasia.
In 1972, Townes and Brocks described a patient Renal Tubular Dysplasia
with imperforate anus, sensorineural deafness, with Disturbance of Embryonic Kidney
minor anomalies of the external ear (preauricular Perfusion
tags and deformity of the superior helix), and mul-
tiple anomalies of the digits including missing Autosomal recessive renal tubular dysgenesis
bones and supernumerary thumbs [62]. Clinical (RTD) is a severe disorder in which development
features of the TBS syndrome are now recognized of renal tubules is compromised by hypoperfusion
to be more complex and include renal hypoplasia/ of the embryonic kidney [67]. Affected fetuses
dysplasia [63]. About half of TBS patients have have oligohydramnios with Potter sequence, peri-
heterozygous mutations of the transcription factor natal death due to pulmonary hypoplasia, and
gene, SALL1, which can sometimes lead to ESRD; refractory arterial hypotension and osseous
dominant negative SALL1 mutations produce a defects of the skull. Although the kidneys may
slightly more severe disease [64]. In developing be hyperechoic, renal size is usually normal. Glo-
mouse kidneys, SALL1 is expressed in the meta- meruli have normal structure and the surrounding
nephric mesenchyme surrounding branches of the tubules stain primarily for collecting duct
ureteric bud; Sall1 knockout mice lack outgrowth markers; proximal tubules are sparse and rudi-
of the ureteric bud and die perinatally of renal mentary, lacking a brush border and devoid of
agenesis [65]. Studies by Basta indicate that typical markers that are largely absent [68]. With
SALL1, like SIX genes, opposes differentiation saline infusion, intensive perinatal support and/or
of renal progenitor cells [66]. Thus, the mutant peritoneal dialysis, 10–15 % may survive the
genes above exemplify settings in which perinatal period. RTD is caused by mutations of
dysregulation of the embryonic renal progenitor genes encoding components of the renin-
cell pool compromises congenital nephron number angiotensin system (RAS): AGT
in humans (Table 3). (angiotensinogen), REN (renin), ACE
124 P. Goodyer and I.R. Gupta

(angiotensin-converting enzyme), and AGTR1 ureteric bud lineage, Hains et al. demonstrated
(angiotensin II receptor type 1) [69]. that a ureteric bud that emerges more cranially
results in ureteral insertion in the bladder neck
and these mice also exhibited VUR [73]. In sum-
Renal Hypoplasia Associated mary, any child with evidence of renal hypopla-
with Lower Tract Anomalies sia warrants a thorough evaluation of the urinary
tract to rule out VUR and/or urinary tract
Mackie and Stephens examined autopsy specimens obstruction, and similarly, any child with evi-
from infants with duplex kidneys and noted an dence of high-grade VUR and/or urinary tract
association between a hypoplastic or dysplastic kid- obstruction is likely to have a malformed kidney
ney and the position of the ureteral orifice within the on the same side as the urinary tract defect.
bladder [70]. Ureters that inserted lateral to the In Japanese perinatal screening studies, renal
trigone or at the bladder neck were usually attached hypoplasia (about 1:400 normal births) was
to a malformed kidney. They then went on to pro- strongly correlated with vesicoureteral reflux [5].
pose the ureteric bud theory in which they postu- Among neonates investigated for vesicoureteral
lated that if the ureteric bud arose from a more reflux, renal hypoplasia (reduced renal length by
caudal location along the nephric duct, the ureter ultrasonography and reduced DMSA uptake by
would insert lateral to the trigone, while if it arose scintigraphy) was frequent, being symmetric in
from a more cranial location, it would insert at the about one third of cases or involving patchy
bladder neck. The ureteric bud is the primordial “scars” of the renal parenchyma in others [47,
tissue that gives rise to both the kidneys and the 48]. Among children evaluated for febrile urinary
ureters; therefore, if the ureteric bud emerges from tract infection in Texas, 15 % showed focal corti-
the wrong location from the nephric duct, it likely cal defects on DMSA scans; an additional 4 % had
does not receive or transmit the proper inductive evidence of reduced renal function without corti-
signals required for the development of these struc- cal defects [74]. In Ireland, DMSA defects were
tures. From human studies, it is known that in seen in 37 % of children with grade IV VUR and
duplicated systems, the ureter that inserts lateral to 67 % of those with grade V VUR [75]. Among
the trigone has a high likelihood of facilitating 173 children with grade IV–V reflux (bilateral or
vesicoureteric reflux. Similarly, the ureter that unilateral) vesicoureteral reflux in Taiwan, half
inserts at the bladder neck is frequently associated had developed CKD II or greater by about
with urinary tract obstruction. While the initial 140 months [76]. One third of children with bilat-
Mackie and Stephens studies were performed on eral high-grade reflux developed hypertension
infants with duplex systems in which two ureteric [76]. Controversy continues as to the best way to
buds emerge, their theory likely applies to infants distinguish congenital renal hypoplasia/dysplasia
with renal hypoplasia and single urinary tracts that from acquired defects attributable to infection, but
exhibit VUR or urinary tract obstruction. some clinicians have suggest a “top-down”
While the ureteric bud theory is plausible, it is approach to assessing renal prognosis, while
difficult to prove. The best evidence that supports focusing on VUR (“down-top” approach) to
the theory comes from the discovery of mouse guide the approach to recurrent infection.
models with vesicoureteric reflux and renal
malformations [71, 72]. From these models, it
has been shown that the ureteric bud does emerge Environmental Causes of Renal
from a more caudal location, and this results in a Hypoplasia/Dysplasia
delay in urinary tract development. The final
ureter has a short intravesical ureter and exhibits In the 1940s and 1950s, Wilson reported that
vesicoureteric reflux. Using a mouse model in severe vitamin A deficiency caused renal agenesis
which the receptor FGFR2 was deleted from the [77]. Observations in rodents indicate that
5 Renal Dysplasia/Hypoplasia 125

Table 4 Polymorphic gene variants associated with congenital renal hypoplasia. Single nucleotide polymorphisms
(SNPs) associated with reduced newborn kidney volume
Gene SNP MAF% SNP type Decrease in newborn kidney volume
PAX2 rs11599825(A) 18.5 % Noncoding 10 %
rs11190688(A)
rs11190702(A)
rs11190739(T) 6.5 % Noncoding 9 %
RET rs1800860(A) 25 % Exonic splice enhancer 10 %
OSR1 rs12329305(T) 6% Exonic splice enhancer 12 %
BMPR1A rs7922846(A) 31.6 % Noncoding 13 %
ALDH1A2 rs7169289(G) 22 % Noncoding +22 %

maternal vitamin A deficiency (50 % reduction in


circulating retinol levels) causes significant reduc- Subtle Renal Hypoplasia
tion (24 %) in postnatal kidney weight associated
with a reduction (20 %) in nephron number Common completely dysfunctional alleles for
[78]. Normally, the fetus acquires retinol from crucial developmental genes are rare, since they
the maternal circulation and converts it to the produce malformations with a major disadvan-
active metabolite, all-trans-retinoic acid, in tage. However, mild mutations of these genes
kidney and other tissues [79]. In cultured fetal exert subtler effects on renal mesenchyme/ureteric
rat kidneys, all-trans-retinoic acid bud interactions and may be fairly common
accelerates new nephron formation two- to three- among normal children (Table 4). If measured by
fold [80, 81]. Vitamin A deficiency is widespread ultrasonography in the newborn period before
in developing countries; in North America and postnatal compensatory hypertrophy is complete,
Europe, vitamin A deficiency has been kidney size was shown to correlate with nephron
documented in unsupplemented newborns born number at autopsy [87]. Using this approach,
prematurely before nephrogenesis is complete. Quinlan et al. identified a signature (AAA) of
In a cohort of 46 pregnant Indian women, several tightly linked intronic PAX2 SNPs, pre-
Goodyer et al. noted that half had retinol levels sent in 18.5 % of normal Canadian newborns, that
below the accepted limit of normal (0.9 umol/L) was associated with a subtle (10 %) reduction in
and newborn kidney volume (adjusted for body newborn kidney size (adjusted for body surface
size) was only 62 % of adjusted newborn kidney area). In a heterozygous renal cell carcinoma line,
size in vitamin A-replete women from the AAA allele was associated with a 50 % reduc-
Montreal [82]. tion in PAX2 transcript level [88]. No common
In rats with streptozotocin-induced diabetes variants of the human GDNF gene affect kidney
mellitus, offspring have renal hypoplasia and size, but a polymorphism of the RET (GDNF
reduced nephron number [83]. In fetal mice receptor) gene (RET1476 G/A, rs1800860) was
exposed, embryonic E14.5 kidneys had 45 % associated with a 10 % reduction in newborn
fewer ureteric bud branches than controls to kidney volume (normalized for body surface
maternal diabetes [84]. Among human offspring area) and 9 % increased serum cystatin C com-
of Type I diabetic mothers, Khalil et al. noted pared to the wild-type (RET1476G) allele
reduced renal reserve in response to amino acid [87]. Although fairly common in the healthy
infusion [85], and Cappuccini et al. found that European Caucasian population (MAF = 25 %),
renal cortex volume was significantly reduced the RET1476A allele alters a splicing enhancer site
compared to controls [86]. in exon 7, permitting frequent transcription of a
126 P. Goodyer and I.R. Gupta

truncated mRNA [87]. Interestingly, among the Evidence is emerging that the common gene
15 % of infants who inherited both a RET1476A variants associated with congenital renal hypopla-
and a PAX2AAA allele, newborn kidney size was sia predict accelerated renal deterioration in the
reduced by 25 %. Taken together, these observa- setting of acquired kidney disease later in life. In a
tions suggest that hypomorphic RET and PAX2 preliminary study, Cosgrove identified a common
gene variants act in combinatorial fashion to affect (MAF = 6.5 %) noncoding PAX2 polymorphic
ureteric bud branching and, thus, newborn kidney variant (rs11190739T) that was associated with
size. In a separate cohort of Polish babies, new- increased serum creatinine in a cohort of patients
born kidney size was reduced by 13 % in infants with Type I diabetes for 10–30 years [94]. Esti-
bearing a common variant of the BMPR1A gene mated GFR was 70.7 ml/min/1.73 m2 in diabetics
(encoding a bone morphogenetic protein receptor with the wild-type allele, but was 60.1 ml/min/
on ureteric bud cells) [89]. Conversely, 22 % of 1.73 m2 (15 % decrease) in diabetics with one or
Canadian newborns inherit a variant of the more rs11190739T alleles. These observations
ALHD1A2 gene [rs7169289 (G)] associated support Brenner’s hypothesis that progressive
with increased production of all-trans-retinoic renal disease may be accelerated among individ-
acid metabolism in fetal tissues; this retinoid is uals with suboptimal nephron number and limited
known to enhance RET expression in the ureteric renal reserve.
bud, and newborns with the G allele were shown
to have a 22 % increase in newborn kidney size
compared to the wild-type allele [90]. Multicystic Dysplastic Kidney (MCDK)
While human nephron number is affected by
genes regulating the extent of ureteric branching, MCDK is a form of severe renal dysplasia in
animal studies also indicate that the size of the which the kidney is occupied by tense
renal progenitor cell pool can also be rate- noncommunicating macrocyst; the largest cyst is
limiting. One of the earliest transcription factors not usually located medially and, typically, there
marking the nephron progenitor cells of the inter- is no functional parenchyma (Fig. 7). The renal
mediate mesoderm is OSR1; Osr1 knockout mice pelvis and calyces cannot usually be identified,
lack nephrogenic mesenchyme and are anephric at and the ureteropelvic junction and ureter are usu-
birth [91]. Recently, a variant of the human OSR1 ally atretic; ipsilateral vesicoureteral reflux is rare.
gene which interferes with an exon 2 splice The incidence of MCDK ranges from 1 per 2200
enhancer site was identified in about 6 % of nor- to 4300 births; it is slightly more common among
mal Caucasians [92]. This OSR1rs12329305(T) vari- males (55 %) and slightly more common on the
ant was associated with a 12 % reduction in right (53 %) than on the left [95–98]. Among
newborn kidney size and 12 % increase in serum males, 13 % have ipsilateral or bilateral unde-
cystatin C level [92]. Again, the effects of the scended testes [97] (Fig. 8). With widely available
OSR1rs12329305(T) were additive with RET1476G antenatal ultrasound screening, MCDK is now
in infants who also inherited both alleles; kidney increasingly diagnosed before birth (Fig. 9), but
volume/body surface area was reduced by 17 % follow-up postnatal ultrasonography is needed to
and serum cystatin C was increased by 22 % distinguish MCDK from high-grade urinary tract
[92]. Recently, Lozic et al. reported association obstruction.
between OSR1rs12329305(T) and stillborn/neonatal Since MCDK is typically unilateral, the long-
death among babies with congenital kidney term renal prognosis depends primarily on the
malformations [93]. Taken together, these obser- integrity of the contralateral kidney. With long-
vations suggest that human nephron endowment term follow-up, 2/40 Italian and 2/34 Indian
may represent a complex polygenic trait deter- MCDK patients were seen to develop end-stage
mined by the additive effects of multiple genes renal failure [18, 99]. In most cases, estimated
regulating either the renal progenitor cell pool or glomerular filtration rate is normal, ranging
the extent of ureteric branching during fetal life. from 86 to 122 ml/min/1.73 m2 in the pediatric
5 Renal Dysplasia/Hypoplasia 127

Fig. 7 Multicystic
dysplastic kidney. (a) Renal
ultrasound; (b) multicystic
kidney (left) and normal
kidney (right) en bloc; (c)
multicystic dysplastic
kidney in situ

Fig. 8 Antenatal magnetic resonance imaging showing the left multicystic dysplastic kidney

period [100]. About one quarter of the contralat- several large series, there are a small number of
eral kidneys have vesicoureteral reflux (most contralateral kidneys (2–5 %) which exhibit
often grade I–III). Although this is occasionally severe reflux, remain small and hyperechoic or
associated with recurrent urinary tract infection, fail to show normal compensatory hypertrophy,
the presence of mild-moderate vesicoureteral and have highest risk of progressing to end-stage
reflux is of little clinical significance and does renal failure [17]. Between 5 % and 30 % of cases
not warrant routine cystourethrogram. Less have low-grade proteinuria [95, 99]. Since the
often, the contralateral kidney has ureteropelvic MCDK is usually nonfunctional, this mild pro-
junction obstruction (3–5 %) and ureterovesical teinuria presumably arises from the contralateral
junction obstruction (2 %) smaller in number. In kidney. Hypertension occurs in a small fraction
128 P. Goodyer and I.R. Gupta

whereas the other son had unilateral MCDK


[103]. In an antenatal cohort of 63 cases with
bilateral hyperechoic kidneys and normal amni-
otic fluid volume, 18 were found to have TCF2
mutations, most commonly a heterozygous TCF2
deletion [104]. However, the presentation is quite
variable and some affected kidneys develop wide-
spread cysts only in the postnatal period,
distinguishing this from typical MCDK. Further-
more, in this form of MCDK there may also be
pancreatic hypoplasia detectable by antenatal
ultrasound [105].
Whereas multicystic kidneys were commonly
resected in the past, most authors currently rec-
ommend a conservative approach. While it
remains important to distinguish MCDK from
cystic Wilms tumors, the risk of malignant trans-
formation in MCDK tissue may approximate the
risk in normal kidneys [100]. Early postnatal
ultrasound should be performed to exclude
obstruction, hyperechogenicity, or other overt
signs of dysplasia in the contralateral kidney.
Serial ultrasonography should document appro-
priate compensatory hypertrophy in the contralat-
eral kidney and involution of the MCKD. Spira
Fig. 9 Ultrasound showing ipsilateral mid-canal unde- et al. showed that renal length of the
scended testis (arrow) hypertrophied kidney should be greater than the
50th percentile for body length, and one third
(1–6 %) of cases [95, 101], usually associated excedes the 95th percentile [17]. In a separate
with minimal residual functional parenchyma cohort of 323 MCDK cases, 10 % of antenatally
(noted by renal scan) in the MCDK. Importantly, diagnosed MCDKs had involuted by birth, 35 %
the hypertension is often reported to resolve when by 2 years, 47 % by 5 years, and 62 % by 10 years
the MCDK is resected [99]. [97]. Blood pressure should be checked in the first
The etiology of MCDK is unclear but presum- years of life; when hypertension is detected, renal
ably involves embryonic disturbance of the mech- scanning is indicated since the presence of resid-
anisms that generate functional nephrons linked to ual functional renal parenchyma may support a
the renal collecting system. Significantly role for nephrectomy. Serum creatinine and urine
increased gene copy number variation is seen in protein excretion should also be monitored in the
>10 % of MCDK patients [102]. In large screens first years of life to detect progressive renal dys-
of CAKUT cohorts, MCDK has been associated function, particularly in those children with evi-
with dominant mutations of CHD1L, ROBO2, dence of overt dysplasia or suboptimal size of the
and SALL1 genes [33]. Interestingly, the MCDK contralateral kidney. Hayes et al. have argued that
phenotype has also been linked to heterozygous costly monitoring might be curtailed after the first
null mutations of the TCF2 (HNF1β) gene. Hasui year in children who exhibit MCDK involution
reported a family in which one son with a TCF2 and normal compensatory hypertrophy and have
mutation presented with bilateral MCDK, no overt complications [97].
5 Renal Dysplasia/Hypoplasia 129

Fig. 10 Horseshoe
kidneys: (a) computerized
tomography scan showing
isthmus of horseshoe
kidney fused in midline,
(b) DMSA scan showing
pancake kidney fused in
midline

Renal Ectopia and Fusion Anomalies kidneys are sometimes associated with poorly
localized pain. Rarely, ectopic kidneys have
During embryogenesis, the two kidneys ascend been reported in the thorax and are identified as
from a lower pelvic position to their normal an incidental nonfunctional chest mass [107].
upper intra-abdominal location. Disturbance of When aberrant ascent brings them into contact,
renal ascent leads to renal ectopia in about 1:900 the two kidneys may fuse. Most commonly, this
births and is usually (90 %) unilateral. The asso- occurs in the midline and is referred to as a horse-
ciated ureter nearly always enters the bladder nor- shoe kidney (Fig. 10). In 95 %, the horseshoe
mally, but about half are associated with kidney is low-lying (L3-L4) where further ascent
hydronephrosis. This may be due to ureteropelvic has been blocked by the inferior mesenteric artery.
junction obstruction (16 %), ureterovesicular The two moieties are fused via a parenchymal
junction obstruction (8 %), or vesicoureteral isthmus bridging their lower poles and may
reflux (25 %) [106]. In about 40 %, the ectopic develop a variety of vascular arrangements, includ-
kidney is found in the pelvis. While most ectopic ing a single artery [108]. About half of horseshoe
kidneys are asymptomatic, lumbar and pelvic kidneys exhibit vesicoureteral reflux and about one
130 P. Goodyer and I.R. Gupta

third report recurrent urinary tract infection [109]. 8. Bertram JF, Cullen-McEwen LA, Egan GF, Gretz N,
About 20 % of horseshoe kidneys develop stones at Baldelomar E, Beeman SC, Bennett KM. Why and
how we determine nephron number. Pediatr Nephrol.
some point in life [110]. There are occasionally 2014;29:575–80.
VACTERL (3 %) and reproductive tract (5 %) 9. Neuringer JR, Brenner BM. Glomerular hyperten-
anomalies [111]. Although there are case reports sion: cause and consequence of renal injury.
of renal cell carcinoma arising within horseshoe J Hypertens Suppl. 1992;10:S91–7.
10. Brenner BM, Garcia DL, Anderson S. Glomeruli and
kidneys, there is no evidence that this occurs blood pressure. Less of one, more the other? Am J
more frequently than in the general population Hypertens. 1988;1:335–47.
[106]. On the other hand, patients have horseshoe 11. Anderson S, Brenner BM. The role of
kidneys have a twofold increased risk of Wilms intraglomerular pressure in the initiation and progres-
sion of renal disease. J Hypertens Suppl. 1986;4:
tumor compared to normal [112]. Between 15 % S236–8.
and 30 % of girls with Turner syndrome have 12. Bates CM. Role of fibroblast growth factor receptor
horseshoe kidneys [113, 114]. signaling in kidney development. Pediatr Nephrol.
Crossed ectopia occurs when one kidney 2011;26:1373–9.
13. Westland R, Schreuder MF, Ket JC, van Wijk
crosses the midline in relationship to the side JA. Unilateral renal agenesis: a systematic review on
where its ureter enters the bladder; in 90 % of associated anomalies and renal injury. Nephrol Dial
cases, the upper pole of the ectopic kidney is Transplant. 2013;28:1844–55.
fused to the lower pole of the orthotopic kidney 14. Dogan CS, Torun Bayram M. Renal outcome of chil-
dren with unilateral renal agenesis. Turk J Pediatr.
but sometimes occurs without fusion, and the two 2013;55:612–5.
kidneys may even exchange sides [106, 15. Schreuder MF, Langemeijer ME, Bokenkamp A,
115]. Abnormalities of the reproductive tracts Delemarre-Van de Waal HA, Van Wijk
are seen in about half of the cases; these include JA. Hypertension and microalbuminuria in children
with congenital solitary kidneys. J Paediatr Child
cryptorchidism or absence of the vas deferens in Health. 2008;44:363–8.
males and vaginal atresia or unilateral uterine 16. Cho JY, Moon MH, Lee YH, Kim KW, Kim
abnormalities in females [106]. SH. Measurement of compensatory hyperplasia of
the contralateral kidney: usefulness for differential
diagnosis of fetal unilateral empty renal fossa. Ultra-
sound Obstet Gynecol. 2009;34:515–20.
References 17. Spira EM, Jacobi C, Frankenschmidt A, Pohl M, von
Schnakenburg C. Sonographic long-term study: pae-
1. Pope JCT, Brock 3rd JW, Adams MC, Stephens FD, diatric growth charts for single kidneys. Arch Dis
Ichikawa I. How they begin and how they end: classic Child. 2009;94:693–8.
and new theories for the development and deteriora- 18. Sanna-Cherchi S, Ravani P, Corbani V, Parodi S,
tion of congenital anomalies of the kidney and urinary Haupt R, Piaggio G, Innocenti ML, Somenzi D,
tract, CAKUT. J Am Soc Nephrol. 1999;10:2018–28. Trivelli A, Caridi G, et al. Renal outcome in patients
2. van Vuuren SH, Damen-Elias HA, Stigter RH, van with congenital anomalies of the kidney and urinary
der Doef R, Goldschmeding R, de Jong TP, Westers P, tract. Kidney Int. 2009;76:528–33.
Visser GH, Pistorius LR. Size and volume charts of 19. Saleh M, Badawy SZ. Unilateral non-communicating
fetal kidney, renal pelvis and adrenal gland. Ultra- cervical atresia in a patient with uterus didelphys and
sound Obstet Gynecol. 2012;40:659–64. unilateral renal agenesis. J Pediatr Adolesc Gynecol.
3. Brace RA, Wolf EJ. Normal amniotic fluid volume 2010;23:e137–40.
changes throughout pregnancy. Am J Obstet Gynecol. 20. Zhou Y, Fu X, Qian H, Lin K, Wang J, Zhou S, Hu X,
1989;161:382–8. Jin H. A Herlyn-Werner-Wunderlich syndrome vari-
4. Phelan JP, Ahn MO, Smith CV, Rutherford SE, ant with ipsilateral renal absence and a contralateral
Anderson E. Amniotic fluid index measurements dur- duplex collecting system in a 26-year-old female.
ing pregnancy. J Reprod Med. 1987;32:601–4. Gynecol Obstet Invest. 2014;77:137–40.
5. Moore TR, Cayle JE. The amniotic fluid index in 21. Heinonen PK. Pregnancies in women with uterine
normal human pregnancy. Am J Obstet Gynecol. malformation, treated obstruction of hemivagina and
1990;162:1168–73. ipsilateral renal agenesis. Arch Gynecol Obstet.
6. Potter EL. Oligohydramnios: further comment. 2013;287:975–8.
J Pediatr. 1974;84:931–2. 22. Han BH, Park SB, Lee YJ, Lee KS, Lee YK. Uterus
7. Potter EL. Bilateral absence of ureters and kidneys: a didelphys with blind hemivagina and ipsilateral renal
report of 50 cases. Obstet Gynecol. 1965;25:3–12. agenesis (Herlyn-Werner-Wunderlich syndrome)
5 Renal Dysplasia/Hypoplasia 131

suspected on the presence of hydrocolpos on prenatal 35. Bienstock JL, Birsner ML, Coleman F, Hueppchen
sonography. J Clin Ultrasound. 2013;41:380–2. NA. Successful in utero intervention for bilateral
23. Herlin M, Hojland AT, Petersen MB. Familial occur- renal agenesis. Obstet Gynecol. 2014;124:413–5.
rence of Mayer-Rokitansky-Kuster-Hauser syn- 36. Roodhooft AM, Birnholz JC, Holmes LB. Familial
drome: a case report and review of the literature. Am nature of congenital absence and severe dysgenesis of
J Med Genet A. 2014;164A:2276–86. both kidneys. N Engl J Med. 1984;310:1341–5.
24. Sahin C, Kalkan M, Yalcinkaya S. Findings 37. Bankier A, de Campo M, Newell R, Rogers JG,
concerning testis, vas deference, and epididymis in Danks DM. A pedigree study of perinatally lethal
adult cases with nonpalpable testes. Int Braz J Urol. renal disease. J Med Genet. 1985;22:104–11.
2011;37:727–32. 38. Humbert C, Silbermann F, Morar B, Parisot M,
25. Cunningham BK, Khromykh A, Martinez AF, Zarhrate M, Masson C, Tores F, Blanchet P, Perez
Carney T, Hadley DW, Solomon BD. Analysis of MJ, Petrov Y, et al. Integrin alpha 8 recessive muta-
renal anomalies in VACTERL association. Birth tions are responsible for bilateral renal agenesis in
Defects Res A Clin Mol Teratol. 2014;100:801–5. humans. Am J Hum Genet. 2014;94:288–94.
26. Saisawat P, Kohl S, Hilger AC, Hwang DY, Yung 39. Barak H, Huh SH, Chen S, Jeanpierre C,
Gee H, Dworschak GC, Tasic V, Pennimpede T, Martinovic J, Parisot M, Bole-Feysot C, Nitschke P,
Natarajan S, Sperry E, et al. Whole-exome Salomon R, Antignac C, et al. FGF9 and FGF20
resequencing reveals recessive mutations in TRAP1 maintain the stemness of nephron progenitors in
in individuals with CAKUT and VACTERL associa- mice and man. Dev Cell. 2012;22:1191–207.
tion. Kidney Int. 2014;85:1310–7. 40. Colquhoun-Kerr JS, Gu WX, Jameson JL, Withers S,
27. Sau T, Chatterjee A, Ghosh K, Dey S. Seizure, deaf- Bode HH. X-linked Kallmann syndrome and renal
ness and renal agenesis: a rare case of Barakat syn- agenesis occurring together and independently in a
drome. Ann Indian Acad Neurol. 2013;16:91–3. large Australian family. Am J Med Genet.
28. Barisic I, Odak L, Loane M, Garne E, Wellesley D, 1999;83:23–7.
Calzolari E, Dolk H, Addor MC, Arriola L, 41. Jeanpierre C, Mace G, Parisot M, Moriniere V,
Bergman J, et al. Fraser syndrome: epidemiological Pawtowsky A, Benabou M, Martinovic J, Amiel J,
study in a European population. Am J Med Genet Attie-Bitach T, Delezoide AL, et al. RET and GDNF
A. 2013;161A:1012–8. mutations are rare in fetuses with renal agenesis or
29. Rosti RO, Kayserili H. Kabuki make-up syndrome other severe kidney development defects. J Med
with unilateral renal agenesis. Turk J Pediatr. Genet. 2011;48:497–504.
2009;51:298–300. 42. Hiraoka M, Tsukahara H, Ohshima Y, Kasuga K,
30. Nathanson J, Swarr DT, Singer A, Liu M, Chinn A, Ishihara Y, Mayumi M. Renal aplasia is the predom-
Jones W, Hurst J, Khalek N, Zackai E, Slavotinek inant cause of congenital solitary kidneys. Kidney Int.
A. Novel FREM1 mutations expand the phenotypic 2002;61:1840–4.
spectrum associated with Manitoba-oculo-tricho-anal 43. Lokmane L, Heliot C, Garcia-Villalba P, Fabre M,
(MOTA) syndrome and bifid nose renal agenesis Cereghini S. vHNF1 functions in distinct regulatory
anorectal malformations (BNAR) syndrome. Am J circuits to control ureteric bud branching and early
Med Genet A. 2013;161A:473–8. nephrogenesis. Development. 2010;137:347–57.
31. Hwang DY, Dworschak GC, Kohl S, Saisawat P, 44. Goncalves A, Zeller R. Genetic analysis reveals an
Vivante A, Hilger AC, Reutter HM, Soliman NA, unexpected role of BMP7 in initiation of ureteric bud
Bogdanovic R, Kehinde EO, et al. Mutations in outgrowth in mouse embryos. PLoS One. 2011;6:
12 known dominant disease-causing genes clarify e19370.
many congenital anomalies of the kidney and urinary 45. Dziarmaga A, Quinlan J, Goodyer P. Renal hypopla-
tract. Kidney Int. 2014;85:1429–33. sia: lessons from Pax2. Pediatr Nephrol.
32. Skinner MA, Safford SD, Reeves JG, Jackson ME, 2006;21:26–31.
Freemerman AJ. Renal aplasia in humans is associ- 46. Sanyanusin P, Schimmenti LA, McNoe TA, Ward TA,
ated with RET mutations. Am J Hum Genet. Pierpont ME, Sullivan MJ, Dobyns WB, Eccles
2008;82:344–51. MR. Mutation of the gene in a family with optic
33. Kohl S, Hwang DY, Dworschak GC, Hilger AC, nerve colobomas, renal anomolies and vesicoureteral
Saisawat P, Vivante A, Stajic N, Bogdanovic R, reflux. Nat Genet. 1996;13:129.
Reutter HM, Kehinde EO, et al. Mild recessive muta- 47. Salomon R, Tellier AL, Attie-Bitach T, Amiel J,
tions in six Fraser syndrome-related genes cause iso- Vekemans M, Lyonnet S, Dureau P, Niaudet P, Gubler
lated congenital anomalies of the kidney and urinary MC, Broyer M. PAX2 mutations in oligomega-
tract. J Am Soc Nephrol. 2014;25:1917–22. nephronia. Kidney Int. 2001;59:457–62.
34. Boyd PA, Tonks AM, Rankin J, Rounding C, 48. Laimutis K, Jackson C, Xu X, Warman B, Sarunas R,
Wellesley D, Draper ES. Monitoring the prenatal Andriuskeviciute I, Birute P, Schimmenti LA, Raca
detection of structural fetal congenital anomalies in G. Typical renal-coloboma syndrome phenotype in a
England and Wales: register-based study. J Med patient with a submicroscopic deletion of the PAX2
Screen. 2011;18:2–7. gene. Am J Med Genet A. 2012;158A:1437–41.
132 P. Goodyer and I.R. Gupta

49. Dziarmaga A, Clark P, Stayner C, Julien JP, Torban E, branchio-oto-renal syndrome by disruption of
Goodyer P, Eccles M. Ureteric bud apoptosis and EYA1-SIX1-DNA complexes. Proc Natl Acad Sci
renal hypoplasia in transgenic PAX2-Bax fetal mice U S A. 2004;101:8090–5.
mimics the renal-coloboma syndrome. J Am Soc 61. Hoskins BE, Cramer CH, Silvius D, Zou D, Raymond
Nephrol. 2003;14:2767–74. RM, Orten DJ, Kimberling WJ, Smith RJ, Weil D,
50. Dziarmaga A, Eccles M, Goodyer P. Suppression of Petit C, et al. Transcription factor SIX5 is mutated in
ureteric bud apoptosis rescues nephron endowment patients with branchio-oto-renal syndrome. Am J
and adult renal function in Pax2 mutant mice. J Am Hum Genet. 2007;80:800–4.
Soc Nephrol. 2006;17:1568–75. 62. Townes PL, Brocks ER. Hereditary syndrome of
51. Dziarmaga A, Hueber PA, Iglesias D, Hache N, imperforate anus with hand, foot, and ear anomalies.
Jeffs A, Gendron N, Mackenzie A, Eccles M, J Pediatr. 1972;81:321–6.
Goodyer P. Neuronal apoptosis inhibitory protein is 63. Sudo Y, Numakura C, Abe A, Aiba S, Matsunaga A,
expressed in developing kidney and is regulated by Hayasaka K. Phenotypic variability in a family with
PAX2. Am J Physiol Renal Physiol. 2006;291: Townes-Brocks syndrome. J Hum Genet.
F913–20. 2010;55:550–1.
52. Barua M, Shieh E, Schlondorff J, Genovese G, 64. Faguer S, Pillet A, Chassaing N, Merhenberger M,
Kaplan BS, Pollak MR. Exome sequencing and Bernadet-Monrozies P, Guitard J, Chauveau
in vitro studies identified podocalyxin as a candidate D. Nephropathy in Townes-Brocks syndrome
gene for focal and segmental glomerulosclerosis. Kid- (SALL1 mutation): imaging and pathological find-
ney Int. 2014;85:124–33. ings in adulthood. Nephrol Dial Transplant.
53. Alur RP, Vijayasarathy C, Brown JD, Mehtani M, 2009;24:1341–5.
Onojafe IF, Sergeev YV, Boobalan E, Jones M, 65. Nishinakamura R, Osafune K. Essential roles of Sall
Tang K, Liu H, et al. Papillorenal syndrome-causing family genes in kidney development. J Physiol Sci.
missense mutations in PAX2/Pax2 result in 2006;56:131–6.
hypomorphic alleles in mouse and human. PLoS 66. Basta JM, Robbins L, Kiefer SM, Dorsett D,
Genet. 2010;6:e1000870. Rauchman M. Sall1 balances self-renewal and differ-
54. Melnick M, Hodes ME, Nance WE, Yune H, entiation of renal progenitor cells. Development.
Sweeney A. Branchio-oto-renal dysplasia and 2014;141:1047–58.
branchio-oto dysplasia: two distinct autosomal dom- 67. Gribouval O, Gonzales M, Neuhaus T, Aziza J, Bieth E,
inant disorders. Clin Genet. 1978;13:425–42. Laurent N, Bouton JM, Feuillet F, Makni S, Ben
55. Chitayat D, Hodgkinson KA, Chen MF, Haber GD, Amar H, et al. Mutations in genes in the renin-
Nakishima S, Sando I. Branchio-oto-renal syndrome: angiotensin system are associated with autosomal reces-
further delineation of an underdiagnosed syndrome. sive renal tubular dysgenesis. Nat Genet. 2005;37:964–8.
Am J Med Genet. 1992;43:970–5. 68. Allanson JE, Hunter AG, Mettler GS, Jimenez
56. Orten DJ, Fischer SM, Sorensen JL, Radhakrishna U, C. Renal tubular dysgenesis: a not uncommon auto-
Cremers CW, Marres HA, Van Camp G, Welch KO, somal recessive syndrome: a review. Am J Med
Smith RJ, Kimberling WJ. Branchio-oto-renal syn- Genet. 1992;43:811–4.
drome (BOR): novel mutations in the EYA1 gene, and 69. Gribouval O, Moriniere V, Pawtowski A, Arrondel C,
a review of the mutational genetics of BOR. Hum Sallinen SL, Saloranta C, Clericuzio C, Viot G,
Mutat. 2008;29:537–44. Tantau J, Blesson S, et al. Spectrum of mutations in
57. Chang EH, Menezes M, Meyer NC, Cucci RA, the renin-angiotensin system genes in autosomal
Vervoort VS, Schwartz CE, Smith RJ. Branchio-oto- recessive renal tubular dysgenesis. Hum Mutat.
renal syndrome: the mutation spectrum in EYA1 and 2012;33:316–26.
its phenotypic consequences. Hum Mutat. 70. Mackie GG, Stephens FD. Duplex kidneys: a corre-
2004;23:582–9. lation of renal dysplasia with position of the ureteral
58. Abdelhak S, Kalatzis V, Heilig R, Compain S, orifice. J Urol. 1975;114:274–80.
Samson D, Vincent C, Levi-Acobas F, Cruaud C, Le 71. Murawski IJ, Maina RW, Malo D, Guay-Woodford
Merrer M, Mathieu M, et al. Clustering of mutations LM, Gros P, Fujiwara M, Morgan K, Gupta IR. The
responsible for branchio-oto-renal (BOR) syndrome C3H/HeJ inbred mouse is a model of vesico-ureteric
in the eyes absent homologous region (eyaHR) of reflux with a susceptibility locus on chromosome 12.
EYA1. Hum Mol Genet. 1997;6:2247–55. Kidney Int. 2010;78:269–78.
59. Kalatzis V, Sahly I, El-Amraoui A, Petit C. Eya1 72. Murawski IJ, Myburgh DB, Favor J, Gupta
expression in the developing ear and kidney: towards IR. Vesico-ureteric reflux and urinary tract develop-
the understanding of the pathogenesis of Branchio- ment in the Pax2 1Neu+/- mouse. Am J Physiol Renal
Oto-Renal (BOR) syndrome. Dev Dyn. Physiol. 2007;293:F1736–45.
1998;213:486–99. 73. Hains D, Sims-Lucas S, Kish K, Saha M, McHugh K,
60. Ruf RG, Xu PX, Silvius D, Otto EA, Beekmann F, Bates CM. Role of fibroblast growth factor receptor
Muerb UT, Kumar S, Neuhaus TJ, Kemper MJ, 2 in kidney mesenchyme. Pediatr Res.
Raymond Jr RM, et al. SIX1 mutations cause 2008;64:592–8.
5 Renal Dysplasia/Hypoplasia 133

74. Snodgrass WT, Shah A, Yang M, Kwon J, Roy A, Pascuet E, et al. A common RET variant is
Villanueva C, Traylor J, Pritzker K, Nakonezny PA, associated with reduced newborn kidney size and
Haley RW, Bush NC. Prevalence and risk factors for function. J Am Soc Nephrol. 2008;19:2027–34.
renal scars in children with febrile UTI and/or VUR: a 88. Quinlan J, Lemire M, Hudson T, Qu H, Benjamin A,
cross-sectional observational study of 565 consecutive Roy A, Pascuet E, Goodyer M, Raju C, Zhang Z,
patients. J Pediatr Urol. 2013;9:856–63. et al. A common variant of the PAX2 gene is associ-
75. Hunziker M, Colhoun E, Puri P. Prevalence and pre- ated with reduced newborn kidney size. J Am Soc
dictors of renal functional abnormalities of high grade Nephrol. 2007;18:1915–21.
vesicoureteral reflux. J Urol. 2013;190:1490–4. 89. Kaczmarczyk M, Goracy I, Loniewska B,
76. Chen MJ, Cheng HL, Chiou YY. Risk factors for renal Kuprjanowicz A, Binczak-Kuleta A, Clark JS,
scarring and deterioration of renal function in primary Ciechanowicz A. Association of BMPR1A polymor-
vesico-ureteral reflux children: a long-term follow-up phism, but not BMP4, with kidney size in full-term
retrospective cohort study. PLoS One. 2013;8: newborns. Pediatr Nephrol. 2012.
e57954. 90. El Kares R, Manolescu DC, Lakhal-Chaieb L,
77. Wilson JG, Roth CB, Warkany J. An analysis of the Montpetit A, Zhang Z, Bhat PV, Goodyer P. A
syndrome of malformations induced by maternal vita- human ALDH1A2 gene variant is associated with
min A deficiency. Effects of restoration of vitamin A increased newborn kidney size and serum retinoic
at various times during gestation. Am J Anat. acid. Kidney Int. 2010;78:96–102.
1953;92:189–217. 91. Mugford JW, Sipila P, McMahon JA, McMahon
78. Lelievre-Pegorier M, Vilar J, Ferrier ML, Moreau E, AP. Osr1 expression demarcates a multi-potent pop-
Freund N, Gilbert T, Merlet-Benichou C. Mild vita- ulation of intermediate mesoderm that undergoes pro-
min A deficiency leads to inborn nephron deficit in the gressive restriction to an Osr1-dependent nephron
rat. Kidney Int. 1998;54:1455–62. progenitor compartment within the mammalian kid-
79. Ross SA, McCaffery PJ, Drager UC, De Luca ney. Dev Biol. 2008;324:88–98.
LM. Retinoids in embryonal development. Physiol 92. Zhang Z, Iglesias D, Eliopoulos N, El Kares R, Chu L,
Rev. 2000;80:1021–54. Romagnani P, Goodyer P. A variant OSR1 allele
80. Vilar J, Gilbert T, Moreau E, Merlet-Benichou which disturbs OSR1 mRNA expression in renal pro-
C. Metanephros organogenesis is highly stimulated genitor cells is associated with reduction of newborn
by vitamin A derivatives in organ culture. Kidney Int. kidney size and function. Hum Mol Genet.
1996;49:1478–87. 2011;20:4167–74.
81. Gilbert T, Merlet-Benichou C. Retinoids and nephron 93. Lozic B, Krzelj V, Kuzmic-Prusac I, Kuzmanic-
mass control. Pediatr Nephrol. 2000;14:1137–44. Samija R, Capkun V, Lasan R, Zemunik T. The
82. Goodyer P, Kurpad A, Rekha S, Muthayya S, OSR1 rs12329305 polymorphism contributes to the
Dwarkanath P, Iyengar A, Philip B, Mhaskar A, development of congenital malformations in cases of
Benjamin A, Maharaj S, et al. Effects of maternal stillborn/neonatal death. Med Sci Monit.
vitamin A status on kidney development: a pilot 2014;20:1531–8.
study. Pediatr Nephrol. 2007;22:209–14. 94. Goodyer PCM, Wang L, Engert J. The PAX2-related
83. Tran S, Chen YW, Chenier I, Chan JS, Quaggin S, SNP rs11190739 is associated with accelerated loss of
Hebert MJ, Ingelfinger JR, Zhang SL. Maternal dia- GFR in Type I diabetes. ISN Forefronts Boston,
betes modulates renal morphogenesis in offspring. Genetic basis of Renal Disease. 2014;68.
J Am Soc Nephrol. 2008;19:943–52. 95. Eickmeyer AB, Casanova NF, He C, Smith EA,
84. Hokke SN, Armitage JA, Puelles VG, Short KM, Wan J, Bloom DA, Dillman JR. The natural history
Jones L, Smyth IM, Bertram JF, Cullen-McEwen of the multicystic dysplastic kidney – Is limited
LA. Altered ureteric branching morphogenesis and follow-up warranted? J Pediatr Urol.
nephron endowment in offspring of diabetic and 2014;10:655–61.
insulin-treated pregnancy. PLoS One. 2013;8: 96. Dogan CS, Torun-Bayram M, Aybar MD. Unilateral
e58243. multicystic dysplastic kidney in children. Turk J
85. Abi Khalil C, Travert F, Fetita S, Rouzet F, Porcher R, Pediatr. 2014;56:75–9.
Riveline JP, Hadjadj S, Larger E, Roussel R, Vexiau P, 97. Hayes WN, Watson AR. Unilateral multicystic dys-
et al. Fetal exposure to maternal type 1 diabetes is plastic kidney: does initial size matter? Pediatr
associated with renal dysfunction at adult age. Diabe- Nephrol. 2012;27:1335–40.
tes. 2010;59:2631–6. 98. Schreuder MF, Westland R, van Wijk JA. Unilateral
86. Cappuccini B, Torlone E, Ferri C, Arnone S, multicystic dysplastic kidney: a meta-analysis of
Troiani S, Bini V, Bellomo G, Barboni G, Di Renzo observational studies on the incidence,
G. Renal echo-3D and microalbuminuria in children associated urinary tract malformations and the con-
of diabetic mothers: a preliminary study. J Dev Ori- tralateral kidney. Nephrol Dial Transplant.
gins Health Dis. 2013;4:285–9. 2009;24:1810–8.
87. Zhang Z, Quinlan J, Hoy W, Hughson MD, 99. Sharada S, Vijayakumar M, Nageswaran P,
Lemire M, Hudson T, Hueber PA, Benjamin A, Ekambaram S, Udani A. Multicystic dysplastic
134 P. Goodyer and I.R. Gupta

kidney: a retrospective study. Indian Pediatr. 107. Stein JP, Kurzrock EA, Freeman JA, Esrig D, Gins-
2014;51:641–3. berg DA, Grossfeld GD, Hardy BE. Right intratho-
100. Hains DS, Bates CM, Ingraham S, Schwaderer racic renal ectopia: a case report and review of the
AL. Management and etiology of the unilateral literature. Tech Urol. 1999;5:166–8.
multicystic dysplastic kidney: a review. Pediatr 108. Glenn JF. Analysis of 51 patients with horseshoe
Nephrol. 2009;24:233–41. kidney. N Engl J Med. 1959;261:684–7.
101. Moralioglu S, Celayir AC, Bosnali O, Pektas OZ, 109. Cascio S, Sweeney B, Granata C, Piaggio G,
Bulut IK. Single center experience in patients with Jasonni V, Puri P. Vesicoureteral reflux and
unilateral multicystic dysplastic kidney. J Pediatr ureteropelvic junction obstruction in children with
Urol. 2014;10:763–8. horseshoe kidney: treatment and outcome. J Urol.
102. Caruana G, Wong MN, Walker A, Heloury Y, 2002;167:2566–8.
Webb N, Johnstone L, James PA, Burgess T, Bertram 110. Raj GV, Auge BK, Assimos D, Preminger
JF. Copy-number variation associated with congenital GM. Metabolic abnormalities associated with renal
anomalies of the kidney and urinary tract. Pediatr calculi in patients with horseshoe kidneys. J
Nephrol. 2014. Endourol. 2004;18:157–61.
103. Hasui M, Kaneko K, Tsuji S, Isozaki Y, Kimata T, 111. Boatman DL, Kolln CP, Flocks RH. Congenital
Nozu Y, Nozu K, Iijima K. Different phenotypes of anomalies associated with horseshoe kidney. J Urol.
HNF1ss deletion mutants in familial multicystic dys- 1972;107:205–7.
plastic kidneys. Clin Nephrol. 2013;79:484–7. 112. Neville HL, Ritchey ML. Wilms’ tumor. Overview of
104. Decramer S, Parant O, Beaufils S, Clauin S, National Wilms’ Tumor Study Group results. Urol
Guillou C, Kessler S, Aziza J, Bandin F, Schanstra Clin North Am. 2000;27:435–42.
JP, Bellanne-Chantelot C. Anomalies of the TCF2 113. Flynn MT, Ekstrom L, De Arce M, Costigan C, Hoey
gene are the main cause of fetal bilateral HM. Prevalence of renal malformation in Turner syn-
hyperechogenic kidneys. J Am Soc Nephrol. drome. Pediatr Nephrol. 1996;10:498–500.
2007;18:923–33. 114. Bilge I, Kayserili H, Emre S, Nayir A, Sirin A,
105. Body-Bechou D, Loget P, D’Herve D, Le Fiblec B, Tukel T, Bas F, Kilic G, Basaran S, Gunoz H,
Grebille AG, Le Guern H, Labarthe C, Redpath M, et al. Frequency of renal malformations in Turner
Cabaret-Dufour AS, Sylvie O, et al. TCF2/HNF- syndrome: analysis of 82 Turkish children. Pediatr
1beta mutations: 3 cases of fetal severe pancreatic Nephrol. 2000;14:1111–4.
agenesis or hypoplasia and multicystic renal dyspla- 115. Bhojwani N, Hartman JB, Ahmed M, Morgan R,
sia. Prenat Diagn. 2014;34:90–3. Davidson JC. Management of ureteral obstruction in
106. Shapiro E. Upper urinary tract anomalies and perina- crossed fused renal ectopia: a case report. Can Urol
tal renal tumors. Clin Perinatol. 2014;41:679–94. Assoc J. 2014;8:E752–4.
Developmental Syndromes
and Malformations of the Urinary Tract 6
Chanin Limwongse

Contents Introduction
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Birth defects involving the kidney and urinary
Prevalence of Urinary Tract Anomalies . . . . . . . . . . 157
system are often encountered and frequently
Approach to the Child with a Urinary Tract occur in association with other structural abnor-
Anomaly . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 158
malities. A congenital urinary tract anomaly may
Overview of Normal Embryogenesis of the provide the first clue to the recognition of
Urinary System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161 multiorgan developmental abnormalities. Never-
Anomalies Involving the Urinary Tract . . . . . . . . . . 163 theless many renal anomalies remain asymptom-
Kidney Defects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163 atic and undiagnosed. Therefore it is critical, not
Ureter Defects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
Bladder Defects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 166
only for pediatric nephrologists but also for pedi-
Urethral Defects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167 atricians in general, to be familiar with the com-
mon anomalies involving the kidney and urinary
Associations and Sequences Involving the
Urinary Tract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167 system and the more complex disorders with
VATER Association . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167 which they may be associated.
CHARGE Syndrome (CHARGE Association) . . . . . 168 The kidney is a pivotal organ in
MURCS Association . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168 dysmorphology. Although the number of single
Oligohydramnios Sequence . . . . . . . . . . . . . . . . . . . . . . . . . 169
Urethral Obstruction Sequence . . . . . . . . . . . . . . . . . . . . . . 169 malformations involving the kidney is limited,
Sirenomelia Sequence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 169 combinations of these malformations in conjunc-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
tion with anomalies involving other organ sys-
tems are found in more than 500 syndromes. In
addition, many well-known sequences and asso-
ciations involve the kidney and urinary tract. This
chapter discusses common malformations,
sequences, and associations involving the kidney
and urinary tract and provides a summary of con-
ditions that have these anomalies as one of their
features. In addition, Tables 1, 2, and 3 summarize
more detailed information about a large number of
C. Limwongse (*) disorders, including their phenotypic features,
Department of Medicine, Division of Medical Genetics,
reported urinary tract anomalies, pattern of inher-
Faculty of Medicine, Siriraj Hospital, Mahidol University,
Bangkoknoi, Bangkok, Thailand itance, causative genes, and related references.
e-mail: chanin.lim@mahidol.ac.th; siclw@mahidol.ac.th These tables can be used both to provide readily
# Springer-Verlag Berlin Heidelberg 2016 135
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_5
136

Table 1 Syndromes and disorders that have urinary tract anomalies as a frequent feature
Urinary tract abnormalities

Other
associated

Renala genesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies anomalies Gene(s) References
Abruzzo-Erickson H Coloboma, cleft palate, XR Unk [48]
hypospadias, deafness,
short stature
Acrocephalopolydactylous + + + Acrodactyly, hand AR Unk [49]
dysplasia (Elejalde hexadactyly, overgrowth,
syndrome) visceromegaly, globular
body, redundant neck skin
Acrorenal (Dieker) 1 E + + + Ua Ectrodactyly, oligodactyly, Sporadic Unk [50]
hypoplastic carpal/tarsal
bones
Acrorenal (Johnson- 1, U, Aphalangy, hemivertebrae, AR Unk [50]
Munson) 2 Ua genital/intestinal/anal
dysgenesis
Acrorenal (Siegler) E + U + Short stature, hypoplastic Uncertain Unk [50]
radii/ulnae/humeri,
oligodactyly
Acrorenal-mandibular 1 + Ectrodactyly, hypoplastic AR Unk [51]
mandible
Acrorenal-ocular 1 E + B Hypoplastic thumb, optic AD SALL4 [52]
coloboma, cleft lip/palate
Adrenoleukodystrophy, See Pseudo-Zellweger
neonatal
Aglossia-adactylia 1 Micrognathia, cranial nerve Sporadic Unk [53]
palsy
Agnathia- H + Arrhinencephaly, situs Sporadic Unk [54]
holoprosencephaly inversus, midline defects
C. Limwongse
6

Alagille 1 + + + Cholestasis, peripheral AD JAG1 [55]


pulmonic stenosis,
characteristic face
Alport + Nephritis, proteinuria, AD, AR, XL COL4 [56]
deafness
Alsing + Nephritis, AR Unk [57]
nephronophthisis, optic
coloboma, hip dislocation
Alstrom + Diabetes mellitus, AR ALMS1 [58]
retinopathy, short stature,
deafness
Amelogenesis imperfecta + Enamel hypoplasia, AR Unk [59]
type IG nephrolithiasis, enuresis
Amyloidosis type 5 + Nephropathy, proteinuria, AD GELSOLIN [60]
cranial nerve palsy, cutis
laxa
Angiotensin-converting + IUGR, oligohydramnios, Sporadic none [61]
enzyme (ACE) inhibitor, patent ductus arteriosus,
maternal use limb anomalies, renal artery
stenosis, progressive renal
failure
Aniridia-Wilms tumor + Aniridia, ambiguous AD WT1 [62]
(WAGR) genitalia, hypospadia, short
Developmental Syndromes and Malformations of the Urinary Tract

stature
Axial mesodermal 1 Bladder exstrophy, Uncertain Unk [63]
dysplasia vertebral anomalies,
Goldenhar-like
Baldellou 1 Hypoparathyroidism, Uncertain Unk [64]
ocular coloboma, MR,
seizures
Baller-Gerold + E + + Craniosynostosis, radial AR RECQL4 [65]
aplasia, malformed ear, anal
atresia
Barakat + Mesangial sclerosis, MR, AD GATA3 [66]
optic atrophy, nystagmus
Bardet-Biedl + + + + + + Obesity, polysyndactyly, AR BBS1-12 [67, 68]
MR, retinopathy,
hypogonadism
137

(continued)
138

Table 1 (continued)
Urinary tract abnormalities

Other
associated

Renala genesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies anomalies Gene(s) References
Beckwith-Wiedemann E + + + + Overgrowth, macroglossia, Sporadic, AD CDKN1C, [69–72]
omphalocele, embryonal NSD1, H19
tumors
Berardinelli + + Insulin resistance, AR BSCL2 [73]
lipodystrophy, acanthosis
nigricans, MR
Brachymesomelia-renal + Micrognathia, corneal Uncertain Unk [74]
opacity, craniofacial
dysmorphism
Branchio-oculo-facial 1 Philtrum hypertrophy, cleft AD TFAP2A [75]
lip/palate, branchial
remnant
Branchio-oto-renal 1, E + + + + + Branchial remnant, AD EYA1, SIX5 [76]
2 preauricular pit/tag,
microtia, deafness
Braun-Bayer + U + Deafness, bifid uvula, Uncertain Unk [77]
digital anomalies
Cat eye See Table 3 Chromosomal
Caudal duplication 1, E + + + Duplication of colon, Sporadic AXIN1 [78]
2 sacrum, genitalia, vertebral
defects
Caudal regression 1, E, + + Ua + Atresia of colon, anus, AD, AR VANGL1 [79, 80]
2 H genitalia, vertebral defects,
TE fistula
Cerebro-hepato-renal + See Zellweger syndrome AR PEX [81]
(Passarge)
C. Limwongse
6

Cerebro-oculo-hepato- + Cerebellar hypoplasia, AR Unk [82]


renal hepatic fibrosis, Leber
amaurosis
Cerebro-osteo-nephro + + Rhizomelic limb AR Unk [83]
dysplasia shortening, cerebral
atrophy, MR, seizures
CHARGE + E + + + + U + See text for details Sporadic, AD CHD7 [33–37]
Chondroectodermal 1 + + + Acromelic dwarfism, AR EVC, EVC2 [84, 85]
dysplasia (Ellis van polydactyly, nail dystrophy,
Creveld) tooth hypoplasia narrow
thorax, CHD
Cocaine, maternal use 1, + + Ua + Vascular disruption Sporadic none [86]
2 anomalies affecting
multiple organs
Cornelia de Lange 1 + + + + SS, microcephaly, limb Sporadic NIPBL, [87]
defect, hirsutism, synophrys SMC1L1
Crossed renal ectopia- E + U Clubbing of fingers, Uncertain Unk [88]
pelvic lipomatosis gynecomastia
Czeizel + U Ectrodactyly, spina bifida, AD Unk [89]
megacystis
Diabetic mother, infant of 1, E, + + + + Ua + Neural tube defect, cardiac/ Sporadic none [90, 91]
2 H limb anomalies, sacral
agenesis
Developmental Syndromes and Malformations of the Urinary Tract

DiGeorge/velocardiofacial 1, E + + + + + See Table 3 Chromosomal


3
Denys-Drash E + + + Pseudohermaphroditism, Sporadic WT1 [92]
Wilms tumor, proteinuria
Down See Table 3 Chromosomal
Ectrodactyly-ectodermal 1 + + + + Ectrodactyly, hypohidrosis, AD Unk [93]
dysplasia-clefting (EEC) sparse hair, cleft lip/palate
Elejalde See
acrocephalopolydactylous
dysplasia
(continued)
139
140

Table 1 (continued)
Urinary tract abnormalities

Other
associated

Renala genesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies anomalies Gene(s) References
Epstein + Thrombocytopenia, nerve AD MYH9 [94]
deafness, cataract
Facio-cardio-renal H + U Cardiomyopathy, AR Unk [95]
conduction defect, MR,
typical face
Fanconi anemia 1 E, + + + + Pancytopenia, limb defects, AR FANCA-N [96, 97]
H leukemia, lymphoma
Fetal alcohol 1 E, + + + + IUGR, DD, microcephaly, Sporadic none [98]
H short palpebral fissure
Fibromatosis, infantile + + Multiple myofibromatosis, AR Unk [99]
myositis ossificans
Fraser cryptophthalmos 1, + U Fused eyelids, ear/genital AR FRAS1 [100]
2 anomalies, syndactyly
Frasier + + Male pseudohermaphrodite, AD WT1 [101]
amenorrhea, ovarian cysts
Goeminne + + Congenital torticollis, XR Unk [102]
keloids, cryptorchidism
Goldenhar (oculo-auriculo- 1 E + + + + + Hemifacial microsomia, ear Sporadic, AD Unk [103]
vertebral) anomalies, vertebral defects
Goldston + Dandy-Walker Uncertain NPHP3 [104]
malformation, cerebellar
malformation
Graham + + Cystic hamartoma of lung Sporadic Unk [105]
and kidney
C. Limwongse
6

Hemifacial microsomia See Goldenhar syndrome


(oculo-auriculo-vertebral)
Hemihyperplasia + + Asymmetry, vascular Sporadic LIT1, H19 [106]
malformation, embryonal
tumors
Hepatic fibrosis + Congenital hepatic fibrosis Sporadic PKHD1 [107]
Holzgreve 2 Potter sequence, cardiac Uncertain Unk [108]
defect, polydactyly, cleft
palate
Hypertelorism-microtia- E + Microcephaly, cleft AR Unk [109]
clefting lip/palate, MR
Ivemark 1 + + + Polysplenia/asplenia, Sporadic, AR Unk [110]
complex CHD, laterality
defects
Jeune + Narrow chest, short limbs, AR EVC, EVC2 [84, 85]
polydactyly,
glomerulosclerosis
Joubert + Vermis aplasia, apnea, jerky AR JBTS1–7 [111]
eyes, retinopathy, ataxia
Juberg-Hayward H + Microcephaly, cleft AR Unk [112]
lip/palate, abnormal
thumbs/toes
Kabuki H + + U + MR, characteristic Kabuki- Uncertain Unk [113]
Developmental Syndromes and Malformations of the Urinary Tract

like face, large ears, cleft


palate
Kallmann 1 + Anosmia, cleft lip/palate, AD, AR, XR KAL1–4 [114]
hypogonadotropic
hypogonadism
Kaufman-McKusick E + + + Ua Hydrometrocolpos, AR GLI3 [115]
polydactyly, anal/urogenital
sinus defect
Kivlin E + Short stature, Peters’ AR B3GALTL [116]
anomaly, MR, genital/
cardiac defects
(continued)
141
Table 1 (continued)
142

Urinary tract abnormalities

Other
associated

Renala genesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies anomalies Gene(s) References
Klippel-Feil 1 E + Short neck, cervical Sporadic, AD Unk [117]
vertebral fusion, low
posterior hairline
Kousseff 1 Sacral meningocele, AR Unk [118]
hydrocephalus, cardiac
defects
Leprechaunism (Donohue) + + Insulin resistance, AR INSR [119]
lipodystrophy, hirsutism
Limb-body wall complex 1 E + U, Lateral body wall defect, Sporadic Unk [120]
Ua limb reduction, CHD
Mammo-renal + Ipsilateral supernumerary Sporadic Unk [121]
breasts/nipples
Marden-Walker + + Microcephaly, AR Unk [122]
blepharophimosis,
micrognathia, contractures
Meckel-Gruber + + U, Encephalocele, cardiac AR MKS1–4 [123]
Ua defects, cleft lip/palate,
polydactyly
Megacystis-microcolon + U Large bladder, intestinal AR Unk [124]
hypoperistalsis,
oligohydramnios
Melnick-Needles + U Bowing long bones, short XR FLNA [125]
osteodysplasty upper limbs, micrognathia
Mendenhall + Insulin resistance, AR INSR [126]
acanthosis nigricans
Microgastria-upper limb 1 E + Hypoplastic spleen, limb Uncertain Unk [127]
anomaly reduction defects
C. Limwongse
6

Miranda + + Brain malformation, liver Uncertain Unk [128]


dysplasia
Moerman 1 + + + + Short limbs, brain Uncertain Unk [129]
malformation, cleft palate
MURCS association 1 E + U See text for details Sporadic Unk
Nager acrofacial dysostosis + + + + Facial bone hypoplasia, AD Unk [130]
cleft eyelid, radial ray defect
Nail-patella + Absent/hypoplastic nails AD LMX1B [131]
and patellae
Neu-Laxova 1 IUGR, lissencephaly, CHD, AR,sporadic Unk [132]
pterygia, ichthyosis
Neurofaciodigitorenal 1 Prominent forehead with Uncertain Unk [133]
vertical groove, MR, ear
anomaly
Neurofibromatosis type I 1 Ua + renal artery stenosis, AD NeUnkro [134]
hypernephroma, cafe-au- fibromin
lait spots
Nezelof + + Arthrogryposis, hepatic AR Unk [135]
impairment, hypotonia,
club feet
Noonan + + + Webbed neck, short stature, AD NS1 [136]
MR, pulmonic stenosis
Developmental Syndromes and Malformations of the Urinary Tract

Occipital horn + B Ua Cranial exostosis, XR cUnkATPase [137]


hyperextensibility, cutis
laxa
Ochoa + + B U, Ua + Facial grimacing with Uncertain Unk [138]
lateral displacement of
mouth
Oculo-auriculo-vertebral See Goldenhar syndrome
Oculo-hepato-encephalo- + + Encephalocele, hepatic AR Unk [82]
renal fibrosis, coloboma
Oculorenal-cerebellar + MR, spastic diplegia, AR Unk [139]
choreoathetosis,
retinopathy
(continued)
143
144

Table 1 (continued)
Urinary tract abnormalities

Other
associated

Renala genesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies anomalies Gene(s) References
Oculorenal See Pierson syndrome
Oculorenal (Karcher) 1 + + Optic nerve coloboma AD Unk [140]
OEIS (omphalocele- + + Ua + Malrotation of colon, sacral Sporadic Unk
exstrophy of bladder- defect, tethered spinal cord,
imperforate anus-spinal pelvic bone abnormalities
dysraphism) complex
Otorenal + + + U Renal pelvis diverticula, AD Unk [141]
nerve deafness
Pallister-Hall 1, E, + + Hypothalamic AD GLI3 [142, 143]
2 H hamartoblastoma,
polydactyly
Pierson + Hypoplastic retina, cataract, AR LAMB2 [144]
anterior chamber anomalies
Penoscrotal transposition E + + + B U Abnormal placement of Sporadic Unk [145]
external genitalia
Perlman + + + + + Early overgrowth, typical AR Unk [146]
face, nephroblastomatosis
Polydactyly-obstructive + Ua + Post-axial polydactyly of Uncertain Unk [147]
uropathy hands and feet
Potter (oligohydramnios) 1, + + Ua See text for details Sporadic Unk
2
Prune belly + + + Ua + See text for details Sporadic Unk
Pseudo-Zellweger + + Hypotonia, seizures, MR, AR PTS1 [148–150]
typical face, FTT,
hepatomegaly
Pyloric stenosis H + + + + Cystic kidney Sporadic Unk [151]
C. Limwongse
6

Raas-Rothschild + + Klippel-Feil anomaly, Uncertain Unk [152]


sacral agenesis,
cryptorchidism
RAPADILINO + Radial/patella hypoplasia, AR RECQL4 [153]
diarrhea, short stature, long
nose
Renal/Mullerian H + + Absent uterus, broad AR Unk [154]
hypoplasia forehead, DD, large
fontanel
Renal dysplasia or 1 E + + + Ua + Abnormal uterus in some AD RET, [155]
adysplasia patients UnkPK3A
Renal-hepatic-pancreatic + Pancreatic cysts, AR EVC, EVC2 [84, 85]
dysplasia extrahepatic biliary atresia,
Caroli disease
Retinoic acid, maternal use + + U Ear anomalies, CHD, cleft Sporadic none [156, 157]
palate, neural tube defect
Roberts 1 H + + Limb reduction, oligo/ AR ESCO2 [158]
syndactyly, CHD,
dysmorphic face
Robson + MR, macrocephaly, XR COL4A [159]
deafness, proteinuria,
Alport-like
Rokitansky-Mayer-Kuster- 1 E + U + Absence of vagina, uterine Sporadic Unk [160]
Developmental Syndromes and Malformations of the Urinary Tract

Hauser anomalies, amenorrhea


Rubella, congenital 1 + + CHD, MR, deafness, Sporadic none [161]
cataract, growth retardation
Rubinstein-Taybi 1 E + + + Ua SS, MR, broad thumbs and Sporadic CREBBP [162, 163]
great toes, typical face
Russell-Silver + Ua SS, triangular face, Sporadic,AD Unk [164]
asymmetry, clinodactyly,
hypoglycemia
Santos 1 Hirschsprung disease, AR Unk [165]
hearing loss, postaxial
polydactyly
(continued)
145
146

Table 1 (continued)
Urinary tract abnormalities

Other
associated

Renala genesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies anomalies Gene(s) References
Say + SS, microcephaly, AD Unk [166]
micrognathia, large ear,
cleft palate
Schimke + SS, spondyloepiphyseal AR SMARCAL1 [167]
dysplasia,
immunodeficiency
Schinzel-Giedion E U + U CHD, distinctive face, AR Unk [168]
figure 8 head shape, eyelid
groove
Senior-Loken + + Nephronophthisis, AR NPHP1,4,5 [169]
tapetoretinal degeneration
Setleis + U Cutis aplasia with temporal AD,AR Unk [170]
scarring, abnormal
eyelashes
Short rib, Beemer Langer + + + U Hydrops, cleft lip, bowed AR Unk [171]
long bones, atretic ear canal
Short rib-polydactyly, type 1 + Ua Urethral fistula, CHD, AR Unk [171, 172]
1–3 cloacal/urogenital sinus
anomalies
Silverman (dyssegmental + U SS, flat face, cleft palate, Uncertain HSPG2 [173]
dwarfism) generalized skeletal
dysplasia
Simopoulos + Hydrocephalus, AR Unk [174]
polydactyly
Simpson-Golabi-Behmel + + + Overgrowth, polydactyly, XR GPC3 [175]
typical face, arrhythmia
C. Limwongse
6

Sirenomelia sequence 1, E + + + + + + + See text for details Sporadic Unk


2
Smith-Lemli-Opitz 1 + + SS, ambiguous genitalia, AR DHCR7 [176, 177]
2–3 toe syndactyly, brain
anomalies
Sommer 1 Iris aplasia, corneal opacity, AD Unk [178]
glaucoma, prominent
forehead
Sorsby (coloboma- 1 Ocular coloboma, AD Unk [179]
brachydactyly) brachydactyly type B, bifid
thumbs
Sotos Ua + Overgrowth, MR, Sporadic NSD1 [180]
embryonal tumors,
advanced bone age
Supernumerary nipples- E + + + + Familial polythelia AD Unk [181]
renal anomalies
Thalidomide, maternal use 1, E, + + + + + Limb reduction, Sporadic none [182, 183]
2 H phocomelia, neural tube
defect
Thymic-renal-anal-lung + + U SS, absent thymus, AR Unk [184]
parathyroid agenesis,
urethral fistula
Tolmie + Lethal multiple pterygia, XR CHRN [185]
Developmental Syndromes and Malformations of the Urinary Tract

long bone abnormalities


Townes-Brock 1 + + Ua + Triphalangeal thumb, AD SALL1 [186]
imperforate anus, skin tag,
deafness
Trimethadione, maternal 1 U SS, CHD, omphalocele, Sporadic none [187]
use distinctive face
Tuberous sclerosis + + MR, seizures, cortical tuber, AD TS1, TS2 [188]
facial angiofibroma
Turner See Table 3 Chromosomal
Ulnar-mammary 1 Oligodactyly, ulnar ray AD TBX3 [189]
defect, nipple aplasia,
genital defects
(continued)
147
148

Table 1 (continued)
Urinary tract abnormalities

Other
associated

Renala genesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies anomalies Gene(s) References
Urogenital adysplasia 1, + Absent uterus, vaginal AD RET, [190]
2 atresia, hydrometrocolpos UnkPK3A
VATER (VACTERL) 1 E, + + + + + + + See text for details
association H
Velocardiofacial 1, E, + + + + + See Table 3
2 H
von Hippel-Lindau + + Cerebello-retinal AD VHL [191, 192]
angiomatosis,
pheochromocytoma
Weinberg-Zumwalt + + Multiple lung cysts, ascites, Uncertain Unk [193]
accessory spleen
Wenstrup 1 + + + U Female Uncertain Unk [194]
pseudohermaphrodite,
imperforate anus
Weyers H + + + U Oligodactyly, pterygia, AR EVC [195]
sternal defect, cleft palate
Williams E + + B, U See Table 3 Chromosomal
U
Wilms tumor-horseshoe H + Possible association Sporadic WT1 [196]
kidney
C. Limwongse
6

Wilms tumor- + Ipsilateral vascular Sporadic WT1 [106]


hemihypertrophy malformation, cafe-au-lait
spots
Wilms tumor-radial aplasia + Hypoplastic fibula/tibia, Sporadic Unk [197]
abnormal thumbs
Winter (oto-renal-genital) 1, + Middle ear anomalies, AR Unk [198]
2 deafness, vaginal atresia
Wolfram (DIDMOAD) + + Diabetes mellitus/insipidus, AR, WFS1–2 [199]
optic atrophy, nerve mitochondrial
deafness
Zellweger 1 + + Hypotonia, seizures, AR PEX [148–150]
hepatosplenomegaly,
growth delay
U ureter, B bladder, Ua urethra, 1 unilateral renal agenesis, 2 bilateral renal agenesis, E ectopia, H horseshoe, ACC agenesis of corpus callosum, SS short stature, MR mental
retardation, DD developmental delay, CHD congenital heart disease, FTT failure to thrive, IUGR intrauterine growth retardation, Unk gene unknown
Developmental Syndromes and Malformations of the Urinary Tract
149
Table 2 Well-known syndromes associated with occasional urinary tract anomalies
150

Inheritance

Renal agenesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies pattern Gene(s) References
Aase Ua Triphalangeal thumb, AD,AR RPS19, 24 [200]
hypoplastic anemia
Achondrogenesis + + Micromelic dwarfism, short AR COL2A1 [201]
trunk, fetal hydrops
Acrocallosal 1 + ACC, macrocephaly, AR GLI3 [202]
polymicrogyria, polydactyly,
CHD
Acro-facial dysostosis H + Abnormal thumb/toe, facial AR Unk [203]
bone defect, ear anomalies
Acromelic frontonasal E + Polydactyly, ACC, Sporadic Unk [204]
dysplasia encephalocele, Dandy-
Walker anomaly
Adrenogenital 1 + + Ambiguous genitalia, AR CYP11,21 [205]
vomiting, salt losing, UPJ
obstruction
Adrenal hypoplasia-MR + U + Aminoaciduria, MR, X-linked NROB1 [206]
muscular dystrophy, visual
abnormality
Antley-Bixler H + Craniosynostosis, radio- AR FGFR2 [207]
humeral synostosis, cardiac
defects
Apert (acrocephalo + + Acrocephaly, AD FGFR2 [208]
syndactyly) craniosynostosis, syndactyly
Bloom + Short stature, telangiectasias, AR BLM [209]
leukemia, lymphoma
Bowen-Conradi H + Micrognathia, AR Unk [210]
arthrogryposis, cloudy
cornea, brain anomaly
C. Limwongse
6

Brachydactyly type E 1 + Vertebral anomalies, narrow Uncertain HOXD13 [211]


auditory canal
3C (Ritscher-Schinzel) + SS, Dandy-Walker anomaly, AR Unk [212]
typical face, CHD
C-trigonocephaly 1 Polysyndactyly, abnormal AR CD96 [213]
ear, hypospadias, dislocated
joints
Campomelic dysplasia 1 + + Tibial bowing, pretibial AD SOX9 [214]
dimples, ambiguous
genitalia
Carbohydrate deficient + FTT, abnormal fat pad, AR PMM2, [215]
glycoprotein hepatosplenomegaly, MPI
neurodegeneration
Carpenter + Aminoaciduria, AD RAB23 [216]
polysyndactyly,
craniosynostosis
CHILD 1,2 + Unilateral erythroderma, X-linked NSDHL [217]
ipsilateral limb defect
Chondrodysplasia + Flat face, microcephaly, AR Unk [218]
punctata, non-rhizomelic cataract, short femora/
humeri, stippled epiphyses
Coffin-Siris 1 + MR, sparse scalp hair, AR Unk [219]
hirsutism, coarse face, thick
Developmental Syndromes and Malformations of the Urinary Tract

lips
Cutis la + a type I B GI tract diverticula, AR FBLN5 [220]
emphysema, diaphragmatic
defect
Disorganization-like + Polydactyly, duplication of Sporadic Unk [221]
lower limbs, skin
appendages
Duane anomaly-radial 1 Limited ocular abduction, AD SALL4 [222]
defects radial defects,
blepharophimosis
Ehlers-Danlos + Joint hypermobility, skin AD, AR, XR COL5A, [223]
hyperextensibility, easy COL1A1
bruising
(continued)
151
152

Table 2 (continued)

Inheritance

Renal agenesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies pattern Gene(s) References
Epidermolysis bullosa + U Skin blistering, pyloric AR LAMA3, [224–245]
stenosis, dystrophic nails, LAMB3
sparse hair
Femoral hypoplasia- 1 Various leg deformities, Sporadic, AD Unk [226]
unusual facies abnormal genitalia, typical
face
Floating-Harbor E SS, typical face, DD, Uncertain Unk [227]
delayed bone age
Focal dermal hypoplasia H Atrophy/linear skin X-linked PORCN [228]
pigmentation, hand/vertebral
anomalies
Freeman-Sheldon + + Whistling face, ulnar AD TNNT3, [229]
deviation of hands, talipes TNNI2
equinovarus
Frontometaphyseal + + Prominent supraorbital X-linked FLNA [230]
dysplasia ridges, contractures,
deafness
Frontonasal dysplasia + E Hypertelorism, broad nasal Sporadic Unk [231]
tip, median cleft nose
Fryns + Digital hypoplasia, AR Unk [232]
diaphragmatic defect, cleft
palate
G (Opitz/BBB) + + + See Opitz (G/BBB)
syndrome
Glutaric aciduria, type II + Cerebral anomalies, AR ETFA, B, [233]
pancreatic dysplasia, biliary DH
dysgenesis
C. Limwongse
6

Grieg cephalopoly + Macrocephaly, polydactyly, AD GLI3 [234]


syndactyly hypertelorism
Hajdu-Cheney + SS, Wormian bones, acro- AD Unk [235]
osteolysis, osteoporosis
Hydrolethalus + Ua Hydrocephalus, polydactyly, AR HYLS1 [236]
polyhydramnios, cleft lip
Jarcho-Levin + Spondylothoracic dysplasia, AR DLL3, [237]
fused ribs, hemivertebrae MESP2
Johanson-Blizzard + Pancreatic insufficiency, AR UnkBR1 [238]
spiky hair, small alae nasi
Killian-Pallister See Table 3 Chromosomal
Lacrimo-auriculo-dento- 1 Nasolacrimal duct stenosis, AD FGFR2, 3 [239]
digital (LADD) malformed ears/enamel/
digits
Larsen 1 + Multiple joint dislocations, AD, AR FLNB [240, 241]
flat face
Lenz microphthalmia 1 + + Ocular coloboma, ear/facial X-linked Unk [242]
anomalies, syndactyly/
camptodactyly
LEOPARD (multiple 1 Hypertelorism, deafness, AD PTPN11, [243]
lentigines) abnormal ECG, genital RAF1
anomalies
Developmental Syndromes and Malformations of the Urinary Tract

Marfan + + Tall thin habitus, aortic root AD FBN1 [244, 245]


dilatation, lens
sublu + ation,
arachnodactyly
Marshall-Smith + MR, FTT, accelerated bone Sporadic Unk [246]
maturation, broad phalanges
Miller-Dieker 1 + See Table 3 Microdeletion LIS1
(lissencephaly)
Moebius-peripheral + Peripheral neuropathy, Sporadic Unk [247]
neuropathy anosmia, hypogonadism
Mohr-Majewski See oro-facio-digital
syndrome type IV
Multiple pterygium + Multiple soft tissue AR CHRNG [248]
contractures, camptodactyly
(continued)
153
154

Table 2 (continued)

Inheritance

Renal agenesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Syndromes Other associated anomalies pattern Gene(s) References
Myotonic dystrophy + Myotonia, muscle weakness, AD DMPK [249, 250]
cataract, arrhythmia,
diabetes
Nijmegen breakage + MR, SS, microcephaly, AR NBS1 [251]
immunodeficiency
Opitz (G/BBB) Hypertelorism, hypospadias, AD, X-linked MID1 [252, 253]
cleft lip/palate, dysphagia
Oro-facio-digital, type I + + Midline cleft lip, multiple X-linked CXORF5 [254]
frenula, polydactyly, tongue
nodules
Oro-facio-digital, type IV 1 + Cleft palate, multiple AR Unk [254, 255]
frenula, polysyndactyly,
lobed tongue
Oro-facio-digital, type VI 1 + Midline cleft lip, multiple AR Unk [254]
frenula, polydactyly, tongue
nodules
Pallister-Killian See Table 3 Chromosomal
Peutz-Jeghers + Hamartomatous intestinal AD STK11 [256, 257]
polyposis, lip
hyperpigmentation
Poland anomaly 1 + + Hypoplastic pectoralis, Sporadic Unk [258, 259]
ipsilateral upper limb
reduction
Restrictive dermopathy U Aplasia cutis, rigid skin, AR LMNA, [260]
contractures, typical face ZMPSTE24
Robinow + + Mesomelic dwarfism, typical AD, AR ROR2 [261]
face, abnormal genitalia
C. Limwongse
6

Rothmund-Thomson + Poikiloderma, alopecia, AR RECQL4 [262]


dysplastic nails,
photosensitivity
Serpentine fibula + Elongated curved fibulae, Uncertain Unk [263]
hirsutism, hypertelorism
Spondylocostal 1 + Sacral agenesis, anal atresia, Uncertain Unk [264]
dysostosis bifid thumb, skin tags
Spondyloepimetaphyseal + U Joint laxity, kyphoscoliosis, AR COL2A1 [265]
dysplasia talipes equinovarus, CHD
Spondylometaphyseal + SS, platyspondyly, co + a AD Unk [266]
dysplasia vara, vertebral/long bone
anomalies
Syndactyly, type V E + Bladder exstrophy, fusion of AD HOXD13 [267]
4th and 5th metacarpal bones
U ureter, B bladder, Ua urethra, 1 unilateral renal agenesis, 2 bilateral renal agenesis, E ectopia, H horseshoe, ACC agenesis of corpus callosum, SS short stature, MR mental
retardation, DD developmental delay, CHD congenital heart disease, FTT failure to thrive, IUGR intrauterine growth retardation, Unk gene unknown
Developmental Syndromes and Malformations of the Urinary Tract
155
Table 3 Chromosomal disorders and their consistent associated urinary tract anomalies
156

Reported

Renal agenesis
Ectopia=horseshoe
Cystic=dysplasia
Duplication
Hypoplasia
Hydronephrosis=ureter
Diverticula
Atresia=stenosis
Reflux
Nephritis=sclerosis
Tumor=nephromegaly
Chromosomal disorders Other associated anomalies familial cases References
3p deletion E, + MR, growth delay, ptosis, postaxial polydactyly, No [270]
H micrognathia
3q duplication H + + + MR, SS, seizures, hirsutism, typical face, cardiac No [271]
defects
Williams syndrome (7q deletion) E + + B, U SS, typical face, supravalvar aortic stenosis, Yes [268]
U hypercalcemia
Trisomy 9 mosaicism + + B MR, joint contractures, cardiac defects, brain No [269]
anomalies
10q duplication H + MR, ptosis, short palpebral fissures, Yes [270]
camptodactyly
Aniridia-Wilms tumor (WAGR) + Ambiguous genitalia, hypospadias, short stature AD [271]
(11p13 deletion)
Pallister-Killian syndrome + SS, MR, hypogonadism, seizures, diaphragmatic No [271]
(tetrasomy 12p) defect
Patau syndrome (trisomy 13) 1,2 H + + + Holoprosencephaly, midline anomalies, cleft No [269]
lip/palate
Miller-Dieker syndrome (17p13 1 + MR, lissencephaly, microgyria, agyria, typical No [272]
deletion) face, seizures
Edward syndrome (trisomy 18) + E, + + + IUGR, CHD, clenched hands, rocker bottom feet Yes [269]
H
18q deletion H SS, MR, microcephaly, narrow external ear Yes [273]
canals, long hands
Down syndrome (trisomy 21) 1 H + + MR, hypotonia, CHD, typical face, clinodactyly Yes [274]
Cat eye syndrome (tetrasomy 22p) 1 H + + U MR, CHD, colobomas, anal/digital anomalies Yes [275]
Velocardiofacial syndrome (22q11 1,2 E, + + + + + + Conotruncal CHD, thymic aplasia, typical face, Yes [276, 277]
deletion) H cleft palate
Turner syndrome (45,+ or 46,+,i 1 E, + + + + + SS, amenorrhea, webbed neck, cubitus valgus, No [278]
(+q)) H hypogonadism
Triploidy H + + Large molar placenta, IUGR, syndactyly of 3rd No [271]
and 4th digit, others
U ureter, B bladder, Ua urethra, 1 unilateral renal agenesis, 2 bilateral renal agenesis, E ectopia, H horseshoe, ACC agenesis of corpus callosum, SS short stature, MR mental retardation, DD developmental
delay, CHD congenital heart disease, FTT failure to thrive, IUGR intrauterine growth retardation
C. Limwongse
6 Developmental Syndromes and Malformations of the Urinary Tract 157

available information about potential urinary tract and phenotypically evolves over time, thus
anomalies for patients with a diagnosed genetic making this designation distinct from
syndrome and to suggest a differential diagnosis “malformation.”
when anomalies are identified. Readers interested Sequence refers to a cascade of abnormalities that
in additional details about a specific syndrome are result from a single initiating anomaly.
referred to standard reference textbooks and data- Sequences can be malformational, deforma-
bases about syndromes and malformations for tional, or disruptive, and they sometimes rep-
further reading (e.g., [5–7]). Online Mendelian resent more than one of these categories.
Inheritance in Man (OMIM; https://www.omim. Obstruction of urine flow at the level of the
org) is currently considered the most updated ureter during early gestation, for example, can
online information for cataloging genetic syn- cause malformation of the kidneys, intestines,
dromes and can lead readers to many informative and abdominal wall – a malformation
sites through its various links. sequence. At the same time, decreased urine
To understand the pathophysiologic basis of flow will produce oligohydramnios, fetal com-
structural abnormalities, it is important to be pression, and multiple deformities of the face,
familiar with the meaning of certain terms as limbs, and chest wall – a deformation
they are used in describing malformations and sequence.
syndromes: Syndrome refers to a consistently observed pattern
of anomalies found in an individual, whether
Malformation refers to a single structural anomaly malformation, deformation, or disruption.
that arises from an error in organogenesis. Anomalies comprising a syndrome are thought
Such an error may be due to the failure of to have a single cause, although in many cases,
cells or tissues to form, to die (programmed their causes are still unknown. Examples
cell death), or to induce others. Examples include Turner syndrome and fetal alcohol
include renal agenesis, horseshoe kidney, and syndrome.
bladder exstrophy. Association refers to a constellation of anomalies
Deformation refers to a single structural anomaly that occur together more often than expected
that arises from mechanical forces, such as by chance alone but cannot be explained by a
intrauterine constraint. Examples include single cause or sequence of events, and so do
many cases of metatarsus adductus, torticollis, not represent a syndrome or sequence. VATER
and congenital scoliosis. The underlying tissue association, which is discussed later in this
may be normal or abnormal, and sometimes a chapter, is a common example.
malformation (e.g., renal agenesis) can predis-
pose patients to a deformation (e.g., Potter’s
sequence from oligohydramnios). Prevalence of Urinary Tract Anomalies
Disruption refers to a single structural anomaly
that results from a destructive event after nor- The true incidence of urinary tract anomalies is
mal morphogenesis. Such events can be caused difficult to ascertain because many anomalies are
by lack of vascular supply, an infectious pro- asymptomatic and therefore undetected. Many
cess, or mechanical factors. Examples include reported statistics have apparent bias of ascertain-
limb amputation from amniotic bands and ment because they are derived from symptomatic
abdominal wall defect from vascular insuffi- individuals. Furthermore, inconsistent terminol-
ciency related to maternal cocaine use. ogy and clustering of data have decreased the
Dysplasia refers to a single structural anomaly power of much of the epidemiologic data. Long-
occurring in one or more organs or tissues term analysis of data collected through major
that result from abnormal organization of national birth defect registries showed increasing
structural components. The term “dysplasia” prevalence of reported statistics for many congen-
frequently implies a process that is dynamic ital birth defects, not only from an actual
158 C. Limwongse

increment but also from increased use of prenatal 9, 10, 11, 12, 13, 14, and 15). For example, it is
screening and increased tendency to report several preferable to search for syndromes with urethral
isolated and associated anomalies [1–4]. For this agenesis (22 syndromes) rather than renal dyspla-
reason, the practical use of the derived prevalence sia (more than 80 syndromes) when the two
seems not to be meaningful. However, there cur- anomalies coexist. A search based on the more
rently are quite a number of reliable estimates for common anomalies can be performed if the first
prevalence of specific isolated anomalies and of search does not reveal a match. Even after careful
those associated with a specific syndrome. A large evaluation, a substantial number of children with
number of European birth cohorts during multiple congenital anomalies remain
1996–1998 (EUROSCAN) was prenatally studied undiagnosed.
and recently reported [4]. Table 4 shows a com- When a suspected syndrome is known to be
parison of prevalence figures from various caused by a gene mutation, confirmatory molecu-
studies. lar genetic testing can be performed. DNA-based
testing is currently available on either a clinical
service or research basis. Knowledge regarding a
Approach to the Child with a Urinary pathogenic mutation specific for each proband
Tract Anomaly may potentially be useful for genetic counseling
and future reproductive options in order to avoid
The approach to the child with a urinary tract intrafamilial recurrence. Tables 1 and 2 list cur-
anomaly is similar to that for other birth defects. rently known causative genetic mutations for a
The initial step is to make a specific diagnosis if number of disorders.
possible, based on a complete history, physical A chromosome analysis is indicated in any
examination, and laboratory investigation. A thor- child who has at least two major congenital anom-
ough family history for both urinary tract anoma- alies or one isolated anomaly that is a pertinent
lies and for any other type of congenital or feature of a well-characterized chromosomal
developmental anomalies that may have occurred abnormality, such as aniridia (microdeletion
in the family should be obtained for at least two 11p). Growth or developmental delay with dys-
generations, if possible. A careful environmental morphic features or lack of familial resemblance
exposure history should be part of this history. should also prompt a chromosome analysis. Chro-
Many genetic disorders have variable expression mosome abnormalities are found in approxi-
even within the same family. A careful physical mately 10–12 % of all renal anomalies [3, 8].
examination looking specifically for major and Table 3 lists common, distinct chromosomal
minor anomalies should be performed. Some- disorders with their associated urinary tract
times, a pattern of multiple anomalies can be anomalies.
recognized immediately as a well-described syn- If a cluster of anomalies is present in a patient
drome. Patterns of anomalies that cannot be rec- but a definitive diagnosis cannot be given, that
ognized may require a literature or database patient is designated an unknown syndrome and
search or referral to an expert in syndrome recog- should be followed periodically. At each follow-
nition, such as a clinical geneticist. The search for up visit, a further literature review may reveal
a specific diagnosis is optimally accomplished by similar cases that will eventually lead to delinea-
identifying the least common and most distinctive tion of a new syndrome. Presently, state-of-the-art
anomalies, for which the list of differential diag- sequencing technology such as whole exome,
noses is limited. Many excellent textbooks, whole genome, or next-generation DNA sequenc-
atlases, and databases are available [5–7]. To aid ing (NGS) can be attempted to delineate patho-
in this effort, refer to Tables 1, 2, and 3 in addition genic culprit mutation in a previously
to the tables listing the differential diagnosis that undiscovered causative gene (see this text,
accompanies the description of each of the major ▶ Chap. 17, “Genomic Methods in the Diagnosis
urinary tract anomalies as noted (Tables 5, 6, 7, 8, and Treatment of Pediatric Kidney Disease”).
6 Developmental Syndromes and Malformations of the Urinary Tract 159

Table 4 Prevalence of urinary tract anomalies detected by various surveys


Rates per 1,000 births
ICHBDS EUROSCAN CBDMP WSBDR MACDP
Anomalies 2011a 1996–1998b 1983–1994c 1987–1989d 1983–1988e
Total renal malformation ~1.6 ~2.31 ~2.33 ~1.5
Unilateral renal agenesis 0.08
Bilateral renal agenesis/ 0.13
dysplasia
Unilateral multicystic 0.14
dysplasia
All renal agenesis /dysplasia 0.2–0.6 0.36 0.48 0.58 0.47
Horseshoe/ectopic kidney 0.04 0.04 0.16 No data
Cystic kidney 0.2–0.5 0.04 0.03 0.05 No data
Obstruction of kidney/ureter No data 0.43 1.27 1.27 0.8
Double ureter No data No data 0.004 0.05 No data
Exstrophy of bladder 0.02 0.03 0.03 0.02 0.03
Cloacal exstrophy 0.005 No data No data No data No data
Obstruction of bladder/urethra No data 0.04 0.16 0.2 0.2
VATER, CHARGE, and No data No data 0.21 No data No data
MURCS associations
Sirenomelia 0.0098 No data 0.09 No data No data
a
International Clearinghouse for Birth Defects Surveillance and Research (total 26,355,094 births) [2]
b
European Renal Anomaly Detection Program (total 709,030 births) [4]
c
California Birth Defect Monitoring Program [1]
d
Washington State Birth Defect Registry
e
Metropolitan Atlanta Congenital Defects Program

For a child with no known urinary tract anom- (see this text, ▶ Chap. 17, “Genomic Methods in
aly, findings that should prompt an evaluation of the Diagnosis and Treatment of Pediatric Kidney
the urinary tract are oligohydramnios, undefined Disease”). The type of anomaly generally guides
abdominal mass, abnormal genitalia, aniridia, treatment. Corrective or reparative treatments are
hypertension, preauricular pits or tags, branchial available for many anomalies (stenosis or atresia,
cleft cyst or sinus, imperforate anus, or symptoms bladder exstrophy, duplication, diverticula, and
indicative of renal dysfunction, urinary tract infec- tumors). Symptomatic treatment for complica-
tion, or obstructive uropathy [3]. For patients with tions is often necessary.
known syndromes, the associated urinary tract Families who have a child with a urinary tract
anomalies are listed in Tables 1 and 2. anomaly should be informed of the diagnosis. A
The best initial evaluation to screen for urinary search for a related anomaly in first-degree rela-
tract anomalies is arenal and bladder ultrasound tives is indicated only when the proband has renal
examination. This study is noninvasive and gives agenesis by ultrasound examination [9]. Other-
good anatomic information about the urinary wise, the decision to investigate family members
tract. It is also the only method routinely used should be based on a thorough family history
for the prenatal diagnosis of urinary tract anoma- and/or physical examination and whether the
lies. Specific imaging studies such as an intrave- child’s disorder is a well-described, inherited syn-
nous pyelogram, voiding cystourethrogram, drome. Genetic counseling should be provided to
radionuclide renal and urinary system scan, and the family and should include a discussion of the
specialized genetic testing may then be manifestations of the disorder, the natural history,
performed, depending on the working diagnosis complications, available treatments, cause, and
160 C. Limwongse

Table 5 Syndromes associated with unilateral renal Table 6 Syndromes associated with unilateral or bilateral
agenesis renal agenesis
Acrocallosal syndrome Acrorenal, Johnson-Munson type
Acrorenal syndrome, Dieker type Alkylating agent, maternal use
Acrorenal-mandibular syndrome Caudal duplication syndrome
Acrorenal-ocular syndrome Caudal regression syndrome
Adrenogenital syndrome Cocaine, maternal use
Aglossia-adactylia syndrome CHARGE syndrome
Alagille syndrome (arteriohepatic dysplasia) Diabetic mother, infant of
Branchio-oto-renal syndrome DiGeorge syndrome
C-trigonocephaly syndrome
Fraser (cryptophthalmos) syndrome
Campomelic dysplasia
Holzgreve syndrome
Cat eye syndrome
Pallister-Hall syndrome
Chondroectodermal dysplasia
Potter (oligohydramnios) sequence
Coffin-Siris syndrome
Sirenomelia sequence
Cornelia de Lange syndrome
Thalidomide embryopathy
Ectrodactyly-ectodermal dysplasia-clefting (EEC) syndrome
Femoral hypoplasia-unusual facies syndrome Urogenital adysplasia
Fetal alcohol syndrome Velocardiofacial syndrome
Goldenhar syndrome Winter syndrome
Ivemark syndrome
Kallmann syndrome
Klippel-Feil anomaly
Lacrimo-auriculo-dento-digital syndrome recurrence risk when these are known. Reproduc-
Larsen syndrome tive options should be discussed in a nondirective
Lenz microphthalmia syndrome fashion. For an isolated anomaly without a family
LEOPARD syndrome (multiple lentigines)
history of similar or related anomalies, an empiric
Limb-body wall complex
risk can be provided. Accurate risk figures can be
Miller-Dieker syndrome
MURCS association
determined for Mendelian disorders, and esti-
Neu-Laxova syndrome mated risks are available for associations.
Oro-facio-digital syndrome, types IV and VI All children with congenital anomalies need
Pfeiffer syndrome long-term, periodic follow-up to detect new
Poland anomaly abnormalities or complications of their birth
Renal dysplasia defects. This is especially the case for children
Roberts syndrome with undiagnosed multiple congenital anomalies,
Rokitansky-Mayer-Kuster-Hauser syndrome for whom follow-up examination may lead to a
Rubella syndrome, congenital
specific diagnosis. Additional relevant family
Rubinstein-Taybi syndrome
information should be specifically sought for any
Russell-Silver syndrome
Short rib-polydactyly syndrome, types 1–3
newly affected member. Finally, for patients with
Smith-Lemli-Opitz syndrome a urinary tract anomaly who reach reproductive
Sorsby coloboma-brachydactyly syndrome age, the recurrence risk for their offspring and
Spondylocostal dysostosis reproductive options should be discussed.
Townes-Brocks syndrome The remainder of this chapter contains descrip-
Trisomy 22 tions of the major types of urinary tract anomalies,
Turner syndrome including the etiology, pathogenesis, and associ-
Ulnar-mammary syndrome ated disorders. First, a review of the embryology
VATER (VACTERL) association
of the normal urinary tract will be useful in under-
Zellweger syndrome
standing structural urinary tract abnormalities.
6 Developmental Syndromes and Malformations of the Urinary Tract 161

Table 7 Syndromes associated with ectopic kidney Table 8 Syndromes associated with horseshoe kidney
Acromelic frontonasal dysplasia Acro-facial dysostosis syndrome
Acrorenal syndrome, Dieker type Agnathia-holoprosencephaly syndrome
Acrorenal syndrome, Siegler type Antley-Bixler syndrome
Acrorenal-ocular syndrome Bowen-Conradi syndrome
Baller-Gerold syndrome Caudal regression syndrome
Beckwith-Wiedemann syndrome Diabetic mother, infant of
Branchio-oto-renal syndrome Fanconi anemia syndrome
Caudal regression syndrome Fetal alcohol syndrome
CHARGE syndrome Focal dermal hypoplasia
Crossed ectopia-pelvic lipomatosis syndrome Juberg-Hayward syndrome
DiGeorge syndrome Kabuki syndrome
Drash (Denys-Drash) syndrome Pallister-Hall syndrome
Fanconi anemia syndrome Pyloric stenosis
Fetal alcohol syndrome Roberts syndrome
Floating-Harbor syndrome Thalidomide embryopathy
Frontonasal dysplasia Trisomy 13, 18, 21, and 22
Goldenhar syndrome Turner syndrome
Kaufman-McKusick syndrome VATER (VACTERL) association
Klippel-Feil anomaly Weyers syndrome
Limb-body wall complex Wilms tumor
MURCS association
Pallister-Hall syndrome
Penoscrotal transposition mesodermal ridge, the urorectal septum that
Renal adysplasia divides the cloaca into the anterior portion, the
Rokitansky-Mayer-Kuster-Hauser syndrome primitive urogenital sinus, and the posterior por-
Rubinstein-Taybi syndrome tion, the cloacal sinus or anorectal canal. The
Schinzel-Giedion syndrome mesonephric ducts open into the urogenital sinus
Sirenomelia sequence and later become the ureters. The urorectal septum
Turner syndrome develops caudally and fuses with the cloacal
VATER (VACTERL) association
membrane, dividing it into the urogenital mem-
Velocardiofacial syndrome
brane (anterior) and the anorectal membrane (pos-
Williams syndrome
terior) by the end of the seventh week. The
primitive perineal body forms at the site of fusion.
The primitive urogenital sinus develops primar-
Overview of Normal Embryogenesis ily into the urinary bladder. The superior portion,
of the Urinary System originally continuous with the allantois, later
becomes a solid fibrous cord, the urachus, or median
Renal organogenesis is reviewed in ▶ Chap. 1, umbilical ligament, which connects the bladder to
“Embryonic Development of the Kidney”. the umbilicus. The inferior portion of the urogenital
Embryogenesis of the lower urinary tract includes sinus in the male divides into a pelvic portion,
development of the mesonephric duct and urogen- containing the prostatic and membranous urethra,
ital sinus. The mesonephric duct from which the and the long phallic portion, containing the penile
ureteric bud arose inserts into the lower allantois, urethra. The inferior portion in the female forms a
just above the terminal part of the hindgut, the small portion of the urethra and the vestibule. At the
cloaca. During the fourth to seventh weeks, meso- same time, the distal portion of the mesonephric
derm proliferates and forms the transverse ducts is incorporated into the endodermal
162 C. Limwongse

Table 9 Syndromes associated with renal dysplasia/cystic Table 9 (continued)


kidney Thalidomide embryopathy
Acrorenal-mandibular syndrome Trisomy 8, 9, 13, 18, 21, and 22
Alagille syndrome (arteriohepatic dysplasia) Tuberous sclerosis
Baller-Gerold syndrome VATER (VACTERL) association
Bardet-Biedl syndrome Von Hippel-Lindau disease
Beckwith-Wiedemann syndrome Zellweger and pseudo-Zellweger syndromes
Branchio-oto-renal syndrome
Campomelic dysplasia
Carbohydrate deficient glycoprotein syndrome Table 10 Syndromes associated with hydronephrosis or
CHARGE syndrome hydroureter
Chondrodysplasia punctata, non-rhizomelic Acrocephalo-polysyndactylous dysplasia
Cloacal exstrophy Acrorenal syndrome, Dieker and Johnson-Munson types
Cornelia de Lange syndrome Bardet-Biedl syndrome
Diabetic mother, infant of Branchio-oto-renal syndrome
Ectrodactyly-ectodermal dysplasia-clefting (EEC) Campomelic dysplasia
syndrome
Caudal duplication and regression syndromes
Fanconi anemia syndrome
CHARGE syndrome
Fetal alcohol syndrome
Cloacal exstrophy
Fraser (cryptophthalmos) syndrome
Coffin-Siris syndrome
Fryns syndrome
Crossed ectopia-pelvic lipomatosis syndrome
Glutaric aciduria, type II
Cornelia de Lange syndrome
Goldenhar syndrome
Diabetic mother, infant of
Hajdu-Cheney syndrome
Ectrodactyly-ectodermal dysplasia-clefting (EEC)
Ivemark syndrome syndrome
Jeune syndrome Fanconi anemia syndrome
Joubert syndrome Fetal alcohol syndrome
Kaufman-McKusick syndrome Goldenhar syndrome
Lenz microphthalmia syndrome Hydrolethalus syndrome
Leprechaunism (Donohue) syndrome Kabuki syndrome
Limb-body wall complex Kaufman-McKusick syndrome
Marden-Walker syndrome Megacystis-microcolon syndrome
Marfan syndrome Noonan syndrome
Marshall-Smith syndrome Ochoa syndrome
Meckel-Gruber syndrome Omphalocele-Sxstrophy of bladder-Imperforate anus-
MURCS association Spinal dysraphism (OEIS) complex
Noonan syndrome Pallister-Hall syndrome
Omphalocele-Exstrophy of bladder-Imperforate anus- Polydactyly-obstructive uropathy syndrome
Spinal dysraphism (OEIS) complex Pyloric stenosis
Oral-facial-digital syndrome, types I and VI Roberts syndrome
Pallister-Hall syndrome Sirenomelia sequence
Pallister-Killian syndrome Schinzel-Giedion syndrome
Potter (oligohydramnios) sequence VATER (VACTERL) association
Prune belly syndrome
Renal adysplasia
Roberts syndrome
vesicoureteral primordium, forming the trigone of
Rokitansky-Mayer-Kuster-Hauser syndrome
Rubella syndrome, congenital
the bladder. A part of the distal end of both meso-
Short rib-polydactyly syndrome nephric ducts just proximal to the trigone develops
Smith-Lemli-Opitz syndrome into the seminal vesicles and ductus deferens in the
(continued) male. Finally, at the end of the 12th week, the
6 Developmental Syndromes and Malformations of the Urinary Tract 163

Table 11 Syndromes associated with duplication of ure- Table 13 Syndromes associated with urethral agenesis
ters or collecting systems
Aase syndrome
Achondrogenesis Acrorenal syndrome, Dieker and Johnson-Munson types
Acromelic frontonasal dysplasia Adrenogenital syndrome
Adrenogenital syndrome Caudal regression syndrome
Antley-Bixler syndrome Cocaine, maternal use
Bardet-Biedl syndrome Diabetic mother, infant of
Bowen-Conradi syndrome Hydrolethalus syndrome
Branchio-oto-ureteral syndrome Kaufman-McKusick syndrome
Braun-Bayer syndrome Limb-body wall complex
Caudal duplication syndrome Meckel-Gruber syndrome
Diabetic mother, infant of Occipital horn syndrome
Drash (Denys-Drash) syndrome Ochoa syndrome
Ectrodactyly-ectodermal dysplasia-clefting (EEC) Omphalocele-Exstrophy of bladder-Imperforate anus-
syndrome Spinal dysraphism (OEIS) complex
Fanconi anemia syndrome Potter (oligohydramnios) Sequence
Fetal alcohol syndrome Prune belly syndrome
Frontometaphyseal dysplasia Renal adysplasia
G (Opitz-Frias) syndrome Russell-Silver syndrome
Goldenhar syndrome Short rib-polydactyly syndrome, types 1–3
Kabuki syndrome Sirenomelia sequence
Kaufman-McKusick syndrome Sotos syndrome
Mammo-renal syndrome Townes-Brocks syndrome
Noonan syndrome Trisomy 21
Ochoa syndrome
Perlman syndrome
Poland anomaly Table 14 Syndromes associated with urethral
Prune belly syndrome duplication
Robinow syndrome Amniotic band disruption sequence
Rubinstein-Taybi syndrome Limb-body wall complex
Trisomy 8, 9, 13, 18, and 21 Omphalocele-Exstrophy of bladder-Imperforate anus-
Turner syndrome Spinal dysraphism (OEIS) complex
Weyers syndrome Prune belly syndrome

epithelium of the superior portion of the prostatic


urethra proliferates to form buds that penetrate the
surrounding mesenchyme. In the male, these buds
form the prostate gland; in the female, they form the
Table 12 Syndromes associated with bladder exstrophy urethral and paraurethral glands.
Axial mesodermal dysplasia
Caudal duplication syndrome
Caudal regression syndrome Anomalies Involving the Urinary Tract
Cloacal exstrophy
Frontonasal dysplasia Kidney Defects
Omphalocele-Exstrophy of bladder-Imperforate anus-
Spinal dysraphism (OEIS) complex
Renal Agenesis
Trisomy 18
By definition, renal agenesis refers to complete
Sirenomelia sequence
Syndactyly, type V
absence of one of both kidneys without identifi-
able rudimentary tissue. Renal agenesis is usually
164 C. Limwongse

Table 15 Syndromes associated with posterior urethral agenesis is made by abdominal ultrasound. Care
valves must be taken to exclude the possibility of ectopic
Acrorenal syndrome, Johnson-Munson type kidney. Intravenous pyelography, computerized
Caudal regression syndrome tomography, and radionuclide studies can be help-
Diabetic mother, infant of ful in equivocal cases.
Kaufman-McKusick syndrome The recurrence risk for renal agenesis can be
Limb-body wall complex provided if the pattern of inheritance is known or
Neurofibromatosis, type I if the proband has a recognizable syndrome. For
Ochoa syndrome
nonsyndromic renal agenesis, an empiric risk of
Omphalocele-Exstrophy of bladder-Imperforate anus-
Spinal dysraphism (OEIS) complex
3 % can be used for families in which renal anom-
Polydactyly-obstructive uropathy syndrome alies in first-degree relatives (siblings, parents)
Potter (oligohydramnios) sequence have been excluded [3]. First-degree relatives of
Prune belly syndrome patients with nonsyndromic renal agenesis have
Renal adysplasia an increased prevalence of related urogenital
Rubinstein-Taybi syndrome anomalies. It has been reported that 9 % of first-
Sirenomelia sequence degree relatives of infants with agenesis or dys-
Townes-Brocks syndrome genesis of both kidneys had a related urogenital
VATER (VACTERL) association anomaly, and 4.4 % had an asymptomatic renal
malformation [13]. In a retrospective review,
empiric risks were 7 % in offspring, 2.5 % in
associated with agenesis of the ipsilateral ureter. siblings, and 4.5 % in parents [14]. Moreover,
The pathogenesis of renal agenesis is failure of offspring of an individual with unilateral agenesis
formation of the metanephros. Causal heterogene- appears to be at an increased risk for bilateral renal
ity has been shown, by both animal studies and agenesis. Therefore renal ultrasound is
human observations [10–12], including failure of recommended for the first-degree relatives of the
ureteric bud formation, failure of the bud to reach proband unless renal agenesis in the proband is
the metanephric blastema, or failure of the bud clearly sporadic or a specific cause without an
and the metanephric blastema to induce interac- increased recurrence risk is identified.
tion. In addition, interruption in vascular supply Tables 5 and 6 list the syndromes commonly
and regression of a multicystic kidney can lead to associated with unilateral and bilateral renal agen-
renal agenesis in the fetal period [11]. esis, respectively. See also Tables 1, 2, and 3 for
Unilateral renal agenesis is usually asymptom- more information about these disorders and other
atic and found incidentally, whereas bilateral renal less known conditions with renal agenesis.
agenesis results in severe oligohydramnios and
fetal or perinatal loss. Renal agenesis can occur Ectopic Kidney
on either side equally. Birth prevalence in the The ectopic kidney derives from an abnormality
United States for renal agenesis/hypoplasia ranges of normal ascent. Most are pelvic kidneys that fail
between 0.30 and 9.61 per 10,000 live births to ascend out of the pelvic cavity. Rare case
[3]. Several studies have demonstrated that unilat- reports of thoracic kidney exist [15]. Ectopic kid-
eral renal agenesis is associated with an increased neys can be unilateral or bilateral. In bilateral
frequency of anomalies in the remaining kidney pelvic kidneys, the kidneys often fuse into a mid-
[9, 13]. Moreover, renal agenesis is often detected line mass of renal tissue, with two pelves and a
in conjunction with anomalies of other organ sys- variable number of ureters, which is referred to as
tems. These anomalies can occur both in contigu- a pancake or discoid kidney. Fused pelvic kidney
ous structures (e.g., vertebrae, genital organs, may in fact be due to fusion of ureteric buds or
intestines, and anus) and also in noncontiguous metanephric blastema. Crossed renal ectopia
structures (e.g., limbs, heart, trachea, ear, and refers to an ectopic kidney whose ureter crosses
central nervous system). The diagnosis of renal the midline. It often fuses with the normal kidney.
6 Developmental Syndromes and Malformations of the Urinary Tract 165

The embryogenesis of crossed renal ectopia is not reported. Horseshoe kidneys also carry an
well understood, but presumably involves abnor- increased risk for renal pelvis carcinoma and
mal migration of the ectopic kidney to the contra- renal squamous cell carcinoma [19].
lateral side. An ectopic kidney is usually Table 8 lists syndromes associated with horse-
hypoplastic and rotated and has numerous anom- shoe kidney. See Tables 1 and 2 for more details of
alous small blood vessels and associated ureteric these disorders.
anomalies. Ectopic kidneys may be asymptomatic
and incidentally found, but complications from Dysplasia and Polycystic Kidney
ureteral obstruction, infection, and calculi can Renal dysplasia is the most common congenital
occur. Ectopic kidney without hypoplasia or urinary tract anomaly and the most common cause
hydronephrosis does not appear to be associated of an abdominal mass in children [3]. (see also in
with an increased frequency of associated anom- this text, ▶ Chap. 5, “Renal Dysplasia/Hypopla-
alies. In such cases, further urologic investigation sia”). Unilateral dysplasia is reported to occur in
is not indicated [16]. Table 7 provides a list of 1:1,000, whereas the prevalence of bilateral dis-
syndromes that include ectopic kidney. These are ease is estimated to be 1:5,000 [20]. It may be
described in Tables 1, 2, and 3. unilateral or bilateral and diffuse, segmental, or
focal. Symptoms are variable and range from a
Horseshoe Kidney symptomatic unilateral or focal dysplasia to the
Horseshoe kidney refers to a condition in which progressive renal dysfunction of diffuse or bilat-
both kidneys are fused at the lower poles with a eral dysplasia. Dysplasia refers to abnormal dif-
renal parenchymal or, less commonly, fibrous ferentiation or organization of cells in the tissue.
isthmus. The embryogenesis of horseshoe kidney Renal dysplasia is characterized histologically by
with parenchymal isthmus is thought to be migra- the presence of primitive ducts and nests of meta-
tion of nephrogenic cells across the primitive plastic cartilage [20, 21]. Although cysts are not
streak before the fifth gestational week. Horse- always present in a dysplastic kidney, the dysplas-
shoe kidney with fibrous isthmus is believed to tic process often results in the formation of cysts
originate from mechanical fusion of the two that are variable in size and number. Several
developing kidneys at or after the fifth week hypotheses have been proposed for the embryo-
before renal ascent [17]. The concept of a narrow genesis of the dysplastic kidney. The most likely
vascular fork leading to approximation and fusion pathogenesis is an error of the inductive interac-
of the two kidneys is no longer considered valid. tion between the ureteric bud and the metanephric
Most horseshoe kidneys are located in the pelvis blastema (see ▶ Chap. 1, “Embryonic Develop-
or at the lower lumbar vertebral level because ment of the Kidney”, in this text). The molecular
ascent is further prevented when the fused kidney pathogenesis of cystic kidney, especially polycys-
reaches the junction of the aorta and inferior mes- tic kidney, has been one of the most extensively
enteric artery. studied aspects of nephrology, and recent studies
Complications of horseshoe kidneys include have discovered distinct genes and pathways crit-
obstructive uropathy related to ureteropelvic junc- ical for renal cyst formation as reviewed in
tion obstruction, calculi, and urinary tract infec- ▶ Chap. 36, “Childhood Polycystic Kidney Dis-
tion. Similar to other urinary tract anomalies, ease”. Dysplastic kidneys are usually identified
horseshoe kidneys are often associated with as enlarged echogenic kidneys on prenatal ultra-
other genitourinary anomalies. In addition, there sonography. If there is associated functional renal
is an increased risk of renal tumors developing in impairment, alteration in amniotic fluid volume
horseshoe kidneys compared with normal kidneys may be detected and signifies a poor prognosis
[18]. Renal cell carcinoma is the most common, [22]. Unilateral dysplasia carries an overall better
but Wilms tumor, adenocarcinoma, transitional postnatal prognosis than that of bilateral disease.
cell carcinoma, malignant teratoma, oncocytoma, However, up to 30–50 % of those with unilateral
angiomyolipoma, and carcinoid have all been dysplasia have associated contralateral urinary
166 C. Limwongse

anomalies [22]. Multicystic renal dysplasia is the has ureters that enter into the bladder separately.
most common cause of renal dysplasia and is Duplication that occurs later results in double
usually unilateral. Polycystic kidney diseases, ureters that may have separate openings into the
both autosomal dominant (ADPKD) and autoso- bladder or may join each other before the opening.
mal recessive (ARPKD) forms, are in general far On rare occasion, one of the ureters may have an
more common than other syndromic causes of ectopic opening into the vagina, vestibule, or ure-
renal dysplasia. See this text, ▶ Chap. 36, “Child- thra. In most double ureters, the two ureters cross
hood Polycystic Kidney Disease”. Table 9 sum- each other, and that from the higher pelvis enters
marizes well-known syndromes with renal the bladder more caudally. Duplication anomalies
dysplasia/cystic kidney. See also Tables 1, 2, are common but usually asymptomatic; therefore
and 3. they often remain undetected. One autopsy study
reported the prevalence of duplication anomalies
Obstruction and Hydronephrosis to be as high as 1 in 25, with females about four
Urinary obstruction is a complication of a pri- times more likely to be affected than males
mary anomaly, which can be stenosis or atresia of [24]. Unilateral duplication is 5–6 times more
the ureteropelvic junction, ureter, or urethra; a common than bilateral duplication [3]. Double
poorly functional bladder causing reflux; a mal- ureters are commonly associated with
formed dilated ureteral end (ureterocele); or vesicoureteral reflux due to their ectopic opening
extrinsic compression by other structures, such into the urinary bladder and/or the ureterocele
as anomalous blood vessels or tumors. [23]. In addition, ureteric obstruction can occur
Hydronephrosis and pyelectasis (dilated renal at the level of vesicoureteric junction or that of
pelvis) are the most common urinary tract abnor- ureteropelvic junction.
malities detected by prenatal ultrasound exami- Table 11 summarizes syndromes associated
nation. Early diagnosis of collecting system with duplication, and Tables 1, 2, and 3 provide
dilatation can be achieved by ultrasound exami- clinical information about these disorders.
nation in the second trimester [23], although
spontaneous remission by birth is not uncom- Hydroureter
mon. Persistent dilatation generally indicates an Hydroureter, or megaloureter, is caused by distal
underlying anomaly. Isolated obstructive obstruction and is usually found with
uropathies diagnosed prenatally may not require hydronephrosis, except in ureteropelvic junction
antenatal or immediate postnatal surgical inter- obstruction. Hydroureter has the same etiology as
vention. Postnatally diagnosed obstructive hydronephrosis (see section “Obstruction and
uropathies are almost always symptomatic and Hydronephrosis”).
require thorough investigation to delineate the
anatomy of the urinary tract and to exclude asso-
ciated anomalies. Bladder Defects
Table 10 provides a list of syndromes com-
monly associated with obstruction and Anomalies of the bladder are rare. These include
hydronephrosis. See also Tables 1, 2, and 3. agenesis, hypoplasia, diverticula, and dilatation
or megacystis with or without distal obstruction.
Agenesis of the bladder is usually associated with
Ureter Defects severe developmental anomalies of the urinary
tract, such as those seen in sirenomelia and cau-
Duplication dal regression syndrome. Hypoplastic bladder
Double ureters or collecting systems are caused can be found in conditions associated with bilat-
by duplication of the ureteric bud. Early duplica- eral renal agenesis because no urine is produced.
tion results in duplicated kidney, which is usually Bladder diverticula have heterogeneous causes.
smaller and fused with the ipsilateral kidney and They result from an intrinsic defect in the bladder
6 Developmental Syndromes and Malformations of the Urinary Tract 167

wall, as seen in cutis laxa (or Ehlers-Danlos), Duplication


Ochoa, occipital horn, and Williams syndromes. Duplication refers to complete or partial duplica-
They can also be caused by increased tion of the urethra, which is a rare anomaly found
intravesicular pressure from distal obstruction only in a few syndromes. Those syndromes asso-
or by a persistent urachus. See Tables 1, 2, and 3 ciated with urethral duplication are listed in
for information about specific syndromes associ- Table 14, and their findings are provided in
ated with bladder diverticula. Tables 1, 2, and 3.

Bladder Exstrophy Posterior Urethral Valves


Bladder exstrophy refers to a urinary bladder that Posterior urethral valves refer to abnormal
is open anteriorly because of the lack of an mucosal folds that function as a valve to obstruct
abdominal wall closure. It is usually associated urine flow. This is the most common childhood
with anomalies of the contiguous structures cause of obstructive uropathy leading to renal
including epispadias and separation of the pubic failure. Posterior urethral valves can be
rami. This anomaly is thought to result from an suspected prenatally when a dilated bladder is
overdeveloped cloacal membrane that interferes seen in association with obstructive uropathy.
with inferolateral abdominal mesenchymal clo- A “keyhole” sign has been demonstrated in pre-
sure. Therefore, when the cloacal membrane rup- natal ultrasound of fetuses with subsequently
tures, the inferior abdominal wall has not confirmed posterior urethral valves [28].
completely closed and the bladder cavity is A voiding cystourethrogram or endoscopy is
exposed. It has been suggested that bladder usually required for a definitive diagnosis. The
exstrophy belongs to the spectrum of embryogenesis of posterior urethral valves is
omphalocele-cloacal exstrophy-imperforate unknown. Proposed hypotheses include an
anus-spinal dysraphism (OEIS) complex overdeveloped posterior urethral fold, a remnant
[25–27]. The extent of anomalies is determined of the mesonephric duct, and an anomalous
by the timing of the cloacal membrane rupture. opening of the ejaculatory duct. Table 15 lists
Rupture that occurs after the separation of cloaca syndromes in which posterior urethral valves can
by the urorectal septum results in bladder be seen, and the other findings in these disorders
exstrophy, whereas one that occurs before the are provided in Tables 1, 2, and 3.
separation results in the more severe cloacal
exstrophy and OEIS complex. Bladder exstrophy
is six times more common in males than females. Associations and Sequences Involving
Table 12 lists syndromes associated with blad- the Urinary Tract
der exstrophy, and Tables 1, 2, and 3 provide
information about these disorders. A number of associations and sequences involve
anomalies of the urinary tract that may be impor-
tant to both diagnosis and management. For this
Urethral Defects reason, such conditions are described in more
detail in this section, in addition to the information
Agenesis and Atresia presented in Tables 1 and 2.
Urethral agenesis is rare, and its predominant
occurrence in males probably reflects the greater
complexity of embryogenesis of the male urethra. VATER Association
Urethral agenesis is often associated with bladder
obstruction sequence. Table 13 lists syndromes VATER association is an acronym used to desig-
associated with urethral agenesis, and clinical nate a nonrandom occurrence of vertebral defects,
information about these disorders is summarized imperforate anus, tracheoesophageal fistula, and
in Tables 1, 2, and 3. radial and renal anomalies [29, 30]. An acronym
168 C. Limwongse

VACTERL has been proposed to broaden the unilateral facial palsy is a common finding. Renal
spectrum of VATER to include cardiac defects anomalies found in CHARGE syndrome may
and limb anomalies. The term VATER is not a include ectopy, dysplasia, renal agenesis, and/or
diagnosis per se, but the designation provides ureteric anomalies. The presence of two or more
clues for potentially associated anomalies and anomalies associated with CHARGE syndrome
for recurrence risk counseling when no specific should prompt a search for the others. To prevent
syndromic diagnosis can be made. Patients with overuse of the term, it has been suggested that at
VATER association need a careful evaluation for least three anomalies should be required for the term
multiorgan anomalies. A specific diagnosis CHARGE to be applied and one of the anomalies
should be sought. Causes of VATER association should be either coloboma or choanal atresia
include: chromosomal disorders, such as trisomy [34]. To date, consistent features in CHARGE syn-
18; genetic syndromes, such as Goldenhar and drome have been ocular coloboma, choanal atresia,
Holt-Oram syndromes; and teratogenic expo- and semicircular canal hypoplasia [37]. Conditions
sures, such as infants of diabetic mothers and with anomalies in the spectrum of CHARGE
fetal alcohol syndrome. A family with a mito- include trisomy 13, trisomy 18, and Wolf-
chondrial DNA mutation was identified in which Hirschhorn (deletion 4p), cat-eye, Treacher-Collins,
the daughter was born with VACTERL associa- velocardiofacial, Apert, Crouzon, and Saethre-
tion, and her mother and sister had classic mito- Chotzen syndromes. Therefore, a careful physical
chondrial cytopathy [31]. Thus all patients examination for malformations and dysmorphic
suspected to have VATER association should features should be conducted. Recently, CHD7
have undergone a chromosomal analysis, a care- mutations have been identified to be the cause of
ful family and prenatal exposure history, and a this syndrome in about 60 % of typical patients, thus
thorough examination for dysmorphic features. making a molecular confirmation possible. In those
The spectrum of anomalies seen in VATER is without a CHD7 mutation, chromosome analysis
broad. Associated renal anomalies usually including specific fluorescence in situ hybridization
include agenesis, ectopy, or obstruction [29, 30]. (FISH) probes for velocardiofacial syndrome (dele-
Because there is causal heterogeneity for tion 22q) and 4p deletion should be performed.
VATER association, the inheritance pattern and Because most cases of CHARGE association are
recurrence risk vary with the cause. VATER asso- sporadic, the empirical recurrence risk in siblings is
ciation is usually sporadic with an empirical recur- low [33, 36].
rence risk of 1–3 % when a specific cause cannot
be identified [32]. Autosomal recessive and
X-linked inheritance have been reported for sub- MURCS Association
sets of patients, such as for VATER with hydro-
cephalus, and recurrence risk in these families can MURCS association refers to a rare occurrence of
be as high as 25 % [32]. Mullerian duct aplasia, renal aplasia, and
cervicothoracic somite dysplasia [38]. Anomalies
include absence of the proximal two-thirds of the
CHARGE Syndrome (CHARGE vagina, uterine hypoplasia or aplasia, unilateral
Association) renal agenesis, ectopic kidney, renal dysplasia,
C5-T1 vertebral anomalies (hypoplasia of verte-
CHARGE syndrome – previously designated as an brae, fusion, hemivertebrae, and butterfly verte-
association but now recognized to have a major brae), and short stature. Additional anomalies are
causative gene – is an acronym used to designate common, including rib defects, facial asymmetry,
an association of coloboma of iris, choroid or retina, limb anomalies, hearing loss, and brain anomalies,
heart defects, atresia choanae, retarded growth and such as encephalocele and cerebellar cyst [39].
development, genital anomalies or hypogonadism, The pathogenesis of MURCS association is
and ear anomalies or deafness [33–36]. In addition, unknown, but is thought to be related to defects in
6 Developmental Syndromes and Malformations of the Urinary Tract 169

the paraxial mesoderm, which gives rise to the autosomal recessive disorder causing bilateral
cervicothoracic somites and the adjoining interme- renal agenesis or dysplasia.
diate mesoderm. Most patients are diagnosed
because of primary amenorrhea or infertility asso-
ciated with normal secondary sexual characteris- Urethral Obstruction Sequence
tics, followed by recognition of reproductive organ
atresia. MURCS association is usually sporadic. A The initial defect in this sequence is obstruction of
report of vertebral and renal anomalies associated the urethra leading to dilation of the proximal
with azoospermia has been proposed to represent a urinary tract, bladder distension, and hydroureter
male version of MURCS association [40]. [3, 6, 41]. Obstruction of urine flow interferes with
normal nephrogenesis, resulting in renal dyspla-
sia. Other potential anomalies related to bladder
Oligohydramnios Sequence distension include cryptorchidism, malrotation of
the colon, persistent urachus, and limb deficiency
Oligohydramnios of any cause leads to a recurrent caused by iliac vessel compression. In addition,
pattern of abnormalities known as the oligohydramnios results from lack of urine and
oligohydramnios sequence [3, 6]. leads to the oligohydramnios sequence.
Oligohydramnios may be caused by decreased pro- Prune belly syndrome [41, 42] is a rare entity
duction of fatal urine from bilateral renal agenesis referring to a constellation of anomalies that
or dysplasia or by urinary obstruction, or it can includes megacystis, abdominal wall muscle defi-
result from amniotic fluid leakage. When the ciency, hydroureter, renal dysplasia, and charac-
oligohydramnios is prolonged and severe, the con- teristic wrinkled abdominal skin. This condition,
dition is lethal because of pulmonary hypoplasia. previously thought to be a form of urethral
Moderate oligohydramnios from amniotic fluid obstruction sequence, is in fact a non-obstructive
leakage may result in a live-born child with multi- cause of bladder distension that results from a
ple congenital anomalies. These anomalies are both malformation, thus now being properly desig-
malformations and deformations. Intrauterine con- nated a syndrome [28].
straint leads to mechanical compression that leads The most common cause of urethral obstruction
to the characteristic flat facial profile (Potter’s is posterior urethral valves, but urethral agenesis/
facies), limb deformities (e.g., talipes equinovarus), atresia or bladder neck obstruction can also be the
and intrauterine growth retardation (IUGR). cause. This anomaly occurs mostly in males, with a
Decreased fetal movement as a result of intrauterine male to female ratio of 20:1. Survival is rare in
constraint causes multiple joint contractures fetuses with complete obstruction, and severe uri-
(arthrogryposis). Breech presentation is common. nary tract dysfunction is always present in those
Pulmonary hypoplasia can be the consequence of that are live-born. Prenatal diagnosis by ultrasound
compression of the chest cavity coupled with examination can detect the abnormally dilated
decreased inspiration of amniotic fluid. Live- bladder at the beginning of the second trimester
borns have respiratory distress caused by pulmo- [43], and intrauterine urinary decompression pro-
nary hypoplasia, and the lungs may have insuffi- cedures, such as vesicoamniotic shunts, are options
cient volume to support life. for treatment in order to decrease the occurrence of
Because the initial defect has many causes, pulmonary hypoplasia, although their benefits have
recurrence risk is based on the underlying defect. not been unequivocal [44, 45].
When oligohydramnios is due to nonsyndromic
bilateral renal agenesis or dysgenesis, related
renal malformations occur at an increased fre- Sirenomelia Sequence
quency in first-degree relatives [13], and recur-
rence risk can be as high as 4–9 %. The Sirenomelia is a malformation characterized by
recurrence risk can be as high as 25 % for an the presence of a single lower extremity with
170 C. Limwongse

posterior alignment of the knees and feet, sacral detection of congenital renal malformations by fetal
agenesis and other lower vertebral defects, imper- ultrasonographic examination: an analysis of 709,030
births in 12 European countries. Eur J Med Genet.
forate anus and rectal agenesis, and absence of 2005;48(2):131–44.
external and internal genitalia [46]. The current 5. Hennekam RCM, Allanson JE, Krantz I. Syndromes
view of the embryogenesis is that sirenomelia of the head and neck. 5th ed. New York: Oxford
results from a vascular steal phenomenon University Press; 2010.
6. Jones KL. Smith’s recognizable patterns of human
[47]. This is supported by the presence of abnor- malformation. 7th ed. Philadelphia: WB Saunders;
mal vasculature in the caudal part of affected 2013.
embryos. A single large vessel originating from 7. Winter RM, Baraitser M. London dysmorphology
the aorta, a derivative of the vitelline artery com- database. London: Oxford University Press; 1995.
8. Nicolaides KH, Cheng HH, Abbas A, et al. Fetal renal
plex, connects the iliac arteries to the placenta defects: associated malformations and chromosomal
rather than the two normal umbilical arteries. defects. Fetal Diagn Ther. 1992;7:1–11.
The area caudal to the origin of this vessel has 9. Robson WLM, Leung AKC, Rogers RC. Unilateral
minimal blood supply because of the lack of aortic renal agenesis. Adv Pediatr. 1995;42:575–92.
10. Ekblom P. Genetics of kidney development. Curr
branches. Therefore, a “vascular steal phenome- Opin Nephrol Hypertens. 1996;5:282–7.
non” is generated, leading to a vascular disruption 11. Mesrobian HG, Rushton HG, Bulas D. Unilateral
sequence. Alternatively, since sirenomelia shares renal agenesis may result from in utero regression of
a number of anomalies with caudal regression multicystic renal dysplasia. J Urol. 1993;150(2 Pt 2):
793–4.
syndrome, it is thought to potentially be causally 12. Atiyeh B, Husmann D, Baum M. Contralateral renal
similar and represent different patterns in the same abnormalities in patients with renal agenesis and
spectrum. noncystic renal dysplasia. Pediatrics. 1993;91:812–15.
Sirenomelia is a rare condition and has a broad 13. Roodhooft AM, Birriholz JC, Holmes LB. Familial
nature of congenital absence and severe dysgenesis of
spectrum of anomalies. Virtually any urinary tract both kidneys. N Engl J Med. 1984;310:1341–5.
anomaly can occur in sirenomelia sequence. 14. McPherson E. Renal anomalies in families of individ-
Renal agenesis occurs in two-thirds of cases, and uals with congenital solitary kidney. Genet Med.
a variable degree of renal dysplasia is present in 2007;9(5):298–302.
15. N’Guessen G, Stephens FD, Pick J. Congenital superior
one-third of cases [3]. Absence of the ureter and ectopic (thoracic) kidney. Urology. 1984;24:219–28.
bladder are common. All cases of sirenomelia are 16. Calisti A, Perrotta ML, Oriolo L, Ingianna D, Miele
sporadic and almost uniformly fatal because of V. The risk of associated urological abnormalities in
pulmonary hypoplasia. Sirenomelia has been children with pre and postnatal occasional diagnosis
of solitary, small or ectopic kidney: is a complete
noted with an increased frequency among mono- urological screening always necessary? World J
zygotic twins in which only one of the twins in Urol. 2008;26:281–4.
usually affected. 17. Domenech-Mateu JM, Gonzalez-Compta
X. Horseshoe kidney: a new theory on its embryo-
genesis based on the study of a 16-mm human
embryo. Anat Rec. 1988;244:408–17.
References 18. Stimac G, Dimanovski J, Ruzic B, Spajic B, Kraus
O. Tumors in kidney fusion anomalies – report of five
1. Schulman J, Edmonds LD, McClearn AB, cases and review of the literature. Scand J Urol
et al. Surveillance for and comparison of birth defect Nephrol. 2004;38(6):485–9.
prevalences in two geographic areas-United States, 19. Mizusawa H, Komiyama I, Ueno Y, Maejima T, Kato
1983–88. Morb Mortal Wkly Rep. 1993;42:1–7. H. Squamous cell carcinoma in the renal pelvis of a
2. International Clearinghouse for Birth Defects Surveil- horseshoe kidney. Int J Urol. 2004;11(9):782–4.
lance and Research. Annual Report 2011 with data for 20. Winyard P, Chitty LS. Dysplastic kidneys. Semin
2009; Rome, ICBDSR, 2011. Fetal Neonatal Med. 2008;13(3):142–51.
3. Van Allen MI. Urinary tract. In: Stevenson RE, Hall 21. Fisher C, Smith JF. Renal dysplasia in nephrectomy
JG, Goodman RM, editors. Human malformations specimens from adolescents and adults. J Clin Pathol.
and related anomalies, vol. 11. New York: Oxford 1975;28:879–90.
University Press; 1993. p. 501–50. 22. Winyard P, Chitty LS. Dysplastic and polycystic kid-
4. Wiesel A, Queisser-Luft A, Clementi M, Bianca S, neys: diagnosis, associations and management. Prenat
Stoll C, EUROSCAN Study Group. Prenatal Diagn. 2001;21:924–35.
6 Developmental Syndromes and Malformations of the Urinary Tract 171

23. Whitten SM, Wilcox DT. Duplex systems. Prenat 43. Cazorla E, Ruiz F, Abad A, et al. Prune belly syn-
Diagn. 2001;21:952–7. drome: early antenatal diagnosis. Eur J Obstet
24. Campbell MF, Harrison JH, editors. Anomalies of the Gynecol Reprod Biol. 1997;72:31–3.
ureter. In: Urology, vol 2. 3rd ed. Philadelphia: WB 44. Freedman AL, Bukowski TP, Smith CA. Fetal therapy
Saunders; 1970. p. 1488–800. for obstructive uropathy: diagnosis specific outcome.
25. Pang D. Sacral agenesis and caudal spinal cord J Urol. 1996;156:720–3.
malformations. Neurosurgery. 1993;32:755–78. 45. Agarwal SK, Fisk NM. In utero therapy for lower
26. Carey JC, Greenbaum B, Hall BD. The OEIS com- urinary tract obstruction. Prenat Diagn. 2001;21:
plex (omphalocoele, imperforate anus, exstrophy and 970–6.
spinal defects). Birth Defects Orig Artic Ser. 1978;14 46. Langer B, Stoll C, Nicolau R, et al. Sirenomelia and
(6B):253–63. situs inversus: case report and review of the literature.
27. Keppler-Noreuil K, Gorton S, Foo F, Yankowitz J, Fetal Diagn Ther. 1996;11:79–84.
Keegan C. Prenatal ascertainment of OEIS complex/ 47. Stevenson RE, Jones KL, Phelan MC, et al. Vascular
cloacal exstrophy – 15 new cases and literature steal: the pathogenetic mechanism producing
review. Am J Med Genet A. 2007;143(18):2122–8. sirenomelia and associated defects of the viscera and
28. McHugo J, Whittle M. Enlarged fetal bladders: soft tissues. Pediatrics. 1986;78:451–7.
aetiology, management and outcome. Prenat Diagn. 48. Abruzzo MA, Erickson RP. A new syndrome of cleft
2001;21:958–63. palate associated with coloboma, hypospadia, deaf-
29. Ueliling DT, Gilbert E, Chesney R. Urologic implica- ness, short stature, and radial synostosis. J Med
tions of the VATER association. J Urol. Genet. 1977;14:76–80.
1983;129:352–4. 49. Nevin NC, Herron B, Armstrong MJ. An 18 week
30. Evans JA, Stranc LC, Kaplan P, et al. VACTERL with fetus with Elejalde syndrome (acrocephalopoly-
hydrocephalus: further delineation of the syndrome dactylous dysplasia). Clin Dysmorphol. 1994;3:
(s). Am J Med Genet. 1989;34:177–82. 180–4.
31. Damian MS, Seibel P, Schachenmayr W, 50. Evans JA, Vitez M, Czeizel A. Pattern of acrorenal
et al. VACTERL with the mitochondrial np 3243 malformation associations. Am J Med Genet.
point mutation. Am J Med Genet. 1996;62:398–403. 1992;44:413–19.
32. Harper PS. Practical genetic counseling. 3rd ed. 51. Halal F, Desgranges M-F, Leduc B, et al. Acro-renal-
London: Butterworth; 1988. mandibular syndrome. Am J Med Genet.
33. Pagon RA, Graham JM, Zonana J, et al. Coloboma, 1980;5:277–84.
congenital heart disease and choanal atresia with mul- 52. Kohlhase J, Schubert L, Liebers M, Rauch A,
tiple anomalies, CHARGE association. J Pediatr. Becker K, Mohammed SN, Newbury-Ecob R,
1981;99:223–7. Reardon W. Mutations at the SALL4 locus on chro-
34. Harris J, Robert E, Kallen B. Epidemiology of mosome 20 result in a range of clinically overlapping
choanal atresia with special reference to the phenotypes, including Okihiro syndrome, Holt-Oram
CHARGE association. Pediatrics. 1997;99:363–7. syndrome, acro-renal-ocular syndrome, and patients
35. Davenport SLH, Hefner MA, Mitchell JA. The spec- previously reported to represent thalidomide
trum of clinical features in CHARGE syndrome. Clin embryopathy. J Med Genet. 2003;40:473–8.
Genet. 1986;29:298–310. 53. Schuhl H. Aglossia-adactylia. A propos of a case.
36. Toriello HV. CHARGE association. J Am Acad Review of the literature. Ann Pediatr (Paris).
Audiol. 1995;6:47–53. 1986;33:137–40.
37. Sanlaville D, Verloes A. CHARGE syndrome: an 54. Pauli RM, Pettersen JC, Arya S. Familial agnathia-
update. Eur J Hum Genet. 2007;15(4):389–99. holoprosencephaly. Am J Med Genet. 1983;14:
38. Herlin M, Højland AT, Petersen MB. Familial occur- 677–98.
rence of Mayer-Rokitansky-K€ uster-Hauser syn- 55. Krantz ID, Piccoli DA, Spinner NB. Alagille syn-
drome: a case report and review of the literature. Am drome. J Med Genet. 1997;34:152–7.
J Med Genet A. 2014;9999:1–11. 56. Kashtan CE. Alport syndrome: an inherited disorder
39. Lin HJ, Cornford ME, Hu B, et al. Occipital of renal, ocular, and cochlear basement membranes.
encephalocele and MURCS association: case report Medicine. 1999;78:338–60.
and review of central nervous system anomalies in 57. Alsing A, Christensen C. A typical macular coloboma
MURCS patients. Am J Med Genet. 1996;61:59–62. (dysplasia) associated with familial juvenile
40. Wellesley DG, Slaney SF. MURCS in a male? J Med nephronophthisis and skeletal abnormality. Ophthal-
Genet. 1995;32:314–15. mic Paediatr Genet. 1988;9:149–55.
41. Terada S, Suzuki N, Uchide K, et al. Etiology of prune 58. Marshall JD, Beck S, Maffei P, Naggert JK. Alstrom
belly syndrome: evidence of megalocystic origin in an syndrome. Eur J Hum Genet. 2007;15:1193–202.
early fetus. Obstet Gynecol. 1994;83:865–8. 59. Hall RK, Phakey P, Palamara J, McCredie
42. Tonni G, Ida V, Alessandro V, Bonasoni MP. The DA. Amelogenesis imperfecta and nephrocalcinosis
Prune belly syndrome. Fetal Pediatr Pathol. 2013; syndrome: case studies of clinical features and ultra-
31(1):13–24. structure of tooth enamel in two siblings. Oral Surg
172 C. Limwongse

Oral Med Oral Pathol Oral Radiol Endod. 75. Milunsky JM, Maher TA, Zhao G, Roberts AE,
1995;79:583–92. Stalker HJ, Zori RT, et al. TFAP2A mutations result
60. DelaChapelle A, Kere J, Sack Jr GH, Tolvanen R, in branchio-oculo-facial syndrome. Am J Hum Genet.
Maury CPJ. Familial amyloidosis, Finnish type: 2008;82(5):1171–7.
G654-to-A mutation of the gelsolin gene in Finnish 76. Chitayat D, Hodgkinson KA, Chen MY, et al.
families and an unrelated American family. Geno- Branchio-oto-renal syndrome: further delineation of
mics. 1992;13:898–901. an underdiagnosed syndrome. Am J Med Genet.
61. Barr Jr M. Teratogen update: angiotensin-converting 1992;43:970–5.
enzyme inhibitors. Teratology. 1994;50:399–409. 77. Braun FC, Bayer JF. Familial nephrosis associated
62. Clericuzio CL. Clinical phenotypes and Wilms’ with deafness and congenital urinary tract anomalies
tumor. Med Pediatr Oncol Suppl. 1993;21:182–7. in siblings. J Pediatr. 1962;60:33–41.
63. Stewart FJ, Nevin NC, Brown S. Axial mesodermal 78. Oates NA, VanVliet J, Duffy DL, Kroes HY, Martin
dysplasia spectrum. Am J Med Genet. 1993;45:426–9. NG, Boomsma DI, et al. Increased DNA methylation
64. Baldellou A, Bone J, Tamparillas M, et al. Congenital at the AXIN1 gene in a monozygotic twin from a pair
hypoparathyroidism, ocular coloboma, unilateral discordant for a caudal duplication anomaly. Am J
renal agenesis and dysmorphic features. Genet Hum Genet. 2006;79:155–62.
Couns. 1991;2:245–7. 79. Kibar Z, Torban E, McDearmid JR, Reynolds A,
65. Van Maldergem L, Siitonen HA, Jalkh N, Chouery E, Berghout J, Mathieu M, et al. Mutations in
De Roy M, Delague V, Muenke M, Jabs EW, Cai J, VANGL1 associated with neural-tube defects. N
Wang LL, Plon SE, Fourneau C, Kestila M, Engl J Med. 2007;356:1432–7.
Gillerot Y, Megarbane A, Verloes A. Revisiting the 80. Adra A, Cordero D, Mejides A, et al. Caudal regres-
craniosynostosis-radial ray hypoplasia association: sion syndrome: etiopathogenesis, prenatal diagnosis,
Baller-Gerold syndrome caused by mutations in the and perinatal management. Obstet Gynecol Surv.
RECQL4 gene. J Med Genet. 2006;43:148–52. 1994;49:508–16.
66. Muroya K, Hasegawa T, Ito Y, Nagai T, Isotani H, 81. Passarge E, MeAdams AJ. Cerebro-hepato-renal syn-
Iwata Y, Yamamoto K, Fujimoto S, Seishu S, drome. A newly recognized hereditary disorder of
Fukushima Y, Hasegawa Y, Ogata T. GATA3 abnor- multiple congenital defects, including sudanophilic
malities and the phenotypic spectrum of HDR syn- leukodystrophy, cirrhosis of the liver, and polycystic
drome. J Med Genet. 2001;38:374–80. kidneys. J Pediatr. 1967;71:691–702.
67. Beales PL, Elcioglu N, Woolf AS, Parker D, Flinter 82. Matsuzaka T, Sakuragawa N, Nakayama H, et al.
FA. New criteria for improved diagnosis of Bardet- Cerebro-oculo-hepato-renal syndrome (Arima’s syn-
Biedl syndrome: results of a population survey. J Med drome), a distinct clinicopathological entity. J Child
Genet. 1999;36:437–46. Neurol. 1986;1:338–46.
68. O’dea D, Parfrey PS, Harnett JD, et al. The impor- 83. Opitz JM, Lowry RE, Holmes TM, et al. Hutterite
tance of renal impairment in the natural history of cerebro-osteo-nephrodysplasia; autosomal recessive
Bardet-Biedl syndrome. Am J Kidney Dis. trait in a Lehrerleut Hutterite family from Montana.
1996;27:776–83. Am J Med Genet. 1985;22:521–9.
69. Elliot M, Bayly R, Cole T, et al. Clinical features and 84. Caraballo A, Lopez Barrios A, Martin Govantes J,
natural history of Beckwith-Wiedemann syndrome: et al. Thoracic asphyxiant dystrophy and renal dis-
presentation of 74 new cases. Clin Genet. ease. An Esp Pediatr. 1977;10:88–95.
1994;46:168–74. 85. Brueton LA, Dillon MJ, Winter RM. Ellis van
70. Mulvihill DM, Mercado MG, Boineau FG. Beckwith- Creveld syndrome, Jeune syndrome, and renal-
Wiedemann syndrome and its association with type hepatic-pancreatic dysplasia: separate entities or dis-
3 polycystic kidney disease. Pediatr Nephrol. ease spectrum? J Med Genet. 1990;27:252–5.
1989;3:286–9. 86. Chasnoff IJ, Chisum GM, Kaplan WE. Maternal
71. Ponte P, Katz SM, FaIkner B. Beckwith-Wiedemann cocaine use and genitourinary tract malformations.
syndrome. Renal pathology at 4 months of age. Clin Teratology. 1988;37:201–4.
Pediatr. 1990;29:393–7. 87. Jackson L, Kline AD, Barr MA, et al. De Lange
72. Watanabe H, Yamanaka T. A possible relationship syndrome: a clinical review of 310 individuals. Am
between Beckwith-Wiedemann syndrome, urinary J Med Genet. 1993;147:940–6.
tract anomaly and prune belly syndrome. Clin 88. Goswami RK, Rangnekar GV, Varshney S, et al.
Genet. 1990;38:410–15. Crossed renal ectopia with pelvic lipomatosis: a new
73. Moller DE, Flier JS. Insulin resistance-mechanisms, syndrome involving chromosome 1. Hum Genet.
syndromes, and implications. N Engl J Med. 1992;89:666–70.
1991;325:938–48. 89. Czeizel A, Losonei A. Split hand, obstructive urinary
74. Langer Jr LO, Nishino R, Yamaguchi A, et al. anomalies and spina bifida or diaphragmatic defect
Brachymesomelia-renal syndrome. Am J Med syndrome with autosomal dominant inheritance. Hum
Genet. 1983;15:57–65. Genet. 1987;77:203–4.
6 Developmental Syndromes and Malformations of the Urinary Tract 173

90. Lynch SA, Wright C. Sirenomelia, limb reduction family with review of the literature. Pediatr Radiol.
defects, cardiovascular malformation, renal agenesis 1993;23:131–3.
in an infant born to a diabetic mother. Clin 108. Thomas IT, Honore GM, Jewett T, et al. Holzgreve
Dysmorphol. 1997;6:75–80. syndrome: recurrence in sibs. Am J Med Genet.
91. Grix Jr A, Curry C, Hall BD. Patterns of multiple 1993;45:767–9.
malformations in infants of diabetic mothers. Birth 109. Verloes A. Hypertelorism-microtia-clefting (HMC
Defects Orig Artic Ser. 1982;18:55–77. syndrome). Genet Couns. 1994;5:283–7.
92. Mueller RF. Syndrome of the month. Denys-Drash 110. Larson RS, Rudloff MA, Liap H, et al. The Ivemark
syndrome. J Med Genet. 1994;31:471–7. syndrome: prenatal diagnosis of an uncommon cystic
93. London R, Heredia RM, Israel J. Urinary tract renal lesion with heterogeneous associations. Pediatr
involvement in EEC syndrome. Am J Dis Child. Nephrol. 1995;9:594–8.
1985;139:1191–3. 111. Cantani A, Lucenti P, Ronzani GA, et al. Joubert syn-
94. Turi S, Kobor J, Erdos A, et al. Hereditary nephritis, drome: review of the fifty-three cases so far published.
platelet disorders and deafness – Epstein’s syndrome. Ann Genet. 1990;33:96–8.
Pediatr Nephrol. 1992;6:38–43. 112. Verloes A, Le Mercer M, Davin J-C, et al. The
95. Nevin NC, Hill AE, Carson DJ. Facio-cardio-renal orocraniodigital syndrome of Juberg and Hayward.
(Eastman-Bixler) syndrome. Am J Med Genet. J Med Genet. 1992;29:262–5.
1991;40:31–3. 113. Niikawa N, Matsuura N, Fukushima Y, et al. The
96. Glaze A, Fraser FC. Spectrum of anomalies in Kabuki-make up syndrome. J Pediatr. 1981;99:565–9.
Fanconi anemia. J Med Genet. 1982;19:412–16. 114. Honiball S, Snadler M. Kallmann syndrome with
97. Joenje H, Patel KJ. The emerging genetic and molec- unilateral renal agenesis: a case report. S Afr Med
ular basis of Fanconi anaemia. Nat Rev Genet. J. 1986;70:489–91.
2001;2:446–57. 115. Schaap C, de Die-Smulder CEM, Kuijten RH,
98. Taylor CL, Jones KL, Jones MC, et al. Incidence of et al. McKusick-Kaufmann syndrome: the diagnostic
renal anomalies in children prenatally exposed to challenge of abdominal distension in the neonatal
ethanol. Pediatrics. 1994;94:209–12. period. Eur J Pediatr. 1992;151:583–5.
99. Davies RS, Carty H, Pierro A. Infantile 116. Hennekam RCM, Van Schooneveld MJ, Ardinger
myofibromatosis – a review. Br J Radiol. HH, et al. The Peters’ plus syndrome: description of
1994;67:619–23. 16 patients and review of the literature. Clin
100. Gattuso J, Patton MA, Baraitser M. The clinical Dysmorphol. 1993;2:283–300.
spectrum of the Fraser syndrome: report of 117. Grise P, Lefort J, Dewald M, et al. Urinary
three new cases and review. J Med Genet. malformations in synostoses of the cervical vertebrae
1987;24:549–55. (Klippel-Feil syndrome). Apropos of 35 cases. Ann
101. Klamt B, Koziell A, Poulat F, Wieacker P, Scambler P, Urol (Paris). 1984;18:232–5.
Berta P, Gessler M. Frasier syndrome is caused by 118. Kousseff EG. Sacral meningocoele and conotruncal
defective alternative splicing of WT1 leading to an heart defects: a possible autosomal recessive trait.
altered ratio of WT1+/KTS splice isoforms. Hum Pediatrics. 1984;74:395–8.
Mol Genet. 1998;7:709–14. 119. Ellis EN, Kemp SF, Frindik JP, et al. Glomerulopathy
102. Goeminne L. A new probably X-linked inherited in patients with Donohue syndrome (Leprechaunism).
syndrome: congenital muscular torticollis, multiple Diabetes Care. 1991;14:413–14.
keloids, cryptorchidism and renal dysplasia. Acta 120. Russo R, D’Armiento M, Angrisami P, et al. Limb
Genet Med Gemeloll. 1968;17:439–67. body wall complex: a critical review and a nosologi-
103. Ritchey NIL, Norbeck J, Huang C, et al. Urologic cal proposal. Am J Med Genet. 1993;47:893.
manifestations of Goldenhar syndrome. Urology. 121. Goeminne L. Synopsis of mammorenal syndrome.
1994;43:88–91. Humangenetik. 1972;14:170–1.
104. Goldston AS, Burke EC, D Agostino A, 122. Schrander-Stumpel C, DeDieSchmulder C,
et al. Neonatal polycystic kidney with brain defect. DeKrom M, et al. Marden-Walker syndrome: case
Am J Dis Child. 1963;106:484–8. report, literature review and nosologic discussion.
105. Graham Jr J, Boyle W, Troxell J, et al. Cystic Clin Genet. 1993;43:303–8.
harmartomata of lung and kidney: a spectrum of 123. Blankenberg TA, Ruebner BH, et al. Pathology of
developmental abnormalities. Am J Med Genet. renal and hepatic anomalies in Meckel-Gruber syn-
1987;27:45–59. drome. Am J Med Genet. 1987;3(Suppl):395–410.
106. Hennessy WT, Cromie WJ, Duckett JW. Congenital 124. Penman DG, Lilford RJ. The megacystis-microcolon
hemihypertrophy and associated abdominal lesions. -intestinal hypoperistalsis syndrome: a fatal autoso-
Urology. 1981;18:576–9. mal recessive condition. J Med Genet. 1989;26:66.
107. Lipschitz B, Berdon WE, Defelice AR, et al. Associ- 125. LaMontagne AE. Urologic manifestations of
ation of congenital hepatic fibrosis and autosomal Melnick-Needles syndrome: a case report and review
dominant polycystic kidney disease. Report of a of the literature. J Urol. 1991;145:1020–1.
174 C. Limwongse

126. Robinson S, Kessling A. Diabetes secondary to 144. Hasselbacher K, Wiggins RC, Matejas V, Hinkes BG,
genetic disorders. Baillieres Clin Endocrinol Metab. Mucha B, Hoskins BE, et al. Recessive missense
1992;6:867–98. mutations in LAMB2 expand the clinical spectrum
127. Cunniff C, Williamson-Kruse L, Olney of LAMB2-associated disorders. Kidney Int.
AH. Congenital microgastria and limb reduction 2006;70:1008–12.
defects. Pediatrics. 1993;91:1192–4. 145. MacKenzie J, Chitayat D, McLorie G, et al.
128. Miranda D, Schjnella RA, Finegold MJ. Familial Penoscrotal transposition: a case report and review.
renal dysplasia. Arch Pathol. 1972;93:483–91. Am J Med Genet. 1994;49:103–7.
129. Moerman P, Vandenberghe K, Fryns JP, et al. A new 146. Henneveld HT, van Lingen RA, Hamel BCJ, Stolte-
lethal chondrodysplasia with spondylocostal Dijkstra I, Van Essen AJ. Perlman syndrome: four
dysostosis, multiple internal anomalies and Dandy- additional cases and review. Am J Med Genet.
Walker cyst. Clin Genet. 1985;27:160–4. 1999;86:439–46.
130. McDonald MT, Gorski JL. Syndrome of the month. 147. Halal F. Distal obstructive uropathy with polydactyly:
Nager acrofacial dysostosis. J Med Genet. a new syndrome? Am J Med Genet. 1986;24:753–7.
1993;30:779–82. 148. Singh I, Johnson GH, Brown III FR. Peroxisomal
131. Rizzo R, Pavone L, Micali G, et al. Familial bilateral disorders Biochemical and clinical diagnostic consid-
antecubital pterygia with severe renal involvement in eration (Review). Am J Dis Child. 1988;142:
nail-patella syndrome. Clin Genet. 1993;44:1–7. 1297–301.
132. King JA, Gardner V, Chen S, et al. Neu-Laxova 149. Moser AB, Rasmussen M, Naldu S, et al. Phenotype
syndrome: pathological evaluation of a fetus and of patients with peroxisomal disorders subdivided
review of the literature. Pediatr Pathol Lab Med. into sixteen complementation groups. J Pediatr.
1995;15:57–79. 1995;127:13–22.
133. Freire-Maia N, Pinheiro M, Opitz JM. The 150. Suzuki Y, Shimozawa N, Takahashi Y,
neurofaciodigitorenal (NFDR) syndrome. Am J Med et al. Peroxisomal disorders: clinical aspects. Ann N
Genet. 1982;11:326–36. Y Acad Sci. 1996;804:442–9.
134. Demierre MF, Gerstein W. Segmental neurofibroma- 151. Bidair M, Kalota SJ, Kaplan GW. Infantile hypertro-
tosis with ipsilateral renal agenesis. Int J Dermatol. phic pyloric stenosis and hydronephrosis: is there an
1996;35:445–7. association? J Urol. 1993;150:153–5.
135. Saraiva JM, Lemos C, Goncalves I, 152. Raas-Rothchild A, Goodman RM, Grunbaum M,
et al. Arthrogryposis multiplex congenita with renal et al. Klippel-Feil anomaly with sacral agenesis: an
and hepatic abnormalities in a female infant. J Pediatr. additional subtype, type IV. J Craniofac Genet Dev
1990;117:761–3. Biol. 1988;8:297–301.
136. George CD, Patton MA, el Sawi M, et al. Abdominal 153. Vargas FR, Carbral de Almeida JC, Llrena Jr JC,
ultrasound in Noonan syndrome: a study in et al. Brief clinical report: RAPADILINO syndrome.
44 patients. Pediatr Radiol. 1993;23:316–18. Am J Med Genet. 1992;44:716–19.
137. Tsukahara M, Imaizumi K, Kawai S, et al. Occipital 154. Davee MA, Moore CA, Bull MJ, et al. New syn-
horn syndrome: report of a patient and review of the drome? Familial occurrence of renal and Mullerian
literature. Clin Genet. 1994;45:32–5. duct hypoplasia, craniofacial anomalies, severe
138. Teebi AS, Hassoon MM. Urofacial syndrome associ- growth and developmental delay. Am J Med Genet.
ated with hydrocephalus due to aqueductal stenosis. 1992;44:293–6.
Am J Med Genet. 1991;40:199–200. 155. Moerman P, Fryns J-P, Sastrowijoto HS, et al. Hered-
139. Hunter AGW, Jurenka S, Thompson D, et al. Absence itary renal adysplasia: new observation and hypothe-
of the cerebellar granular layer, mental retardation, ses. Pediatr Pathol. 1994;14:405–10.
tapetoretinal degeneration and progressive 156. Rosa FW, Wilk AL, Ketsey FO. Teratogen update:
glomerulopathy: an autosomal recessive oculo-renal- vitamin A congeners. Teratology. 1986;34:366.
cerebellar syndrome. Am J Med Genet. 157. Rothman M, Moore LL, Singer MR, et al. Teratoge-
1982;11:383–95. nicity of high vitamin A intake. N Engl J Med.
140. Weaver RG, Cashwell LP, Lorentz W, et al. Optic 1995;333:1369–73.
nerve coloboma associated with renal disease. Am J 158. VanDenBerg DJ, Francke U. Roberts syndrome: a
Med Genet. 1988;29:597–605. review of 100 cases and a new rating system for
141. Morse MJ, Lirenman DS, Johnson HW. The association severity. Am J Med Genet. 1993;47:1104–23.
of renal pelviocaliceal dysmorphism with sensorineural 159. Robson WLM, Lowry RB, Leung AM. X-linked
deafness: a new syndrome. J Urol. 1981;125:625–7. recessive nephritis with mental retardation, sensori-
142. Hall JG, Pallister M, Clarren SK, et al. Congenital neural hearing loss and macrocephaly. Clin Genet.
hypothalamic hamartoblastoma, hypopituitarism, 1994;45:314–17.
imperforate anus and postaxial polydactyly – a new 160. Strubbe EH, Cremers CWRJ, Willensen P, et al. The
syndrome. Am J Med Genet. 1980;7:47–83. Mayer-Rokitansky-Kuster-Hauser syndrome without
143. Biesecker LG, Graham Jr JM. Pallister-Hall syn- and with associated features: two separate entities.
drome. J Med Genet. 1996;33:585–9. Clin Dysmorphol. 1994;3:192–9.
6 Developmental Syndromes and Malformations of the Urinary Tract 175

161. Menser MA, Robertson SE, Dorman DC, et al. Renal 178. Sommer A, Rathbun MA, Battles ML. A syndrome
lesions in congenital rubella. Pediatrics. 1967;40: with partial aniridia, unilateral renal agenesis and
901–4. mild psychomotor retardation. J Pediatr. 1974;85:
162. Prosperi P, Ricotti CG. Rubinstein-Taybi syndrome with 870–2.
renal malformations. Minerva Pediatr. 1975;27:2230–8. 179. Thompson EM, Baraitser M. Sorsby syndrome: a
163. Wallerstein R, Anderson CE, Hay B, et al. Submicro- report on further generations of the original family. J
scopic deletions at 16p13.3 in Rubinstein-Taybi syn- Med Genet. 1988;25:313–21.
drome: frequency and clinical manifestations in a 180. Sotos JF. Syndromes and other disorders associated
North American population. J Med Genet. 1997;34: with overgrowth. Clin Pediatr. 1997;36:89–103.
203–6. 181. Jejart G, Seres E. Supernumerary nipples and renal
164. Patton MA. Syndrome of the month. Russell-silver anomalies. Int Urol Nephrol. 1994;26:141–4.
syndrome. J Med Genet. 1988;25:557–60. 182. McBride WG. Thalidomide embryopathy. Teratol-
165. Santos H, Mateus J, Leal MJ. Hirschsprung disease ogy. 1977;16:79–82.
associated with polydactyly, unilateral renal agenesis, 183. Smithells BW, Newman CGH. Recognition of thalid-
hypertelorism and congenital deafness: a new autoso- omide defects. J Med Genet. 1992;29:716–23.
mal recessive syndrome. J Med Genet. 1988;25: 184. Rudd NL, Curry C, Chen KTK, et al. Thymic-renal-
204–8. anal-lung dysplasia in sibs: a new autosomal recessive
166. Abu-Libdeh B, Fujimoto A, Ehinger M. Syndrome of error of early morphogenesis. Am J Med Genet.
cleft palate, microcephaly, large ears and short stature 1990;37:401–5.
(Say syndrome). Am J Med Genet. 1993;45:358–60. 185. Tolmie JL, Patrick A, Yates JRW. A lethal multiple
167. Ludman MD, Cole DEC, Crocker JFS, et al. Schimke pterygium syndrome with apparent X-linked inheri-
immuno-osseous dysplasia: case report and review. tance. Am J Med Genet. 1987;27:913–19.
Am J Med Genet. 1993;47:793–6. 186. O’Callaghan M, Young ID. Syndrome of the month.
168. Okamoto N, Takeuchi M, Kitajinia H, et al. A patient Townes-Brocks syndrome. J Med Genet. 1990;27:
with Schinzel-Giedion syndrome and a review of 457–61.
20 patients. Jpn J Hum Genet. 1995;40:189–93. 187. Kelly TE. Teratogenicity of anticonvulsant drugs. I:
169. Cantani A, Bamonte G, Ceccoli D, et al. Familial review of the literature. Am J Med Genet. 1984;19:
juvenile nephronophthisis. A review and differential 413–34.
diagnosis. Clin Pediatr. 1986;25:90–5. 188. Bernstein J, Robbins TO. Renal involvement in tuber-
170. Clark RD, Golabi M, Lacassie Y, et al. Expanded ous sclerosis. Ann N Y Acad Sci. 1991;615:36–49.
phenotype and ethnicity in Setleis syndrome. Am J 189. Bamshad M, Root S, Carey JC. Clinical analysis of a
Med Genet. 1989;34:354–7. large kindred with the Pallister ulnar-mammary syn-
171. Tsai Y-C, Chang J-M, Changchien CC, et al. Unusual drome. Am J Med Genet. 1996;65:325–31.
short rib-polydactyly syndrome. Am J Med Genet. 190. Battin J, Lacombe B, Leng JJ. Familial occurrence of
1992;44:31–6. hereditary renal adysplasia with Mullerian anomalies.
172. Silence D, Kozlowski K, Bar-Ziv J, et al. Perinatally Clin Genet. 1993;43:23–4.
lethal short rib-polydactyly syndromes 1. Variability 191. Neumann HP, Zbar B. Renal cyst, renal cancer and
in known syndromes. Pediatr Radiol. 1987;17: von Hippel-Lindau disease. Kidney Int. 1997;51:
474–80. 16–26.
173. Aleck KA, Grix A, Clericuzio C, et al. Dyssegmental 192. Maher ER, Yates JRW, Harries R, et al. Clinical fea-
dysplasia: clinical, radiographic and morphologic evi- tures and natural history of von Hippel-Lindau dis-
dence of heterogeneity. Am J Med Genet. 1987;27: ease. Q J Med. 1990;77:1151–63.
295–312. 193. Weinberg AG, Zumwalt RE. Bilateral nephromegaly
174. Simoupoulos A, Brennan GC, Alivan A, et al. and multiple pulmonary cysts. Am J Clin Pathol.
Polycystic kidneys, internal hydrocephalus, 1976;67:284–8.
polydactylism in newborn siblings. Pediatrics. 194. Wenstrup RJ, Pagon RA. Female pseudohermaph-
1967;39:931–4. roditism with anorectal, Mullerian duct, and urinary
175. Verloes A, Massart B, Dehalleux I, et al. Clinical tract malformations: report of four cases. J Pediatr.
overlap of Beckwith-Wiedermann, Perlman and 1985;107:751–4.
Simpson-Golabi-Behmel syndromes: a diagnostic pit- 195. Turnpenny PD, Dean JCS, Dufty P, et al. Syndrome of
fall. Clin Genet. 1995;47:257–62. the month: Weyers’ ulnar ray/oligodactyly and the
176. Cunniff C, Kratz LE, Moser A, et al. Clinical and association of midline malformations and ulnar ray
biochemical spectrum of patients with RSH/Smith- defects. J Med Genet. 1992;29:659–62.
Lemli-Opitz syndrome and abnormal cholesterol 196. DelPapa M, Attardo S, Mobilli M, et al. Critical con-
metabolism. Am J Med Genet. 1997;68:263–9. siderations on a case of nephroblastoma in a horse-
177. Curry JCR, Carey JC, Holland JS, et al. Smith-Lemli- shoe kidney. G Chir. 1991;12:549–52.
Opitz syndrome type II: multiple congenital anoma- 197. Hewitt M, Lunt PW, Oalchill A. Wilms’ tumor and a
lies with male pseudohermaphroditism and frequent de novo (1;7) translocation in a child with bilateral
early lethality. Am J Med Genet. 1987;26:45–57. radial aplasia. J Med Genet. 1991;28:411–12.
176 C. Limwongse

198. King LA, Sanchez-Ramos L, Talledo OE, 217. Cullen SI, Harris DE, Carter CH, et al. Congenital
et al. Syndrome of genital, renal and middle ear unilateral ichthyosiform erythroderma. Arch
anomalies: a third family and report of a pregnancy. Dermatol. 1969;199:724–9.
Obstet Gynecol. 1986;69:891–3. 218. Nuoffer J, Pfammatter JP, Spahr A, et al.
199. Monller Gisberg J, Tano Pino F, Rodriguez Chondrodysplasia punctata with a mild clinical
Arteaga G, et al. Urologic manifestations in Wolfram course. J Inherit Metab Dis. 1994;17:60–6.
syndrome. Acta Urol Esp. 1996;20:474–7. 219. Levy P, Baraitser M. Syndrome of the month. Coffin-
200. Hing AV, Dowton SB. Aase syndrome: novel radio- Siris syndrome. J Med. 1991;28:338–41.
graphic feature. Am J Med Genet. 1993;45:413–15. 220. Janik JS, Shandling B, Mancer K. Cutis laxa and
201. Schulte JM, Lenz W, Vogel M. Lethale hollow viscus diverticula. J Pediatr Surg.
Achondrogenesis: Eine Ubersicht uber 56 Falle. 1982;17:318–20.
Klin Padiatr. 1978;191:327–40. 221. Woods CG, Treleaven S, Betheras FR, et al. Disorga-
202. Casamassima AC, Beneck D, Gewitz MH, nization-like syndrome with 47, XXY and unilateral
et al. Acrocallosal syndrome: additional manifesta- narrowing of the common iliac artery. Clin
tions. Am J Med Genet. 1989;32:311. Dysmorphol. 1995;4:82–6.
203. Petit P, Moerman P, Fryns J-P. Acrofacial dysostosis 222. Cullen P, Rodgers CS, Callen CF, et al. Association of
syndrome type Rodriguez: a new lethal MCA syn- familial Duane anomaly and urogenital abnormalities
drome. Am J Med Genet. 1992;42:343–5. with a bisatellited marker derived from chromosome
204. Toriello HV, Radecki LL, Sharda J, et al. Frontonasal 22. Am J Med Genet. 1993;47:925–30.
‘dysplasia’, cerebral anomalies and polydactyly: 223. Levard G, Aigrain Y, Ferkadji L, et al. Urinary blad-
report of a new syndrome and discussion from a der diverticula and the Ehlers-Danlos syndrome in
developmental field perspective. Am J Med children. J Pediatr Surg. 1989;24:1184–6.
G. 1986;2(Suppl):89–96. 224. Berger TG, Detlefs RL, Donatucci CF. Junctional
205. McMillan DD, McArthur RG, Williams JA, epidermolysis bullosa, pyloric atresia, and genitouri-
et al. Upper urinary tract anomalies in children with nary disease. Pediatr Dermatol. 1986;3:130–4.
adrenogenital syndrome. J Pediatr. 1976;89:953–5. 225. Eklof O, Parkkulainen K. Epidermolysis bullosa
206. Petrykowski WV, Beckmann R, Bohm N, et al. Adre- dystrophica with urinary involvement. J Pediatr
nal insufficiency, myotonic hypotonia, severe psycho- Surg. 1984;19:215–17.
motor retardation, failure to thrive, constipation, and 226. Burn J, Winter RM, Baraitser M, et al. The femoral
bladder ectasia in 2 brothers: Adrenomyodystrophy. hypoplasia-unusual facies syndrome. J Med Genet.
Helv Paediatr Acta. 1982;37:387–400. 1984;21:331–40.
207. Hassell S, Butler MG. Antley-Bixler syndrome: 227. Patton RA, Hurst J, Donnai D, et al. Syndrome of the
report of a patient and review of the literature. Clin month. Floating-Harbor syndrome. J Med Genet.
Genet. 1994;46:372–6. 1991;28:201–4.
208. Cohen Jr MM, Kreiborg S. Visceral anomalies in the 228. Suskan E, Kurkcuoglu N, Uluoglu O. Focal dermal
Apert syndrome. Am J Med Genet. 1993;45:758–60. hypoplasia (Goltz syndrome) with horseshoe kidney
209. Cairney AEL, Andrews M. Wilms’ tumor in three abnormality. Pediatr Dermatol. 1990;7:283–6.
patients with Bloom syndrome. J Pediatr. 229. Hashemi C, Mahloudji M. The whistling face syn-
1987;111:414–16. drome: report of a case with a renal anomaly. Birth
210. Bowen P, Conradi GJ. Syndrome of skeletal and Defects Orig Artic Ser. 1974;10:265–7.
genitourinary anomalies with unusual facies and fail- 230. Kanemura T, Orii T, Olitani M. Frontometaphyseal
ure to thrive in Hutterite sibs. Birth Defects Orig Artic dysplasia with congenital urinary tract malformations.
Ser. 1976;12(6):101–8. Clin Genet. 1979;16:399–404.
211. Temtamy SA. The genetics of hand malformations. 231. Roizenblatt J, Wajntal A, Diament AJ. Median cleft
Congenital Anomalies. 1985;25:73–92. face syndrome or frontonasal dysplasia: a case report
212. Kosaki K, Curry C, Roeder E, et al. Ritscher-Schinzel with associated renal malformations. J Pediatr
(3C) syndrome: delineation of the phenotype. Am J Ophthalmol Strabismus. 1979;16:16–20.
Med Genet. 1997;68:421–7. 232. Siebert JR, Benjamin DR, Jutil S, et al. Urinary tract
213. Antley RM, Hwang DS, Theopold W, et al. Further anomalies associated with congenital diaphragmatic
delineation of the C (trigonocephaly) syndrome. J defects. Am J Med Genet. 1990;37:1–5.
Med Genet. 1981;9:147–63. 233. Hockey A, Knowles S, Davies D, et al.
214. Houston CS, Opitz JM, Spranger JW, et al. The Glutaric aciduria type II, an unusual cause of
campomelic syndrome. J Med Genet. 1983;15:3–28. prenatal polycystic kidneys: report of prenatal
215. Jaeken J, Carchon R. The carbohydrate deficient gly- diagnosis and confirmation of autosomal recessive
coprotein syndromes: an overview. J Inherit Metab inheritance. Birth Defects Orig Artic Ser. 1993;29:
Dis. 1993;16:813–20. 373–82.
216. Robinson LK, James HE, Mubarak SJ, et al. Carpen- 234. Gallop TR, Fontes LR. The Grieg cephalopoly-
ter syndrome: natural history and clinical spectrum. syndactyly syndrome: report of a family and review
Am J Med Genet. 1985;20:461–9. of the literature. Am J Med Genet. 1985;22:59–68.
6 Developmental Syndromes and Malformations of the Urinary Tract 177

235. Kaplan P, Ramos F, Zackai EH, et al. Cystic kidney 253. Cappa M, Borrelli P, Marini R, et al. The Opitz syn-
disease in Hajdu-Cheney syndrome. Am J Med drome: a new designation for clinically indistinguish-
Genet. 1995;56:25–30. able BBB and G syndromes. Am J Med Genet.
236. Salonen R, Herva R. Syndrome of the month: 1987;28:303–10.
hydrolethalus syndrome. J Med Genet. 1990;27: 254. Toriello HV. Review Oral-facial-digital syndromes,
756–9. 1992. Clin Dysmorphol. 1993;2:95–105.
237. Devos EA, Leroy JG, Braeckman JJ, et al. 255. Meinecke P, Hayek H. Orofaciodigital syndrome type
Spondylocostal dysostosis and urinary tract anomaly: IV (Mohr-Majewski) with severe expression
definition and review of the entity. Eur J Pediatr. expanding the known spectrum of anomalies. J Med
1978;128:7–15. Genet. 1990;27:200–2.
238. Gershoni-Baruch R, Lemer A, Braun J, 256. Kieselstein M, Herman G, Wahrman J, et al. Muco-
et al. Johanson-Blizzard syndrome: clinical spectrum cutaneous pigmentation and intestinal polyposis
and further delineation of the syndrome. Am J Med (Peutz-Jeghers syndrome) in a family of Iraqi Jews
Genet. 1990;35:546–51. with polycystic kidney disease, with a chromosome
239. Horn D, Wakowski R. Phenotype and counseling in study. Isr J Med Sci. 1969;5:81–90.
lacrimo-auriculodento-digital (LADD) syndrome. 257. Kuwada SK, Burt RW. The clinical features of the
Genet Couns. 1993;4:305–9. hereditary and nonhereditary polyposis syndromes.
240. Raff ML, Byers PH. Joint hypermobility syndromes. Surg Oncol Clin N Am. 1996;5:553–67.
Curr Opin Rheumatol. 1996;8:459–66. 258. Bouvet J, Maroteaux P, Briand-Guillemot M. Le syn-
241. Silverman FN. Larsen syndrome: congenital disloca- drome de Poland: Etudes clinique et genetique con-
tion of the knees and other joints, distinctive facies, siderations physiopathologiques. Presse Med.
and frequently, cleft palate. Ann Radiol. 1972; 1976;5:185–90.
15:297–328. 259. Bamforth JS, Fabian C, Machin G, et al. Poland anom-
242. Traboulsi EI, Lenz W, GonzalesRamos M, et al. The aly with a limb body wall disruption defect: case report
Lenz microphthalmia syndrome. Am J Ophthalmol. and review. Am J Med Genet. 1992;43:780–4.
1988;105:40–5. 260. Verloes A, Mulliez N, Gonzales M, et al. Restrictive
243. Swanson SL, Santen RJ, Smith DW. Multiple dermopathy, a lethal form of arthrogryposis multiplex
lentigines syndrome: new findings of hypogonado- with skin and bone dysplasias: three new cases and
trophism, hyposmia, and unilateral renal agenesis. J review of the literature. Am J Med Genet.
Pediatr. 1971;78:1037–9. 1992;43:539–47.
244. Sbar GR, Venkataseshan VS, Huang Z, et al. Renal 261. Butler MG, Wadlington WB. Robinow syndrome:
disease in Marfan syndrome. Am J Nephrol. report of two patients and review of the literature.
1996;16:320–6. Clin Genet. 1987;31:77.
245. Biermann CW, Rutishauser G. Polycystic kidneys 262. Vennos EM, Collins M, James WD. Rothmund-
associated with Marfan syndrome in an adult. Scand Thomson syndrome: review of the world literature. J
J Urol Nephrol. 1994;28:295–6. Am Acad Dermatol. 1992;27:750–62.
246. Sharon A, Gillerot Y, Van Maldergem L, et al. The 263. Majewski F, Enders H, Ranke MB, et al. Serpentine
Marshall-Smith syndrome. Eur J Pediatr. 1990;150: fibula-polycystic kidney syndrome and Melnick-
54–5. Needles syndrome are different disorders. Eur J
247. Kawai M, Momoi T, Fujii T, et al. The syndrome of Pediatr. 1993;152:916–21.
Moebius sequence, peripheral neuropathy and 264. Murr MM, Waziri MH, Schelper RL, et al. Case of
hypogonadotropic hypogonadism. Am J Med Genet. multivertebral anomalies, cloacal dysgenesis, and
1990;37:578–82. other anomalies presenting prenatally as cystic kid-
248. Willems PJ, Colpaert C, Vaerenbergh M, neys. Am J Med Genet. 1992;42:761–5.
et al. Multiple pterygium syndrome with body asym- 265. Beighton P. Syndrome of the month. Spondyloepime-
metry. Am J Med Genet. 1993;47:106–11. taphyseal dysplasia with joint laxity (SEMWL). J
249. Roig M, Balliu PR, Navarro C, et al. Presentation, Med Genet. 1994;31:136–40.
clinical course and outcome of the congenital form of 266. Carter P, Burke JR, Searle J. Renal abnormalities and
myotonic dystrophy. Pediatr Neurol. 1994;11:208–13. spondylometaphyseal dysplasia. Aust Paediatr J.
250. Emery AEH, Oleesky S, Williams RT. Myotonic dys- 1985;21:115–17.
trophy and polycystic disease of the kidneys. J Med 267. Robinow M, Johnson GF, Broock W. Syndactyly type
Genet. 1967;4:26–8. V. Am J Med Genet. 1982;11:475–82.
251. Taalman RDHM, Hustinx TWJ, Weemaes CM, 268. Pankau R, Partsch CJ, Winter M, et al. Incidence and
et al. Further delineation of the Nijmegen breakage spectrum of renal abnormalities in Williams-Beuren
syndrome. Am J Med Genet. 1989;32:425–31. syndrome. Am J Med Genet. 1996;63:301–4.
252. Robin NH, Opitz JM, Muenke M. Opitz/GBBB syn- 269. Sandoval R, Sepulveda W, Gutierrez J, Be C, Altieri
drome: clinical comparisons of families linked to E. Prenatal diagnosis of nonmosaic trisomy 9 in a
Xp22 and 22q, and review of the literature. Am J fetus with severe renal disease. Gynecol Obstet
Med Genet. 1996;62:305–17. Invest. 1999;48:69–72.
178 C. Limwongse

270. Barakat AY, Butler MG. Renal and urinary tract 274. Kupferman JC, Stewart CL, Kaskel FJ, et al. Posterior
abnormalities associated with chromosome aberra- urethral valves in patients with Down syndrome.
tions. Int J Pediatr Nephrol. 1987;8:215–26. Pediatr Nephrol. 1996;10:143–6.
271. Jones KL. Smith’s recognizable patterns of human 275. VanBuggenhout GJ, Verbruggen J, Fryns JP. Renal
malformation. 6th ed. Philadelphia: WB Saunders; agenesis and trisomy 22: case report and review. Ann
2005. Genet. 1995;38:44–8.
272. Van Zelderen-Bhola SL, Breslau-Siderius EJ, 276. Goldberg R, Motzkin B, Scambler PJ, et al. Velo-
Beveratocks GC, et al. Prenatal and postnatal investi- cardio-facial syndrome: a review of 120 patients.
gation in a case of Miller-Dieker syndrome due to a Am J Med Genet. 1993;45:313–19.
familial cryptic translocation t(17;20) (p13.3;q13.3) 277. Greenberg F. DiGeorge syndrome: a historical review
detected by prenatal fluorescence in situ hybridiza- of clinical and cytogenetic features. J Med Genet.
tion. Prenat Diagn. 1997;17:173–9. 1993;30:803–6.
273. Gorlin RJ, Cohen MM, Hennekam RCM. Syndromes 278. Flynn MT, Ekstrom L, DeArce M, et al. Prevalence of
of the head and neck. 4th ed. New York: Oxford renal malformation in Turner syndrome. Pediatr
University Press; 2001. Nephrol. 1996;10:498–500.
Part II
Homeostasis
Physiology of the Developing Kidney:
Sodium and Water Homeostasis and Its 7
Disorders

Nigel Madden and Howard Trachtman

Contents Sodium Balance Disturbances: Deficit and


Excess . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182 Sodium Deficit . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
Body Fluid Compartments and Their Sodium Excess . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
Composition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182 Water Balance Disturbances: Deficit and
Total Body Water and Its Compartments . . . . . . . . . . . . 182 Excess . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
Composition of Body Water Compartments . . . . . . . . 183 Hyponatremia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207
Serum Osmolality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184 Hypernatremia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 210
Maintenance Sodium and Water References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 212
Requirements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184
Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 184
Water . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
Distinct Roles of Sodium and Water
in Body Fluid Homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . 187
Sensor Mechanisms: Sodium and Water . . . . . . . . . 189
Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 189
Water . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
Efferent Mechanisms: Sodium and Water . . . . . . . 191
Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 191
Water . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 196
Effector Mechanisms: Sodium and Water . . . . . . . . 197
Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 197
Water . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
Laboratory Assessment of Sodium and Water
Balance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
Water . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202
Overview of the Evaluation of Fluid and Water
Abnormalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202

N. Madden (*) • H. Trachtman (*)


NYU Langone Medical Center and NYU School of
Medicine, New York, NY, USA
e-mail: howard.trachtman@nyumc.org

# Springer-Verlag Berlin Heidelberg 2016 181


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_6
182 N. Madden and H. Trachtman

Introduction Total Body Water


1:2
Let there be work, bread, water, and salt for all.
Nelson Mandela ECW ICW

Salt and water are the stuff of life [1, 2]. The
ancient and the modern voices have been
invoked in the past to demonstrate that human
beings intuitively appreciate the critical role
played by sodium and water balance for the
integrity of the plasma compartment and for
continued existence on land. The third time
around, I turn to a towering contemporary figure Sodium sequestering, water
independent portion of the interstitial
of our age as inspiration for this chapter. The compartment

material will review the physiological mecha- IVS INT


nisms involved in the control of sodium and
water homeostasis. This knowledge will provide
a basis for the analysis of the diseases that arise 1:3

when these systems malfunction and a guide to Fig. 1 Graphic illustration of the total body water com-
the optimal therapy of conditions associated partments and the relative size of the intracellular water
with excessive or deficient total body sodium (ICW) to the extracellular water (ECW) spaces and intra-
and water. vascular (IVS) to interstitial water space

Total body water is divided into two principle


Body Fluid Compartments and Their components – the intracellular (ICW) and the
Composition extracellular water (ECW) spaces [4]. These
spaces are apportioned in a 2:1 ratio. When there
Total Body Water and Its is an increase in the total body water, this is
Compartments clinically manifested by an increase in the ECW
space because the ICW compartment is not acces-
Water is vital for the maintenance of life. It pro- sible to direct assessment. The ECW compartment
vides an aqueous environment for cytosolic chem- is further divided into the interstitial and intravas-
ical reactions; a solvent for elimination of waste cular spaces, which are separated in a 3:1 ratio.
products; a medium for transport of nutrients, key Thus, the intravascular space constitutes 1/12 of
molecules, and gases; and thermoregulation via the total body water, i.e., 1/3 X 1/4 (Fig. 1). A
sweat production [1]. On average, water com- component of the ECW, namely, the interstitial
prises 60 % of the total body weight in adults. fluid in the skin and connective, may serve as a
This proportion is higher in infants and even reservoir that can mobilize water into the plasma
greater in babies born prematurely and very low- volume to sustain circulation during conditions of
birth-weight neonates. It declines during early hypovolemia (see below [5]). Finally, there are
infancy and reaches the adult value by the end of separate transcellular water compartments, such
the first year [3]. The percentage of body weight as the gastrointestinal lumen or cerebrospinal
that is water is lower in postpubertal girls because fluid. They are not in direct contact with the
they have a higher percentage of body mass that is rest of the fluid spaces and are separated by an
fat. It is altered in disease states that are associated epithelial membrane. Water and electrolytes enter
with altered salt handling such as cystic fibrosis these spaces via tightly regulated active transport
and endocrinopathies. processes.
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 183

Composition of Body Water Besides distinctive membrane permeability


Compartments characteristics for specific solutes, the unequal
distribution of ions across the membrane is in
All of the major fluid compartments in the body part due to the Gibbs-Donnan effect, which arises
are separated by semipermeable membranes because of the presence of impermeant, nega-
[4, 5]. This type of barrier permits free passage tively charged proteins, primarily albumin, in the
of the aqueous solvent but limits movement of intravascular space [3].
selective solutes across the membrane. Water Recent findings underscore the importance of
moves down its concentration gradient to ensure nonvascular body compartments in the regulation
that the osmolality of the solution is the same on of total body sodium content and balance, inde-
both sides of the membrane. Water channels pendent of water, specifically the interstitial space
called aquaporins are selectively expressed in spe- in the skin and muscle. The skin interstitium has
cific cell membranes such as the erythrocytes and been identified as a storage site for sodium using
distinct nephron segments and facilitate water animal models and sodium-MRI measurements of
movement in response to an osmotic gradient sodium content in humans [11]. This storage
[6]. Upregulation of aquaporin-1 channels can occurs without compensatory water retention by
occur in disease states such as peritoneal dialysis, the kidneys. This implies that there is a need for
resulting in increased water permeability [7]. alternate mechanisms of electrolyte regulation in
Because of the presence of active transporters the skin (Fig. 1).
and selective channels for various solutes within Sodium storage in the skin is directly related to
the cell membrane, there is an uneven distribution the production of negatively charged, sulfated gly-
of solutes in the ICW and ECW compartments. cosaminoglycans (GAGs). These negatively
The presence of the Na-K ATPase pump in the charged molecules trap cations without water, pro-
basolateral membrane domain ensures that potas- ducing a hypertonic environment in the skin
sium and sodium are the principle cations in the interstitium relative to blood plasma. Macrophages
ICW and ECW spaces, respectively [8]. The sec- in the blood and tissue contain a tonicity response
ondary movement of Na+ and K+ through other element and sense this hypertonicity. They migrate
pathways such as the apical amiloride-sensitive to the skin interstitium where they release vascular
epithelial sodium channel (ENaC) or ROMK endothelial growth factor-C (VEGF-C). VEGF-C
channel is driven by the primary operation of the induces the hyperplasia of the cutaneous lymph
Na-K-ATPase [9]. Limitations in membrane per- capillary network and thus enhances the local clear-
meability to chloride and bicarbonate confine ance of skin electrolytes [12].
these anions almost exclusively to the ECW Disturbances in this extrarenal mechanism for
space, while proteins and phosphate comprise sodium storage can lead to salt-sensitive hyper-
the major intracellular anions. The differences in tension, emphasizing the role of this regulatory
cell permeability and binding characteristics of system in local hemostatic control of electrolyte
specific solutes are reflected in the coefficients composition. The importance of interstitial
that are used to determine their volume of distri- sodium content to body homeostasis is likely to
bution. For example, because of the permeability have expanding clinical relevance as methods are
of cell membranes to water, the volume of distri- developed to assay sodium in this compartment.
bution for sodium is equal to the entire body water Sodium-MRI studies have demonstrated that
compartment even though sodium is confined to hemodialysis can remove tissue sodium from
the ECW space. In contrast, the volume of distri- this compartment in the skin and muscle. Patient
bution of bicarbonate is 0.3  total body water. age and levels of circulating VEGF-C determined
These considerations are important in formulating the extent of removal during a standard hemodi-
therapeutic regimens to treat specific disorders of alysis session. Older individuals had more sodium
sodium and water homeostasis [10]. storage in the skin and muscle and lower levels of
184 N. Madden and H. Trachtman

VEGF-C. Additionally, patients with lower levels chemistry laboratories. If the calculated serum
of VEGF-C showed higher skin sodium storage osmolality is significantly lower than the value
after dialysis than those with higher VEGF-C obtained by measurement with an osmometer,
levels [13]. there is an “osmolal gap” and reflects the accumu-
lation of unmeasured osmoles. Clinically relevant
examples are the organic solutes that are produced
after an ingestion of ethanol or ethylene glycol
Serum Osmolality
(antifreeze) [14, 15].
Despite the significant differences in the compo-
sition of the various body water compartments,
Maintenance Sodium and Water
under equilibrium conditions, there is
Requirements
electroneutrality and tonicity or osmolality, i.e.,
the sum of all osmotically active particles, is equal
Sodium
in all body water compartments. Under normal
conditions, the serum osmolality is 286 
The total body sodium content in adults is approx-
4 mosm/kg water. Because sodium is the major
imately 80 mmol/kg of fat-free body weight. This
cation in the ECW, osmolality can be closely
proportion is higher in newborns, infants, and
estimated by the formula:
young children. Sodium is an essential dietary
Serum osmolality  2  ½serum sodium concentration component in newborns that is required for nor-
(1) mal growth, especially in very low-birth-weight
infants. In adults, 30–50 % of sodium is contained
A reflection coefficient of 1.0 indicates a totally in the skeleton. Because this proportion is smaller
non-permeant solute, while freely permeable mol- in infants, a positive sodium balance is required to
ecules have a reflection coefficient of zero. The build the skeleton and promote adequate growth
reflection coefficient for urea is approximately [16]. Sodium stimulates cell growth and prolifer-
0.4. Similarly, in the absence of insulin, the reflec- ation, possibly through the Na+ -H+ antiporter-
tion coefficient for glucose is 0.5. When there is a mediated alkalinization of the cell interior. It
pathological elevation in the serum urea nitrogen, causes protein synthesis by promoting nitrogen
e.g., acute kidney injury (AKI), or glucose con- retention, thereby increasing muscle mass, and is
centration, e.g., diabetic ketoacidosis, these sol- necessary for proper neural development
utes will contribute to osmolality, albeit less [17]. The growth-permissive role of sodium has
effectively than sodium. Therefore, the following been studied in animal models, which demon-
formula should be used to more accurately calcu- strate that deprivation of NaCl results in a reduc-
late serum osmolality: tion in weight, height, muscle mass, and brain
protein, RNA, and lipid content [18]. This effect
Serum osmolality ¼ is independent of the protein or calorie content of
the diet. Interestingly, in contrast to chronic potas-
2  ½serum sodium concentration
(2) sium deficiency induced by diuretics,
þ ½serum urea nitrogen=2:8 compromised sodium intake does not cause struc-
þ ½serum glucose concentration=18 tural damage to the kidney [19]. Although sodium
supplementation restored the rate of growth to
This formula is based on the molecular weights of normal and reversed the brain abnormalities, it
urea nitrogen (28 Da) and glucose (180 Da) and was not possible for these animals to catch up to
the standard practice of reporting the serum con- the healthy controls [20]. The impact of sodium
centrations as mg/100 ml. The calculated serum depletion has also been studied in children with
osmolality is normally within 1–2 % of the value salt-wasting renal diseases and in premature
obtained by direct osmometry in clinical infants, both of which are particularly vulnerable
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 185

Table 1 Mean daily sodium intake and recommended do or do not have CKD. Therefore, it is not pos-
amounts sible to define a daily sodium intake during child-
Recommended hood that will prevent cardiovascular and renal
Age group Actual sodium sodium intake disease in adulthood.
(years) intake (g/day) (g/day)
Under normal circumstances, the principal
1–5 4 2
anion that accompanies sodium is chloride. The
6–10 6 4
identity of the anion that accompanies sodium and
11–20 8 5
a variety of other dietary constituents impacts on
the adverse consequences of excessive sodium
to hyponatremia. In these cases, salt supplemen- intake such as hypertension [27]. In certain dis-
tation is necessary to promote normal growth and ease states such as renal tubular acidosis, meta-
cognitive development [21]. bolic acidosis associated with CKD, or
Balance studies indicate that the daily sodium urolithiasis, it may be advisable to provide a por-
requirement is 2–3 mmol/kg body weight. This tion of the daily sodium requirement as the bicar-
quantity is nearly two to threefold higher in term bonate or the citrate salt.
and very low-birth-weight premature infants
[22]. This reflects the immaturity in renal tubular
function coupled with the increased need for Water
sodium to achieve the high rate of growth during
early life. It will be exaggerated by intrinsic (diar- The daily requirement for water is traditionally
rhea, increased losses via chronic peritoneal dial- expressed as ml per metabolic kg [28]. However,
ysis, genetic defect in tubular sodium transport) or in clinical practice, this is a very cumbersome and
exogenous (administration of diuretics) factors impractical method and all calculations are based
that promote sodium loss. In most developed on body weight or body surface area (BSA). There
countries, the daily sodium intake in childhood are three methods that are currently utilized to
is in excess of the amount needed to promote estimate the daily fluid requirement. The first is a
growth or maintain body function. Table 1 sum- direct extension of the use of metabolic kilogram
marizes the difference between actual sodium and utilizes the following formula:
intake and current recommendations in young
children and adolescents. 1. Daily water requirement = 100 ml/kg for a
There has been extensive discussion about the child weighing less than 10 kg + 50 ml/kg for
daily sodium intake that yields optimal health in each additional kg up to 20 kg + 20 ml/kg for
adults. An emerging consensus is that a daily salt each kg in excess of 20 kg
intake in the range of 3–6 g is desirable to prevent The second method is based on BSA and
hypertension and minimize cardiovascular mor- utilizes the following formula:
bidity and mortality [23–25]. A J curve, with 2. Daily water requirement = 1500 ml/m2 BSA
extremely low or high sodium intake leading to The last method is a refinement of the sec-
worsening cardiovascular outcomes, appears to ond and utilizes the following formula:
define the relationship in adults. This recommen- 3. Daily water requirement = Urine output +
dation is based on large international cohort stud- insensible water losses
ies with extended follow-up, accurate assessment
of sodium intake, and detailed characterization of Based on clinical experience, under normal
the clinical outcomes. It is worth noting that even circumstances, urine output is approximately
in adults with nondiabetic chronic kidney disease 1,000 ml/m2 per day and insensible losses amount
(CKD), reducing daily salt intake to below the to 500 ml/m2 per day.
3–6 g range is not associated with improved Example: For a child weighing 30 kg and
renal or cardiovascular outcomes [26]. There are 123 cm in height with a BSA of 1.0 m2, according
no comparable studies in pediatric patients who to the first method, the daily water requirement is
186 N. Madden and H. Trachtman

1,700 ml, while the second method yields of sensible and insensible water loss. Addition-
1,500 ml per day. The first method is easier to ally, acutely ill children often present with
apply, but it tends to overestimate the water overproduction of AVP, which causes free water
requirement as body weight increases. The third retention and renders the patient vulnerable to
method is the most precise and should be applied hyponatremia [37]. Adding hypotonic solution in
in more complicated circumstances such as the the setting of inappropriate AVP secretion may
patient in the intensive care unit with oliguria place the child at even greater risk of serious
secondary to AKI or the child with increased complications of low serum sodium including
insensible losses, e.g., diarrhea, increased ambient headache, nausea, vomiting, muscle cramps,
temperature, tachypnea, burns, or cystic fibrosis depressed reflexes, disorientation, seizures, respi-
[29]. In addition to the daily energy requirement ratory arrest, and cerebral edema, which can lead
and insensible losses that are represented in the to permanent brain damage and death [38]. Chil-
formulas, the amount of water excreted by dren are at even greater risk of developing neuro-
the kidney on a daily basis is dependent upon logical symptoms from hyponatremia because the
the solute load. Because urine has a minimum size of their brain relative to their skull is greater
osmolality, approximately 50 mosm/kg H2O, than adults. Opponents of the use of hypotonic
even in the absence of arginine vasopressin maintenance fluids have documented the occur-
(AVP), increased dietary intake of solute will rence of hyponatremia and neurological compli-
result in a larger obligatory urine volume to cations in hospitalized children who receive these
accommodate the larger solute load [30, 31]. fluids parenterally [37, 39]. To prevent cerebral
Provision of adequate water intake is espe- edema and neurological consequences of
cially important in select medical conditions hyponatremia, they advocate routine administra-
such as urolithiasis and CKD where higher water tion of isotonic saline (0.9 % NaCl), with or with-
intake can prevent disease recurrence or progres- out dextrose, to all pediatric patients who require
sion, respectively [32]. intravenous maintenance fluids [40]. Moritz and
The daily sodium and water requirement are Ayus report more than 50 cases of neurologic
generally provided enterally. Intravenous admin- damage and death from 1993 to 2003 as a result
istration of fluids and electrolytes should be of iatrogenic hyponatremia due to administration
resorted to only under clinical circumstances that of hypotonic maintenance fluid [37]. In large ran-
interfere with normal feeding such a persistent domized control trials of critically ill and postop-
vomiting, gastrointestinal tract surgery, or states erative pediatric patients, Choong et al. [41, 42]
of altered consciousness. support this view by demonstrating that the use of
The choice of parenteral fluid for maintenance hypotonic parenteral maintenance solution results
therapy in children has long been debated in the in significantly increased risk of hyponatremia as
literature and by clinicians. Holliday and Segar compared to isotonic fluids.
developed early guidelines based on a linkage In a meta-analysis of randomized control trials,
between metabolic needs and fluid requirements hypotonic maintenance fluids were associated
in healthy children. They suggested that a hypo- with an increased risk of hyponatremia in ICU
tonic maintenance solution containing 0.2 % and postoperative patients [43]. Opponents to iso-
saline in 5 % dextrose water should be adminis- tonic saline point to the risk of hypernatremia with
tered to sick children [33]. This guideline has been isotonic solutions, yet results from Choong
repeated in several recent reports [34–36]. Since et al. suggest that isotonic maintenance fluids do
then, many nephrologists have criticized this rec- not result in increased risk for hypernatremia
ommendation claiming that the Holliday and [41, 42]. Several groups in the UK, Canada, and
Segar guidelines were developed based on the the USA, including the Institute for Safe Medica-
study of healthy children. They assert that it does tion Practices, have warned against the use of
not apply to children with acute illness who often hypotonic saline in the pediatric population. This
have reduced energy expenditure and lower levels issue will require well-designed interventional
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 187

Fig. 2 This scheme Clinical Disorders of


illustrates the importance of Sodium/Water Homeostasis
independent assessment of
sodium and water handling
in managing patients with Sodium Disturbance Water Disturbance
clinical disorders of
sodium-water homeostasis
Altered Sodium Balance Altered Water Distrubution

Abnormal ECF Volume Abnormal ICW/ECW Ratio

Clinical Assesment Laboratory Testing

Integrate Evaluation

studies to supplement practice guidelines [44]. The regulation of sodium and water homeostasis
Finally, it is worth noting that in a prospective, represents two distinct processes with discrete
open-label pilot study in 760 adults, implementa- sensing and effector mechanisms. Although
tion of a strategy to restrict chloride content of these systems overlap, from a physiological and
intravenous maintenance fluids resulted in a clinical perspective, a complete understanding of
reduced incidence of AKI and the need for renal body fluids and electrolytes mandates separate
replacement therapy [45]. The impact of chloride- evaluation of sodium and water (Fig. 2).
containing solutions on kidney function in pedi- Because sodium is the principal cation in the
atric patients has not been studied. In the interim, ECW compartment, disturbances in total body
it is important to emphasize that in the face of sodium content are reflected by expansion or con-
clinically significant acute ECW contraction, traction of this space. Adequacy of the ECW
there is universal agreement that isotonic fluids compartment is essential to maintain the intravas-
are necessary to effectively replete the intravascu- cular space and sustain perfusion of vital organs.
lar compartment. Careful clinical and laboratory In terrestrial mammals living in an environment
monitoring is key to ensuring good outcomes in where ECW volume depletion is a constant threat,
all children who are given maintenance fluids and the kidney is designed for maximal sodium
electrolytes parenterally. reabsorption as the default mode unless physio-
logical signals instruct it to respond otherwise.
The primary step in the pathogenesis of distur-
Distinct Roles of Sodium and Water bances in ECW compartment size is a perturba-
in Body Fluid Homeostasis tion in sodium balance. When total body water
and sodium content are within the normal range,
Sodium and water are inextricably linked in the the net sodium balance is zero and the daily intake
determination of the serum sodium concentration. of sodium is matched by losses in the urine, stool,
However, it is critical to recognize that sodium and insensible losses. Provided kidney function is
and water serve two distinct functions within the normal, the daily dietary sodium intake can be as
body. Sodium is instrumental in the maintenance low as 0.1 mmol/kg or in excess of 10 mmol/kg
of the size of the ECF space and the vascular without any derangement in ECF compartment
perfusion compartment, while water is critical to size. If the alterations in diet are not abrupt, then
the maintenance of the size of individual cells. sodium balance is maintained, even when kidney
188 N. Madden and H. Trachtman

Table 2 Clinical diseases of sodium and water homeosta- deuterium dilution [48]. Fluid overload often indi-
sis: relationship between ECW size and tonicity cates worsening disease severity in many clinical
Tonicity conditions including CKD, heart failure, and liver
ECW volume Low Normal High cirrhosis. As such, a reliable and efficient tool for
Low Addison’s Isotonic Hypertonic measuring volume status is important in the man-
disease diarrheal diarrheal
agement of these diseases [47]. Measurement of
Salmonella Dehydration Dehydration
diarrhea ECW fluid content is particularly important in
Mannitol Diabetes patients undergoing dialysis. Blood pressure reg-
infusion insipidus ulation in these individuals has proven difficult
Normal SIADH No disease Acute and abnormalities, including both hypotension
sodium and hypertension, are common. Bioimpedance
bicarbonate
infusion
has been established as a useful tool in monitoring
High Acute renal Nephrotic Salt
the body fluid content of patients on dialysis and
failure syndrome intoxication yields important information about their volume
Nephrotic Salt-water status, which can be used to reduce the cardiovas-
syndrome drowning cular risks associated with chronic renal replace-
Cirrhosis ment therapy [49].
Congestive Water homeostasis is a prerequisite for the nor-
heart
mal distribution of fluid between the ICW and
Failure
ECW compartments. Cell function is dependent
on stabilization of cell volume in order to keep the
cytosolic concentration of enzymes, cofactors,
function is markedly impaired [46]. In contrast, if and ions at the appropriate level and to prevent
the daily input of sodium exceeds losses, there is molecular crowding. Perturbations in water bal-
expansion of the ECF space with edema, while if ance result in fluctuations in serum osmolality.
the input does not match the daily losses, there Because cell membranes permit free movement
will be symptoms and signs related to ECW space of water down its osmolal gradient, this causes
contraction. These disturbances are not associated obligatory shifts in water between the cell and
with any obligatory parallel changes in the serum the ECW space. Any disorder that alters the 2:1
sodium concentration (Table 2). ratio of water volume in the ICW/ECW spaces
The determination of ECW compartment size will be reflected by changes in cell size and sub-
and adequacy has generally been based on a clin- sequent cellular dysfunction. Under hypoosmolal
ical assessment (see below). Recent advances conditions, water will move from the intravascular
have highlighted new technological methods to compartment into the cell, causing relative or
measure this space that may facilitate this evalua- absolute cell volume expansion. Conversely, if
tion and provide greater accuracy and validity. the serum osmolality is elevated, water will exit
Bioelectrical impedance is an emerging technique from the cell to the ECW space resulting in abso-
for the measurement of body fluid content. It is an lute or relative cellular contraction [50].
easy, affordable, and noninvasive method and Disturbances in cell function related to abnor-
potentially has great utility in many clinical malities in cell size are most prominent in cerebral
areas. This tool applies an electrical current of cells for two reasons. First, the blood-brain barrier
various frequencies to measure the reactance and limits the movement of solute between the ICWand
resistance of the space between electrodes placed ECW compartments while permitting unrestricted
on the skin. The reactance value is proportional to flow of water down an osmolal gradient [51]. Sec-
body mass and the resistance is inversely related ond, the brain is contained within the skull, which is
to total body water (TBW) [47]. The correlation a closed, noncompliant space, and is tethered to the
between bioimpedance analysis and TBW is cranial vault by bridging blood vessels, which
strong as assessed by measurements made by limits its tolerance of cell swelling or contraction.
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 189

Thus, alterations in water balance and serum osmo- perceived as an obligatory expansion of the ECW
lality are dominated by clinical findings of central space. These receptors in the venous (low-
nervous system dysfunction, including lethargy, pressure) and arterial (high-pressure) circulation,
seizures, and coma [50]. which are influenced by the filling pressure within
In the same way that disturbances in sodium the circulation, are called baroreceptors or mech-
balance do not necessarily predict specific abnor- anoreceptors. These signals are supplemented in
malities in serum sodium concentration, the pres- certain instances by chemoreceptors that respond
ence of a disturbance in water balance and serum directly to changes in the serum sodium concen-
osmolality is not linked to a specific abnormality in tration and trigger adaptive modifications in renal
the ECW compartment size. The independent sodium handling. These receptors may effect
nature of disturbances in sodium and water balance change by altering nervous system activity or by
is illustrated in Table 2. Alterations in ECW size activating upstream promoter elements and stim-
can occur in patients with hypotonicity, isotonicity, ulating the expression of relevant genes [53].
or hypertonicity. Similarly, each alteration in serum Atrial receptors: Within the right atrium, sen-
osmolality can develop in patients with contraction sors possess the distensibility and compliance
or expansion of the ECW compartment. needed to detect alterations in intrathoracic
The presence of disturbances in sodium and blood volume provoked by increasing negative
water homeostasis must be addressed separately intrathoracic pressure or head-out water immer-
in the clinical evaluation of patients with derange- sion. Both of these maneuvers, which increase the
ments in ECF volume or tonicity. This assessment central blood volume and raise central venous and
must be integrated to obtain a comprehensive right atrial pressures, are followed by a brisk
view of what is abnormal and determine how to natriuresis and diuresis [54]. These relative
effectively restore sodium and water homeostasis changes are triggered even in the absence of a
with minimal side effects (Fig. 2). concomitant change in the total ECW space size.
Neural receptors that respond to mechanical
stretch or changes in right or left atrial pressure
Sensor Mechanisms: Sodium convey the signal via the vagus nerve [54, 55].
and Water Hepatic receptors: The enhanced renal sodium
excretion triggered by saline infusions directly into
For both sodium and water homeostasis, the sen- the hepatic vein versus the systemic circulation
sor mechanisms that maintain the equilibrium suggests that there are low-pressure sensors within
state are primarily designed to be responsive to the portal vein or hepatic vasculature. The hepatic
the consequences of abnormalities in sodium or responses to changes in sodium balance have been
water balance, i.e., changes in ECW and cell size, divided into two categories [56]. The “hepatorenal
respectively, rather than measuring the primary reflex” involves direct activation of sodium chemo-
variable. In this regard, they differ from the acid- receptors and mechanoreceptors in the hepatoportal
base sensor that is directly responsive to changes region, via the hepatic nerve, and causes a reflex
in pH [52]. They operate using negative feedback decrease in renal nerve activity. The “hepatoin-
loops in which deviations from normal are testinal reflex” utilizes chemoreceptors to respond
detected, counter-regulatory mechanisms are acti- to changes in sodium concentration and modulate
vated that antagonize the initiating event, and the intestinal absorption of sodium via signals con-
system is restored to the normal state. veyed along the vagus nerve. Activation of these
hepatic volume sensors may contribute to the
sodium retention and edema states that develop
Sodium secondary to chronic liver disease and cirrhosis
with the associated intrahepatic hypertension.
The net sodium deficit is detected as a decrease in Pulmonary receptors: There may also be pres-
ECW space size, while the net sodium excess is sure sensors within the pulmonary circulation that
190 N. Madden and H. Trachtman

are activated by changes in pulmonary perfusion perceived underfilling of the arterial tree may
or mean airway pressure [57]. The receptors in the occur despite significant venous distention [40].
lung may be located in the interstitial spaces and Similarly, women who develop edema during
influence the physical forces that modulate pregnancy may have primary peripheral vasodila-
paracellular absorption of sodium and water. tation and excess total body sodium [62, 63].
They resemble receptors in the renal interstitium
that also influence paracellular absorption of fluid
and solutes along the nephron, especially in the Water
proximal tubule segment [58].
Carotid arch receptors: There are also volume- The receptors that are responsible for regulating
dependent sensors on the high-pressure arterial water homeostasis are primarily osmoreceptors
side of the circulation including the carotid arch, and are sensitive to alterations in cell size
the brain, and the renal circulation. Thus, occlu- [63, 64]. These osmoresponsive cells are located
sion of the carotid leads to increased sympathetic in the circumventricular organs and anterolateral
nervous system activity and alterations in renal regions of the hypothalamus, adjacent to but dis-
sodium handling [55]. The responsiveness of the tinct from the supraoptic nuclei. They shrink or
carotid arch receptors may be modulated by swell in response to increases or decreases in
chronic changes in ECF volume. For example, a plasma tonicity and this change in cell size trig-
head-down bed position and a high salt diet blunt gers the release of AVP and/or the sensation of
carotid baroreceptor activity [59]. thirst. The anatomic configuration of these cells
Cerebral receptors: Increases in the sodium enables them to be exposed to circulating peptides
concentration of the CSF or brain arterial plasma that are involved in water homeostasis.
promote renal sodium excretion [60]. Lesions in AVP: AVP is a peptide containing nine amino
discrete anatomic areas of the brain such as the acids and has a molecular weight of 1,099 Da. It is
anteroventral third ventricle alter renal sodium synthesized by magnocellular neurons in the
reabsorption, confirming that there are central hypothalamus, transported down the axon, and
mechanisms of sensing changes in sodium bal- stored in the posterior pituitary in conjunction
ance and ECF volume. Derangements in the sens- with larger proteins, called neurophysins
ing system within the brain in patients with long- [65]. The gene for AVP is located on chromosome
standing central nervous system diseases may 20 and has a cAMP response element in the pro-
contribute to the cerebral salt-wasting syndrome. moter region. Prolonged stimulation of AVP
Intracerebral expression of angiotensin- release leads to upregulation of the AVP gene;
converting enzyme isoforms contributes to however, the synthesis does not keep up with the
peripheral sodium and water handling [61]. need for the peptide because pituitary levels of
If arterial sensors perceive underfilling of the AVP are usually depleted in states such as chronic
vascular space, this activates counter-regulatory salt loading and hypernatremia [66].
mechanisms to restore the ECW compartment size The principle solute that provokes the release
even if the receptors in the venous system detect of AVP is sodium. Infusion of sodium chloride to
adequate or even overfilling of the venous tree. increase plasma osmolality results in increased
This implies that despite normal or even excess secretion of AVP in the absence of parallel
total body sodium and net positive sodium bal- changes in ECW volume. This underscores the
ance, there are conditions in which the body per- primary role of plasma osmolality per se in stim-
ceives an inadequate circulating plasma volume. ulating AVP release [64]. Mannitol, an exogenous
This has given rise to the notion of the “effective” solute that is used in clinical practice to treat
intravascular volume, a concept that is applicable increased intracranial pressure, is nearly as effec-
in the edema states such as congestive heart fail- tive as sodium in stimulating AVP release. Urea
ure, cirrhosis, and nephrotic syndrome [62]. For and glucose are less than 50 % as effective as
example, in patients with cardiac pump failure, sodium in provoking AVP secretion because
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 191

they are more permeable than sodium and cause Table 3 Factors that increase AVP release
less pronounced changes in osmoreceptor cell "Plasma osmolality
volume. However, in disease states such as AKI Hemodynamic
or diabetic ketoacidosis, in which urea or glucose, #Blood volume
respectively, acts as osmotically active molecules #Blood pressure
or following the exogenous administration of Emesis
mannitol, these solutes also stimulate increases Hypoglycemia
in AVP release. There is coupling between Stress
mechanical changes in membrane structure and Elevated body temperature
hormone release. However, the exact mechanism Angiotensin II
and the neurotransmitters that mediate the actions Hypoxia
Hypercapnia
of the osmoreceptors on the cells of the posterior
Drugs
pituitary have not been identified.
A variety of non-osmotic stimuli to AVP
release may contribute to water handling in vari- are generally the same as those for AVP release,
ous disease states [64, 65]. Vomiting and acute with hypernatremia being the most potent trigger.
hypoglycemia promote AVP release by neural- The osmotic threshold for thirst in humans
hormonal pathways that are not well defined. appears to be higher than for AVP secretion,
Stress associated with pain or emotional anxiety, namely, 295 mosm/kg. The sensing mechanism
physical exertion, high body temperature, acute that leads to this increase in water intake is even
hypoxia, and acute hypercapnia are other condi- more obscure than that for AVP release. It is likely
tions that lead to increased secretion of AVP in the that changes in ECW volume are also involved in
absence of a primary disturbance in water balance. this process because angiotensin II, which rises in
Numerous drugs directly influence the hypotha- states of ECW volume contraction, is a potent
lamic release of AVP including carbamazepine, dipsogen [68]. Recent studies suggest that the
cyclophosphamide, and vincristine. Finally, day-to-day regulation of thirst by osmoreceptors
hemodynamic changes arising from primary alter- is under the control of dopamine-mu opioid neu-
ations in sodium balance and the ECW space can rotransmitters in the brain, while angiotensin II
trigger AVP release. If the ECW volume distur- may be activated under more stressful conditions.
bance is mild, then the stimulation of AVP release
is modest. However, in the face of severe ECW
volume contraction, there is marked secretion of Efferent Mechanisms: Sodium
AVP. Under these circumstances, the imperative and Water
to protect the effective circulating blood volume
takes precedence over the need to maintain The efferent mechanisms involved in maintaining
plasma osmolality and ECW volume is restored sodium and water balance include the neural and
at the expense of hypoosmolality. This clinical endocrine-humoral systems. There often is an
observation indicates the intellectual attempts to overlap in the action of these effectors, with an
separate sodium and water homeostatic mecha- individual effector having distinctive effects on
nisms; these two factors are closely linked both sodium and water balance.
in vivo and there can be significant overlap in
sensor and effector mechanisms in the regulation
of ECW and ICW compartment size. Table 3 sum- Sodium
marizes the factors that modulate AVP release.
In addition to AVP release, the osmoreceptor Renin-angiotensin-aldosterone axis: The major
cells also respond to the changes in serum osmo- components of this system – renin,
lality in an independent manner to stimulate thirst angiotensinogen, and angiotensin-converting
and increase drinking [67]. The stimuli for thirst enzyme (ACE) – are found within the kidney
192 N. Madden and H. Trachtman

and the vasculature of most organs. These ele- increased filtration of liver-derived
ments are linked in a large feedback loop involv- angiotensinogen rather than conversion of
ing the liver, kidney, and lung as well as smaller angiotensinogen produced within the kidney [73].
loops within individual organs. This accounts for Aldosterone: The effects of aldosterone on the
the often disparate data about plasma renin activ- renal tubule include an immediate effect to
ity (PRA) and the expression of individual com- increase apical membrane permeability to sodium
ponents within the kidney during disturbances in and more extended effects that involve enhanced
ECW compartment size. gene transcription and de novo synthesis of Na-K-
Angiotensin II is the major signal generated by ATPase. Aldosterone stimulates the synthesis of
this axis [69]. There are two distinct forms of ACE other enzymes involved in renal cell bioenergetics
and the ACE2 isoform may metabolize angioten- such as citrate synthase that are needed to sustain
sin II to non-pressor breakdown products that maximal tubular sodium transport [74]. Aldoste-
react with specific receptors and that are less likely rone induces a state of glucose-6-phosphate dehy-
to promote the development of hypertension drogenase deficiency in endothelial cells which
[70]. This introduces another layer of complexity may contribute to oxidant stress and altered reac-
in the regulation of sodium balance by the renin- tivity of blood vessels in response to disturbances
angiotensin axis. Angiotensin II interacts with two in sodium balance [75]. It is important to note that
different receptors, and most of its biological increased salt intake can activate the mineralocor-
activity is mediated by the angiotensin type ticoid receptor via a pathway that is independent
1 (AT1) receptor. The AT2 receptor is more prom- of aldosterone. In salt-sensitive animal strains,
inently expressed in the fetal kidney; however, high dietary salt intake activates Rac1, a member
interaction of angiotensin II with the AT2 receptor of the Rho-guanine triphosphate hydroxylase
postnatally stimulates the release of molecules family, which stimulates activity of the mineralo-
such as nitric oxide that counteract the primary corticoid receptor. This may represent a novel
action of the peptide [71]. In addition, angiotensin pathway by which high salt intake promotes
I can be processed to the heptapeptide angiotensin hypertension, vascular injury, and cardiovascular
1-7, which interacts with a separate mas receptor disease [76].
and modulates the biological effects of angioten- Endothelin: This vasoactive molecule is part of
sin II [70]. More research is needed to elucidate a family of three peptides of which endothelin-1
the role of alternate forms of angiotensin such as (ET-1) is the most important in humans [77]. It is
angiotensin 1-7 on sodium and water balance in converted in two steps from an inactive precursor
children. to a biologically active 21-amino acid peptide.
The best-known effects of angiotensin II Endothelins react with two receptors, ETA and
include peripheral vasoconstriction to preserve ETB, and cause vasoconstriction, resulting in a
organ perfusion and stimulation of adrenal syn- decrease in renal blood flow and GFR. With
thesis of aldosterone to enhance renal sodium regard to sodium balance, the primary effect of
reabsorption. Angiotensin II also has direct endothelin is sodium retention mediated by the
actions on tubular function and stimulates both reduction in GFR. This suggests that endothelin
proximal and distal sodium reabsorption. The acts in concert with angiotensin II to protect ECW
proximal tubule cells contain all of the elements compartment size under conditions of sodium
needed to synthesize angiotensin II locally and the deficit. However, the situation may be more com-
peptide increases the activity of the sodium- plicated because direct exposure of proximal
hydrogen exchanger [72]. In the distal tubule, tubule and medullary collecting duct cells to
angiotensin II modulates this exchanger as well endothelin in vitro inhibits sodium absorption.
as the amiloride-sensitive sodium channel [69]. It Renal nerves: There is abundant sympathetic
is worth noting that in the context of glomerular nervous innervation of the renal vasculature and
disease and podocyte injury, the major source of all tubular segments of the nephron [78]. The
increased renal angiotensin II is derived from efferent autonomic fibers are postganglionic and
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 193

originate in splanchnic nerves. The renal innerva- Atrial natriuretic peptide (ANP): ANP is a
tion is primarily adrenergic and involves α1 28-amino acid peptide that is a member of a
adrenoreceptors on blood vessels and both α1 group of proteins that includes C-type natriuretic
and α2 receptors along the basolateral membrane peptide [83]. It is synthesized as a prohormone
of the proximal tubule. Renal sympathetic ner- that is stored in granules in the cardiac atria. There
vous activity is inversely proportional to dietary are other molecular isoforms of the hormone
salt intake [78]. Renal sympathetic nervous sys- including brain natriuretic peptide (BNP) whose
tem activity contributes to preservation of ECF circulating levels are altered and which can be
volume by [1] promoting renal vasoconstriction monitored at diagnosis and in response to treat-
and lowering GFR and [2] increasing sodium ment in conditions such as congestive heart
reabsorption. Among the catecholamines failure [84].
involved in adrenergic transmission, norepineph- Increases in right atrial pressure provoke cleav-
rine exerts an antinatriuretic effect. Dopamine, age and release of the mature peptide. For each
another sympathetic nervous system neurotrans- 1 mmHg rise in central venous pressure, there is a
mitter, promotes a natriuresis, suggesting that corresponding 10–15 pmol/L increase in circulat-
there is internal regulation of the effect of nerve ing ANP levels. Conversely, declines in atrial
activation on renal sodium handling [79]. Some of pressure secondary to sodium depletion or hem-
the genes involved in dopamine metabolism orrhage inhibit ANP release. There are two recep-
including dopamine receptors and their regula- tors for ANP and both are coupled to guanylate
tors, G-protein-coupled receptor kinase 4 in the cyclase. The activation of this enzyme results in
proximal tubule cell, may contribute to salt sensi- cytosolic accumulation of cGMP, which in turn
tivity in patients with hypertension and the sus- diminishes agonist-stimulated increases in intra-
ceptibility to CKD [80]. cellular calcium concentration. The principle
The sympathetic nervous system also influ- effects of ANP are to promote an increase in
ences sodium handling in the distal nephron. In GFR, diuresis, and, most importantly, natriuresis.
animal models of salt-sensitive hypertension, acti- The augmented renal sodium excretion is, in part,
vation of the sympathetic nervous system leads to mediated by an increased filtered load secondary
reduced expression of WNK4 (see below). Alter- to the rise in GFR. However, ANP also exerts
ation in this regulatory protein results in enhanced direct actions on renal tubular cells to diminish
activity of the sodium-chloride co-transporter sodium reabsorption including inhibition the Na-
in the distal convoluted tubule leading to K-Cl co-transporter in the loop of Henle and the
hypertension [76]. amiloride-sensitive ENaC in the medullary
Drug-induced sodium retention and volume- collecting duct. Finally, ANP antagonizes the
dependent hypertension, e.g., with the use of action of several antinatriuretic effectors, includ-
cyclosporine, is mediated in part by activation of ing sympathetic nervous system activity, angio-
the sympathetic nervous system [81]. Increased tensin II, and endothelin. The overall effects of
adrenergic nervous signaling within the kidney is ANP to counteract increases in ECW compart-
instrumental in the initiation of hypertension in ment have been demonstrated by short-term stud-
experimental animals by causing a right shift ies in which acute infusions of ANP improved
in the pressure-natriuresis curve [79]. Alterations cardiac status in patients with congestive heart
in WNK4 activity may be operative in calcineurin failure and promoted a diuresis in patients with
inhibitor-induced hypertension [82]. However, acute renal failure [85].
sodium balance is normal and ECF volume is Prostaglandins: The kidney contains the
maintained in the denervated transplanted kidney, enzymes required for constitutive (COX-1) and
implying that the role of the sympathetic nervous inducible (COX-2) cyclooxygenase activity that
system in maintaining sodium homeostasis is convert arachidonic acid to prostaglandins
redundant and can be taken over by other regula- [86]. The major products of these pathways are
tory mechanisms [78]. PGE2, PGF2α, PGD2, prostacyclin (PGI2), and
194 N. Madden and H. Trachtman

thromboxane (TXA2). In the cortical regions, and causes vasodilatation and an increase in GFR.
PGE2 and PGI2 predominate, while PGE2 is the In addition to its effect on renal blood flow and
major prostaglandin metabolite in the medulla. GFR, NO directly inhibits Na-K-ATPase in cul-
These two compounds increase GFR and promote tured proximal tubule and collecting duct cells
increased urinary sodium excretion. In addition, [89, 90]. The specific isoform of NOS that is
they antagonize the action of AVP. These actions responsible for modulating urinary sodium excre-
may mediate the adverse effects of hypercalcemia tion is not as well defined. Studies with inducible
and hypokalemia on renal tubular function NOS, neuronal NOS, and endothelial NOS
[86]. The natriuretic effects of prostaglandins, in knockout mice suggest that only the first two
response to normal alterations in dietary sodium isoforms are involved in the regulation of sodium
intake, are unclear. The role of prostaglandins as and water reabsorption in the proximal tubule
efferent signals is more apparent in conditions [64]. A role of NO in maintaining sodium balance
associated with increased vasoconstrictor tone under normal conditions is suggested by the
such as congestive heart failure or reduced renal observation that alterations in dietary salt intake
perfusion, where prostaglandins counteract the are associated with parallel changes in urinary
vasoconstrictor and sodium-retaining effects of excretion of nitrite, the metabolic byproduct of
high circulating levels of angiotensin II and nor- NO [91]. In normotensive Wistar-Kyoto rats and
epinephrine. Inhibition of prostaglandins with spontaneously hypertensive rats, increased die-
cyclooxygenase inhibitors is associated with dra- tary sodium intake is associated with a modest
matic declines in GFR and profound sodium increase in urinary nitrite excretion [92]. This
retention and edema [87]. effect is not well documented in pediatric patients.
Prostaglandins also modulate sodium handing Along with ANP and bradykinin, NO is part of the
in the collecting tubule. Inactivation of the B1 defense system against sodium excess and expan-
proton pump in β-intercalated cells results in sion of the ECW compartment. Derangements in
increased urinary losses of NaCl, potassium, and renal NO synthesis and responsiveness to cGMP
water leading to hypovolemia, hypokalemia, and may be instrumental in the pathogenesis of salt-
polyuria. Inhibition of PGE2 synthesis restores dependent hypertension in experimental animals
activity of the ENaC and reverses the electrolyte [92]. There are no drugs in current use that mod-
abnormalities [88]. These novel findings under- ulate sodium handling based on alteration of
score the importance of prostaglandins in the reg- intrarenal NO production.
ulation of sodium homeostasis. Moreover, they Kinins: Kinins are produced within the kidney
indicate that in the collecting duct, both principal and act via B1 and B2 receptors. Because the half-
and intercalated cells are instrumental in life of kinins in the plasma is very short, in the
maintaining sodium balance. range of 20–40 s, it is likely that their actions in
Nitric oxide (NO): The kidney contains all the kidney are regulated locally through produc-
three isoforms of nitric oxide synthase (NOS) – tion and proteolytic processing in the tissue
neuronal NOS in the macula densa, inducible [93]. Their principal action is to promote renal
NOS in renal tubules and mesangial cells, and vasodilatation and natriuresis. The kinins act pri-
endothelial NOS in the renal vasculature – marily in the distal tubule to reduce sodium
involved in NO synthesis. The neuronal and endo- reabsorption [93, 94].
thelial isoforms are calcium-dependent enzymes Insulin: Insulin promotes sodium reabsorption
and produce small, transient increases in NO syn- in the proximal and distal segments of the neph-
thesis. The inducible isoform is upregulated by ron. It has direct effects on the Na-H antiporter in
various cytokines and inflammatory mediators, the proximal convoluted tubule [95] and on the
resulting in large sustained elevations in NO NaCl co-transporter in the distal tubule [96]. Part
release. of the effect of insulin may be mediated by mod-
Activation of eNOS within the kidney ulation of the sympathetic nervous system and
increases the activity of soluble guanylate cyclase WNK kinase activity [97]. The antinatriuretic
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 195

Fig. 3 Graphic illustration


Disturbance in sodium
summarizing sensor and balance
effector mechanisms Clinical assessment
involved in the regulation of
sodium balance and ECF
volume. ANP atrial
natriuretic peptide, NO Volume sensing
nitric oxide, PG mechanisms
prostaglandins Arterial and venous

EFFECTOR SYSTEMS

Renin- Circulating mediators


Sympathetic
Angiotensin- Endothelin, insulin,
nervous system
Aldosterone Axis ANP, NO, PG, kinins

Restoration of sodium balance


Resolution of hypovolemia or
edema

effect of insulin contributes to the hypertension increased synthesis of NO. The resultant natriure-
observed in children with hyperinsulinemia and sis secondary to the increase in GFR is accompa-
the metabolic syndrome. In the context of insulin nied by direct inhibition of tubular sodium
and sodium, it is worth noting that a new class of reabsorption. Its role in sodium balance is under
drugs that have been introduced for the treatment investigation.
of diabetes, namely, sodium-glucose The WNK, with no lysine protein kinase fam-
co-transporter inhibitors, is associated with ily, are serine threonine kinases. They are respon-
increased natriuresis and a modest reduction in sible for the regulation of several membrane
blood pressure [98]. proteins in the kidney that mediate Na+ handling
FGF23: Recent data suggest that FGF23, an in the distal tubule. This family senses intracellu-
important bone-derived hormone involved in lar and extracellular ion concentrations and
phosphate homeostasis, may contribute to sodium accordingly alters the activity of channels and
balance [99]. It appears to act in the distal tubule transport proteins in the nephron via SPS1-related
to promote sodium reabsorption by the Na-Cl proline/alanine-rich kinase (SPAK) or oxidative
co-transporter via a pathway that involves the stress-responsive kinase 1 (OSR1) [101]. Of par-
FGF23 receptor/Klotho complex, extracellular ticular importance are the cation-chloride
signal-regulated kinase 1/2, serum co-transporters (CCCs) found throughout the
glucocorticoid-regulated kinase 1, and WNK4. tubule. The WNK family serves to help maintain
This observation may account, in part, for the appropriate blood pressure by acting on these
association of FGF23 and cardiovascular disease co-transporters. WNK inhibition acts to lower
in children with CKD. blood pressure by decreasing NaCl reabsorption
Adrenomedullin: Adrenomedullin is a in the distal collecting tubule and thick ascending
52-amino acid peptide that was isolated from limb while maintaining serum K+ levels by reduc-
human pheochromocytoma cells [100]. It reacts ing K+ secretion [101]. Figure 3 summarizes the
with a G-protein cell receptor and causes vasodi- sensor and effector mechanisms that are involved
latation, an effect that may be mediated by in the regulation of sodium homeostasis.
196 N. Madden and H. Trachtman

Water acting as a potent dipsogen and stimulating drink-


ing [68]. Its role in water handling within the
AVP: The primary efferent mechanism in the kidney is minor and may be related to modulation
maintenance of water homeostasis is AVP. This of the renal response to AVP. However, it is a
peptide fosters water retention by the kidney and potent dipsogen and stimulates thirst and drinking
stimulates thirst. The plasma AVP concentration behavior.
is approximately 1–2 pg/ml under basal condi- Thirst: Thirst, or the consciously perceived
tions [64, 65]. It is unknown whether there is desire to drink, is a major efferent system in
tonic release of AVP or whether there is pulsatile water homeostasis [67]. It is estimated that for
secretion in response to minute fluctuations in each 1 pg/mL increase in the circulating plasma
plasma osmolality. The set point, or osmotic AVP level, there is parallel rise of 100 mosm/kg
threshold for AVP release, ranges from 275 to H2O in urinary concentration. If the basal plasma
290 mosm/kg H2O. The circulating hormone con- osmolality and AVP concentration are approxi-
centration rises approximately 1 pg/mL for each mately 280 mosm/kg H2O and 2 pg/mL, respec-
1 % increase in plasma osmolality. The sensitivity tively, and the steady state urine osmolality is
of the osmoreceptors in promoting AVP release 200 mosm/kg H2O, then as soon as the plasma
varies from person to person with some individ- osmolality and AVP concentration reach
uals capable of responding to as small as a 0.5 290 mosm/kg H2O and 12 pg/mL, respectively,
mosm/kg H2O increase in osmolality and others the urine is maximally concentrated. Beyond this
requiring greater than a 5 mosm/kg H2O incre- point, the only operational defense against a fur-
ment to stimulate AVP release. Patients with ther rise in plasma osmolality is increased free
essential hypernatremia possess osmoreceptors water intake, underscoring the essential role of
that have normal sensitivity but the osmotic thirst as an efferent mechanism in water homeo-
threshold for AVP release is shifted to the right. stasis. It highlights the increased risk of
Because the relative distribution of water between hyperosmolality in patients who do not have free
the ECW and ICW compartments is undisturbed, access to water such as infants, the physically or
these patients are unaffected by their abnormally mentally incapacitated, or the elderly [69].
high serum sodium concentration. Although there Thirst is a biological function that is difficult to
may be sex-related differences in AVP secretion in quantitate because it is an expression of a drive
response to abnormal water homeostasis with rather than an actual behavior. At present, visual
increased sensitivity in women, this is not a rele- analog scales using colors or faces are the most
vant clinical concern in prepubertal children. useful tools for quantitating thirst under controlled
Reliable measurement of AVP is hindered by condition. There can be dissociation between
several factors including high level (>90 %) of water intake and the sensation of thirst as in
binding to platelets. Thus, plasma AVP measure- patients with psychogenic polydipsia (e.g.,
ments are influenced by the number of platelets, schizophrenia, neurosis). It is not known whether
incomplete removal of platelets, or pre-analytical specific drugs directly stimulate the dipsogenic
processing steps. Copeptin (CTproAVP), a response. The role of diet, e.g., high salt intake,
39-amino acid glycopeptide, is a C-terminal part in the regulation of thirst is also unknown. The
of the precursor pre-provasopressin (pre- osmotic control of thirst may be suboptimal in
proAVP). Activation of the AVP system stimu- newborn infants and in the elderly [103].
lates copeptin secretion into the circulation from Thirst and drinking behavior are stimulated by
the posterior pituitary gland in equimolar amounts significant contraction of the ECF space or by
with AVP. Therefore, CTproAVP directly reflects hypotension. In addition, thirst and drinking
AVP concentration and it has been used clinically behavior are modulated by signals that originate
as a surrogate biomarker of AVP secretion [102]. in the oropharynx and upper gastrointestinal tract.
Angiotensin: Angiotensin II serves as an effer- Animals with hypernatremia who are given access
ent system in water homeostasis primarily by to water as the sole means of correcting the
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 197

Fig. 4 Graphic illustration


summarizing sensor and Disturbance in serum osmolality
effector mechanisms Laboratory determination
involved in the regulation of
water homeostasis,
osmolality, and cell volume.
AVP arginine vasopressin Hypothalamic sensors
Peripheral sodium sensors

EFFECTOR MECHANISMS

Urinary concentrating
mechanism Thirst
AVP

Normalization of serum osmolality and cell


volume
Resolution of cell shrinkage or expansion

hyperosmolal state stop drinking sooner than alterations in sodium balance. This is accom-
animals corrected in part with supplemental intra- plished by glomerular-tubular balance in which
venous fluid. This is most likely due to oropha- load-dependent proximal tubule sodium absorp-
ryngeal stimuli that curtail drinking prior to tion and delivery of filtrate to the distal tubule is
complete normalization of plasma osmolality modulated in response to GFR [105]. Changes in
[104]. Figure 4 summarizes the sensor and effec- GFR also lead to changes in the oncotic pressure
tor mechanisms that are involved in the regulation in the peritubular capillaries that influence sodium
of sodium homeostasis. reabsorption [106]. Thus, an increased GFR is
associated with higher hydrostatic pressures in
the peritubular capillary network that retard fluid
Effector Mechanisms: Sodium and solute reabsorption in the proximal tubule.
and Water Finally, tubuloglomerular feedback is activated
by alterations in solute delivery to the distal neph-
The kidney is the principal organ that acts in ron to bring GFR in line with alterations in tubular
response to sensory input, delivered via neural or function. Many of the efferent signals including
humoral signals, to restore ECW volume size to renin, angiotensin, NO, adenosine, and prosta-
normal following the full range of clinical prob- glandins participate in this particular pathway.
lems. Absorption of sodium across the gastroin- The release of these effector molecules is acti-
testinal tract, modulated by chemoreceptors in the vated via myogenic stretch receptors and chemo-
hepatic vasculature, plays a minor in body sodium receptors located in the macula densa region of the
homeostasis. distal nephron. Even in children with
compromised renal function (GFR <30 ml/min/
1.73 m2), glomerulotubular balance is maintained
Sodium in the face of gradual changes in GFR. However,
they are unable to respond to abrupt changes in
GFR: In children with normal kidney function, sodium balance and ECF volume changes as rap-
changes in GFR are associated with parallel idly as healthy children and are susceptible to
198 N. Madden and H. Trachtman

volume contraction or hypervolemia if sodium synthesized angiotensin II stimulate the activity


intake is substantially reduced or increased over of the sodium-hydrogen antiporter and promote
a short period of time [46]. sodium reabsorption in conditions associated with
Neural and humoral factors that lower GFR act decreased ECF volume. Conversely, ANP and the
predominantly on the vascular tone of the afferent kinins act on proximal tubular cells to inhibit
arteriole and reduce renal blood flow and the sodium reabsorption and limit expansion of the
filtration fraction. Agents in this category include ECW space.
adrenergic nerve stimulation and endothelin. In Distal nephron including collecting duct: This
contrast, angiotensin II acts primarily on efferent portion of the nephron is responsible for the
arteriolar tone. This tends to preserve GFR more reabsorption of approximately 10–25 % of the fil-
than renal blood flow and the filtration fraction tered sodium and water load. Under most circum-
(RBF/GFR) is increased. This pattern is most stances, it adapts to changes in delivery arising
evident in states of compromised effective perfu- from alterations in proximal tubule function. This
sion such as congestive heart failure, cirrhosis, segment of the nephron is responsive to virtually all
and nephrotic syndrome [62, 63, 107]. The critical of the humoral efferent signals and accomplishes
role of angiotensin II in maintaining GFR and the final renal homeostatic response to fluctuations
sodium excretion in these conditions is manifest in sodium balance. Sodium reabsorption in the
during the acute reversible functional decline in distal tubule and connecting segment is responsive
GFR that occurs after the administration of ACE to circulating levels of aldosterone [74]. In the
inhibitors [108]. This phenomenon also explains collecting tubule, mineralocorticoid-responsive
the reduction in kidney function and sodium sodium reabsorptive pathways in the principal cell
retention that are observed in patients with a crit- achieve the final modulation of sodium excretion in
ical renal artery stenosis in a kidney transplant response to alterations in sodium intake. Aldoste-
following initiation of ACE inhibitor rone enhances sodium reabsorption by inducing a
therapy [108]. number of transport proteins whose synthesis is
Proximal tubule: Nearly 60–70 % of the fil- triggered by activation of SGK1, serum, and
tered sodium and water load are reabsorbed in the glucocorticoid-inducible kinase [109]. The most
proximal tubule. Sodium and fluid reabsorption prominent of these is the epithelial sodium channel
are isosmotic in this nephron segment. These pro- (ENaC). This transepithelial protein is composed of
cesses are driven by Na-K-ATPase activity along three distinct chains – α, β, and γ – each of which is
the basolateral membrane surface with secondary encoded by a separate gene [110]. The complete
active transport of solute across the apical mem- protein has two membrane-spanning domains with
brane. The bulk of sodium reabsorption is driven an amino and carboxyl terminus within the cell.
by the sodium-hydrogen exchanger, with a lesser The α-chain appears to constitute the actual
contribution by other co-transport systems for sodium-conducting pathway, while the β- and
glucose, phosphate, organic anions, and amino γ-chains represent regulatory components that con-
acids. The linkage between disturbances in ECF trol the open/closed status of the channel. Genetic
volume and sodium reabsorption in the proximal defects in each individual component have been
tubule is created, in part, by changes in the phys- described and linked to human disease. Thus,
ical forces that govern fluid and solute movement. pseudohypoaldosteronism has been mapped to
These include changes in peritubular capillary mutations in the α, β, and γ chains, and Liddle’s
hydrostatic pressure, peritubular capillary protein syndrome has been attributed to truncation in the
concentration, and oncotic pressure and changes β-chain with increased ubiquitinylation and
in renal interstitial pressure that modulate water proteasomal degradation of the abnormal protein
and solute movement across cells (transcellular) [109–111]. As noted above, recent evidence indi-
and along the paracellular pathway. cates that β-intercalated cells in the collecting duct
Sympathetic nervous stimulation, norepineph- also modulate sodium, potassium, and water
rine release, and both filtered and locally excretion [88].
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 199

Water are nine known members of this group of proteins,


all of which contain six membrane-spanning
AVP: AVP acts along several segments of the domains. The first member to be identified was
nephron. However, its primary site of action for aquaporin-1 (originally called channel-forming
maintenance of water homeostasis is the integral membrane protein of 28 kD or CHIP-28),
collecting tubule [112]. In that segment of the which mediates water movement across the eryth-
nephron, AVP reacts with the V2 receptor, a rocyte membrane and along the proximal tubule.
371-amino acid protein that is coupled to a Mice that do not express AQP-1 have a normal
heterotrimeric G protein, along the basolateral phenotype and concentrate their urine normally.
membrane of the distal tubule and collecting Aquaporin-2 (AQP-2) is the major AVP-sensitive
duct cells. The V2 receptor gene has been local- water channel in the collecting tubule
ized to region 28 of the X chromosome. This [117]. Immunogold electron microscopy studies
epithelial cell receptor is distinct from the V1 have confirmed that AQP-2 represents the water
receptor in the vasculature that is linked to Ca channel in the cytosolic vesicles which fuse with
activation of the inositol triphosphate cascade the apical membrane following exposure of princi-
and which mediates vasoconstrictor response to pal cells to AVP. The contribution of AQP-3 and
the hormone [112]. AQP-4 to the normal urinary concentrating mech-
The binding of AVP to the V2 receptor acti- anism has been confirmed in mice that have been
vates basolateral adenylate cyclase and stimulates genetically manipulated and which do not express
the formation of cAMP within the cytosol. This these two proteins [118].
intracellular second messenger then interacts with The importance of AQP-2 in mediating the
the cytoskeleton, specifically microtubules and normal response to AVP has been verified by the
actin filaments, and promotes fusion of discovery of mutations in the AQP-2 gene in
intramembrane particles that contain preformed children with non-X-linked, autosomal-recessive
water channels with the apical membrane of prin- forms of nephrogenic diabetes insipidus [119].
cipal cells in the collecting duct. The Moreover, alterations in AQP-2 protein expres-
AVP-induced entry of preformed water channels sion have been documented in other states associ-
involves clathrin-coated pits. The withdrawal of ated with a urinary concentrating defect such as
AVP leads to endocytosis of the membrane seg- lithium exposure, urinary tract obstruction, hypo-
ment containing the water channels into vesicles kalemia, and hypercalcemia [116]. There are per-
that are localized to the submembrane domain of sistent challenges in the development of better
the cell. This terminates the hormone signal. modulators of the aquaporins including
Recycling of water channels from vesicles to the druggability of the target and the suitability of
apical membrane and then back into vesicles has the assay methods used to identify the
been demonstrated in freeze-fracture studies of modulators [120].
cells exposed to AVP [112]. The importance of Water reabsorption in the collecting duct is not
the V2 receptor in water homeostasis is confirmed completely dependent upon the presence of AVP.
by genetic mutations and corresponding abnor- In animals that are genetically deficient in AVP
malities in protein structure in children with (Brattleboro rats) or in patients with central dia-
X-linked congenital nephrogenic diabetes betes insipidus, urinary osmolality increases
insipidus [113]. Conversely, activating mutations slightly above basal levels in the face of severe
in the V2 receptor lead to increased water absorp- ECF volume contraction. This may be the conse-
tion and hyponatremia in the absence of AVP, quence of reduction in urinary flow rate along the
resulting in a condition called nephrogenic syn- collecting duct that enables some passive equili-
drome of inappropriate antidiuresis [114, 115]. bration between the luminal fluid and the hyper-
The water channels that mediate transmembrane tonic medullary interstitium.
movement of water across the collecting tubule in Although the collecting duct is the primary site
response to AVP are called aquaporins [116]. There of regulation of net water reabsorption, the
200 N. Madden and H. Trachtman

proximal tubule contributes to water balance Table 4 Factors that contribute to the countercurrent
under circumstances of decreased ECW compart- mechanism
ment size. Whereas the proximal tubule normally Na-Cl-K-mediated solute absorption in the medullary
reabsorbs approximately 60 % of the filtered thick ascending limb of Henle
water load, this proportion may exceed 70 % Low water permeability of the distal tubule and
connecting segment
when the ECF volume is diminished. Further-
High water permeability in the descending limb of Henle
more, by decreasing fluid delivery to the distal Vasa recta and elimination of interstitial water volume
nephron, this enhances the AVP-independent AVP responsiveness of the collecting tubule
reabsorption of water along the collecting tubule.
These effects may explain the clinical benefit
achieved by the administration of thiazide that contain hairpin loop-shaped blood vessels.
diuretics to patients with nephrogenic diabetes This facilitates efficient removal of water that
insipidus [121]. The utility of hydrochlorothiazide exits the descending limb of Henle from the med-
in the management of nephrogenic diabetes ullary interstitium without washing out the solute
insipidus also reflects increased synthesis and gradient that passively drives water reabsorption
expression of aquaporins and sodium transporters in the collecting tubule. The final effector mecha-
such as ENaC and sodium-chloride nism is the alteration in the water permeability of
co-transporter [122]. the collecting tubule in response to AVP and gen-
eration of a concentrated urine or the excretion of
Countercurrent Mechanism solute-free water in the absence of AVP (Table 4).
The primary locus of the urinary concentrating Conditions that disturb the normal architectural
mechanism is the medulla and involves the thin relationship between the vasculature and the
descending limb of Henle, medullary thick tubules in the medulla such as chronic allograft
ascending limb of Henle, cortical thick ascending nephropathy, acute interstitial nephritis, and
limb of Henle, and collecting duct [123]. Sodium obstructive uropathy disrupt the countercurrent
and water reabsorption are isosmotic in all seg- mechanism and lead to a urinary concentrating
ments of the nephron proximal to the loop of defect.
Henle. In order to concentrate or dilute the urine, Osmoprotective molecule (compatible
water and solute must be separated to enable osmolytes): Besides the presence of effector
excretion of free water or urine that is mechanisms to maintain water balance, cells pos-
hyperosmolal relative to plasma. This process sess a wide range of adaptive mechanisms to
begins in the medullary and cortical thick ascend- counteract the undesirable movement of water
ing limb of Henle where NaCl is reabsorbed inde- between the cell and the ECW during hypotonic
pendently of water, generating a hypotonic and hypertonic states and to prevent neurological
luminal fluid. This action is linked in series to dysfunction. These include early response genes
the low water permeability of the distal tubule that mediate the prompt accumulation of chaper-
and the connecting segment, which together with one molecules to counteract the adverse effects of
continued sodium reabsorption, enhances the altered cell size on protein function [50]. This is
hypotonicity of the urine in this segment. In a followed temporally by the uptake or extrusion of
secondary step, the permeability to water along electrolytes as an acute response to altered size
this segment of the nephron is much lower than in cell. Because there are inherent limits on the abil-
the descending limb of the loop of Henle. This ity to regulate cell volume exclusively with inor-
enables water to move down its osmolal gradient ganic electrolytes, the more extended response
from the tubule lumen into the interstitium as it involves membrane transport and/or synthesis/
enters the medulla in the descending limb. Finally, degradation of a variety of compatible solutes,
the third critical component of the countercurrent called osmolytes, whose cytosolic concentration
mechanism is the presence of vasa recta, which can be safely altered without perturbing enzy-
perfuse the inner medulla via vascular bundles matic activity and cell function. These
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 201

osmoprotective molecules include carbohydrates demyelinating syndrome following rapid reversal


(sorbitol, myoinositol), amino acids (taurine, glu- of hyponatremia, dialysis disequilibrium syn-
tamate), and methylamines (betaine, glyceropho- drome after the initiation of dialysis in patients
sphorylcholine) [50]. They accumulate in the with AKI or CKD, and cerebral edema and brain
cytosol to preserve cell function during chronic herniation in patients with a first episode of acute
osmolal disturbances. The cell volume regulatory diabetic ketoacidosis [127, 128].
response can be activated by electrolytes such as
sodium or neutral molecules, e.g., urea and glu-
cose [50]. The adequacy of the cell volume regu- Laboratory Assessment of Sodium
latory response and the accumulation of and Water Balance
osmoprotective molecules in cerebral and renal
cells depend on the rate of rise in osmolality as There are no normal values for sodium and water
well as the magnitude of the absolute intake or excretion, a reflection of the wide range
change [124]. of normal daily dietary intake for both sodium and
Experimental data in animals and clinical water. Healthy individuals are in balance and the
experience in premenopausal women suggest excretion of sodium and water matches the daily
that estrogens may impair the cell volume regula- intake. Therefore, laboratory assessment of
tory response to disturbances in plasma osmolal- sodium and water homeostasis is confined to dis-
ity. This increases the risks associated with both ease states in which the clinician must determine
the untreated abnormalities and therapy whether renal sodium and water handling are
[125]. The cell volume regulatory adaptation is appropriate for the clinical circumstances.
fully operational during maturation. The accumu-
lation of osmoprotective molecules in the face of
chronic hypernatremia is normal in pre-weanling Sodium
rats, albeit with a higher set point to preserve the
increased brain cell water content [126]. The urine sodium concentration is not a valid
The failure to adequately account for the cell index of sodium balance because the value may
volume regulatory response to osmolal disorders vary depending upon the volume and concentra-
contributes to some of the adverse effects associ- tion of the sample. Therefore, renal handling of
ated with inappropriately rapid correction of sodium is best evaluated using the fractional
abnormalities in plasma osmolality. Under these excretion of sodium (FENa). After obtaining a
circumstances, there is disruption in the blood- random urine sample and a simultaneous blood
brain barrier and activation of glial cells in the sample and measuring the sodium and creatinine
central nervous system. These lead to neurologi- concentrations in both specimens, the FENa is
cal dysfunction, specifically seizures, during calculated using the following formula:
the treatment of hypernatremia, osmotic

FENa ¼ excreted sodium=filtered sodium


¼ urinary sodium concentration  urine flow rate=plasma sodium concentration  GFR
urine sodium concentration=plasma sodium concentration (3)
¼
urine creatinine concentration=plasma creatinine concentration

This formula is based on the insertion of the denominator. The determination of the FENa is
creatinine clearance as a measurement of GFR in useful in clinical practice because it can be done
the second equation and the cancelation of the using spot samples and does not require timed
urine flow rate term in the numerator and urine collections. In healthy individuals, the
202 N. Madden and H. Trachtman

FENa varies depending upon the daily sodium upon the water intake in the past 2–4 h. Therefore,
intake. However, in patients with ECF volume the assessment of water handling is best judged by
contraction who are responding appropriately, determining these values under more controlled
the FENa is less than 1 % (<3 % in neonates). conditions such as in the water-deprived state or
Conversely, in patients with ECW compartment following administration of a water load (10–20
expansion, the FENa will exceed 3 % unless there ml/kg body weight) to evaluate the urinary con-
is concomitant renal disease. centrating or diluting capacity, respectively.
The functional aspects of renal water handling
are best assessed by determining the free water
Water clearance. This represents the amount of solute-
free water excreted by the kidney. It is calculated
The determination of the urine specific gravity or using the following formula:
osmolality in a random sample varies depending

Free water clearance ¼ urine volume  osmolal clearance


(4)
¼ urine volume  ½urine osmolality  urine flow rate=plasma osmolality

If the free water clearance is a positive number, body fluid homeostasis, the distinct regulatory
then the urine/plasma osmolality ratio is less than mechanisms activated to control these factors,
1, the urine is dilute, and the kidney is in a diuretic and the varied therapeutic strategies that must be
mode. When a water diuresis is maximal, the free employed to restore sodium and water balance in
water clearance measures the capacity of the kid- disease states, it is essential that disturbances in
ney to excrete free water. Under these circum- sodium-waterand water balance be evaluated sep-
stances, urine volume is directly related to the arately. These dual assessments are done in paral-
dietary solute intake [31]. In contrast, patients lel, a reflection of the body’s own method of
who are in an antidiuretic mode have a urine/ operation.
plasma osmolality ratio greater than 1 and a neg- When confronted with a child with a sodium
ative free water clearance. As the solute excretion and water disturbance, the first question is
rate increases, both the maximum values for free whether the size of the ECF volume is altered –
water clearance and free water reabsorption is it decreased or expanded? – to the extent that
increase. At any given solute excretion rate, the perfusion of vital organs or pulmonary gas
free water clearance greatly exceeds the free water exchange is jeopardized. Depending on the sever-
reabsorption. This indicates that the renal water ity of the problem, such patients have a life-
homeostatic mechanisms designed to protect threatening condition and may require emergency
against overhydration and dilution of the ECW therapy such as volume resuscitation or acute
are more robust than those used to defend against dialysis.
water deficit and dehydration. After making this acute determination and
instituting appropriate emergency therapy, it is
important to grade the magnitude of the distur-
Overview of the Evaluation of Fluid bance in ECF volume. There is no single or com-
and Water Abnormalities bination of laboratory tests that substitute for
clinical judgment. Because acute changes in
In practice, the clinical information and laboratory body weight always reflect alterations in sodium
data used to evaluate patients overlap with one balance and ECW compartment size, serial mea-
another. However, in view of the different physi- surements in body weight are the most reliable
ological roles of sodium and water balance in indicator of the presence and severity of
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 203

disturbances in ECF volume. However, these Table 5 Steps in the initial evaluation and treatment of a
measurements are often unavailable or will have child with a disturbance in sodium and/or water balance
been done in different locations using different Step 1: determine if there is a life-threatening alteration in
equipment. Therefore, the evaluation of sodium ECF volume
balance is based upon a wide range of clinical Volume resuscitation if there is ECF volume
contraction
findings including changes in mental status, level
Consider dialysis if there is ECF volume expansion
of alertness, irritability, presence of thirst, pulse Step 2: grade severity of defect in sodium balance
rate, blood pressure, orthostatic changes, fullness Clinical determination of ECF volume
of the anterior fontanel in infants, presence of Step 3: determine if there is a defect in water balance
tears, dryness of the mucus membranes, skin Laboratory measurement of plasma osmolality
color, elasticity of the skin or tenting, capillary
refill or turgor, peripheral edema, shortness of
breath, and presence of rales on auscultation of sections describe the individual clinical entities
the chest. Capillary refill may be the most useful responsible for derangements in sodium and
test to rapidly and accurate assess ECF volume water balance and outline the general treatment
and the response to treatment [129]. Other find- of these conditions.
ings on clinical examination include urinary spe-
cific gravity and central venous pressure.
Laboratory investigations include BUN, serum Sodium Balance Disturbances: Deficit
creatinine, and bicarbonate concentrations. and Excess
It is important to emphasize that assessment of
ECF volume is a clinical determination. There is Sodium Deficit
no single or combination of laboratory tests that is
a valid surrogate marker. Moreover, despite the Diagnosis and evaluation: Sodium deficits and
frequency of clinical disturbances in sodium bal- ECF volume contraction are dangerous because
ance, especially ECF volume contraction, no suit- decreased size of the intravascular space leads to
able scoring has been devised to accurately and reduced perfusion and ischemia of vital organs,
reliably distinguish different degrees of ECF vol- namely, the brain, heart, and kidneys. In children,
ume contraction or expansion. This contrasts to the absence of concomitant atherosclerosis dis-
the Glasgow coma score or the PRISM score that ease or endothelial dysfunction secondary to
have been successfully applied to the initial essential hypertension, smoking, hyperlipidemia,
assessment of patients with acute neurological or and diabetes decreases this risk. However, there
multisystem organ failure. are pediatric patients who are more susceptible to
The third step in the evaluation of a patient the adverse consequences of hypovolemia includ-
with a sodium and water disturbance is to measure ing newborn babies in whom high circulating
the plasma osmolality, which indicates an abnor- levels of vasoconstrictor hormones and impaired
mal distribution of water between the ECW and autoregulation render the glomerular microcircu-
ICW compartments. The most likely symptoms lation sensitive to reduced perfusion [130]. In
include confusion, irritability, lethargy, addition, underlying diseases or medications
obtundation, and seizures. These manifestations may hinder the counter-regulatory responses to
overlap significantly in patients with ECF volume contraction and heighten the risks
hyperosmolality or hypoosmolality. In contrast of hypovolemia.
to disorders of ECF volume, disturbances in Diseases that cause sodium deficiency can
water balance and distribution require a labora- originate outside the kidney or within the kidney.
tory determination for confirmation and grading. It is unfortunate that the word dehydration is rou-
The steps involved in the initial evaluation of a tinely used to describe these states because it
child with a disturbance in sodium and water suggests that water deficit is the major pathophys-
balance are summarized in Table 5. The following iological problem. It deflects attention from the
204 N. Madden and H. Trachtman

primary defect, namely, a net negative sodium bal- Table 6 Causes of net sodium deficit
ance [131, 132]. The critical role of the ECF space Renal causes
and sodium balance in the pathogenesis of clinical Compromised GFR
states of volume contraction is highlighted by com- Acute decrease in sodium intake or increased losses
paring the situation that occurs in patients with Tubular disorders
diabetes insipidus. When sodium balance is Osmotic diuresis
perturbed, >30 % of the fluid loss is derived from Diabetic ketoacidosis
the ECW compartment provoking the rapid onset Renal tubular acidosis
of symptoms. In contrast, only 1/12 or 8 % of the Renal Fanconi syndrome
pure water loss that occurs in diabetes insipidus is Pseudohypoaldosteronism
derived from the ECF (1/3 X 1/4), accounting for Obstructive uropathy
Bartter’s syndrome
the rare evidence of ECF volume contraction in
Renal dysplasia/hypoplasia
children with central or nephrogenic diabetes
Central nervous system
insipidus (Fig. 1). The term “denatration” may
Cerebral salt wasting
provide a more accurate depiction of what is occur-
CSF drainage procedures
ring in patients with primary deficits in sodium Hepatobiliary system
balance and contraction of the ECF volume [131]. Biliary tract drainage
Extrarenal causes can occur from losses of Gastrointestinal tract
sodium in any body fluid or across any epithelial Infectious diarrhea
surface including the CSF, pleural fluid, biliary Chloride diarrhea
tree, gastrointestinal losses, or skin. They can be Laxative abuse
the result of medical treatment. CKD can cause Malignancy (carcinoid, tumor related)
sodium deficit because the lower GFR compro- Adrenal diseases
mises the homeostatic capacity of the renal Salt-losing congenital adrenal hyperplasia
tubules. Alternatively, there may be primary Addison’s disease
renal sodium loss that is not the consequence of Skin losses
a decrease in kidney function. Finally, renal Cystic fibrosis
sodium reabsorption may be diminished because Neuroectodermal diseases
of reduced circulating levels or unresponsiveness Burns
to aldosterone. The major causes of sodium defi-
ciency are summarized in Table 6.
Identifying the cause of a disturbance in If the losses are extrarenal, then renal sodium
sodium balance is accomplished based on a thor- reabsorptive mechanisms will be activated and the
ough history and physical examination. Previ- urinary specific gravity will be >1.015 and the
ously, the degree of ECF volume contraction FENa will be low, generally <1 % except in
was categorized as mild, moderate, or severe if infants. The failure to increase the urine concen-
the changes in body weight were estimated to be tration and lower FENa in the face of clinical signs
<5 %, 5–10 %, or >10 %, respectively. Life- of ECF volume contraction points toward a renal
threatening ECF volume contraction was thought or adrenal cause for the disorder. A renal ultra-
to represent >15 % decrease in weight. Recent sound documenting the presence of small,
data, based upon systematic body weights at the misshaped kidneys or hydronephrosis may be
time of hospitalization and immediately after cor- indicative of congenital abnormalities of the kid-
rection of the sodium deficit, suggest that these ney such as dysplasia or obstructive uropathy.
numbers overestimate the degree of sodium defi- Adrenal insufficiency is suggested by concomi-
cit and that the ECF volume contraction is better tant hyponatremia and hyperkalemia.
estimated to be <3 %, 3–6 %, and >6 % with Treatment: If the ECW compartment size is so
>9 % change in body weight representing an severely contracted that vital organ perfusion is
emergency [133]. compromised, based upon an altered mental
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 205

status, orthostatic changes, and azotemia, then be given to correcting sodium and electrolyte
fluid resuscitation must be initiated on an emer- deficits with oral rehydration solutions (ORS).
gent basis. This is necessary to prevent the devel- In general, patients can be repleted with a rapid
opment of AKI, which may occur if there is (1–2 h) intravenous infusion to restore ECF
sustained hypotension and renal ischemia second- volume followed immediately by initiation of
ary to ECF volume contraction. The risk of AKI is ORS [137]. The only conditions that represent
higher in children with preexisting renal disease, contraindications to the use of ORS are impaired
who are receiving nephrotoxic medications or neurological status, persistent vomiting, or dis-
who have hemoglobinuria or myoglobinuria, e. eases associated with mucosal damage in the gas-
g., crush injury or compartment syndrome. If trointestinal lumen.
there is no evidence of cardiac or pulmonary dis- ORS fluids introduced by the World Health
ease, then the optimal therapy under these condi- Organization contain sodium 90, potassium
tions is infusion of isotonic crystalloid (0.9 % 20, bicarbonate 30, chloride, and glucose
NaCl, Ringer’s lactate), 20 ml/kg body weight. 111 mmmol/L (20 g/L). The sodium and glucose
This fluid is appropriate regardless of the initial are present in a molar ratio that maximizes the
serum sodium or osmolality. Concerns about secondary active uptake of these solutes via the
infusing Ringer’s lactate are misplaced in view sodium-glucose co-transporter across the gastro-
of the low potassium concentration (4 mmol/L) intestinal epithelium. These transporters retain
in this solution. Transfusion of whole blood is activity in the context of secretory diarrhea and
optimal for treatment of hemorrhagic shock. only become impaired when there is extensive
However, in all other circumstances, infusion of mucosal damage. Water is absorbed passively
crystalloid solutions avoids difficulties caused by down its osmolal gradient. The presence of other
extravasation of the colloid into the interstitial solutes such as potassium and bicarbonate are not
compartment. A systematic review of the litera- critical to the successful utilization of ORS. These
ture does not support the use of colloid solutions fluids have been utilized for over 30 years. They
for volume replacement in critically ill patients can be administered ad libitum in response to the
[134]. Thus, in general, there is no proven benefit child’s own thirst and they are effective and safe
of providing colloid versus crystalloid solutions with minimal occurrence of hypernatremia or
for the initial treatment of shock and organ hyperkalemia. Various alternatives to glucose
hypoperfusion [135]. In addition, a meta-analysis such as rice-syrup or amylase-resistant starch
has demonstrated that the use of synthetic colloids may facilitate sodium and water reabsorption
such as hydroxyethyl starch is associated with a from ORS, decrease fecal fluid loss, and shorten
higher risk of AKI and the need for implementa- recovery time after an episode of cholera [138,
tion of renal replacement therapy compared to any 139]. Clinical studies are needed to confirm the
other alternative fluid [136]. The infusion should utility of these additives because they may
be as rapid as possible and the fluid dose should be increase the cost and decrease the shelf life of
repeated as often as necessary to achieve some ORS. These are important considerations in
evidence of clinical improvement such as developing countries where there is a high inci-
improved mental status, decrease in pulse, rise in dence of infectious diarrhea in infants and
blood pressure, or improved capillary refill. A children.
catheter should be placed in the bladder to facili- When parenteral therapy is required to correct
tate monitoring of urine output, especially in sodium and water deficits, the following guide-
patients with preexisting renal or cardiopulmo- lines can be applied when devising a therapeutic
nary disease who may be more sensitive to sudden plan. First, in the absence of reliable data regard-
changes in ECF volume. ing the acute weight loss, it is easiest to calculate
After correcting life-threatening maintenance and deficit therapy based on the cur-
hypoperfusion, a fluid repletion plan should be rent weight and the clinical estimate of the per-
initiated as soon as possible. Preference should centage decrease in body weight. Second, it is
206 N. Madden and H. Trachtman

advisable to discount any emergency fluid ther- exogenous addition of sodium or abnormal reten-
apy, such as bolus infusions of isotonic saline, in tion of endogenous sodium. Because the normal
computing the total corrective fluid prescription. kidney has the adaptive capacity to rapidly excrete
Third, if the clinical problem has developed in less a sodium load, in order for the excess sodium to
than 48 h, then it should be considered an acute cause clinical symptoms and signs, there must be
process and the sodium and fluid losses are a concomitant factor that limits natriuresis.
derived from the ICW and ECW in the ratio of Common causes of excess exogenous sodium
80–20 %. If the patient has been ill for more than are salt-water drowning, ingestion of a diet abnor-
48 h and the process is chronic, then the sodium mally high in sodium, or therapeutic infusion of
and fluid losses are derived from the ICW and sodium-containing intravenous solutions, such as
ECW in the ratio of 60–40 %. Under most condi- sodium bicarbonate during the resuscitation after
tions in which the sodium and water losses are a cardiopulmonary arrest. Examples of dietary
isotonic, the ECW portion of the loss, in liters, can excess of sodium include errors in the preparation
be multiplied by 140 mmol/L to determine the of infant formulas. The widespread use of
absolute sodium loss. Similarly, the ICW portion premixed baby formulas may decrease the inci-
of the loss, in liters, can be multiplied by dence of these accidents.
140 mmol/L to determine the absolute potassium Conditions associated with excessive endoge-
deficit. If the ECW and ICW fluid losses together nous sodium retention include AKI and the edema
with the respective sodium and potassium deficits states, namely, nephrotic syndrome, cirrhosis,
are added to the maintenance requirements, then a congestive heart failure, and gastrointestinal dis-
total fluid volume can be determined. After ease (malabsorption or protein-losing enteropa-
selecting a fluid that most closely approximates thy). In the first condition, which may occur due
the total sodium and water losses, the total fluid to glomerulonephritis or acute tubular necrosis,
volume is divided by the time frame of the cor- the sodium retention is directly related to the
rection to determine the intravenous infusion rate. decrease in GFR and diminished filtration of
It is important to monitor the child and replace sodium. In the later four states, renal sodium and
ongoing losses to insure that there is full resolu- water reabsorption are activated by a combination
tion of the underlying clinical problem. of mechanisms that are termed, underfill and over-
Example: If a 10 kg child is judged to have a fill. A critical review of the evidence in patients
5 % decrease in body weight over 36 h, then the with nephrotic syndrome suggests that those dis-
total fluid deficit of 500 ml can be divided into two eases that are associated with an inflammatory
components – 400 ml ICW with 56 mmol potas- infiltrate in the kidney develop primary sodium
sium and 100 ml ECW containing 14 mmol retention and overfilling of the ECF volume. This
sodium. The daily maintenance water and sodium histopathological feature is absent in minimal
requirements are 1,000 ml and 30 mmol sodium. change nephrotic syndrome and the underfill
Adding these two quantities together results in a mechanism predominates [140]. Thus, in
fluid that closely resembles 0.25 % saline nephrotic syndrome, total body sodium excess
(37 mmol NaCl/L) containing 30 mmol KCl/L may be coupled with a diminished, normal, or
and the infusion rate is approximately 60 ml/h if expanded effective ECF volume. This explains
the correction is designed to occur over 24 h. the normal distribution of PRA values and wide
range of measured plasma volume in these
patients. The causes of net total body sodium
Sodium Excess excess are summarized in Table 7.
The major clinical problems that accompany
Diagnosis and evaluation: These conditions, these conditions are related to pulmonary venous
which generally are less common than sodium congestion, impaired gas exchange, and dyspnea.
deficit, are evidenced by clinical signs of ECF In idiopathic nephrotic syndrome, the intra-
volume expansion. They can arise secondary to alveolar pressure is often sufficient to prevent
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 207

Table 7 Causes of net sodium excess cases, acute dialysis using peritoneal or hemodi-
Exogenous sodium alysis techniques may be necessary to foster
Salt-water drowning rapid clearance of excess sodium. Finally, contin-
Errors in formula preparation uous hemofiltration may be a safe and effective
Infusion of hypertonic sodium solutions means to rapidly remove sodium and water in
For example, after cardiopulmonary arrest severely ECF-overloaded children with nephrotic
Endogenous sodium syndrome [142].
Acute renal failure (glomerulonephritis, ATN)
Nephrotic syndrome
Cirrhosis
Congestive heart failure Water Balance Disturbances: Deficit
and Excess

frank pulmonary edema. However, in other cir- Hyponatremia


cumstances, intrinsic capillary leak together with
lowered plasma oncotic pressure promotes the Diagnosis and evaluation: Patients with
development of pulmonary interstitial fluid. hyponatremia have relative or absolute expansion
Peripheral edema may be associated with skin of the ICW compartment. If renal function is
infection, peritonitis, or thromboembolic events. normal, the excess free water is rapidly eliminated
Conditions associated with sodium excess within 2–4 h. Therefore, for hyponatremia to
should be evident on physical examination. The occur, there must be continued AVP release that
FENa will be high if there is sodium loading and is inappropriate to the serum sodium concentra-
renal function is normal. In contrast, in the states tion and the patient must have continued access to
characterized by retention of endogenous sodium, free water. The symptoms and signs of
the FENa will be very low. The FENa is more hyponatremia, which generally involve central
useful than the urinary specific gravity because nervous system dysfunction, are vague and
the later is likely to be elevated in all cases of nonspecific. Laboratory confirmation is required
sodium excess. to diagnose this abnormality. The presence of
Therapy: The optimal therapy is targeted at hyponatremia is not informative about the status
correcting the underlying disease. This is most of the ECF volume, and this later issue must be
important in patients with cirrhosis, congestive assessed clinically to determine whether the child
heart failure, or gastrointestinal disease. Ancillary has hypovolemic hypotonicity, isovolemic hypo-
therapies include administration of diuretics to tonicity, or hypervolemic hypotonicity.
facilitate urinary sodium excretion. Although thi- Hyponatremia is not uncommon in newborns,
azide diuretics may be adequate, more potent loop especially in preterm very low-birth-weight
diuretic may be required if the GFR is diminished. infants. Most often, hyponatremia is a result of
Potassium-sparing diuretic such as amiloride or impaired water excretion caused by excessive
spironolactone may be added to attenuate arginine vasopressin (AVP). It can also be caused
diuretic-induced hypokalemia. Patients with by the immaturity of the renal tubular sodium
nephrotic syndrome may require combinations reabsorption mechanisms, which result in exces-
of diuretic agents that act in the proximal (e.g., sive urinary sodium loss [143]. Hyponatremia is
metolazone) and distal (e.g., furosemide) seg- dangerous in newborns because sodium is a per-
ments of the nephron to promote an adequate missive growth factor and chronic deficiency is
natriuresis and diuresis. Infusions of albumin associated with impaired muscle, skeletal, and
(1 g/kg body weight) can augment the neural tissue growth and development [144].
medication-induced diuresis, especially in those Hyponatremia has also been linked to cerebral
with severe ECF volume contraction, reduced palsy, sensorineural hearing impairment, and
GFR, and azotemia [141]. In the most severe intracranial hemorrhages. When the hyponatremia
208 N. Madden and H. Trachtman

is severe, less than 120 mEq/L, there is a risk of All biochemical analyzers currently in use at
hyponatremic encephalopathy and possibly death large medical centers assay sodium concentration
[143, 145]. As such, it is important to recognize in the aqueous phase of serum and yield an accu-
hyponatremia in infants early and institute the rate determination of the sodium level. Therefore,
appropriate treatment to ensure proper growth the entity called pseudohyponatremia is no longer
and neurocognitive development. clinically relevant. In contrast, the reduced serum
In older children, hyponatremia represents one sodium concentration noted in patients with
of the common electrolyte disorders in hospital- increased circulating levels of an impermeant sol-
ized patients. The incidence ranges from 12 % in ute such as mannitol, urea, contrast media, or
patients with intracranial tumors who undergo glucose in patients with diabetic ketoacidosis is
neurosurgical procedures [146] to 38 % in patients genuine and reflects osmotic redistribution of
with cirrhosis and refractory ascites [147]. In all water from the ICW to the ECW space. The phe-
clinical circumstances hyponatremia is an indica- nomenon is reflected in the following formula
tor of a heightened risk for adverse neurological which enables adjustment of the serum sodium
outcomes and mortality. in patients with severe hyperglycemia:

“Physiological” sodium concentration ¼ measured serum sodium concentration þ 1:6 


(5)
½each 100 mg=dl increment in serum glucose above 100 mg=dl

The most common clinical causes of hyponatremia. In practice, diagnosing SIADH


hyponatremia are classified by the concomitant requires comparison of the urine specific
ECF volume status in Table 8. In some diseases, gravity or osmolality with the concurrent serum
such as congestive heart failure, the degree of osmolality. The urine should normally be
hyponatremia reflects circulating AVP levels and maximally dilute if the serum sodium concentra-
sympathetic nervous system activation and pro- tion is <130 mmol/L or the plasma osmolality
vides a marker of disease severity. A relationship <270 mosm/kg H2O. A urinary sodium concen-
between the degree of hyponatremia and the clin- tration >40 mmol/L is an adequate evidence
ical disturbance has not been demonstrated in against ECF volume contraction.
patients with nephrotic syndrome or cirrhosis. Treatment: In all cases, the first line of therapy
The syndrome of inappropriate AVP or ADH should be directed at the underlying cause of the
(SIADH) release causes hyponatremia with mild low serum sodium concentration. However,
to modest ECF volume expansion. It can occur as hyponatremia often warrants specific corrective
a consequence of central neurological lesions, treatment. Much ink has been spilled in detailing
pulmonary disease, or tumors. In addition, drugs the appropriate therapy of this electrolyte abnor-
can result in abnormal secretion or action of AVP mality. At times, this issue has been quite conten-
and lead to chronic hyponatremia. A list of these tious and the world has been divided into two
agents is provided in Table 9. The diagnosis of supposedly distinct camps – those who advocate
SIADH requires confirmation that the urine is “rapid” versus “slow” correction of
excessively concentrated relative to the plasma hyponatremia. The former group asserts that
osmolality without evidence of ECF volume con- hyponatremia has direct adverse effects on central
traction or adrenal or thyroid insufficiency. These nervous system function including impaired oxy-
two hormones are required to maintain the low genation that can lead to seizures or cardiopulmo-
water permeability of the collecting duct in the nary collapse prior to initiation of therapy
absence of AVP. Deficiencies of either hormone [148]. Such a sequence of events has been
impair free water clearance leading to euvolemic documented in experimental animals with acute
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 209

Table 8 Causes of hyponatremia Table 9 Drugs that cause water retention and the syn-
drome of inappropriate AVP release according to mode of
Hypovolemic: ECF volume contraction
action
Renal
Mineralocorticoid deficiency Increasing water permeability of the nephron
Mineralocorticoid resistance AVP (arginine or lysine vasopressin)
Diuretics Vasopressin analogs, e.g., 1-deamino, 8-D-arginine
vasopressin (DDAVP)
Polyuric acute renal failure
Oxytocin
Salt-wasting renal disease
Promoting AVP release
Renal Fanconi syndrome
Barbiturates
Renal tubular acidosis
Carbamazepine
Metabolic alkalosis
Clofibrate
Bartter’s syndrome/Gitelman’s syndrome
Colchicine
Gastrointestinal
Isoproterenol
Diarrheal dehydration
Nicotine
Gastrointestinal suction
Vincristine
Intestinal fistula
Inhibition of prostaglandin synthesis
Laxative abuse
Salicylates
Transcutaneous
Indomethacin
Cystic fibrosis
Acetaminophen (paracetamol)
Heat exhaustion
Other nonsteroidal anti-inflammatory drugs
“Third-space” loss with inadequate fluid replacement
Potentiation of the action of AVP
Burns
Chlorpropamide
Major surgery, trauma
Cyclophosphamide
Septic shock
Euvolemic: normal ECF volume
Glucocorticoid deficiency
Hypothyroidism hyponatremia is more closely related to the acuity
Mild hypervolemia: ECF volume expansion of the change rather than the absolute size of
Reduced renal water excretion the drop in serum sodium concentration [151].
Antidiuretic drugs Thus, therapy should be guided by the time
Inappropriate secretion of ADH
frame in which hyponatremia has developed. If
Hypervolemic: ECF volume expansion
the hyponatremia is acute, i.e., <12 h in duration,
Acute renal failure (glomerulonephritis, ATN)
then the brain will behave as a perfect osmometer
Chronic renal failure
leading to potentially life-threatening cerebral cell
Nephrotic syndrome
Cirrhosis
swelling. Under these circumstances, there is an
Congestive heart failure urgent need to rapidly reverse the hyponatremia to
Psychogenic polydipsia/compulsive drinking counteract cell swelling. Clinical experience indi-
cates that infusion of a 3 % NaCl solution
(513 mmol/L) in a volume designed to raise the
hyponatremia [149]. This risk may be especially sodium concentration by 3–5 mmol/L is sufficient
prominent in premenopausal women. In contrast, to halt central nervous system dysfunction
there are others who emphasize the cerebral cell [152]. The benefits and lack of adverse effects of
volume regulatory response to hyponatremia and acute correction have been confirmed in a series of
highlight the risk of brain cell dehydration and 34 infants and children with acute water intoxica-
osmotic demyelinating syndrome in patients who tion caused by the ingestion of dilute infant
are corrected too quickly [150]. formula [153]. After this is achieved, the
Taking into account the entire literature on the hyponatremia can be corrected more slowly.
subject, current evidence suggests that the risk of There is preclinical evidence that administration
210 N. Madden and H. Trachtman

of urea but not steroids can attenuate the adverse Finally, non-peptide vasopressin receptor
neurological consequences of rapid correction of antagonists, the vaptans that bind to the V2 recep-
hyponatremia with hypertonic saline or a vaso- tor in the collecting duct and block AVP action,
pressin antagonist [154]. This requires confirma- have been developed to treat chronic
tion in children with hyponatremia. hyponatremia [156, 157]. This results in diuresis
Example: If a 6-year-old child weighing 20 kg without electrolyte loss, termed aquaresis, reduc-
develops a seizure after a tonsillectomy and is ing total body water and raising the serum
noted to have a serum sodium concentration of concentration of sodium, thus correcting
115 mmol/L, then 36–60 mmol of sodium are hyponatremia.
needed to raise the sodium concentration by 3–5 Vaptans have been approved for use in the
mmol/L. This is accomplished by infusing treatment of hypervolemic or euvolemic
72–120 ml of the hypertonic saline infusion. hyponatremia in adults. They are not indicated in
If hyponatremia has developed over more than patients with hypovolemic hyponatremia. Vaptans
12 h or if the duration of the problem is unclear, have proven useful in the treatment of congestive
especially in the absence of signs of neurological heart failure and chronic liver failure, both of
dysfunction, then slow correction is the prudent which are frequently associated with
course of action [150, 151]. The current definition hypervolemic hyponatremia, and SIADH when
of slow correction includes two features: (1) the associated with euvolemic hyponatremia.
rate of rise in serum sodium concentration should Conivaptan was the first FDA-approved agent in
be <0.6 mmol/L throughout the correction phase this class and is useful for short-term intravenous
and (2) the total increment and/or the final serum use. Tolvaptan is an oral agent that has been
sodium concentration after 48 h of treatment demonstrated to safely correct hyponatremia in
should not exceed 25 mmol/L or 130 mmol/L, several dose-response studies involving adults
respectively. The more cautious criterion should with euvolemic hyponatremia or hypervolemic
be applied depending on the initial serum sodium hyponatremia [158]. Safety and efficacy of these
level. If a child develops acute changes in mental drugs have not been assessed in pediatric patients.
status or new neurological findings, during or Randomized control trials of the use of vaptans in
shortly after the fluid treatment, then a serum children with euvolemic hyponatremia are under-
sodium concentration should be checked. Imag- way. The results may demonstrate that vaptans
ing studies, specifically an MRI of the brain, may have an important role in treating newborns and
reveal the changes of osmotic demyelinating children with hyponatremia due to excessive AVP.
syndrome.
There are several therapeutic options for
patients with SIADH whose underlying cause Hypernatremia
cannot be corrected. Restriction of free water
intake to match insensible losses and urine output Diagnosis and evaluation: Hypernatremia may
may be adequate to stabilize the serum sodium arise due to excessive intake of sodium and ECF
concentration. If this is not well tolerated, then the volume expansion. However, excessive water loss
administration of furosemide 1–2 mg/kg per day relative to the sodium deficit with hypovolemia
to promote a hypotonic diuresis together with oral is far more common. In a recent survey of
administration of NaCl 1–2 g per day may correct hypernatremia in hospitalized children, the vast
the hyponatremia. If these measures fail, consid- majority had significant underlying medical prob-
eration can be given to treatment with lithium or lems and 76 % of the cases were secondary to
demeclocycline, two drugs that interfere with inadequate water intake [159]. The prevalence of
AVP action in the collecting tubule to foster excre- this electrolyte abnormality is much lower than
tion of free water and raise the serum sodium hyponatremia. One of the common causes is diar-
concentration [155]. rheal illness in infants; however, the reduction in
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 211

the sodium concentration of most baby formulas pitched cry, and a doughy skin texture. Because
to match the level in human breast milk has the hyperosmolality of the ECW compartment
resulted in a dramatic decrease in the incidence provokes movement of water from the ICW
of hypernatremic dehydration. Nonetheless, down its osmolal gradient, these patients preserve
changes in medical practice with early discharge ECF volume until late in the disease course. Their
of newborn infants after delivery have resulted in illness is usually chronic and there is a greater
the steady occurrence of hypernatremic dehydra- contribution of the ICW to the water and electro-
tion in breastfed babies [160]. Patients with lyte deficits. Assessment of the FENa is useful in
hypernatremia have relative or absolute contrac- assessing the ECF volume in these patients. The
tion of the ICW compartment. Similar to the situ- causes of hypernatremia are listed in Table 10.
ation with hyponatremia, the clinical clues to the Treatment: Because children with
presence of hypernatremia are nonspecific and hypovolemic hypernatremia are usually very ill,
laboratory confirmation is mandatory to diagnose they often require bolus infusions of isotonic
this abnormality. Moreover, like hyponatremia, saline to restore organ perfusion. Once this is
hypernatremia must be evaluated in light of the accomplished, then the fluid regimen should
clinically determined ECF volume status. include the maintenance fluids, the estimated def-
Patients with hypernatremia and ECF volume icit with the assumption that 60 % is derived from
expansion are easy to diagnose. The children who the ECW and 40 % from the ICW. In addition,
represent a serious problem are those with there is a free water deficit. This can be calculated
hypovolemia. They may have some distinct fea- from the following formula:
tures including a marked irritability, a high-

Water deficitðin mlÞ ¼ 0:6  body weight  ½actual serum sodium concentration=140  1 (6)

This formula may overestimate the water deficit increase in serum sodium concentration that will
and it has been recommended that the following be achieved following the infusion of 1 l of a
alternative equation be used to estimate the given solution [161]:

Change in sodium concentration ¼ ½infusate Naþ  serum Naþ =total body water þ 1 (7)

Finally, with regard to the rate of correction, the expansion and clinical signs of cerebral edema,
standard practice is to correct hypovolemic findings that have been confirmed in NMR spec-
hypernatremia gradually over at least 48 h. After troscopy studies of the brain [164]. The experi-
the groundbreaking work of Finberg et al. [162], mental observations were confirmed in a
the risk of cerebral edema following rapid correc- randomized trial, which demonstrated that the
tion of chronic hypernatremia is now attributed to safest and most effective fluid therapy for
the inability of brain cells to extrude the hypovolemic hypernatremia is 0.18 % NaCl
osmoprotective solutes that accumulate during given slowly over 48 h compared to 0.45 % saline
sustained hyperosmolal conditions in parallel given slowly or rapidly over the same time period
with the decline in plasma osmolality during [165]. Because hyperosmolality impairs insulin
fluid therapy [163, 164]. Therefore, the osmolal and PTH release, patients should be monitored
gradient will be reversed with plasma osmolality for hyperglycemia and hypocalcemia during the
lower than cerebral cell osmolality leading to ICW correction period.
212 N. Madden and H. Trachtman

Table 10 Causes of hypernatremia preterm infant with respiratory distress syndrome.


J Pediatr. 1986;109:509–14.
Hypovolemic: ECF volume contraction
4. Skorecki KL, Brenner BM. Body fluid homeostasis in
Gastrointestinal (diarrhea and vomiting) man. Am J Med. 1981;70:77–88.
Evaporative (high fever, high ambient temperature) 5. Holliday MA. Extracellular fluid and its proteins:
Hypothalamic diabetes insipidus (ADH deficiency) dehydration, shock, and recovery. Pediatr Nephrol.
Head trauma 1999;13:989–95.
6. Nielsen S, Kwon TH, Frokiaer J, Agre P. Regulation
Infarction (Sheehan’s syndrome)
and dysregulation of aquaporins in water balance
Tumors (e.g., craniopharyngioma) disorders. J Intern Med. 2007;261:53–64.
Histiocytosis 7. Devuyst O. Water transport across biological
Degenerative brain diseases membranes: Overton, water channels, and peritoneal
Infections dialysis. Bull Mem Acad R Med Belg. 2010;165:
Hereditary central diabetes insipidus (usually 250–5.
dominant) 8. Kaplan JH. Biochemistry of Na,K-ATPase. Annu Rev
Biochem. 2002;71:511–35.
Idiopathic
9. Huang CL, Kuo E. Mechanism of disease: WNK-ing
Nephrogenic diabetes insipidus (ADH resistance) at the mechanism of salt-sensitive hypertension. Nat
Chronic renal failure Clin Pract Nephrol. 2007;3:623–30.
Hypokalemia 10. Trachtman H, Futterweit S, Tonidandel W, Gullans
Hypercalcemia SR. The role of organic osmolytes in the cerebral cell
volume regulatory response to acute and chronic renal
Damage to renal medulla
failure. J Am Soc Nephrol. 1993;12:1913–9.
Sickle cell disease 11. Kopp C, Linz P, Dahlmann A, et al. 23Na magnetic
Nephronophthisis resonance imaging determined tissue sodium in
Renal papillary necrosis healthy subjects and hypertensive patients. Hyperten-
Chronic pyelonephritis (reflux nephropathy) sion. 2013;61:635–40.
Euvolemic: normal ECF volume 12. Titze J, Dahlmann A, Lerchl K, Kopp C, Rakova N,
Schroder A, Luft FC. Spooky sodium balance. Kid-
Unconscious patients
ney Int. 2014;85(4):759–67.
Infants 13. Dahlmann A, Dorfelt K, Eicher F, Linz P, et al. Mag-
Lack of access to water (lost in the desert) netic resonance-determined sodium removal from tis-
Primary adipsia sue stores in hemodialysis patients. Kidney Int
Essential hypernatremia (osmoreceptor destruction or 2015;87:434–41.
malfunction) 14. Glaser DS. Utility of the serum osmolal gap in the
diagnosis of methanol or ethylene glycol ingestion.
Hypervolemic: ECF volume expansion
Ann Emerg Med. 1996;27:343–6.
Inappropriate IV fluid therapy 15. Schelling JR, Howard RL, Winter SD, et al. Increased
Salt poisoning osmolal gap in alcoholic ketoacidosis and lactic aci-
Mineralocorticoid excess dosis. Ann Intern Med. 1990;113:580–2.
16. Lava SAG, Biachetti MG, Simonetti GD. Sodium
intake in children and its consequence on blood pres-
Acknowledgment The author wishes to thank Laura sure. Pediatr Nephrol. 2014; online.
Malaga-Dieguez, MD, PhD, for her careful review of this 17. Haycock GB. The influence of sodium on growth in
chapter. infancy. Pediatr Nephrol. 1993;7(6):871–5.
18. Wassner SJ. Altered growth and protein turnover in
rats fed sodium-deficient diets. Pediatr Res. 1989;26:
608–13.
References 19. Reungjui S, Hu H, Mu W, Roncal CA, Croker BP,
Patel JM, Nakagawa T, Srinivas T, Byer T, Simoni J,
1. Thomas DR, Cote TR, Lawhorne L, Levenson SA, Sitprija V, Johnson RJ. Thiazide-induced subtle renal
Rubenmstein LZ, Smith DA, Stefanacci RG, injury not observed in states of equivalent hypokale-
Tangalos EG, Morley JE. Understanding clinical mia. Kidney Int. 2007;72:1483–92.
dehydration and its treatment. J Am Med Dir Assoc. 20. Wassner SJ. The effect of sodium repletion on growth
2008;9:292–301. and protein turnover in sodium-depleted rats. Pediatr
2. Fatehi P. Salt and water: a brief natural history. Kid- Nephrol. 1991;5:501–4.
ney Int. 2008;64:3–4. 21. Al-Dahhan J, Jannoun L, Haycock G. Effect of salt
3. Shaffer SG, Bradt SK, Hall RT. Postnatal changes in supplementation of newborn premature infants on
total body water and extracellular volume in the neurodevelopmental outcome at 10–13 years of age.
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 213

Arch Dis Child Fetal Neonatal Ed. 2002;86(2): 33. Holliday MA, Segar ME. The maintenance need for
F120–3. water in parenteral fluid therapy. Pediatrics.
22. Sulyok E, Varga F, Nemeth M, Tenyi I, Csaba IF, 1957;19:823–32.
Ertl T, Gyory E. Furosemide-induced alterations in 34. Holliday MA, Friedman AL, Segar WE, Chesney R,
electrolyte status, the function of the renin- Finberg L. Acute hospital-induced hyponatremia in
angiotensin-aldosterone system, and the urinary children: a physiologic approach. J Pediatr.
excretion of prostaglandins in newborn infants. 2004;145:584–7.
Pediatr Res. 1980;14:765–8. 35. Ray PE. Neurological complications from
23. O’Donnell M, Mente A, Rangarajan S, McQueen MJ, dysnatremias in children: a different point of view.
Wang X, Liu L, Yan H, Lee SF, Mony P, Devanath A, Pediatr Nephrol. 2006;21:1048–9.
Rosengren A, Lopez-Jaramillo P, Diaz R, Avezum A, 36. Holliday MA, Ray PE, Friedman AL. Fluid therapy
Lanas F, Yusoff K, Iqbal R, Ilow R, for children: facts, fashions, and questions. Arch Dis
Mohammadifard N, Gulec S, Yusufali AH, Child. 2007;92:546–50.
Kruger L, Yusuf R, Chifamba J, Kabali C, 37. Moritz ML, Ayus JC. Prevention of hospital-acquired
Dagenais G, Lear SA, Teo K, Yusuf S, PURE Inves- hyponatremia: a case for using isotonic saline. Pedi-
tigators. Urinary sodium and potassium excretion, atrics. 2003;111:227–30.
mortality, and cardiovascular events. N Engl J Med. 38. Adrogue HJ, Madias NE. Hyponatremia. N Engl J
2014;371:612–23. Med. 2000;342:1581–9.
24. Mente A, O’Donnell MJ, Rangarajan S, McQueen 39. Achinger SG, Moritz ML, Ayus JC. Dysnatremias:
MJ, Poirier P, Wielgosz A, Morrison H, Li W, why are patients still dying? South Med J. 2006;
Wang X, Di C, Mony P, Devanath A, Rosengren A, 99:353–62.
Oguz A, Zatonska K, Yusufali AH, Lopez-Jaramillo P, 40. Moritz ML, Ayus JC. Hospital-acquired
Avezum A, Ismail N, Lanas F, Puoane T, Diaz R, hyponatremia – why are hypotonic parenteral fluids
Kelishadi R, Iqbal R, Yusuf R, Chifamba J, still being used? Nat Clin Pract Nephrol. 2007;3:
Khatib R, Teo K, Yusuf S, PURE Investigators. Asso- 374–82.
ciation of urinary sodium and potassium excretion 41. Choong K, Bohn D. Maintenance parenteral fluids in
with blood pressure. N Engl J Med. 2014;371:601–11. the critically ill child. J Pediatr (Rio J). 2007;83
25. Mozaffarian D, Fahimi S, Singh GM, Micha R, (2 Suppl):S3–10.
Khatibzadeh S, Engell RE, Lim S, Danaei G, 42. Choong K, Arora S, Cheng J, Farrokhyar F, Reddy D,
Ezzati M, Powles J, Global Burden of Diseases Nutri- Thabane L, et al. Hypotonic versus isotonic mainte-
tion and Chronic Diseases Expert Group. Global nance fluids after surgery for children: a randomized
sodium consumption and death from cardiovascular controlled trial. Pediatrics. 2011;128:857–66.
causes. N Engl J Med. 2014;371:624–34. 43. Foster BA, Tom D, Hill V. Hypotonic versus isotonic
26. Fan L, Tighiouart H, Levey AS, Beck GJ, Sarnak fluids in hospitalized children: a systematic review and
MJ. Urinary sodium excretion and kidney failure in meta-analysis. J Pediatr. 2014;165(1):163–9. e162.
nondiabetic chronic kidney disease. Kidney Int. 44. Ince C, Johan Groeneveld AB. The case for 0.9 %
2014;86:582–8. NaCl: is the undefendable, defensible? Kidney Int.
27. Holmes E, Loo RL, Stamler J, Bictash M, Yap IKS, 2014;86:1087–95.
Chan Q, Ebbels T, De Iorio M, Brown IJ, Veselkov 45. Yunos NM, Bellomo R, Hegarty C, Story D, Ho L,
KA, Daviglus ML, Kesteloot H, Ueshima H, Zhao L, Bailey M. Association between a chloride-liberal vs
Nicholson JK, Elliott P. Human metabolic phenotype chloride-restrictive intravenous fluid administration
diversity and its association with diet and blood pres- strategy and kidney injury in critically ill adults.
sure. Nature. 2008;453:396–400. JAMA. 2012;308:1566–72.
28. Holliday M. The evolution of therapy for dehydra- 46. Danovitch GM, Bourgoignie J, Bricker
tion: should deficit therapy still be taught? Pediatrics. NS. Reversibility of the “salt losing” tendency of
1996;98:171–7. chronic renal failure. N Engl J Med. 1976;296:14–9.
29. Bell EF, Neidich GA, Cashore WJ, et al. Combined 47. Di Somma S, Navarin S, Giordano S, Spadini F,
effect of radiant warmer and phototherapy on insen- Lippi G, Cervellin G, et al. The emerging role of
sible water loss in low-birth weight infants. J Pediatr. biomarkers and bio-impedance in evaluating hydra-
1979;94:810–3. tion status in patients with acute heart failure. Clin
30. Schoorlemmer GH, Evered MD. Water and solute Chem Lab Med. 2014;50(12):2093–105.
balance in rats during 10 h water deprivation and 48. Kushner RF, Schoeller DA, Fjeld CR, Danford L. Is
rehydration. Can J Physiol Pharmacol. 1993;71: the impedance index (ht2/R) significant in predicting
379–86. total body water? Am J Clin Nutr. 1992;56(5):835–9.
31. Berl T. Impact of solute intake on urine flow and water 49. Hyun SH, Choi JY, Cho JH, Park SH, Kim CD, Kim
excretion. J Am Soc Nephrol. 2008;19:1076–8. YL. Assessment of fluid and nutritional status using
32. Wang CJ, Grantham JJ, Wetmore JB. The medicinal multifrequency bioelectrical impedance analysis in
use of water in renal disease. Kidney Int. 2013;84: peritoneal dialysis patients. Blood Purif. 2014;37(2):
45–53. 152–62.
214 N. Madden and H. Trachtman

50. Trachtman H. Cell volume regulation: a review of 68. Blair-West JR, Carey KD, Denton DA, Weisinger RS,
cerebral adaptive mechanisms and implications for Shade RE. Evidence that brain angiotensin II is
clinical treatment of osmolal disturbances. Pediatr involved in both thirst and sodium appetite in
Nephrol. 1991;5:743–52 (Part I) and 1992;6: baboons. Am J Physiol. 1998;275:R1639–46.
104–112 (Part II). 69. Ichikawa I, Harris RC. Angiotensin actions in the
51. Kleeman CR. Metabolic coma. Kidney Int. kidney: renewed insight into the old hormone. Kidney
1989;36:1142–58. Int. 1991;40:583–9.
52. Huang AL, Chen X, Hoon MA, Chandrashekar J, 70. Santos RA, Ferreira AJ, Simoes E, Silva AC. Recent
Guo W, Trankner D, Ryba NJP, Zuker CS. The cells advances in the angiotensin converting enzyme2-
and logic for mammalian sour taste detection. Nature. angiotensin(1-7)-Mas axis. Exp Physiol.
2006;442:934–8. 2008;93:519–27.
53. Morita H, Tanaka K, Hosomi H. Chemical inactiva- 71. Tsutsumi Y, Matsubara H, Masaki H, et al. Angioten-
tion of the nucleus tractus solitarius abolished sin II type 2 receptor overexpression activates the
hepatojejunal reflex in the rat. J Auton Nerv Syst. vascular kinin system and causes vasodilatation. J
1994;48:207–12. Clin Invest. 1999;104:925–35.
54. Firth JD, Raine AEG, Ledingham JGG. Raised 72. Liu FY, Cogan MG. Role of angiotensin II in
venous pressure: a direct cause of renal sodium reten- glomerulotubular balance. Am J Physiol. 1990;259:
tion in edema. Lancet. 1988;331:1033–5. F72–9.
55. Fater D, Schultz HD, Sundet WD, et al. Effects of left 73. Matsusaka T, Niimura F, Pastan I, Shintani A,
atrial stretch in cardiac-denervated and intact con- Nishiyama A, Ichikawa I. Podocyte injury enhances
scious dogs. Am J Physiol. 1982;242:H1056–64. filtration of liver-derived angiotensinogen and renal
56. Levy M, Wexler MJ. Sodium excretion in dogs with angiotensin II generation. Kidney Int. 2014;85:
low-grade caval constriction: role of hepatic nerves. 1068–77.
Am J Physiol. 1987;253:F672–8. 74. Verrey F. Early aldosterone action: toward filling the
57. Paintal AS. Vagal sensory receptors and their reflex gap between transcription and transport. Am J
effects. Physiol Rev. 1973;53:159–227. Physiol. 1999;277:F319–27.
58. Kirchheim HR, Ehmke H, Hackenthal E, 75. Leopold JA, Dam A, Maron BA, Scribner AW,
et al. Autoregulation of renal blood flow, glomerular Liao R, Handy DE, Stanton RC, Pitt B, Loscalzo
filtration rate, and renin release in conscious dogs. J. Aldosterone impairs vascular reactivity by decreas-
Pflugers Arch. 1987;410:441–9. ing glucose-6-phosphate dehydrogenase activity. Nat
59. Quail AW, Woods RL, Korner PI. Cardiac and arterial Med. 2007;13:189–97.
baroreceptor influences in release of vasopressin and 76. Fujita T. Mechanism of salt sensitive hypertension:
renin during hemorrhage. Am J Physiol. 1987;252: focus on adrenal and sympathetic nervous system. J
H1120–6. Am Soc Nephrol. 2014;25:1148–55.
60. Harrigan MR. Cerebral salt wasting syndrome: a 77. Hunley TE, Kon V. Update on endothelins: biology
review. Neurosurgery. 1996;38:152–60. and clinical implications. Pediatr Nephrol.
61. Feng Y, Yue X, Xia H, Bindom SM, Hickman PJ, 2001;16:752–62.
Filipeanu CM, Wu G, Lazartigues E. Angiotensin 78. DiBona GF. Neural control of the kidney: function-
converting enzyme 2 overexpression in the ally specific renal sympathetic nerve fibers. Am J
subfornical organ prevents angiotensin II-mediated Physiol. 2000;279:R1517–24.
pressor and drinking responses and is associated 79. Jose PA, Eisner GM, Felder RA. Role of dopamine
with angiotensin II type I1 receptor downregulation. receptors in the kidney in the regulation of blood pres-
Circ Res. 2008;102:729–36. sure. Curr Opin Nephrol Hypertens. 2002;11:87–92.
62. Schrier RW. Pathogenesis of sodium and water reten- 80. Armando I, Konkalmatt P, Felder RA, Jose PA. The
tion in high-output and low-output cardiac failure, renal dopaminergic system: novel diagnostic and ther-
nephrotic syndrome, cirrhosis, and pregnancy. N apeutic approaches in hypertension and kidney dis-
Engl J Med. 1988;319:1065–72. & 1127–1134. ease. Transl Res. 2014. doi:10.1016/j.
63. Schrier RW. Hormones and hemodynamics in heart trsl.2014.07.006. pii: S1931-5244(14)00269-2.
failure. N Engl J Med. 1999;341:577–85. 81. Carvalho MJ, Van den Meiracker AH, Boomsma F,
64. Share L, Kimura T, Matsui K, Shade RE, Crofton JT. et al. Role of sympathetic nervous system in
Metabolism of vasopressin. Fed Proc. 1985;44:59–61. cyclosporine-induced rise in blood pressure. Hyper-
65. Hays RM. Antidiuretic hormone. N Engl J Med. tension. 1999;34:102–6.
1976;295:659–65. 82. Hoorn EJ, Walsh SB, McCormick JA, F€ urstenberg A,
66. Robinson AG, Roberts MM, Evron WA, et al. Yang CL, Roeschel T, Paliege A, Howie AJ, Conley J,
Hyponatremia in rats induces downregulation of Bachmann S, Unwin RJ, Ellison DH. The calcineurin
vasopressin synthesis. J Clin Invest. 1990;86:1023–9. inhibitor tacrolimus activates the renal sodium chlo-
67. Robertson GL. Abnormalities of thirst regulation. ride cotransporter to cause hypertension. Nat Med.
Kidney Int. 1984;25:460–9. 2011;17:1304–9.
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 215

83. Levin ER, Gardner DG, Samson WK. Natriuretic 98. Boyle LD, Wilding JP. Emerging sodium/glucose
peptides. N Engl J Med. 1998;339:321–8. co-transporter 2 inhibitors for type 2 diabetes. Expert
84. Martinez-Rumayor A, Richards AM, Burnett JC, Opin Emerg Drugs. 2013;18:375–91.
Januzzi Jr JL. Biology of natriuretic peptides. Am J 99. Andrukhova O, Slavic S, Smorodchenko A, Zeitz U,
Cardiol. 2008;101:3–8. Shalhoub V, Lanske B, Pohl EE, Erben RG. FGF23
85. Cody RJ, Atlas SA, Laragh JH. Atrial natriuretic regulates renal sodium handling and blood pressure.
factor in normal subjects and heart failure patients: EMBO Mol Med. 2014;6:744–59.
plasma levels and renal, hormonal, and hemodynamic 100. Taylor MM, Samson WK. Adrenomedullin and the
responses to peptide infusion. J Clin Invest. 1986;78: integrative physiology of fluid and electrolyte bal-
1362–74. ance. Microsc Res Tech. 2002;57:105–9.
86. Smith WL, Garavito RM, DeWitt DL. Prostaglandin 101. Alessi DR, Zhang J, Khanna A, Hochdörfer T,
endoperoxide H synthases (cyclooxygenases)-1 and Shang Y, Kahle KT. The WNK-SPAK/OSR1 path-
–2. J Biol Chem. 1996;271:33157–60. way: master regulator of cation-chloride
87. Patrono C, Dunn MJ. The clinical significance of co-transporters. Sci Signal. 2014;7(334):re3.
inhibition of renal prostaglandin synthesis. Kidney doi:10.1126/scisignal.2005365.
Int. 1987;32:1–12. 102. Bolignano D, Cabassi A, Fiaccadori E, Ghigo E,
88. Gueutin V, Vallet M, Jayet M, Pet-Peterdi J, Pasquali R, Peracino A, Peri A, Plebani M,
Corniere N, Leviel F, Sohet F, Wagner CA, Santoro A, Settanni F, Zoccali C. Copeptin
Eladari D, Chambrey R. Renal β-intercalated cells (CTproAVP), a new tool for understanding the role of
maintain body fluid and electrolyte balance. J Clin vasopressin in pathophysiology. Clin Chem Lab Med.
Invest. 2013;123:4219–31. 2014. doi:10.1515/cclm-2014-0379. pii: /j/cclm.
89. Wang T. Role of iNOS and eNOS in modulating ahead-of-print/cclm-2014-0379/cclm-2014-0379.xml.
proximal tubule transport an acid-base balance. Am 103. Phillips PA, Rolls BJ, Ledingham JGG, et al. Reduced
J Physiol. 2002;283:F658–62. thirst after water deprivation in healthy elderly men. N
90. Stoos BA, Carretero OA, Garvin JL. Endothelial- Engl J Med. 1984;311:753–9.
derived nitric oxide inhibits sodium transport by 104. Thompson CJ, Burd JM, Baylis PH. Acute suppres-
affecting apical membrane channels in cultured sion of plasma vasopressin and thirst after drinking in
collecting duct cells. J Am Soc Nephrol. 1994;4: hypernatremic humans. Am J Physiol. 1987;252:
1855–60. R1138–42.
91. Schultz PJ, Tolins JP. Adaptation to increased dietary 105. Dirks JH, Cirkensa WJ, Berliner RW. The effect of
salt intake: role of endogenous nitric oxide. J Clin saline infusion on sodium reabsorption by the proxi-
Invest. 1992;91:642–50. mal tubule of the dog. J Clin Invest. 1965;44:
92. Kagota S, Tamashiro A, Yamaguchi Y, et al. 1875–85.
Downregulation of soluble guanylate cyclase induced 106. Brenner BM, Troy JL, Daugharty TM. On the mech-
by high salt intake in spontaneously hypertensive rats. anism of inhibition of fluid reabsorption by the renal
Br J Pharmacol. 2001;134:737–44. proximal tubule of the volume-expanded rat. J Clin
93. Phipps JA, Feener EP. The kallikrein-kinin system in Invest. 1971;50:1596–602.
diabetic retinopathy: lessons for the kidney. Kidney 107. Better OS, Schrier RW. Disturbed volume homeosta-
Int. 2008;73:1114–9. sis in patients with cirrhosis of the liver. Kidney Int.
94. Tonel J, Madrid MI, Garcia-Salom M, Wirth KJ, 1983;23:303–11.
Fenoy FJ. Role of kinins in the control of renal pap- 108. Hricik D, Broning PJ, Kopelman R, Goorno WE,
illary blood flow, pressure natriuresis, and arterial Madias NE, Dzau VJ. Captopril-induced functional
pressure. Circ Res. 2000;86:589–95. renal insufficiency in patients with bilateral renal
95. Fuster DG, Bobulescu IA, Zhang J, Wade J, Moe artery stenosis or renal artery stenosis in a solitary
OW. Characterization of the regulation of renal Na+/ kidney. N Engl J Med. 1983;308:373–6.
H+ exchanger NHE3 by insulin. Am J Physiol Renal 109. Verry F, Fakitsas P, Adam G, Staub O. Early tran-
Physiol. 2007;292:F577–85. scriptional control of ENaC (de)ubiquitylation by
96. Chávez-Canales M, Arroyo JP, Ko B, Vázquez N, aldosterone. Kidney Int. 2008;73:691–6.
Bautista R, Castañeda-Bueno M, Bobadilla NA, Hoo- 110. Stokes JB. Disorders of the epithelial sodium
ver RS, Gamba G. Insulin increases the functional channel: insights into the regulation of extracellular
activity of the renal NaCl cotransporter. J Hypertens. volume and blood pressure. Kidney Int. 1999;56:
2013;31:303–11. 2318–33.
97. Nishida H, Sohara E, Nomura N, Chiga M, Alessi 111. Staub O, Verrey F. Impact of Nedd4 proteins and
DR, Rai T, Sasaki S, Uchida S. Phosphatidylinositol serum and glucocorticoid-induced kinases on epithe-
3-kinase/Akt signaling pathway activates the lial Na + transport in the distal nephron. J Am Soc
WNK-OSR1/SPAK-NCC phosphorylation cascade Nephrol. 2005;16:3167–74.
in hyperinsulinemic db/db mice. Hypertension. 112. Harris Jr HW, Strange K, Zeidel ML. Current under-
2012;60:981–90. standing of the cellular biology and molecular
216 N. Madden and H. Trachtman

structure of the antidiuretic hormone-stimulated water 127. Duck SC, Wyatt DT. Factors associated with brain
transport pathway. J Clin Invest. 1991;88:1–8. herniation in the treatment of diabetic ketoacidosis.
113. Knoers NV, Deen PM. Molecular and cellular defects J Pediatr. 1988;113:10–4.
in nephrogenic diabetes insipidus. Pediatr Nephrol. 128. Harris GD, Fiordalisi I. Physiologic management of
2001;16:1146–52. diabetic ketoacidemia: a 5-year prospective experi-
114. Feldman BJ, Rosenthal SM, Vargas GA, Fenwick ence in 231 episodes. Arch Pediatr Adolesc Med.
RG, Huang EA, Matsuda-Abedini M, Lustig RH, 1994;148:1046–52.
Mathias RS, Portale AA, Miller WL, Gitelman 129. Saavedra JM, Harris GD, Finberg L. Capillary refill
SE. Nephrogenic syndrome of inappropriate (skin turgor) in the assessment of dehydration. Am J
antidiuresis. N Engl J Med. 2005;352:1884–90. Dis Child. 1991;145:296–8.
115. Rochdi MD, Vargas GA, Carpentier E, Oligny- 130. Prevot A, Mosig D, Guignard JP. The effects of
Longpré G, Chen S, Kovoor A, Gitelman SE, losartan on renal function in the newborn rabbit.
Rosenthal SM, von Zastrow M, Bouvier Pediatr Res. 2002;51:728–32.
M. Functional characterization of vasopressin type 131. Trachtman H. Volume depletion state: dehydration or
2 receptor substitutions (R137H/C/L) leading to denatration? Pediatr Nephrol. 1991;5:271–2.
nephrogenic diabetes insipidus and nephrogenic syn- 132. Mange K, Matsuura D, Cizman B, et al. Language
drome of inappropriate antidiuresis: implications for guiding therapy: the case of dehydration versus
treatments. Mol Pharmacol. 2010;77:836–45. volume depletion. Ann Intern Med. 1997;127:
116. Kozono D, Yasui M, King LS, Agre P. Aquaporin 848–53.
water channels: atomic structure and molecular 133. Mackenzie A, Barnes G, Shann F. Clinical signs of
dynamics meet clinical medicine. J Clin Invest. dehydration in children. Lancet. 1989;334:605–7.
2002;109:1395–9. 134. Schierhout G, Roberts I. Fluid resuscitation with col-
117. Deen PM, Verdijk MA, Knoers NV, Wieringa B, loid or crystalloid solutions in critically ill patients: a
Monnens LA, Van Os CH, Van Oost systematic review of randomized trials. BMJ. 1998;
BA. Requirement of human renal water channel 316:961–4.
aquaporin-2 for vasopressin-dependent concentration 135. Patel A, Laffan MA, Waheed U, Brett
of urine. Science. 1994;264:92–5. SJ. Randomised trials of human albumin for adults
118. Ma T, Song Y, Yang B, et al. Nephrogenic diabetes with sepsis: systematic review and meta-analysis with
insipidus in mice lacking aquaporin-3 water channel. trial sequential analysis of all-cause mortality. BMJ.
Proc Natl Acad Sci USA. 2000;97:4386–91. 2014;349:g4561. doi:10.1136/bmj.g4561.
119. Morello JP, Bichet DG. Nephrogenic diabetes 136. Mutter TC, Ruth CA, Dart AB. Hydroxyethyl
insipidus. Annu Rev Physiol. 2001;63:603–30. starch (HES) versus other fluid therapies: effects on
120. Verkman AS, Anderson MO, Papadopoulos kidney function. Cochrane Database Syst Rev.
MC. Aquaporins: important but elusive drug targets. 2013;7, CD007594. doi:10.1002/14651858.CD00
Nat Rev Drug Discov. 2014;13(4):259–77. 7594.pub3.
121. Kirchlechner V, Koller DY, Seidl R, Waldhauser 137. Holliday MA, Friedman AL, Wassner SJ. Extracellu-
F. Treatment of nephrogenic diabetes insipidus with lar fluid restoration in dehydration: a critique of rapid
hydrochlorothiazide and amiloride. Arch Dis Child. versus slow. Pediatr Nephrol. 1999;13:292–7.
1999;80:548–52. 138. Pizarro D, Posada G, Sandi L, Moran JR. Rice-based
122. Kim GH, Lee JW, Oh YK, Chang HR, Joo KW, Na electrolyte solutions for the management of infantile
KY, Earm JH, Knepper MA, Han JS. Antidiuretic diarrhea. N Engl J Med. 1991;324:517–21.
effect of hydrochlorothiazide in lithium-induced 139. Ramakrishna BS, Venkataraman S, Srinivasan P,
nephrogenic diabetes insipidus is associated with et al. Amylase-resistant starch plus oral rehydration
upregulation of aquaporin-2, Na-Cl co-transporter, solution for cholera. N Engl J Med. 2000;342:
and epithelial sodium channel. J Am Soc Nephrol. 308–13.
2004;15:2836–43. 140. Rodriguez-Iturbe B, Herrera-Acosta J, Johnson
123. Berliner RW. Mechanisms of urine concentration. RJ. Interstitial inflammation, sodium retention, and
Kidney Int. 1982;22:202–11. the pathogenesis of nephrotic edema: a unifying
124. Cai Q, Michea L, Andrews P, Zhang Z, Rocha G, hypothesis. Kidney Int. 2002;62:1379–84.
Dmitrieva N, Burg MB. Rate of increase of osmolality 141. Haws RM, Baum M. Efficacy of albumin and diuretic
determines osmotic tolerance of mouse inner medul- therapy in children with nephrotic syndrome. Pediat-
lary epithelial cell. Am J Physiol. 2002;283:F792–8. rics. 1993;91:1142–6.
125. Ayus JC, Arieff AI. Chronic hyponatremic encepha- 142. Forni LG, Hilton PJ. Continuous hemofiltration in the
lopathy in postmenopausal women: association of treatment of acute renal failure. N Engl J Med.
therapies with morbidity and mortality. J Am Med 1997;336:1303–9.
Assoc. 1999;281:2299–304. 143. Marcialis MA, Dessi A, Pintus MC, Irmesi R,
126. Trachtman H, Yancey PH, Gullans SR. Cerebral cell Fanos V. Neonatal hyponatremia: differential diagno-
volume regulation during hypernatremia in develop- sis and treatment. J Matern Fetal Neonatal Med.
ing rats. Brain Res. 1995;693:155–62. 2011;24 Suppl 1:75–9.
7 Physiology of the Developing Kidney: Sodium and Water Homeostasis and Its Disorders 217

144. Modi N. Hyponatraemia in the newborn. Arch Dis with vasopressin antagonist or hypertonic saline. Kid-
Child Fetal Neonatal Ed. 1998;78(2):F81–4. ney Int. 2015:87(2):323–31.
145. Suarez-Rivera M, Bonilla-Felix M. Fluid and electro- 155. Miller M. Inappropriate antidiuretic hormone secre-
lyte disorders in the newborn: sodium and potassium. tion. Curr Ther Endocrinol Metab. 1994;5:186–9.
Curr Pediatr Rev. 2014;10(2):115–22. 156. Fujisawa G, Ishikawa S, Tsuboi Y, et al. Therapeutic
146. Williams CN, Belzer JS, Riva-Cambrin J, Presson AP, efficacy of non-peptide ADH antagonist OPC-31260
Bratton SL. The incidence of postoperative in SIADH rats. Kidney Int. 1993;44:19–23.
hyponatremia and associated neurological sequelae 157. Shimizu K. aquaretic effect of the nonpeptide V2
in children with intracranial neoplasms. J Neurosurg antagonist OPC-31260 in hydropenic humans. Kid-
Pediatr. 2014;13:283–90. ney Int. 1995;48:220–6.
147. Sersté T, Gustot T, Rautou PE, Francoz C, Njimi H, 158. Lehrich RW, Greenberg A. When is it appropriate to
Durand F, Valla D, Lebrec D, Moreau R. Severe use vasopressin receptor antagonists? J Am Soc
hyponatremia is a better predictor of mortality than Nephrol. 2008;19:1054–8.
MELDNa in patients with cirrhosis and refractory 159. Moritz ML, Ayus JC. The changing pattern of
ascites. J Hepatol. 2012;57(2):274–80. hypernatremia in hospitalized children. Pediatrics.
148. Fraser CL, Arieff AI. Epidemiology, pathophysiol- 1999;104:435–9.
ogy, and management of hyponatremic encephalopa- 160. Cooper WO, Atherton HD, Kahana M, Kotagal
thy. Am J Med. 1997;102:67–77. UR. Increased incidence of severe breastfeeding mal-
149. Ayus CC, Armstrong D, Arieff AI. Hyponatremia nutrition and hypernatremia in a metropolitan area.
with hypoxia: effects on brain adaptation, perfusion, Pediatrics. 1995;96:957–60.
and histology in rodents. Kidney Int. 2006;69: 161. Adrogue HJ, Madias NE. Hypernatremia. N Engl J
1319–25. Med. 2000;342:1493–9.
150. Sterns RH. Severe hyponatremia: the case for 162. Finberg L, Luttrell C, Redd H. Pathogenesis of lesion
conservative management. Crit Care Med. 1992;20: in the nervous system in hypernatremic states.
534–9. II. Experimental studies of gross anatomic changes
151. Berl T. Treating hyponatremia: damned if we do and and alterations of chemical composition of the tissues.
damned if we don’t. Kidney Int. 1990;37:1006–18. Pediatrics. 1959;23:46–53.
152. Sarnaik AP, Meert K, Hackbarth R, et al. Manage- 163. Lee JH, Arcinue E, Ross BD. Organic osmolytes in
ment of hyponatremic seizures in children with hyper- the brain of an infant with hypernatremia. N Engl J
tonic saline: a safe and effective strategy. Crit Care Med. 1994;334:439–42.
Med. 1991;19:758–62. 164. Lien YHH, Shapiro JI, Chan L. Study of brain electro-
153. Keating JP, Schears GJ, Dodge PR. Oral water intox- lytes and organic osmolytes during correction of chronic
ication in infants: an American epidemic. Am J Dis hyponatremia: implications for the pathogenesis of cen-
Child. 1991;145:985–90. tral pontine myelinolysis. J Clin Invest. 1991;88:303–9.
154. Gankam Kengne F, Couturier BS, Soupart A, Decaux 165. Banister A, Matin-Siddiqi SA, Hatcher G. Treatment
G. Urea minimizes brain complications following of hypernatremic dehydration in infancy. Arch Dis
rapid correction of chronic hyponatremia compared Child. 1975;50:179–86.
Physiology of the Developing Kidney:
Potassium Homeostasis and Its Disorder 8
Lisa M. Satlin and Detlef Bockenhauer

Contents Potassium is the most abundant intracellular


Potassium Homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 219 cation. Approximately 98 % of the total body
potassium content is located within cells, primar-
Regulation of Internal Potassium Balance . . . . . . . 220
Plasma Potassium Concentration . . . . . . . . . . . . . . . . . . . . 222
ily muscle, where its concentration ranges from
Hormones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 222 100 to 150 mEq/l; the remaining 2 % resides in the
Acid–Base Balance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 223 extracellular fluid, where the potassium concen-
Other Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224 tration is tightly regulated within a narrow
Regulation of External Potassium Balance . . . . . . . 224 range (3.5–5.0 mEq/l in the adult). The ratio of
Renal Contribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 224 the intra- to extracellular potassium concentration
Sites of Potassium Transport Along the Nephron . . . 225
determines, in large part, the resting membrane
Luminal and Peritubular Factors Affecting potential and is thus critical for normal function of
Potassium Transport . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231 electrically excitable cells, including nerve and
Sodium Delivery and Absorption . . . . . . . . . . . . . . . . . . . 231
Tubular Flow Rate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232 muscle. Maintenance of a high intracellular
Potassium Intake and Cellular Potassium potassium concentration is essential for many
Content . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 232 cellular processes, including DNA and protein
Hormones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 233 synthesis, cell growth and apoptosis, mitochon-
Acid–Base Balance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
Circadian Control of Renal Potassium Excretion . . . 235 drial enzyme function, and conservation of cell
volume and pH [1–7]. Because of the many vital
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
processes dependent on potassium homeostasis,
multiple complex and efficient mechanisms have
developed to regulate the internal distribution of
potassium between the intra- and extracellular
compartments and the external balance between
intake and excretion by the kidney and GI tract.
L.M. Satlin (*)
Department of Pediatrics, Division of Pediatric
Nephrology, Icahn School of Medicine at Mount Sinai, Potassium Homeostasis
New York, NY, USA
e-mail: lisa.satlin@mssm.edu
Total body potassium content reflects the balance
D. Bockenhauer between intake and output, the latter regulated
Great Ormond Street Hospital, Institute of Child Health,
primarily by renal and, to a lesser extent, fecal
University College London, London, UK
e-mail: d.bockenhauer@ucl.ac.uk; excretion; the amount of potassium lost through
detlef.bockenhauer@nhs.net sweat is normally negligible. The homeostatic
# Springer-Verlag Berlin Heidelberg 2016 219
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_7
220 L.M. Satlin and D. Bockenhauer

goal of the healthy adult is to remain in zero from mother to fetus [19]. This notion is further
potassium balance. Thus, ~90 % of the daily supported by studies in dog [20] and rat [21] that
potassium intake, which typically averages show that fetal plasma potassium concentrations
1 mEq/kg body weight, is eliminated by the kid- are maintained during maternal hypokalemia, an
neys and the residual ~10 % is lost through the adaptation proposed to be due to directed potas-
stool [8]. Approximately 85 % of ingested potas- sium transport towards the fetus.
sium is reabsorbed by the gut in the adult [9].
Total body potassium content increases from
approximately 8 mEq/cm body height at birth to Regulation of Internal Potassium
>14 mEq/cm body height by 18 years of age [10, Balance
11] (Fig. 1), with the rate of accumulation of body
potassium per kilogram body weight in the infant The daily dietary intake of potassium in the adult
exceeding that in the older child and adolescent. (~50–100 mEq) approaches or exceeds the total
The increase in total body potassium content cor- potassium normally present within the extracellular
relates with an increase in cell number and potas- fluid space (~70 mEq in 17 L of extracellular
sium concentration (at least in skeletal muscle) fluid with a potassium concentration averaging ~4
with advancing age [11–13]. The robust somatic mEq/l). This potassium load must ultimately be
growth early in life requires the maintenance of a excreted. However, potassium excretion by the
state of positive potassium balance [14, 15], as has kidney is sluggish. Only 50 % of an oral or intra-
been demonstrated in growing infants greater than venous load of potassium is excreted during the
approximately 30 weeks of gestational age first 4–6 h after it is administered [22, 23]. Life-
(GA) [16, 17]. The tendency to retain potassium threatening hyperkalemia is not generally observed
early in postnatal life is reflected, in part, in the during this period because of the rapid (within
higher plasma potassium values in infants and minutes) hormonally mediated translocation of
particularly in preterm neonates [17, 18]. ~80 % of the retained potassium load from the
Studies in rats suggest that the accumulation of extracellular space into cells [24]. The buffering
potassium in the growing fetus is facilitated by the capacity of the combined cellular storage
active transport of potassium across the placenta reservoirs, which includes muscle, bone, liver, and

Fig. 1 Relationship
between total body
MALES
potassium (gms) and height 100
FEMALES
(cm) for infants and
children. The rate of
accretion of body potassium
in the neonate is faster than
Total Body K (gms)

in later childhood, likely


reflecting both an increase
in cell number and
potassium concentration, at
least in skeletal muscle, 10
with advancing age (From
Flynn et al. [11])

1
10 20 30 40 50 60 70 80 90 100 110 120 130 140 150 160 170 180 190
Height (cm)
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 221

red blood cells (RBCs), is capable of sequestering Regulation of internal potassium balance in the
up to approximately 3,500 mEq of potassium and is neonate is influenced by developmental stage-
vast compared with the extracellular pool [8]. specific expression of potassium transporters,
Cells must expend a significant amount of including the Na-K-ATPase, as well as channels,
energy to maintain the steep potassium (and receptors, and signal transduction pathways [30,
sodium) concentration gradients across their cell 31]. Thirty to fifty percent of very low-birth
membranes. This is accomplished by the ubiqui- weight and premature infants <28 weeks GA
tous Na-K-ATPase which transports three sodium exhibit nonoliguric hyperkalemia during the first
ions out of and two potassium ions into the cell at 72 h of life, defined as a serum potassium concen-
the expense of the hydrolysis of adenosine tration of >7.0 mEq/l, in the presence of urinary
triphosphate (ATP). The unequal cation exchange output of >1 ml/kg/h, despite the intake of negli-
ratio produces a charge imbalance across the cell gible amounts of potassium [32–37]. This phe-
membrane, and thus the Na-K-ATPase is defined nomenon, not observed in mature infants or
as an electrogenic pump. Positively charged VLBW infants after 72 h, has been proposed to
potassium ions, present in high concentration reflect failure of the Na-K pump as well as a
within the cell, passively leak out of cells down limited secretory capacity of the kidney for potas-
a concentration gradient through ubiquitously sium [35, 36, 38]. Prenatal steroid treatment may
expressed potassium-selective channels. A steady prevent this nonoliguric hyperkalemia via induc-
state is reached at which the outward movement of tion of Na-K-ATPase activity (see below) in the
positively charged potassium is opposed by the fetus [39, 40].
negative cell potential (generally, 30 to 70 The chemical, physical, and hormonal factors
mV, cell interior negative, in mammalian cells). that acutely influence the internal balance of
At this cell equilibrium potential, the net trans- potassium are listed in Table 1 and are discussed
membrane flux of potassium is zero. below. Potassium uptake into cells is acutely stim-
The basic functional unit of the Na-K-ATPase ulated by insulin, β2-adrenergic agonists, and
is comprised of a catalytic α- and β-subunits; the alkalosis and is impaired by α-adrenergic ago-
β-subunit acts as a molecular chaperone that nists, acidosis, and hyperosmolality. Generally,
directs the correct membrane insertion of the deviations in extracellular potassium concentra-
α-subunit [25]. Na-K-ATPase is regulated by tion arising from fluctuations in internal
changes in its intrinsic activity, subcellular distri-
bution, and cellular abundance. Long-term stimu-
Table 1 Factors that regulate internal potassium balance
lation of pump activity is generally mediated by
Effect on cell uptake of
changes in gene and protein expression, whereas
potassium
short-term regulation typically results from Physiologic factors
changes in the intracellular sodium concentration, Plasma K
alterations in the phosphorylation status of the concentration
pump and/or interaction with regulatory Increase Increase
proteins, or changes in membrane trafficking of Decrease Decrease
preexisting pumps [26–28]. The α/β-heterodimer Insulin Increase
can complex with the γ-subunits phospholemman Catecholamines
in heart and skeletal muscle [26] or FXYD2 in α-Agonists Decrease
the kidney [29], which modulate pump activity. β-Agonists Increase
The cardiac glycoside digoxin binds to the Pathologic factors
catalytic α-subunit of the enzyme, inhibiting its Acid–base balance
activity. Thus, digoxin overdose can be associated Acidosis Decrease
Alkalosis Increase
with hyperkalemia, especially in the presence of a
Hyperosmolality Enhances cell efflux
concomitant perturbation of potassium
Cell breakdown Enhances cell efflux
homeostasis.
222 L.M. Satlin and D. Bockenhauer

distribution are self-limited as long as the endo- adipocytes, and brain, a response that is
crine regulation of internal balance and mecha- independent of the hormonal effects on glucose
nisms responsible for regulation of external metabolism [42]. The mechanism of insulin
balance are intact. action in these tissues differs, in part, because of
differences in the isoform composition of the
catalytic α-subunit of the pump. Insulin stimulates
Plasma Potassium Concentration Na-K-ATPase activity by promoting the
translocation of preformed pumps from intracel-
Active cellular potassium uptake by the ubiqui- lular stores to the cell surface [43–47] and/or
tous Na-K-ATPase in large part determines the increasing cytoplasmic sodium content [48–50]
intracellular pool of potassium. An increase in or the apparent affinity of the enzyme for
potassium input into the extracellular fluid space, sodium [51].
which may arise from exogenous and endogenous Basal insulin secretion is necessary to maintain
sources or result from a chronic progressive loss fasting plasma potassium concentration within the
of functional renal mass, decreases the concentra- normal range [22]. An increase in plasma potas-
tion gradient against which the Na-K-ATPase sium in excess of 1.0 mEq/l in the adult induces a
must function and thus favors an increase in cel- significant increase in peripheral insulin levels to
lular potassium uptake. Sources of exogenous aid in the rapid disposal of the potassium load, yet
potassium input may not be readily apparent and a more modest elevation of approximately 0.5
include not only diet but also potassium- mEq/l is without effect [22, 52, 53]. In the setting
containing drugs (potassium penicillin G), other of insulin deficiency (diabetes), there is a reduc-
parenteral potassium supplements (including total tion in uptake of potassium by muscle and
parenteral nutrition), salt substitutes, herbal med- liver [24].
ications, and packed RBCs [41]. The extracellular Catecholamines enhance the cell uptake of
fluid potassium concentration can also increase in potassium via stimulation of Na-K-ATPase activ-
the absence of change in total body potassium ity in skeletal muscle and hepatocytes through β2-
content in response to endogenous release of adrenergic receptors [54, 56]. The effect of epi-
potassium during tissue breakdown (intravascular nephrine on potassium balance in the adult is
hemolysis, rhabdomyolysis, tumor lysis syn- biphasic and is characterized by an initial
drome) and severe exercise, the latter mediated increase, followed by a prolonged fall in plasma
by ATP depletion and opening of potassium concentration to a final value below
ATP-dependent potassium channels. In epithelial baseline. The initial transient rise in plasma potas-
cells of the kidney and colon specifically respon- sium results from α-adrenergic receptor stimula-
sible for potassium secretion, an increase in intra- tion which causes release of potassium from
cellular potassium maximizes the concentration hepatocytes and impairs cell uptake of potassium
gradient between cell and tubular lumen, thereby [54, 57–59]. β2-Receptor stimulation, via stimu-
promoting potassium diffusion into the urinary lation of adenylate cyclase leading to generation
space and thus potassium excretion. of the second messenger cyclic adenosine
monophosphate (cAMP) and activation of down-
stream protein kinases, stimulates the sodium
Hormones pump and thus promotes enhanced uptake of
potassium by skeletal and cardiac muscle, effects
Insulin, the most important hormonal regulator of that are inhibited by nonselective β-blockers
internal potassium balance, stimulates Na-K- including propranolol and labetalol [54,
ATPase-mediated cellular potassium uptake and 59–63]. The observation that the potassium-
thus the rapid transfer of potassium from the lowering effects of insulin and epinephrine are
extracellular to the intracellular fluid space of additive suggests that their responses are mediated
insulin-responsive cells in liver, skeletal muscle, by different signaling pathways.
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 223

Plasma Potassium,
potassium secretion in the distal nephron (see
mmol/L below) or, in the presence of renal insufficiency,
by the gut (using cation exchange resins) or by
dialysis.
Bicarbonate
0 Aldosterone is best known for its effect on
transporting tissue, increasing potassium secre-
Epinephrine
tion in distal segments of the nephron and colon
(see below). Triiodothyronine (T3) also promotes
−0.5
Na-K-ATPase-mediated potassium cellular
uptake in skeletal muscle [27]. Whereas T3 had
Insulin/glucose
been thought to act as a direct transcriptional
−1.0 activator of target genes, nongenomic effects are
also important, including the stimulation of trans-
Dialysis location of Na-K-ATPase to the plasma mem-
−1.5 p < 0.01 brane by a pathway that requires activation of
MAPK and phosphatidylinositol 3-kinase (PI3K)
0 10 20 30 40 50 60 [73, 74]. The postnatal increases in Na-K-ATPase
expression in kidney, brain, and lung depend on
Fig. 2 Changes in plasma potassium concentration normal thyroid hormone status [75].
(mmol/L) during intravenous infusion of bicarbonate
(8.4 %), epinephrine, or insulin in glucose and during
hemodialysis in adult patients on maintenance hemodialysis
(From Blumberg et al. [83]) Acid–Base Balance

It is well known that the transcellular distribution


of potassium and acid–base balance are interre-
The effects of these hormones on the distribu- lated [76–79]. Whereas acidemia (increase in
tion of potassium between the intracellular and extracellular hydrogen ion concentration) is asso-
extracellular compartments have been exploited ciated with a variable increase in plasma potas-
to effectively treat disorders of homeostasis sium secondary to potassium release from the
(Fig. 2). Administration of β2-adrenoreceptor ago- intracellular compartment, alkalemia (decrease in
nists (albuterol or salbutamol via nebulizer), extracellular hydrogen ion concentration) results
which promote potassium uptake by cells, has in a shift of potassium into cells and a consequent
been used to treat acute hyperkalemia in neonates, decrease in plasma potassium. However, the
children, and adults [64–66]. A single dose of reciprocal changes in plasma potassium that
nebulized albuterol can lower serum potassium accompany acute changes in blood pH differ
by as much as 0.5 mEq/l [67, 68]. Transient side widely among the four major acid–base disorders;
effects associated with this class of drugs include metabolic disorders cause greater disturbances in
mild tachycardia, tremor, and mild vasomotor plasma potassium than do those of respiratory
flushing [69]. Administration of glucose alone origin, and acute changes in pH result in larger
(to induce endogenous insulin release) or glucose changes in plasma potassium than do chronic
plus insulin is efficacious, although patients must conditions [76].
be monitored for complications of hyperglycemia Acute metabolic acidosis after administration
(without insulin) or hypoglycemia (with insulin), of a mineral acid that includes an anion that does
especially in neonates [70–72]. It should be kept not readily penetrate the cell membrane, such as
in mind that the treatment options discussed thus the chloride of hydrochloric acid or ammonium
far are only temporizing. To remove potassium chloride, consistently results in an increase in
from the body, potassium excretion must be plasma potassium. As excess extracellular pro-
enhanced, either by the kidney by stimulating tons, unaccompanied by their impermeant anions,
224 L.M. Satlin and D. Bockenhauer

enter the cell where neutralization by intracellular Other Factors


buffers occurs, potassium is displaced from the
cells, thus maintaining electroneutrality and lead- A number of other pathologic perturbations alter
ing to hyperkalemia. However, comparable the internal potassium balance. An increase in
acidemia induced by acute organic anion acidosis plasma osmolality secondary to severe dehydra-
(lactic acid in lactic acidosis, acetoacetic and tion or administration of osmotically active agents
β-hydroxybutyric acids in uncontrolled diabetes causes water to shift out of cells. The consequent
mellitus) may not elicit a detectable change in increase in intracellular potassium concentration
plasma potassium [76, 80, 81]. In organic exaggerates the transcellular concentration gradi-
acidemia, the associated anion diffuses more ent and favors movement of this cation out of
freely into the cell and thus does not require a cells. The effect of hyperosmolality on potassium
shift of potassium from the intracellular to the balance becomes especially problematic in dia-
extracellular fluid. betic patients with hyperglycemia, in whom the
In respiratory acid–base disturbances, in which absence of insulin exacerbates the hyperkalemia.
carbon dioxide and carbonic acid readily perme- Succinylcholine, a depolarizing paralytic agent
ate cell membranes, little transcellular shift of and an agonist of nicotinic acetylcholine recep-
potassium occurs because protons are not tors, which are found predominantly in skeletal
transported in or out in association with potassium myocyte membranes, may lead to efflux of potas-
moving in the opposite direction [76]. sium from myocytes into the extracellular fluid
Changes in plasma bicarbonate concentration, under certain pathologic states associated with
independent of the effect on extracellular pH, can upregulation and redistribution of the receptors,
reciprocally affect plasma potassium concentra- including states characterized by physical or
tion [82]. Movement of bicarbonate (out of the chemical upper or lower motor neuron denerva-
cell at a low extracellular bicarbonate concentra- tion, immobilization, infection, muscle trauma or
tion and inward at a high extracellular bicarbonate inflammation, and burn injury [84, 85].
concentration) between the intracellular and Finally, parenteral administration of cationic
extracellular compartments may be causally amino acids such as lysine, arginine, or epsilon-
related to a concomitant transfer of potassium. aminocaproic acid (used to improve hemostasis in
This relationship may account for the less marked patients undergoing cardiac surgery) may lead to
increase in plasma potassium observed during electroneutral exchange of cell potassium for the
acute respiratory acidosis, a condition cationic amino acid in skeletal myocytes and thus
characterized by an acid plasma pH with an ele- hyperkalemia [86–88].
vated serum bicarbonate (hence inward net bicar-
bonate and potassium movement), as compared
with acute metabolic acidosis with a low serum Regulation of External Potassium
bicarbonate concentration (hence outward net Balance
bicarbonate and potassium movement). Though
recommended as a mainstay of therapy, alkalini- Renal Contribution
zation of the extracellular fluid with sodium bicar-
bonate to promote the rapid cellular uptake of The kidney is the major excretory organ for potas-
potassium may not be useful in patients on main- sium. In the adult, urinary potassium excretion
tenance hemodialysis for end-stage renal disease generally parallels dietary intake. However, as
(Fig. 2) [83]. However, this maneuver remains stated earlier, the renal adaptation to variations in
valuable if metabolic acidosis is at all responsible dietary intake is rather sluggish. Extreme adjust-
for the hyperkalemia. The major toxicities of ments in the rate of renal potassium conservation
bicarbonate therapy include sodium overload cannot be achieved as rapidly as for sodium, nor
and precipitation of tetany in the face of are the adjustments as complete; whereas urinary
preexisting hypocalcemia. sodium can be virtually eliminated within
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 225

3–4 days of sodium restriction, there is a minimum than older animals [95, 96]. Comparison of the
urinary potassium loss of about 10 mEq/day in the fractional delivery of potassium to the early distal
adult, even after several weeks of severe potassium tubule with that present in the final urine reveals
restriction [89]. An increase in dietary potassium that the distal nephron of the young (2-week-old)
intake is matched by a parallel increase in renal rat secretes approximately fivefold less potassium
potassium excretion within hours, yet maximal than the older (5-week-old) rat [97]. Similarly,
rates of potassium excretion are not attained for clearance studies in saline-expanded dogs also
several days after increasing potassium intake. provide indirect evidence of a diminished secre-
Children and adults ingesting an average tory and enhanced reabsorptive capacity of the
American diet that contains sodium in excess of distal nephron to potassium early in life [98]. Fur-
potassium excrete urine with a sodium-to-potas- thermore, premature neonates studied weekly
sium ratio greater than one [17, 90]. Although after birth exhibited a 50 % reduction in the frac-
breast milk and commercially available infant for- tional excretion of potassium between 26 and
mulas generally provide a sodium-to-potassium 30 week GA, in the absence of significant change
ratio of approximately 0.5–0.6, the urinary in absolute urinary potassium excretion [16]. To
sodium-to-potassium ratio in the newborn up to the extent that the filtered load of potassium
4 months of age generally exceeds 1. This high increases almost threefold during this same time
ratio may reflect the greater requirement of potas- interval, the constancy of renal potassium excre-
sium over sodium for growth. In fact, some pre- tion could be best explained by a developmental
mature (<34 weeks GA) newborns may excrete increase in the capacity of the kidney for potas-
urine with a sodium-to-potassium ratio greater sium reabsorption [16]. It is important to note that,
than 2, a finding suggesting significant salt in general, the limited potassium secretory capac-
wasting and a relative hyporesponsiveness of the ity of the immature kidney becomes clinically
neonatal kidney to mineralocorticoid activity. relevant only under conditions of potassium
This is consistent with the physiological postnatal excess.
reduction in relative size of the extracellular fluid
compartment from approximately 60 % of body
weight at 23 weeks of gestation to 40 % at term Sites of Potassium Transport Along
[17, 91, 92]. the Nephron
Net urinary potassium excretion in the fully
differentiated kidney reflects the sum of three Proximal Tubule
processes: glomerular filtration, reabsorption Potassium is freely filtered at the glomerulus.
along the proximal tubule and the loop of Henle, Thus, the concentration of potassium entering
and bidirectional transport (secretion and the proximal convoluted tubule (PCT) is similar
reabsorption) in the distal nephron [8, 93]. Most to that of plasma. Approximately 65 % of the
of the factors known to modulate potassium filtered load of potassium is reabsorbed along the
excretion do so by altering the rate of potassium initial two-thirds of the proximal tubule in the
secretion. adult, a fractional rate of reabsorption similar to
Renal potassium clearance is low in newborns, that of sodium and water (Fig. 3) [100–102].
even when it is corrected for their low glomerular A similar fraction (50–60 %) of the filtered load
filtration rate [16–18]. Infants, like adults, can of potassium is reabsorbed along the proximal
excrete potassium at a rate that exceeds its glo- tubules of suckling (13–15-day-old) rats
merular filtration, reflecting the capacity for net [97, 103].
tubular secretion. However, they are unable to Reabsorption of potassium along the early
excrete an exogenously administered potassium proximal tubule is passive, closely following that
load as efficiently as the adult [94]; specifically, of sodium and water [104], and has been proposed
the rate of potassium excretion normalized to to occur by solvent drag via the paracellular path-
body weight or kidney weight is less in newborn way and diffusion [105–107]. Solvent drag
226 L.M. Satlin and D. Bockenhauer

Fig. 3 Potassium transport along the nephron. (a) The models of potassium transport along the nephron, showing
percentages of filtered potassium reabsorbed along the apical transporters unique to discrete nephron segments
proximal tubule and thick ascending limb of the loop of and cells therein and basolateral transporters which are
Henle (TALH) are indicated for the adult (A). Arrows show similar in all nephron segments (Adapted from Giebisch
direction of net potassium transport (reabsorption or secre- [99], Fig. 3. Publisher: Nature Publishing Group Date: Nov
tion). GFR glomerular filtration rate. (b) Simplified cell 1, 2002)

depends on active sodium reabsorption, which modulation of proximal reabsorption of this cat-
generates local hypertonicity in the paracellular ion can be accounted for by alterations in sodium
compartment, providing an osmotic force driving transport.
water reabsorption that entrains potassium in the Electrogenic sodium-coupled entry of sub-
reabsorbate. Potassium diffusion is driven by the strates such as amino acids and glucose across
lumen-positive transepithelial voltage in the sec- the luminal cell membrane of the proximal tubule
ond half of the proximal tubule and the slightly as well as bicarbonate exit across the basolateral
elevated concentrations of potassium in the cell membrane is driven by the potential differ-
lumen [108]. ences across the respective cell membranes,
In the proximal tubule, as in all other nephron which are maintained by potassium flux through
segments discussed below, transepithelial sodium potassium channels [109]. Electrophysiological
reabsorption requires the coordinated function of studies in isolated perfused proximal tubules sug-
apical sodium transport proteins and the gest that potassium movement from the cell to
basolateral Na-K-ATPase which actively extrudes lumen maintains the electrical driving force for
intracellular sodium into the interstitium and sodium-coupled cotransport in the proximal
thereby maintains the low intracellular sodium tubule. Immunohistochemical studies reveal that
concentration and steep sodium concentration KCNE1, a modulatory β-subunit, and the pore-
gradient critical to the driving force for apical forming α-subunit KCNQ1, which together con-
sodium entry (Fig. 3). There is no evidence for stitute the slowly activated component of the
specific regulation of potassium reabsorption delayed rectifying potassium current in the heart,
along the proximal tubule, and most observed colocalize in the luminal membrane of the
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 227

proximal tubule in mouse kidney, as does the ensures that a continuous supply of substrate is
cyclic nucleotide-gated, voltage-activated potas- available for the apical cotransporter. Moreover,
sium channel KCNA10 [110]. The observation potassium secretion into the urinary space creates
that KCNE1 knockout mice exhibit an increased a lumen-positive transepithelial potential differ-
fractional excretion of fluid (with accompanying ence, which in turn provides a favorable electrical
volume depletion), sodium, chloride, and glucose driving force that facilitates paracellular
compared to their wild-type littermates supports a reabsorption of sodium, potassium, calcium, and
role for KCNE1 in repolarizing the membrane magnesium.
potential in proximal tubule in response to The luminal potassium secretory channel in the
sodium-coupled transport [111]. However, TALH has been identified as ROMK, a channel
although mutations in KCNQ1 [112] and originally cloned from the TALH in the rat outer
KCNE1 [113, 114] give rise to the long QT syn- medulla [115–117]; this low-conductance
drome, affected patients have not been described ATP-sensitive potassium channel is encoded by
to have proximal tubular transport defects, the KCNJ1 gene. Loss-of-function mutations in
suggesting that the role of these channels in ROMK lead to antenatal Bartter syndrome
human kidney is not clinically significant. (type 2), also known as the hyperprostaglandin E
syndrome, which is characterized by severe renal
Thick Ascending Limb of the Loop salt and fluid wasting, electrolyte abnormalities
of Henle (TALH) (hypokalemia, hypomagnesemia, and
Approximately 10 % of the filtered load of potas- hypercalciuria), and elevated renin and aldoste-
sium reaches the early distal tubule of the adult rat rone levels [118]. The clinical picture observed
[100], an observation that implies that significant is similar to that of chronic administration of loop
reabsorption of this cation occurs beyond the ini- diuretics. The typical presentation of antenatal
tial two-thirds of the proximal tubules that are Bartter syndrome includes polyhydramnios,
accessible to micropuncture. Sites responsible premature delivery, life-threatening episodes of
for this additional avid potassium reabsorption dehydration during the first week of life, and
are the later, deeper part of the proximal tubule, profound growth failure [119]. It should be
which is inaccessible to micropuncture analysis, noted that mutations in either NKCC2
and the TALH where potassium reabsorption is (SLC12A1), or the basolateral chloride channel
mediated, at least in part, by the apical Na-K-2Cl CLC-Kb (CLCNKB), or its associated subunit
cotransporter (NKCC2) that translocates a single Barttin (BSND) can also give rise to Bartter syn-
potassium ion into the cell accompanied by one dromes types 1, 3, and 4, respectively [119–121].
sodium and two chloride ions (Fig. 3). This sec- In contrast to the situation in the adult, up to
ondary active transport is ultimately driven by the 35 % of the filtered load of potassium reaches the
low intracellular sodium concentration, early distal tubule of the young (2-week-old) rat
established by the basolateral Na-K-ATPase, [97], suggesting that the TALH undergoes a sig-
which drives sodium entry from the lumen into nificant postnatal increase in its capacity for
the cell. The diuretics furosemide and bumetanide reabsorption. Consistent with this premise are
specifically inhibit NKCC2 and thus block the observations from studies in rats of significant
sodium, potassium, and chloride reabsorption at increases in the (a) fractional reabsorption of
this site. potassium along the TALH, expressed as a per-
Critical to the function of NKCC2 is the pres- centage of delivered load, between the second and
ence of a secretory potassium channel in the uri- sixth weeks of postnatal life [97], and
nary membrane which provides a pathway for (b) osmolarity of early distal fluid between the
potassium, taken up into the cell via the second and fourth weeks of life [122]. While
cotransporter, to recycle back into the lumen. these findings provide compelling evidence for a
This “recycling” of potassium counteracts net developmental maturation of potassium absorp-
potassium reabsorption through NKCC2 but tive pathways and diluting capacity of the
228 L.M. Satlin and D. Bockenhauer

TALH, respectively, direct functional analysis of functional and morphologic characteristics. In


the transport capacity of this segment in the devel- general, CNT/principal cells reabsorb sodium
oping nephron has not been performed. and secrete potassium, whereas intercalated cells
Molecular studies in rat kidney indicate that regulate acid–base balance but can reabsorb
mRNA encoding NKCC2, absent at birth, is first potassium in response to dietary potassium
expressed on postnatal day 8 in rat, coincident restriction or metabolic acidosis (Fig. 3) [133,
with completion of nephrogenesis [31], and 134]. Thus, the direction and magnitude of net
increases, at least in medulla, between postnatal potassium transport in these segments depend on
days 10 and 40 [123]. Na-K-ATPase activity in the the metabolic needs of the organism and reflect
TALH of the neonatal rabbit is only 20 % of that the balance of potassium secretion and absorption.
measured in the adult when expressed per unit of
dry weight [124]. The postnatal increase in pump Potassium Secretion
activity is associated with a parallel increase in Potassium secretion in the CNT and CCD is crit-
expression of medullary Na-K-ATPase mRNA ically dependent on the reabsorption of filtered
[123]. Although the balance of the studies sum- sodium delivered to these segments. Sodium pas-
marized above identifies a functional immaturity sively diffuses into the CNT/principal cell from
of the TALH early in life and would thus predict the urinary fluid down a favorable concentration
limited effects of inhibitors of NKCC2 on gradient through the luminal amiloride-sensitive
transepithelial transport, administration of furose- epithelial Na channel (ENaC) and is then
mide (2 mg/kg) to newborn lambs leads to a transported out of the cell at the basolateral mem-
natriuretic response equivalent to that observed brane in exchange for the uptake of potassium via
in adult animals [125]. Similarly, furosemide is the basolateral Na-K-ATPase (Fig. 3). The accu-
one of the most commonly prescribed medica- mulation of potassium within the cell and the
tions in premature neonates with a demonstrated lumen-negative voltage, created by movement of
natriuretic response [126]. sodium from the tubular lumen into the cell and its
electrogenic extrusion, creates a favorable electro-
Distal Nephron chemical gradient for intracellular potassium to
Within the aldosterone-sensitive distal nephron diffuse into the urinary space through apical
(ASDN) of the fully differentiated kidney, the potassium-selective channels. The magnitude of
late distal convoluted tubule (DCT), connecting potassium secretion is determined by its electro-
tubule (CNT), and cortical collecting duct (CCD) chemical gradient and the apical permeability to
are considered to be the primary sites of potassium this cation. Basolateral potassium channels in
secretion, and thus urinary potassium excretion, these same cells provide a route for intracellular
which can approach 20 % of the filtered load [100, potassium ions to recycle back into the
102, 127, 128] (Fig. 3). The DCT secretes a con- interstitium, thereby maintaining the efficiency
stant small amount of potassium into the urinary of the Na-K pump. Any factor that increases the
fluid [129]. An elevation in plasma potassium electrochemical gradient across the apical mem-
concentration inhibits the thiazide-sensitive brane or increases the apical potassium permeabil-
NaCl cotransporter NCC expressed at the apical ity will promote potassium secretion. An apical
membrane of the DCT [130, 131], thereby electroneutral potassium chloride cotransporter
increasing the delivery of fluid and sodium into has also been functionally identified in the CCD
the distal nephron. This observation may account [135, 136].
for the ability of high potassium diets to lower Two apical potassium-selective channels have
blood pressure, in a fashion similar to that of been functionally identified in the distal nephron:
thiazide administration [132]. the small-conductance secretory potassium
Regulated bidirectional potassium transport (SK) channel and the high-conductance BK chan-
occurs in the CNT and CCD, comprised of two nel. The density of these channels appears to be
major populations of cells, each with distinct greater in the CNT than in the CCD [137].
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 229

The SK channel, restricted to the of this channel [152], thereby negating the effect
CNT/principal cell, mediates baseline constitutive of the enhanced electrochemical gradient on stim-
potassium secretion [138, 139]. ROMK, origi- ulation of net potassium secretion. Mutations in
nally cloned from the TALH (described in the the genes encoding WNK1 or WNK4 lead to
section above on the TALH), is considered to pseudohypoaldosteronism type II (PHA II; famil-
constitute the SK channel. Indeed, there is ial hyperkalemic hypertension, or FHHt), an auto-
absence of SK channel activity in apical mem- somal dominant disorder characterized by
branes from both TALH and collecting duct in hypertension sensitive to thiazide diuretics,
ROMK null mice [140]. This is also consistent hyperkalemia, and metabolic acidosis
with the clinical observation of initial [153]. WNK4 harboring PHAII-type mutations
hyperkalemia in neonates with Bartter syndrome lead to increased apical expression of NCC and
type 2 due to loss-of-function of ROMK [141]. A stimulation of sodium absorption in the DCT
complex interplay of hormones, second messen- [151, 154]. The consequent reduction in sodium
gers, and kinases/phosphatases regulates the delivery to the CNT and CCD would be expected
SK/ROMK channel in the distal nephron, thereby to reduce potassium secretion. However, the same
allowing the kidney to respond appropriately to mutations in WNK4 that relieve the inhibition of
the metabolic needs of the organism [142]. Protein NCC further decrease surface expression of
kinase A (PKA)-induced phosphorylation of the ROMK and thus reduce potassium secretion in
channel is essential for its activity [143, 144] and the CCD. Together, these effects explain the car-
may account for the well-documented stimulatory dinal symptoms of PHAII (hypertension and
effect of vasopressin on renal potassium secretion hyperkalemia). WNK1 suppresses the activity of
[145]. Protein tyrosine kinase (PTK) mediates the WNK4. Therefore, a gain-in-function mutation in
endocytosis of ROMK channels in the rat CCD in WNK1 will also produce the clinical signs and
the face of dietary potassium restriction [146, symptoms of PHA II [153]. Finally, a kidney-
147]. Tyrosine phosphorylation of ROMK specific short form of WNK1, KS-WNK1,
enhances channel internalization and thus the inhibits WNK1 and can thereby abrogate
removal of channels from the plasma membrane WNK-1-mediated inhibition of WNK4
[148], leading to a reduction in surface expression [155]. More recently, mutations in ubiquitin-
of channels and net potassium secretion. Tyrosine ligase elements Kelch-like 3 and Cullin 3 have
phosphorylation of ROMK channels decreases in been identified as further genetic causes of PHAII
response to dietary potassium loading [149]. [156, 157], and these proteins are thought to be
The “with-no-lysine” kinases, or WNK involved in ubiquitination and degradation of
kinases, comprise a recently discovered family WNK4 [158].
of serine/threonine kinases that act as molecular The BK channel, present in CNT/principal and
switches that direct differential effects on down- intercalated cells, is considered to mediate flow-
stream ion channels, transporters, and the stimulated potassium secretion [159, 160]. In the
paracellular pathway to allow either maximal CNT and CCD, the density of conducting BK
sodium chloride reabsorption or maximal potas- channels is greater in intercalated than in
sium secretion in response to hypovolemia or CNT/principal cells [161, 162]. The BK channel
hyperkalemia, respectively [150]. WNK4 inhibits is comprised of two subunits: a channel pore-
sodium and chloride absorption in the DCT by forming α-subunit and a regulatory β-subunit.
reducing the surface expression of the apical This channel is rarely open at the physiologic
thiazide-sensitive NaCl cotransporter NCC resting membrane potential but can be activated
[151], an effect that would be expected to increase by cell depolarization, membrane stretch, and
sodium delivery to and reabsorption by the CCD, increases in intracellular Ca2+ concentration
in turn augmenting the driving force for potassium elicited by increases in urinary flow rate
secretion. However, WNK4 decreases surface [162–165]. The role of the BK channel in flow-
expression of ROMK by enhancing endocytosis stimulated urinary potassium secretion has been
230 L.M. Satlin and D. Bockenhauer

confirmed in mouse models with targeted deletion is mediated by an H-K-ATPase, localized to the
of the BKα pore-forming subunit or accessory β1 apical membrane of acid–base transporting inter-
subunit; the fractional excretion of potassium in calated cells, that couples potassium reabsorption
knockout mice subjected to high distal urinary to proton secretion [133, 174–176]. Two isoforms
flow rates is significantly lower than that observed of the H-K-ATPase are found in the kidney: the
in wild-type mice [166, 167]. gastric isoform, HKAg, is normally found in gas-
The BK channel assumes great importance in tric parietal cells and is responsible for acid secre-
regulating potassium homeostasis under condi- tion into the lumen whereas the colonic HKAc
tions where SK/ROMK channel-mediated potas- isoform is a structurally related H-K-ATPase
sium secretion is limited. Thus, adult animals with found in distal colon that mediates active potas-
genetic ablation of ROMK (i.e., Bartter pheno- sium reabsorption [175]. Expression of the apical
type) are not hyperkalemic, as would be expected gastric-like H-K-ATPase in the rat and rabbit
in the absence of a primary potassium secretory intercalated cell is increased in response to dietary
channel, but instead lose urinary potassium potassium restriction and metabolic acidosis [133,
[168]. The sensitivity of distal potassium secre- 174, 177, 178].
tion in this rodent model of Bartter syndrome to A reduction in potassium intake leads to a fall
iberiotoxin, a specific inhibitor of BK channels, in potassium secretion by the distal nephron
presumably reflects recruitment of the latter chan- within 5–7 days in rat [179]. This adaptation is
nels to secrete potassium in response to high distal associated with a decrease in the number of apical
flow rates consequent to loss-of-function of the SK/ROMK [180] and BK [181] channels and
TALH NKCC2 cotransporter activity [168]. Simi- stimulation of H-K-ATPase-mediated potassium
larly, although neonates with antenatal Bartter reabsorption in intercalated cells in the distal
syndrome due to loss-of-function mutations in nephron [182]. The reduction in number of
ROMK (Bartter type 2) may exhibit severe SK/ROMK channels in potassium-restricted ani-
hyperkalemia during the first few days of life mals is mediated by the effect of dietary potassium
[169], the hyperkalemia is not sustained. In fact, on circulating levels of aldosterone and other
these patients typically exhibit modest hypokale- effectors, such as PTK, as described above. Stim-
mia beyond the neonatal period [170, 171]. Of ulation of luminal H-K-ATPase activity in inter-
note is that increases in renal tubular flow rate in calated cells results not only in potassium
the CCD stimulate PGE2 release and inhibitors of retention but also in urinary acidification and met-
PGE2 production (indomethacin) inhibit flow- abolic alkalosis.
induced potassium secretion in the CCD
[172]. Indomethacin has long been used as a ther- Developmental Regulation of Distal
apeutic option in polyuric loop disorders, such as Potassium Transport
Bartter Syndrome, to mitigate the salt and volume Potassium secretion in the distal nephron, and spe-
losses [280]. cifically in the cortical collecting duct (CCD) stud-
Emerging evidence now suggests that WNK ied in vitro, is low early in life and cannot be
kinases regulate apical BK channel expression in stimulated by high urinary flow rates [128]. Indeed,
the ASDN. Specifically, WNK4 reduces expres- basal potassium secretion cannot be detected in the
sion of the α-subunit of the BK channel, targeting rabbit CCD until after the third week of postnatal
the protein for degradation through an ubiquitin- life, just about the time of weaning, with potassium
dependent pathway [173]. secretory rates increasing thereafter to reach adult
levels by 6 weeks of age [128]. Consistent with the
Potassium Absorption relatively undifferentiated state of the newborn
In response to dietary potassium restriction or CCD are the ultrastructural and morphometric find-
metabolic acidosis, the distal nephron may reverse ings in neonatal principal cells of few organelles,
the direction of net potassium transport from mitochondria, and basolateral infoldings, the site of
secretion to absorption. Potassium reabsorption Na-K-ATPase [183, 184].
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 231

The limited capacity of the CCD for potassium determined by the transcriptional/translational
secretion early in life could be explained by either regulation of expression of BK channels.
an unfavorable electrochemical gradient across While the neonatal distal nephron is limited in
the apical membrane or a limited apical perme- its capacity for potassium secretion, indirect evi-
ability to this ion. Cumulative evidence suggests dences suggest that this nephron segment absorbs
that the electrochemical gradient is not limiting potassium. As indicated above, saline-expanded
for potassium secretion in the neonate. Activity of newborn dogs absorb 25 % more of the distal
the Na-K-ATPase, present along the basolateral potassium load than do their adult counterparts
membrane of corticomedullary collecting ducts in [95]. Functional analysis of the rabbit collecting
the neonatal rabbit [185], is 50 % of that measured duct has shown that the activity of apical H-K-
in the mature nephron; the observation that the ATPase in neonatal intercalated cells is equivalent
intracellular potassium concentration in this seg- to that in mature cells [133]. The latter data alone
ment is similar in the neonate and adult presum- do not predict transepithelial potassium absorp-
ably reflects a relative paucity of membrane tion under physiologic conditions, as the balance
potassium channels in the distal nephron early in of transport will depend on the presence and
life [124, 184, 186]. Concordant with the mea- activity of apical and basolateral potassium con-
surements of sodium pump activity, the rate of ductances and the potassium concentration of the
sodium absorption in the CCD at 2 weeks of age tubular fluid delivered to this site. The high distal
is approximately 60 % of that measured in the tubular fluid potassium concentrations, as mea-
adult [128]. However, electrophysiologic analysis sured in vivo in the young rat, may facilitate
has confirmed the absence of functional lumen-to-cell potassium absorption mediated by
SK/ROMK channels in the luminal membrane of the H-K-ATPase [97].
the neonatal rabbit CCD with the number of open
channels per patch increasing progressively after
the second week of life [187]. Thus, the postnatal Luminal and Peritubular Factors
increase in the basal potassium secretory capacity Affecting Potassium Transport
of the distal nephron appears to be due primarily
to a developmental increase in number of The major factors that influence the external bal-
SK/ROMK channels, reflecting an increase in ance of potassium are listed in Table 2 and are
transcription and translation of functional channel discussed in the following sections.
proteins [187–189].
The appearance of flow-stimulated net potas-
sium secretion is a relatively late developmental Sodium Delivery and Absorption
event. Flow-stimulated potassium secretion can-
not be elicited in rabbit CCDs until the fifth week The magnitude of sodium reabsorption in the dis-
of postnatal life, which is approximately 2 weeks tal nephron determines the electrochemical
after basal net potassium secretion is first detected
[128, 190]. The absence of flow-stimulated potas-
sium secretion early in life is not due to a limited Table 2 Factors that regulate external potassium balance
flow-induced rise in net sodium absorption and/or Renal factors
intracellular calcium concentration, each of which Potassium intake/plasma potassium concentration
is equivalent to that detected in the adult by the Tubular (urinary) flow rate
second week of postnatal life [190]. The observa- Distal sodium delivery and transepithelial voltage
tion that mRNA encoding the BK channel Hormones (mineralocorticoids, vasopressin)
α-subunit and immunodetectable channel protein Acid–base balance
Gastrointestinal tract factors
cannot be demonstrated until the fourth and fifth
Stool volume
weeks of postnatal life, respectively [190], sug-
Hormones (mineralocorticoids)
gests that flow-dependent potassium secretion is
232 L.M. Satlin and D. Bockenhauer

driving force favoring potassium secretion into electronegativity is maintained, thereby eliciting
the luminal fluid, as described above. Processes more potassium secretion than what occurs with a
that enhance distal sodium delivery and increase comparable sodium load delivered with
tubular fluid flow rate, such as extracellular vol- chloride [199].
ume expansion or administration of a variety of
diuretics (osmotic diuretics, carbonic anhydrase
inhibitors, loop and thiazide diuretics), lead to an Tubular Flow Rate
increase in urinary excretion of both sodium and
potassium. The kaliuresis is due not only to the High rates of urinary flow in the mature, but not
increased delivery of sodium to the distal nephron the neonatal or weanling, distal nephron, as
but also to the increase in tubular fluid flow rate, elicited by extracellular fluid volume expansion
which maximizes the chemical driving forces, as or administration of diuretics or osmotic agents,
described below, favoring potassium secretion. stimulate potassium secretion [128]. There are a
Processes that decrease sodium delivery to less number of factors responsible for the flow stimu-
than 30 mM in the distal tubular fluid [191, 192] lation of potassium secretion. First, increases in
and/or sodium reabsorption sharply reduce potas- tubular fluid flow rate in the distal nephron
sium secretion in the CCD and can lead to enhance sodium reabsorption due to an increase
hyperkalemia. In vivo measurements of the in the open probability of ENaC (time the channel
sodium concentration in distal tubular fluid gen- spends in the open state), which augments the
erally exceed 35 mEq/l both in healthy adult and electrochemical gradient favoring potassium
suckling rats and thus should not restrict distal secretion [195, 200]. Second, the higher the uri-
sodium secretion [97, 122, 191, 193]. However, nary flow rate in the distal nephron, the slower the
in edema-forming states, including congestive rate of rise of tubular fluid potassium concentra-
heart failure, cirrhosis, and nephrotic syndrome, tion because secreted potassium is rapidly diluted
the urinary sodium concentration typically falls to in urine of low potassium concentration
less than 10 mEq/l; a reduction in potassium [201]. Maintenance of a low tubular fluid potas-
excretion in these patients may be ascribed to the sium concentration maximizes the potassium
low rates of distal sodium delivery as well as concentration gradient (and thus the chemical
urinary flow. The potassium-sparing diuretics, driving force) favoring net potassium secretion.
amiloride and triamterene, inhibit ENaC and Finally, increases in luminal flow rate transduce
thus block sodium absorption, thereby mechanical signals (circumferential stretch,
diminishing the electrochemical gradient favoring shear stress, hydrodynamic bending moments on
potassium secretion [194, 195]. Trimethoprim and the cilium decorating the apical surface of
pentamidine can also limit urinary potassium virtually all renal epithelial cells) into increases
secretion via the same mechanism [196, 197]. in intracellular calcium concentration, which in
Sodium delivered to the distal nephron is gen- turn activate apical BK channels to secrete
erally accompanied by chloride. Chloride potassium, thereby enhancing urinary potassium
reabsorption occurs in large part via the excretion [160, 190].
paracellular pathway. The movement of the nega-
tively charged chloride out of the lumen dissipates
the lumen-negative potential, creating a less Potassium Intake and Cellular
favorable driving force for luminal potassium Potassium Content
secretion [198]. When sodium delivered to the
distal nephron is accompanied by an anion less The kidney adjusts potassium excretion to match
reabsorbable than chloride, such as bicarbonate input, in large part by regulating the magnitude of
(in proximal renal tubular acidosis), potassium secretion and reabsorption in the distal
β-hydroxybutyrate (in diabetic ketoacidosis), or nephron. Thus, for example, an increase in dietary
carbenicillin (during antibiotic therapy), luminal potassium intake stimulates whereas a decrease in
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 233

intake reduces net potassium secretion [102]. An liver, into the circulation and its uptake by
increase in potassium concentration in the extra- kidney [206].
cellular fluid space increases potassium entry into Chronic potassium loading leads to potassium
principal cells via the basolateral Na-K-ATPase, adaptation, an acquired tolerance to an otherwise
which in turn maximizes the concentration gradi- lethal acute potassium load [8]. Potassium adap-
ent favoring apical potassium secretion into the tation, which begins after a single potassium-rich
urinary fluid. Simultaneously, the increase in meal, includes increases in the rate of skeletal
circulating levels of plasma aldosterone that muscle uptake of potassium from the extracellular
accompanies potassium loading enhances the fluid [56] due to stimulation of Na-K-ATPase
electrochemical driving force favoring potassium activity [210] and secretion of potassium by the
secretion in the distal nephron by stimulation of distal nephron and colon. The process is facili-
ENaC-mediated sodium absorption and its elec- tated by the increase in circulating levels of aldo-
trogenic absorption via the Na-K-ATPase. Within sterone elicited by the increase in serum
6 h of an increase in dietary potassium intake, the potassium concentration [211]. A similar adaptive
density of apical ROMK channels increases in response is seen in renal insufficiency such that
principal cells in rats due to activation of a previ- potassium balance is maintained during the course
ously “silent” pool of channels or closely associ- of many forms of progressive renal disease, as
ated proteins [202]. Chronic potassium loading long as potassium intake is not excessive
also increases BK channel message, apical pro- [212]. The molecular mechanisms underlying
tein, and function in the distal nephron this adaptation in the distal nephron (and colon)
[181]. Finally, the inhibition of proximal tubule include not only an increase in the density of
and loop of Henle salt and water reabsorption in apical membrane potassium channels but also an
response to a potassium load [203, 204] increases increase in the number of conducting ENaC chan-
tubule fluid flow rate which in turn stimulates nels and activity of the basolateral Na-K pump.
potassium secretion via activation of BK channels The latter two processes result in increases in
and increased distal delivery of sodium to the transepithelial voltage and the intracellular potas-
distal nephron. sium concentration, events that enhance the driv-
The trigger for the renal adaptation to dietary ing force favoring potassium diffusion from the
potassium loading remains uncertain. It is now cell into the urinary fluid.
believed that after a potassium-rich meal, a reflex
increase in potassium excretion is initiated by
sensors in the splanchnic bed (gut, portal circula- Hormones
tion, and/or liver) that respond to local increases in
potassium concentration that occur in the absence Mineralocorticoids are key regulators of renal
of changes in plasma potassium concentration or sodium absorption and potassium excretion and
before changes in plasma potassium concentra- thus of circulating volume, blood pressure, and
tion are detected [205, 206]. In support of the sodium and potassium homeostasis. There are two
notion of potassium sensing, intraportal delivery major physiologic stimuli for aldosterone release
of potassium chloride to rats leads to an increase from the zona glomerulosa in the adrenal gland
in hepatic afferent nerve activity and urinary [213]: angiotensin II (AII) and elevation in serum
potassium excretion, responses that are unaccom- potassium concentration [214]. AII, generated in
panied by increases in plasma potassium concen- response to intravascular volume depletion by
tration [207]. Increases in the intraportal activation of the renin-angiotensin system, binds
concentration of glucagon, following ingestion to the type 1 AII receptor (AT1) to increase aldo-
of a protein- and potassium-rich meal, also sterone production. ACE inhibitors, by reducing
increase renal excretion of potassium [208, 209], the conversion of angiotensin I to angiotensin II
a response proposed to be due to the release of and thus aldosterone secretion by the adrenal
cAMP, the second messenger of glucagon in the gland, may lead to hyperkalemia as the ability of
234 L.M. Satlin and D. Bockenhauer

the distal nephron to secrete potassium is and proximal tubular sodium and fluid
impaired. AII receptor blockers (ARBs), by com- reabsorption [227]. Sgk1/ mice exhibit an
petitively binding to AT1 receptors and thus impaired upregulation of renal potassium excre-
antagonizing the action of AII on aldosterone tion in response to potassium loading, presumably
release, may have similar effects. due to the impact of the loss of SGK1 function on
Aldosterone stimulates sodium reabsorption ENaC and/or Na-K-ATPase activity and thus the
and potassium secretion in principal cells of the electrochemical gradient favoring potassium
fully differentiated ASDN [215, 216]. Circulating secretion [228]. Corticosteroid hormone-induced
mineralocorticoids bind to their cytosolic recep- factor (CHIF) is another aldosterone-induced pro-
tors in the ASDN; the aldosterone antagonists tein that is expressed in the basolateral membrane
spironolactone and eplerenone competitively of the collecting duct where it increases the affin-
inhibit this binding. The hormone–receptor com- ity of Na-K-ATPase for sodium [229–232].
plex translocates to the nucleus where it promotes Plasma aldosterone concentrations in prema-
the transcription of aldosterone-induced physio- ture infants and newborns are higher than those
logically active proteins (e.g., Na-K-ATPase). measured in the adult [17, 233]. Yet, clearance
Among the cellular and molecular effects of an studies in fetal and newborn animals demonstrate
increase in circulating levels of aldosterone are a relative insensitivity of the immature kidney to
increases in density of ENaC channels, achieved the hormone [17, 234–236]. The density of aldo-
by the recruitment to and retention of intracellular sterone binding sites, receptor affinity, and degree
channels at the apical membrane, de novo synthe- of nuclear binding of hormone receptor are
sis of new ENaC subunits, and activation of believed to be similar in mature and immature
preexistent channels, as well as stimulation rats [236].
of Na-K-ATPase activity by translocation of The transtubular potassium gradient (TTKG)
preformed transporters to the membrane and provides an indirect, semiquantitative measure of
translation of new sodium pump subunits the renal response to mineralocorticoid activity in
[217–222]. The sum effect of the stimulation of the aldosterone-sensitive cortical distal nephron
apical sodium entry and Na-pump-mediated and is calculated using the equation: TTKG =
reabsorption is an increase in lumen-negative U[K]/P[K] divided by Uosmolality/Posmolality, where
transepithelial voltage and thus electrochemical [K] equals the potassium concentration in mEq/l
driving force favoring potassium exit across the [237–239]. Measurements of TTKG have been
apical membrane [217, 220, 223]. reported to be lower in 27- than 30-week GA
The effects of aldosterone on ENaC and, to preterm infants followed over the first 5 weeks
some extent, the Na-K pump appear to be indirect, of postnatal life [240]. The low TTKG has been
mediated by aldosterone-induced proteins, attributed to a state of relative hypoaldosteronism
including serum and glucocorticoid-inducible [240] but may also reflect the absence of potas-
kinase (sgk). Aldosterone rapidly induces Sgk1 sium secretory transport pathways (i.e., channel
in the distal nephron [224]. Phosphorylated Sgk proteins).
stimulates sodium reabsorption, in large part by
inhibiting ubiquitin-ligase Nedd4-2-mediated
endocytic retrieval of ENaCs from the luminal Acid–Base Balance
membrane [225]. Renal water and electrolyte
excretion is indistinguishable in sgk1-knockout Disorders of acid–base homeostasis induce
(sgk1/) and wild-type (sgk1+/+) mice fed a nor- changes in tubular potassium secretion in the dis-
mal diet [226], indicating that the kinase is not tal nephron [198]. In general, acute metabolic
necessary for basal sodium absorption. However, acidosis causes the urine pH and potassium excre-
dietary sodium restriction reveals an impaired tion to decrease, whereas both acute respiratory
ability of sgk1/ mice to reduce sodium excre- alkalosis and metabolic alkalosis increase urine
tion despite increases in plasma aldosterone levels pH and potassium excretion [198, 241]. Chronic
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 235

metabolic acidosis has variable effects on urinary afternoon emerges, even when potassium intake
potassium excretion. ingestion is spread evenly throughout the day and
Acute metabolic acidosis reduces cell potas- night [247, 248]. As with other circadian rhythms,
sium concentration and leads to a reduction in evidence suggests that the periodicity depends on
urine pH, which in turns inhibits activity of the both central and peripheral pacemakers. Notably,
SK/ROMK channel and thereby limits potassium the DCT and CCD express several regulatory
secretion in the distal nephron [139, “clock” genes, indicating operation of a local
241–243]. This makes physiological sense, as intrarenal pacemaker. ENaC, aquaporin 2, and
the electrical balance for sodium reabsorption by vasopressin receptors have marked circadian
either potassium or proton secretion is thus rhythms, which could influence indirectly the
maintained by increased proton secretion to excretion of potassium [249, 250]. Moreover,
restore acid–base homeostasis. recent data from space flight simulation suggest
The effect of chronic metabolic acidosis on that there is also an infradian rhythmicity to aldo-
potassium secretion is more complex and may be sterone secretion [251]. It is unknown whether a
influenced by modifications of the glomerular fil- circadian cycle of urinary potassium excretion
trate (e.g., chloride and bicarbonate concentra- prevails in infancy.
tions), tubular fluid flow rate, and circulating
aldosterone levels [8, 198]. The latter two factors Contribution of the Gastrointestinal
may lead to an increase rather than a decrease in Tract
potassium secretion and excretion. Under normal conditions in the adult, 5–10 % of
The alkalosis-induced stimulation of potas- daily potassium intake is excreted in the stool. The
sium secretion reflects two direct effects on prin- colon is considered to be the main target for reg-
cipal cells: (1) an increase in net sodium ulation of intestinal potassium excretion
absorption [244], which enhances the electro- [252]. Potassium transport in the colon represents
chemical gradient for net potassium secretion, the balance of secretion and absorption
and (2) an increase in the permeability of the [253]. Under baseline conditions, net potassium
apical membrane to potassium resulting from an secretion predominates over absorption in the
increase in duration of time the potassium- adult, whereas the neonatal gut is poised for net
selective channels remain open [8]. Alkalosis potassium absorption [252].
also decreases acid secretion in intercalated cells, Potassium secretion requires potassium uptake
thereby reducing H-K-ATPase-mediated by the Na-K-ATPase and furosemide-sensitive
countertransport of potassium. Na-K-2Cl cotransporter located in the basolateral
Potassium deficiency stimulates proton secre- membrane of colonocytes; potassium is then
tion in the distal nephron, increases the production secreted across the apical membrane through
of the urinary buffer ammonia [245], and may potassium channels, including a calcium-
stimulate bicarbonate generation by increasing activated BK channel similar to that found in the
expression of H-K-ATPase in the distal distal nephron [254–258]. Potassium absorption
nephron [246]. is mediated by two H-K-ATPases localized to the
apical membrane of the distal colon [259].
Stool potassium content can be enhanced by
Circadian Control of Renal Potassium any factor that increases colonic secretion, includ-
Excretion ing aldosterone, epinephrine, and prostaglandins
[24, 260, 261]. Indomethacin and dietary potas-
Renal potassium excretion varies during the 24-h sium restriction reduce potassium secretion by
day in the adult [247, 248]. Although some of this inhibiting the basolateral transporters and apical
variation can be attributed to timing of meals, a potassium channels. Diarrheal illnesses are typi-
pattern of low rates of potassium excretion at cally associated with hypokalemia, presumably
night and in the morning and high rates in the due to the presence of nonreabsorbed anions
236 L.M. Satlin and D. Bockenhauer

(which obligate potassium), an enhanced electro- true plasma level but occur during or after blood
chemical gradient established by active chloride drawing. This is a common cause of
secretion, and secondary hyperaldosteronism due hyperkalemia, especially in small children,
to volume contraction [262]. where obtaining a free-flowing blood specimen
Potassium adaptation in the colon is demon- is difficult. Small needle size, squeezing of the
strated by increased fecal potassium secretion extremity, and use of a tourniquet can all contrib-
after potassium loading and in the face of renal ute to cell lysis with subsequent release of potas-
insufficiency. Fecal potassium excretion may sium [268–270]. In addition, release of potassium
increase substantially to account for 30–50 % of after phlebotomy due to delayed processing of the
potassium excretion in patients with severe specimen, the presence of an increased cellular
chronic renal insufficiency [24, 263–265]. The component of blood, or altered potassium trans-
enhanced colonic potassium secretion character- port activity of the cell membrane can lead to
istic of renal insufficiency requires induction pseudohyperkalemia [271, 272]. Most laborato-
and/or activation of apical BK channels in surface ries will alert a healthcare provider as to the pres-
colonic epithelial cells [266]. ence of hemolysis. A repeat sample with prompt
Net colonic potassium absorption is signifi- processing will establish a normal plasma level in
cantly higher in young compared to adult most cases. In the interim, an ECG may help
rats [252]. The higher rate of potassium absorp- assess the degree of hyperkalemia, as judged by
tion during infancy is due to robust activity of the height of the T-waves, although the sensitivity
apical K-ATPases and limited activity of the of this tool is reportedly poor [273].
basolateral transporters that mediate secretion
[252, 256, 267]. Altered Internal Balance
Hyperkalemia due to an altered internal balance
Mendelian Disorders Associated should be suspected if there are suggestive factors,
with Altered Plasma Potassium Levels such as metabolic acidosis and/or poorly con-
There are several inherited disorders, which can trolled diabetes mellitus (see also Table 1). Treat-
affect plasma potassium levels either by altering ment in these cases should focus on correcting the
internal potassium balance between the intra- and underlying abnormality.
extracellular fluid compartments or renal potas-
sium handling. These disorders are summarized in Altered External Balance
Table 3. Most of these disorders are discussed in Hyperkalemia due to an altered external balance
detail in their respective chapters. may be due to excessive potassium intake,
impaired renal potassium secretion, or a combina-
Clinical Approach to the Patient tion of both. With normal kidney function, even a
with Hyperkalemia very high potassium intake should not lead to
Cardiac arrhythmias are feared consequences of hyperkalemia, due to the rapid hormonally medi-
severe hyperkalemia and constitute a medical ated translocation of potassium into cells, unless
emergency. Immediate treatment usually focuses administered as an acute bolus [274]. Commonly
on enhancing potassium uptake by cells. This can used tools to assess renal potassium secretion
be accomplished by administration of insulin/glu- include the fractional excretion of potassium
cose or β-sympathomimetics, such as albuterol. In (FEK), the ratio of urinary potassium to sodium,
asymptomatic patients, more time is available to and the TTKG, described earlier in this chapter.
consider potential causes, allowing for specific The TTKG is considered to be specific for the
treatment. assessment of distal mineralocorticoid-sensitive
potassium secretion independent of the glomeru-
Pseudohyperkalemia lar filtration rate [237, 238, 275]. In one study in
Pseudohyperkalemia refers to elevated in vitro children, a TTKG value of 4.1 corresponded to the
potassium concentrations that do not reflect the 3rd percentile for TTKG results and 13.1 to the
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 237

Table 3 Genetic disorders associated with altered plasma potassium levels


Disease OMIM# Gene(s) Mode Mechanism
Hyperkalaemic disorders
Hyperkalaemic periodic paralysis 170500 SCN4A AD Acute release of potassium from muscle
cells
Pseudohypoaldosteronism type 1 264350 SCNN1A AR Impaired renal potassium secretion
SCNN1B AR
SCNN1G AR
600983 NR3C2 AD
Pseudohypoaldosteronism type 614492 WNK1 AD Impaired renal potassium secretion
2 (Fhht; Gordon syndrome) 614491 WNK4 AD
614495 KLH3 AD
614496 CUL3 AD
or
AR
Congenital adrenal hyperplasia 201910 CYP21A2 AR Impaired renal potassium secretion due to
(salt wasting forms) deficiency in mineralocorticoids
Hypokalaemic disorders associated with low or normal blood pressure
Hypokalaemic periodic paralysis 170400 CACNA1S AD Acute potassium uptake by muscle cells
613345 SCN4A AD
Renal Fanconi syndromes 134600 ? AD Increased renal potassium secretion
613388 SLC34A1 AR
615605 EHHADH AD
Bartter syndrome 601678 SLC12A1 AR Increased renal potassium secretion
241200 KCNJ1 AR
607364 CLCKNB AR
602522 BSND AR
Gitelman syndrome 263800 SLC12A3 AR Increased renal potassium secretion
EAST (SeSAME) syndrome 612780 KCNJ10 AR Increased renal potassium secretion
Hypokalaemic disorders associated with elevated blood pressure
Congenital adrenal hyperplasia 202010 CYP11B1 AR Increased renal potassium secretion due to
(salt retaining forms) excess mineralocorticoids
Liddle syndrome 177200 SCNN1G AD Increased renal potassium secretion
Apparent mineralocorticoid excess 218030 HSD11B2 AR Increased renal potassium secretion
Familial hyperaldosteronism 605635 ? AD Increased renal potassium secretion
613677 KCNJ5 AD
Glucocorticoid-remediable 103900 CYP11B1 AD Increased renal potassium secretion
hyperaldosteronism
Distal renal tubular acidosis 267300 ATP6V1B1 AR Increased renal potassium secretion
ATP6V0A4 AR
179800 SLC4A1 AD
Listed are Mendelian disorders associated with altered plasma potassium levels, their respective OMIM numbers, mode of
inheritance and the key mechanism for dyskalemia. The disorders listed in the upper panel of the table are associated with
hyperkalemia, whereas those in the lower panels with hypokalemia, the latter grouped according to expected blood
pressure
Not listed are disorders leading to end-stage kidney disease and associated hyperkalemia, nor metabolic disorders with
secondary tubular dysfunction (e.g., cystinosis, mitochondrial cytopathies), nor intestinal salt wasting disorders with
secondary hyperaldosteronism (e.g., congenital chloride diarrhea)
AR autosomal recessive, AD autosomal dominant, ? gene unidentified yet
238 L.M. Satlin and D. Bockenhauer

97th percentile [240]. Accordingly, in Altered External Balance


hyperkalemia, a TTKG value below 4.1 would Hypokalemia due to an altered external balance
be inappropriate and indicate low mineralocorti- can be either from excessive potassium loss from
coid activity, suggesting impaired renal potassium the intestine or kidney, insufficient intake, or a
secretion. Conversely, a TTKG value above 13.1 combination of these factors. As with
is consistent with high mineralocorticoid activity hyperkalemia, assessment of the urinary potas-
and would thus suggest excessive potassium sium excretion can clarify the role of the kidney
intake as the underlying etiology of the in the etiology. A low TTKG suggests renal potas-
hyperkalemia. sium conservation and thus points towards intes-
Treatment varies depending on the etiology of tinal losses and/or insufficient intake. Again,
the hyperkalemia. Intake of potassium should be treatment should focus on the underlying abnor-
reduced if indicated. In those with impaired potas- mality. Conversely, a high TTKG points to renal
sium secretion, the cause should be identified. If potassium wasting. In the context of volume
clinical evidence of volume depletion is present, depletion, this finding would likely reflect
hyperkalemia may simply reflect inadequate distal aldosterone-mediated attempts to maintain extra-
sodium delivery and treatment should be aimed at cellular volume at the expense of potassium
volume expansion. This approach would also be homoeostasis and volume repletion is the appro-
appropriate for the rare patients with type I priate treatment. Hypokalemia with high TTKG in
pseudohypoaldosteronism (PHA), who may the presence of hypervolemia and hypertension
require supplementation with sodium chloride or suggests primary hyperaldosteronism or the rare
sodium bicarbonate and, possibly, treatment with forms of primary increased distal sodium
potassium-binding resins [276]. In patients who reabsorption/potassium secretion (“pseudohyper-
present with hyperkalemia and hypertension and aldosteronism”) as seen in some inherited disor-
low TTKG, PHAII (FHHt, see above) should be ders, such as Liddle syndrome or apparent
suspected. Administration of calcineurin inhibi- mineralocorticoid excess (see Table 3). This
tors can result in a clinical picture similar to pseudohyperaldosteronism can also be acquired,
PHAII and, like PHAII, responds to thiazide e.g., with licorice abuse [279]. Treatment should
diuretics [277]. Type III PHA is typically transient focus on the underlying defect. In the short term,
and secondary to other pathologies such as or if the underlying problem cannot be targeted,
obstructive uropathy or urinary tract distal potassium secretion can be reduced either
infection [278]. by administration of inhibitors of ENaC
(amiloride) and/or mineralocorticoid receptor
Clinical Approach to the Patient (spironolactone or eplerenone).
with Hypokalemia
Cardiac arrhythmias, muscle weakness, and even
paralysis are major morbidities associated with References
severe hypokalemia, and emergency treatment usu-
1. Adler S, Fraley DS. Potassium and intracellular
ally consists of potassium administration. In asymp- pH. Kidney Int. 1977;11(6):433–42.
tomatic patients, more time is available to consider 2. Bygrave FL. The ionic environment and metabolic
potential causes, allowing for specific treatment. control. Nature. 1967;214(5089):667–71.
3. Lubin M. Intracellular potassium and macromolecu-
lar synthesis in mammalian cells. Nature.
Altered Internal Balance 1967;213(5075):451–3.
Hypokalemia due to an altered internal balance 4. Lopez-Rivas A, Adelberg EA, Rozengurt
should be suspected if there are suggestive factors, E. Intracellular K+ and the mitogenic response of
such as metabolic alkalosis or evidence of 3T3 cells to peptide factors in serum-free medium.
Proc Natl Acad Sci U S A. 1982;79(20):6275–9.
increased insulin activity (see also Table 1). Treat- 5. Lang F, et al. Ion channels in cell proliferation
ment in these cases should focus on correcting the and apoptotic cell death. J Membr Biol.
underlying abnormality. 2005;205(3):147–57.
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 239

6. Schmidt-Nielsen B. Comparative physiology of cel- 25. Geering K. The functional role of beta subunits in
lular ion and volume regulation. J Exp Zool. oligomeric P-type ATPases. J Bioenerg Biomembr.
1975;194(1):207–19. 2001;33(5):425–38.
7. Weiner MW, et al. Renal mitochondrial enzymes 26. Benziane B, Chibalin AV. Frontiers: skeletal muscle
in potassium depletion. Am J Physiol. sodium pump regulation: a translocation paradigm.
1971;221(2):613–7. Am J Physiol Endocrinol Metab. 2008;295(3):
8. Giebisch G. Renal potassium transport: mechanisms E553–8.
and regulation. Am J Physiol. 1998;274(5 Pt 2): 27. Clausen T. Na+-K+ pump regulation and skeletal
F817–33. muscle contractility. Physiol Rev. 2003;83
9. Holbrook JT, et al. Sodium and potassium intake and (4):1269–324.
balance in adults consuming self-selected diets. Am J 28. Therien AG, Blostein R. Mechanisms of sodium
Clin Nutr. 1984;40(4):786–93. pump regulation. Am J Physiol Cell Physiol.
10. Butte NF, et al. Body composition during the first 2000;279(3):C541–66.
2 years of life: an updated reference. Pediatr Res. 29. Geering K. FXYD proteins: new regulators of Na-K-
2000;47(5):578–85. ATPase. Am J Physiol Renal Physiol. 2006;290(2):
11. Flynn MA, et al. Total body potassium in normal F241–50.
children. Pediatr Res. 1972;6(4):239–45. 30. Horster M. Embryonic epithelial membrane trans-
12. Dickerson JW, Widdowson EM. Chemical changes in porters. Am J Physiol Renal Physiol. 2000;279(6):
skeletal muscle during development. Biochem F982–96.
J. 1960;74:247–57. 31. Schmitt R, et al. Developmental expression of sodium
13. Rutledge MM, et al. A longitudinal study of total entry pathways in rat nephron. Am J Physiol.
body potassium in normal breastfed and bottle-fed 1999;276(3 Pt 2):F367–81.
infants. Pediatr Res. 1976;10(2):114–7. 32. Brion LP, et al. Early hyperkalaemia in very low
14. Dorup I, Clausen T. Effects of potassium deficiency birthweight infants in the absence of oliguria. Arch
on growth and protein synthesis in skeletal Dis Child. 1989;64(2):270–2.
muscle and the heart of rats. Br J Nutr. 33. Leslie GI, Carman G, Arnold JD. Early neonatal
1989;62(2):269–84. hyperkalaemia in the extremely premature newborn
15. Wilde WS. Potassium. In: Bronner CLCF, editor. infant. J Paediatr Child Health. 1990;26(1):58–61.
Mineral metabolism: an advanced treatise, vol. II. 34. Gruskay J, et al. Nonoliguric hyperkalemia in the
New York: Academic; 1962. p. 73–107. premature infant weighing less than 1000 grams.
16. Delgado MM, et al. Sodium and potassium clearances J Pediatr. 1988;113(2):381–6.
by the maturing kidney: clinical-molecular correlates. 35. Lorenz JM, Kleinman LI, Markarian K. Potassium
Pediatr Nephrol. 2003;18(8):759–67. metabolism in extremely low birth weight infants in
17. Sulyok E, et al. Relationship between maturity, elec- the first week of life. J Pediatr. 1997;131(1 Pt 1):81–6.
trolyte balance and the function of the renin- 36. Sato K, et al. Internal potassium shift in premature
angiotensin-aldosterone system in newborn infants. infants: cause of nonoliguric hyperkalemia. J Pediatr.
Biol Neonate. 1979;35(1–2):60–5. 1995;126(1):109–13.
18. Satlin LM. Regulation of potassium transport 37. Shaffer SG, et al. Hyperkalemia in very low birth
in the maturing kidney. Semin Nephrol. weight infants. J Pediatr. 1992;121(2):275–9.
1999;19(2):155–65. 38. Stefano JL, et al. Decreased erythrocyte Na+, K+-
19. Mohammed T, et al. Mechanisms of potassium trans- ATPase activity associated with cellular potassium
fer across the dually perfused rat placenta. Am J loss in extremely low birth weight infants
Physiol. 1993;265(2 Pt 2):R341–7. with nonoliguric hyperkalemia. J Pediatr.
20. Serrano CV, Talbert LM, Welt LG. Potassium defi- 1993;122(2):276–84.
ciency in the pregnant dog. J Clin Invest. 39. Uga N, et al. Antenatal steroid treatment prevents
1964;43:27–31. severe hyperkalemia in very low-birthweight infants.
21. Dancis J, Springer D. Fetal homeostasis in maternal Pediatr Int. 2003;45(6):656–60.
malnutrition: potassium and sodium deficiency in 40. Omar SA, et al. Effect of prenatal steroids on potas-
rats. Pediatr Res. 1970;4(4):345–51. sium balance in extremely low birth weight neonates.
22. DeFronzo RA, et al. Influence of basal insulin and Pediatrics. 2000;106(3):561–7.
glucagon secretion on potassium and sodium metab- 41. McCaughan D. Hazards of non-prescription potas-
olism. Studies with somatostatin in normal dogs and sium supplements. Lancet. 1984;1(8375):513–4.
in normal and diabetic human beings. J Clin Invest. 42. Zierler KL, Rabinowitz D. Effect of very small con-
1978;61(2):472–9. centrations of insulin on forearm metabolism. Persis-
23. DeFronzo RA, et al. Impaired renal tubular potassium tence of its action on potassium and free fatty acids
secretion in sickle cell disease. Ann Intern Med. without its effect on glucose. J Clin Invest.
1979;90(3):310–6. 1964;43:950–62.
24. Bia MJ, DeFronzo RA. Extrarenal potassium homeo- 43. Hundal HS, et al. Insulin induces translocation of the
stasis. Am J Physiol. 1981;240(4):F257–68. alpha 2 and beta 1 subunits of the Na+/K+-ATPase
240 L.M. Satlin and D. Bockenhauer

from intracellular compartments to the plasma mem- 60. Rosa RM, et al. Adrenergic modulation of extrarenal
brane in mammalian skeletal muscle. J Biol Chem. potassium disposal. N Engl J Med. 1980;302
1992;267(8):5040–3. (8):431–4.
44. Lavoie L, et al. Insulin-induced translocation of Na+- 61. Angelopoulous M, et al. In vitro analysis of the Na+-
K+-ATPase subunits to the plasma membrane is K+ ATPase activity in neonatal and adult red blood
muscle fiber type specific. Am J Physiol. 1996;270 cells. Biol Neonate. 1996;69(3):140–5.
(5 Pt 1):C1421–9. 62. Gillzan KM, Stewart AG. The role of potassium
45. Omatsu-Kanbe M, Kitasato H. Insulin stimulates the channels in the inhibitory effects of beta
translocation of Na+/K+-dependent ATPase mole- 2-adrenoceptor agonists on DNA synthesis in human
cules from intracellular stores to the plasma mem- cultured airway smooth muscle. Pulm Pharmacol
brane in frog skeletal muscle. Biochem J. 1990;272 Ther. 1997;10(2):71–9.
(3):727–33. 63. Clausen T, Flatman JA. The effect of catecholamines
46. Marette A, et al. Insulin increases the Na+-K+- on Na-K transport and membrane potential in rat
ATPase alpha 2-subunit in the surface of rat skeletal soleus muscle. J Physiol. 1977;270(2):383–414.
muscle: morphological evidence. Am J Physiol. 64. Mandelberg A, et al. Salbutamol metered-dose inhaler
1993;265(6 Pt 1):C1716–22. with spacer for hyperkalemia: how fast? How safe?
47. Kanbe M, Kitasato H. Stimulation of Na, K-ATPase Chest. 1999;115(3):617–22.
activity of frog skeletal muscle by insulin. Biochem 65. Singh BS, et al. Efficacy of albuterol inhalation in
Biophys Res Commun. 1986;134(2):609–16. treatment of hyperkalemia in premature neonates.
48. Lytton J. Insulin affects the sodium affinity of the rat J Pediatr. 2002;141(1):16–20.
adipocyte (Na+, K+)-ATPase. J Biol Chem. 1985;260 66. Helfrich E, de Vries TW, van Roon EN. Salbutamol
(18):10075–80. for hyperkalaemia in children. Acta Paediatr.
49. Sargeant RJ, Liu Z, Klip A. Action of insulin on Na+- 2001;90(11):1213–6.
K+-ATPase and the Na+-K+-2Cl- cotransporter in 67. Allon M. Hyperkalemia in end-stage renal disease:
3T3-L1 adipocytes. Am J Physiol. 1995;269(1 Pt 1): mechanisms and management. J Am Soc Nephrol.
C217–25. 1995;6(4):1134–42.
50. Sweeney G, Klip A. Regulation of the Na+/K+- 68. McClure RJ, Prasad VK, Brocklebank JT. Treatment
ATPase by insulin: why and how? Mol Cell Biochem. of hyperkalaemia using intravenous and nebulised
1998;182(1–2):121–33. salbutamol. Arch Dis Child. 1994;70(2):126–8.
51. Feraille E, et al. Insulin enhances sodium sensitivity 69. Semmekrot BA, Monnens LA. A warning for the
of Na-K-ATPase in isolated rat proximal treatment of hyperkalaemia with salbutamol. Eur J
convoluted tubule. Am J Physiol. 1994;267(1 Pt 2): Pediatr. 1997;156(5):420.
F55–62. 70. Shortland D, Trounce JQ, Levene
52. Hiatt N, Davidson MB, Bonorris G. The MI. Hyperkalaemia, cardiac arrhythmias, and cerebral
effect of potassium chloride infusion on lesions in high risk neonates. Arch Dis Child. 1987;62
insulin secretion in vivo. Horm Metab Res. (11):1139–43.
1972;4(2):64–8. 71. Yeh TF, et al. Renal response to frusemide in preterm
53. Pettit GW, Vick RL, Swander AM. Plasma K plus and infants with respiratory distress syndrome during the
insulin: changes during KCl infusion in normal and first three postnatal days. Arch Dis Child. 1985;60
nephrectomized dogs. Am J Physiol. 1975;228 (7):621–6.
(1):107–9. 72. Lui K, et al. Treatment with hypertonic dextrose and
54. DeFronzo RA, Bia M, Birkhead G. Epinephrine and insulin in severe hyperkalaemia of immature infants.
potassium homeostasis. Kidney Int. 1981;20 Acta Paediatr. 1992;81(3):213–6.
(1):83–91. 73. Lei J, Mariash CN, Ingbar DH. 3,30 ,5-Triiodo-L-
55. Brown RS. Extrarenal potassium homeostasis. Kid- thyronine up-regulation of Na, K-ATPase activity
ney Int. 1986;30(1):116–27. and cell surface expression in alveolar epithelial
56. Salem MM, Rosa RM, Batlle DC. Extrarenal potas- cells is Src kinase- and phosphoinositide 3-kinase-
sium tolerance in chronic renal failure: implications dependent. J Biol Chem. 2004;279(46):47589–600.
for the treatment of acute hyperkalemia. Am J Kidney 74. Lei J, et al. T3 increases Na-K-ATPase activity via a
Dis. 1991;18(4):421–40. MAPK/ERK1/2-dependent pathway in rat adult alve-
57. Todd EP, Vick RL. Kalemotropic effect of epineph- olar epithelial cells. Am J Physiol Lung Cell Mol
rine: analysis with adrenergic agonists and antago- Physiol. 2008;294(4):L749–54.
nists. Am J Physiol. 1971;220(6):1964–9. 75. Nakhoul F, Thompson CB, McDonough
58. Williams ME, et al. Impairment of extrarenal potas- AA. Developmental change in Na, K-ATPase alpha1
sium disposal by alpha-adrenergic stimulation. N and beta1 expression in normal and hypothyroid rat
Engl J Med. 1984;311(3):145–9. renal cortex. Am J Nephrol. 2000;20(3):225–31.
59. Williams ME, et al. Catecholamine modulation of 76. Adrogue HJ, Madias NE. Changes in plasma potas-
rapid potassium shifts during exercise. N Engl J sium concentration during acute acid–base distur-
Med. 1985;312(13):823–7. bances. Am J Med. 1981;71(3):456–67.
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 241

77. Burnell JM, et al. The effect in humans of extracellu- 95. Lorenz JM, Kleinman LI, Disney TA. Renal response
lar pH change on the relationship between serum of newborn dog to potassium loading. Am J Physiol.
potassium concentration and intracellular potassium. 1986;251(3 Pt 2):F513–9.
J Clin Invest. 1956;35(9):935–9. 96. McCance R, Widdowson EM. The response of the
78. Magner PO, et al. The plasma potassium concentra- new-born piglet to an excess of potassium. J Physiol.
tion in metabolic acidosis: a re-evaluation. Am J 1958;141(1):88–96.
Kidney Dis. 1988;11(3):220–4. 97. Lelievre-Pegorier M, et al. Developmental
79. Oster JR, Perez GO, Vaamonde CA. Relationship pattern of water and electrolyte transport in rat
between blood pH and potassium and phosphorus superficial nephrons. Am J Physiol. 1983;245(1):
during acute metabolic acidosis. Am J Physiol. F15–21.
1978;235(4):F345–51. 98. Kleinman LI, Banks RO. Segmental nephron sodium
80. Fulop M. Serum potassium in lactic acidosis and potassium reabsorption in newborn and adult
and ketoacidosis. N Engl J Med. 1979;300 dogs during saline expansion. Proc Soc Exp Biol
(19):1087–9. Med. 1983;173(2):231–7.
81. Graber M. A model of the hyperkalemia produced by 99. Giebisch GH. A trail of research on potassium. Kid-
metabolic acidosis. Am J Kidney Dis. 1993;22 ney Int. 2002;62:1498–512. doi:10.1046/j.1523-
(3):436–44. 1755.2002.t01-2-00644.x.
82. Fraley DS, Adler S. Correction of hyperkalemia by 100. Malnic G, Klose RM, Giebisch G. Micropuncture
bicarbonate despite constant blood pH. Kidney Int. study of distal tubular potassium and sodium transport
1977;12(5):354–60. in rat nephron. Am J Physiol. 1966;211(3):529–47.
83. Blumberg A, et al. Effect of various therapeutic 101. Malnic G, Klose RM, Giebisch G. Microperfusion
approaches on plasma potassium and major regulat- study of distal tubular potassium and sodium transfer
ing factors in terminal renal failure. Am J Med. in rat kidney. Am J Physiol. 1966;211(3):548–59.
1988;85(4):507–12. 102. Malnic G, Klose RM, Giebisch G. Micropuncture
84. Cooperman LH. Succinylcholine-induced study of renal potassium excretion in the rat. Am J
hyperkalemia in neuromuscular disease. JAMA. Physiol. 1964;206:674–86.
1970;213(11):1867–71. 103. Solomon S. Absolute rates of sodium and potassium
85. Martyn JA, Richtsfeld M. Succinylcholine-induced reabsorption by proximal tubule of immature rats.
hyperkalemia in acquired pathologic states: etiologic Biol Neonate. 1974;25(5–6):340–51.
factors and molecular mechanisms. Anesthesiology. 104. Bomsztyk K, Wright FS. Dependence of ion fluxes on
2006;104(1):158–69. fluid transport by rat proximal tubule. Am J Physiol.
86. Hertz P, Richardson JA. Arginine-induced 1986;250(4 Pt 2):F680–9.
hyperkalemia in renal failure patients. Arch Intern 105. Kibble JD, et al. Effect of barium on potassium diffu-
Med. 1972;130(5):778–80. sion across the proximal convoluted tubule of the
87. Perazella MA, Biswas P. Acute hyperkalemia associ- anesthetized rat. Am J Physiol. 1995;268(4 Pt 2):
ated with intravenous epsilon-aminocaproic acid ther- F778–83.
apy. Am J Kidney Dis. 1999;33(4):782–5. 106. Weinstein AM. Modeling the proximal tubule: com-
88. Ponce SP, et al. Drug-induced hyperkalemia. Medi- plications of the paracellular pathway. Am J Physiol.
cine (Baltimore). 1985;64(6):357–70. 1988;254(3 Pt 2):F297–305.
89. Ethier JH, et al. The transtubular potassium 107. Wilson RW, Wareing M, Green R. The role of active
concentration in patients with hypokalemia transport in potassium reabsorption in the proximal
and hyperkalemia. Am J Kidney Dis. 1990;15 convoluted tubule of the anaesthetized rat. J Physiol.
(4):309–15. 1997;500(Pt 1):155–64.
90. Rodriguez-Soriano J, et al. Renal handling of water 108. Fromter E, Gessner K. Free-flow potential profile
and sodium in infancy and childhood: a study using along rat kidney proximal tubule. Pflugers Arch.
clearance methods during hypotonic saline diuresis. 1974;351(1):69–83.
Kidney Int. 1981;20(6):700–4. 109. Edelman A, et al. Determination of intracellular K+
91. Hartnoll G, Betremieux P, Modi N. Body water con- activity in rat kidney proximal tubular cells. Pflugers
tent of extremely preterm infants at birth. Arch Dis Arch. 1978;378(1):37–45.
Child Fetal Neonatal Ed. 2000;83(1):F56–9. 110. Yao X, et al. Expression of KCNA10, a voltage-gated
92. Modi N, et al. Postnatal weight loss and contraction K channel, in glomerular endothelium and at the
of the extracellular compartment is triggered by apical membrane of the renal proximal tubule. J Am
atrial natriuretic peptide. Early Hum Dev. Soc Nephrol. 2002;13(12):2831–9.
2000;59(3):201–8. 111. Vallon V, et al. Role of KCNE1-dependent K+ fluxes
93. Gurkan S, et al. Potassium transport in the maturing in mouse proximal tubule. J Am Soc Nephrol.
kidney. Pediatr Nephrol. 2007;22(7):915–25. 2001;12(10):2003–11.
94. Tudvad F, Mc NH, Barnett HL. Renal response of 112. Wang Q, et al. Positional cloning of a novel potassium
premature infants to administration of bicarbonate channel gene: KVLQT1 mutations cause cardiac
and potassium. Pediatrics. 1954;13(1):4–16. arrhythmias. Nat Genet. 1996;12(1):17–23.
242 L.M. Satlin and D. Bockenhauer

113. Schulze-Bahr E, et al. KCNE1 mutations cause jervell 126. Mirochnick MH, et al. Renal response to furosemide
and Lange-Nielsen syndrome. Nat Genet. 1997;17 in very low birth weight infants during chronic admin-
(3):267–8. istration. Dev Pharmacol Ther. 1990;15(1):1–7.
114. Splawski I, et al. Mutations in the hminK gene cause 127. Imai M, Nakamura R. Function of distal convoluted
long QT syndrome and suppress IKs function. Nat and connecting tubules studied by isolated nephron
Genet. 1997;17(3):338–40. fragments. Kidney Int. 1982;22(5):465–72.
115. Ho K, et al. Cloning and expression of an inwardly 128. Satlin LM. Postnatal maturation of potassium trans-
rectifying ATP-regulated potassium channel. Nature. port in rabbit cortical collecting duct. Am J Physiol.
1993;362(6415):31–8. 1994;266(1 Pt 2):F57–65.
116. Zhou H, Tate SS, Palmer LG. Primary structure and 129. Schnermann J, Steipe B, Briggs JP. In situ studies of
functional properties of an epithelial K channel. Am J distal convoluted tubule in rat. II. K secretion. Am J
Physiol. 1994;266(3 Pt 1):C809–24. Physiol. 1987;252(6 Pt 2):F970–6.
117. Boim MA, et al. ROMK inwardly rectifying 130. Frindt G, Palmer LG. Effects of dietary K on cell-
ATP-sensitive K+ channel. II. Cloning and distribu- surface expression of renal ion channels and trans-
tion of alternative forms. Am J Physiol. 1995;268(6 Pt porters. Am J Physiol Renal Physiol. 2010;299(4):
2):F1132–40. F890–7.
118. International Collaborative Study Group for Bartter- 131. Vallon V, et al. Expression and phosphorylation of the
like Syndromes, c.o.G.K.L., Konrad M, Na+-Cl- cotransporter NCC in vivo is regulated by
Köckerling A, Ziegler A, Zimmermann DK, Roth B, dietary salt, potassium, and SGK1. Am J Physiol
Wieg C, Grzeschik K-H, Koch MC, Seyberth HW, Renal Physiol. 2009;297(3):F704–12.
Group 2: Vargas R, Forestier L, Jean G, Deschaux M, 132. Huang CL, Kuo E. Mechanisms of disease: WNK-ing
Rizzoni GF, Niaudet P, Antignac C, Group 3: at the mechanism of salt-sensitive hypertension. Nat
Feldmann D, Lorridon F, Cougoureux E, Laroze F, Clin Pract Nephrol. 2007;3(11):623–30.
Alessandri J-L, David L, Saunier P, Deschenes G, 133. Constantinescu A, Silver RB, Satlin LM. H-K-
Group 4: Hildebrandt F, Vollmer M, Proesmans W, ATPase activity in PNA-binding intercalated cells of
Brandis M, Group 5: van den Heuvel LP, Lemmink newborn rabbit cortical collecting duct. Am J Physiol.
HH, Nillesen W, Monnens LAH, Knoers NVAM, 1997;272(2 Pt 2):F167–77.
Group 6: Guay-Woodford LM, Wright CJ, 134. Satlin LM, Matsumoto T, Schwartz GJ. Postnatal mat-
Madrigal G, Hebert SC. Mutations in the gene uration of rabbit renal collecting duct. III. Peanut
encoding the inwardly-rectifying renal potassium lectin-binding intercalated cells. Am J Physiol.
channel, ROMK, cause the antenatal variant of 1992;262(2 Pt 2):F199–208.
Bartter syndrome: evidence for genetic heterogeneity. 135. Velazquez H, Ellison DH, Wright FS. Chloride-
International Collaborative Study Group for dependent potassium secretion in early and late renal
Bartter-like Syndromes. Hum Mol Genet. 1997;6 distal tubules. Am J Physiol. 1987;253(3 Pt 2):
(1):17–26. F555–62.
119. Simon DB, et al. Bartter’s syndrome, hypokalaemic 136. Wingo CS. Reversible chloride-dependent potassium
alkalosis with hypercalciuria, is caused by mutations flux across the rabbit cortical collecting tubule. Am J
in the Na-K-2Cl cotransporter NKCC2. Nat Genet. Physiol. 1989;256(4 Pt 2):F697–704.
1996;13(2):183–8. 137. Frindt G, Palmer LG. Apical potassium channels in
120. Simon DB, et al. Mutations in the chloride channel the rat connecting tubule. Am J Physiol Renal
gene, CLCNKB, cause Bartter’s syndrome type III. Physiol. 2004;287(5):F1030–7.
Nat Genet. 1997;17(2):171–8. 138. Frindt G, Palmer LG. Low-conductance K channels in
121. Birkenhager R, et al. Mutation of BSND causes apical membrane of rat cortical collecting tubule. Am
Bartter syndrome with sensorineural deafness and J Physiol. 1989;256(1 Pt 2):F143–51.
kidney failure. Nat Genet. 2001;29(3):310–4. 139. Wang WH, Schwab A, Giebisch G. Regulation of
122. Zink H, Horster M. Maturation of diluting capacity in small-conductance K+ channel in apical membrane
loop of Henle of rat superficial nephrons. Am J of rat cortical collecting tubule. Am J Physiol.
Physiol. 1977;233(6):F519–24. 1990;259(3 Pt 2):F494–502.
123. Yasui M, et al. Development of urinary concentrating 140. Lu M, et al. Absence of small conductance K+ chan-
capacity: role of aquaporin-2. Am J Physiol. nel (SK) activity in apical membranes of thick ascend-
1996;271(2 Pt 2):F461–8. ing limb and cortical collecting duct in ROMK
124. Schmidt U, Horster M. Na-K-activated ATPase: (Bartter’s) knockout mice. J Biol Chem.
activity maturation in rabbit nephron segments 2002;277(40):37881–7.
dissected in vitro. Am J Physiol. 1977;233(1): 141. Kleta R, Bockenhauer D. Bartter syndromes and other
F55–60. salt-losing tubulopathies. Nephron Physiol.
125. Smith FG, Abraham J. Renal and renin responses 2006;104(2):73–80.
to furosemide in conscious lambs during 142. Wang WH. Regulation of ROMK (Kir1.1) channels:
postnatal maturation. Can J Physiol Pharmacol. new mechanisms and aspects. Am J Physiol Renal
1995;73(1):107–12. Physiol. 2006;290(1):F14–9.
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 243

143. Wang WH, Giebisch G. Dual modulation of renal 160. Woda CB, et al. Flow-dependent K+ secretion in the
ATP-sensitive K+ channel by protein kinases A and cortical collecting duct is mediated by a maxi-K chan-
C. Proc Natl Acad Sci U S A. 1991;88(21):9722–5. nel. Am J Physiol Renal Physiol. 2001;280(5):
144. Xu ZC, Yang Y, Hebert SC. Phosphorylation of the F786–93.
ATP-sensitive, inwardly rectifying K+ channel, 161. Li D, et al. Inhibition of MAPK stimulates the Ca2+-
ROMK, by cyclic AMP-dependent protein kinase. dependent big-conductance K channels in cortical
J Biol Chem. 1996;271(16):9313–9. collecting duct. Proc Natl Acad Sci U S A. 2006;103
145. Schafer JA, Troutman SL. Effect of ADH on rubid- (51):19569–74.
ium transport in isolated perfused rat cortical 162. Pacha J, et al. Apical maxi K channels in intercalated
collecting tubules. Am J Physiol. 1986;250(6 Pt 2): cells of CCT. Am J Physiol. 1991;261(4 Pt 2):
F1063–72. F696–705.
146. Lin DH, et al. Protein tyrosine kinase is expressed and 163. Liu W, et al. Effect of flow and stretch on the [Ca2+]i
regulates ROMK1 location in the cortical collecting response of principal and intercalated cells in cortical
duct. Am J Physiol Renal Physiol. 2004;286(5): collecting duct. Am J Physiol Renal Physiol.
F881–92. 2003;285(5):F998–1012.
147. Lin DH, Sterling H, Wang WH. The protein tyrosine 164. Taniguchi J, et al. Pressure- and parathyroid-
kinase-dependent pathway mediates the effect of K hormone-dependent Ca2+ transport in rabbit
intake on renal K secretion. Physiology (Bethesda). connecting tubule: role of the stretch-activated
2005;20:140–6. nonselective cation channel. J Membr Biol.
148. Zeng WZ, et al. Evidence for endocytosis of ROMK 1994;140(2):123–32.
potassium channel via clathrin-coated vesicles. Am J 165. Palmer LG, Frindt G. High-conductance K channels
Physiol Renal Physiol. 2002;283(4):F630–9. in intercalated cells of the rat distal nephron. Am J
149. Wang W. Regulation of renal K transport by dietary K Physiol Renal Physiol. 2007;292(3):F966–73.
intake. Annu Rev Physiol. 2004;66:547–69. 166. Pluznick JL, et al. Renal fluid and electrolyte handling
150. Kahle KT, Ring AM, Lifton RP. Molecular physiol- in BKCa-beta1-/- mice. Am J Physiol Renal Physiol.
ogy of the WNK kinases. Annu Rev Physiol. 2003;284(6):F1274–9.
2008;70:329–55. 167. Rieg T, et al. The role of the BK channel in potassium
151. Wilson FH, et al. Molecular pathogenesis of inherited homeostasis and flow-induced renal potassium excre-
hypertension with hyperkalemia: the Na-Cl tion. Kidney Int. 2007;72(5):566–73.
cotransporter is inhibited by wild-type but not 168. Bailey MA, et al. Maxi-K channels contribute to
mutant WNK4. Proc Natl Acad Sci U S A. urinary potassium excretion in the ROMK-deficient
2003;100(2):680–4. mouse model of Type II Bartter’s syndrome and in
152. Kahle KT, et al. WNK4 regulates the balance between adaptation to a high-K diet. Kidney Int. 2006;70
renal NaCl reabsorption and K+ secretion. Nat Genet. (1):51–9.
2003;35(4):372–6. 169. Finer G, et al. Transient neonatal hyperkalemia in the
153. Wilson FH, et al. Human hypertension caused by antenatal (ROMK defective) Bartter syndrome.
mutations in WNK kinases. Science. 2001;293 J Pediatr. 2003;142(3):318–23.
(5532):1107–12. 170. Rodriguez-Soriano J. Bartter and related syndromes:
154. Xie J, et al. Role of with-no-lysine [K] kinases in the the puzzle is almost solved. Pediatr Nephrol. 1998;12
pathogenesis of Gordon’s syndrome. Pediatr Nephrol. (4):315–27.
2006;21(9):1231–6. 171. Simon DB, et al. Genetic heterogeneity of Bartter’s
155. Subramanya AR, et al. Dominant-negative regulation syndrome revealed by mutations in the K+ channel,
of WNK1 by its kidney-specific kinase-defective iso- ROMK. Nat Genet. 1996;14(2):152–6.
form. Am J Physiol Renal Physiol. 2006;290(3): 172. Flores D, et al. Flow-induced prostaglandin E2
F619–24. release regulates Na and K transport in the collecting
156. Boyden LM, et al. Mutations in kelch-like 3 and cullin duct. Am J Physiol Renal Physiol. 2012;303(5):
3 cause hypertension and electrolyte abnormalities. F632–8.
Nature. 2012;482(7383):98–102. 173. Wang Z, et al. Regulation of large-conductance Ca2+-
157. Louis-Dit-Picard H, et al. KLHL3 mutations cause activated K+ channels by WNK4 kinase. Am J
familial hyperkalemic hypertension by impairing ion Physiol Cell Physiol. 2013;305(8):C846–53.
transport in the distal nephron. Nat Genet. 2012;44 174. Buffin-Meyer B, et al. K depletion modifies the prop-
(4):456–60. S1–3. erties of Sch-28080-sensitive K-ATPase in rat
158. Shibata S, et al. Kelch-like 3 and Cullin 3 regulate collecting duct. Am J Physiol. 1997;272(1 Pt 2):
electrolyte homeostasis via ubiquitination and F124–31.
degradation of WNK4. Proc Natl Acad Sci U S A. 175. Codina J, DuBose Jr TD. Molecular regulation and
2013;110(19):7838–43. physiology of the H+, K+ -ATPases in kidney. Semin
159. Liu W, et al. Ca2+ dependence of flow-stimulated K Nephrol. 2006;26(5):345–51.
secretion in the mammalian cortical collecting duct. 176. Zhou X, et al. Increased CO2 stimulates K/Rb
Am J Physiol Renal Physiol. 2007;293(1):F227–35. reabsorption mediated by H-K-ATPase in CCD of
244 L.M. Satlin and D. Bockenhauer

potassium-restricted rabbit. Am J Physiol Renal 193. Aperia A, Elinder G. Distal tubular sodium
Physiol. 2001;281(2):F366–73. reabsorption in the developing rat kidney. Am J
177. Ahn KY, et al. Effects of chronic hypokalemia on Physiol. 1981;240(6):F487–91.
renal expression of the “gastric” H+-K+-ATPase 194. Garty H, Palmer LG. Epithelial sodium channels:
alpha-subunit gene. Am J Physiol. 1996;270(4 Pt 2): function, structure, and regulation. Physiol Rev.
F557–66. 1997;77(2):359–96.
178. Silver RB, Mennitt PA, Satlin LM. Stimulation of 195. Satlin LM, et al. Epithelial Na+ channels are regulated
apical H-K-ATPase in intercalated cells of cortical by flow. Am J Physiol Renal Physiol. 2001;280(6):
collecting duct with chronic metabolic acidosis. Am F1010–8.
J Physiol. 1996;270(3 Pt 2):F539–47. 196. Kleyman TR, Roberts C, Ling BN. A mechanism for
179. Palmer LG, Frindt G. Regulation of apical K channels pentamidine-induced hyperkalemia: inhibition of dis-
in rat cortical collecting tubule during changes in tal nephron sodium transport. Ann Intern Med.
dietary K intake. Am J Physiol. 1999;277(5 Pt 2): 1995;122(2):103–6.
F805–12. 197. Schlanger LE, Kleyman TR, Ling BN. K+-sparing
180. Wang W, et al. Protein tyrosine kinase regulates the diuretic actions of trimethoprim: inhibition of Na+
number of renal secretory K channels. Am J Physiol channels in A6 distal nephron cells. Kidney Int.
Renal Physiol. 2000;278(1):F165–71. 1994;45(4):1070–6.
181. Najjar F, et al. Dietary K+ regulates apical membrane 198. Muto S. Potassium transport in the mammalian
expression of maxi-K channels in rabbit cortical collecting duct. Physiol Rev. 2001;81(1):85–116.
collecting duct. Am J Physiol Renal Physiol. 199. Stapleton FB, et al. Hypokalemia associated
2005;289(4):F922–32. with antibiotic treatment. Evidence in children with
182. Silver RB, Soleimani M. H+-K+-ATPases: regulation malignant neoplasms. Am J Dis Child.
and role in pathophysiological states. Am J Physiol. 1976;130(10):1104–8.
1999;276(6 Pt 2):F799–811. 200. Morimoto T, et al. Mechanism underlying flow stim-
183. Evan AP, et al. Postnatal maturation of rabbit renal ulation of sodium absorption in the mammalian
collecting duct. II. Morphological observations. Am J collecting duct. Am J Physiol Renal Physiol.
Physiol. 1991;261(1 Pt 2):F91–107. 2006;291(3):F663–9.
184. Satlin LM, et al. Postnatal maturation of the rabbit 201. Malnic G, Berliner RW, Giebisch G. Flow depen-
cortical collecting duct. Pediatr Nephrol. 1988;2 dence of K+ secretion in cortical distal tubules of the
(1):135–45. rat. Am J Physiol. 1989;256(5 Pt 2):F932–41.
185. Minuth WW, et al. Expression of the alpha-subunit 202. Frindt G, et al. Dissociation of K channel density and
of Na/K-ATPase in renal collecting duct epithelium ROMK mRNA in rat cortical collecting tubule during
during development. Kidney Int. 1987;31 K adaptation. Am J Physiol. 1998;274(3 Pt 2):
(5):1104–12. F525–31.
186. Constantinescu AR, et al. Na+-K+-ATPase-mediated 203. Brandis M, Keyes J, Windhager EE. Potassium-
basolateral rubidium uptake in the maturing rabbit induced inhibition of proximal tubular fluid
cortical collecting duct. Am J Physiol Renal Physiol. reabsorption in rats. Am J Physiol. 1972;222
2000;279(6):F1161–8. (2):421–7.
187. Satlin LM, Palmer LG. Apical K+ conductance in 204. Sufit CR, Jamison RL. Effect of acute potassium load
maturing rabbit principal cell. Am J Physiol. on reabsorption in Henle’s loop in the rat. Am J
1997;272(3 Pt 2):F397–404. Physiol. 1983;245(5 Pt 1):F569–76.
188. Benchimol C, Zavilowitz B, Satlin LM. 205. Rabinowitz L. Aldosterone and potassium homeosta-
Developmental expression of ROMK mRNA in sis. Kidney Int. 1996;49(6):1738–42.
rabbit cortical collecting duct. Pediatr Res. 206. Youn JH. Gut sensing of potassium intake and its role
2000;47(1):46–52. in potassium homeostasis. Semin Nephrol. 2013;33
189. Zolotnitskaya A, Satlin LM. Developmental expres- (3):248–56.
sion of ROMK in rat kidney. Am J Physiol. 207. Morita H, et al. Hepatoportal bumetanide-sensitive K
1999;276(6 Pt 2):F825–36. +-sensor mechanism controls urinary K+ excretion.
190. Woda CB, et al. Ontogeny of flow-stimulated potas- Am J Physiol Regul Integr Comp Physiol. 2000;278
sium secretion in rabbit cortical collecting duct: func- (5):R1134–9.
tional and molecular aspects. Am J Physiol Renal 208. Ahloulay M, et al. Influence of glucagon on GFR and
Physiol. 2003;285(4):F629–39. on urea and electrolyte excretion: direct and indirect
191. Good DW, Wright FS. Luminal influences on potas- effects. Am J Physiol. 1995;269(2 Pt 2):F225–35.
sium secretion: sodium concentration and fluid flow 209. Pullman TN, Lavender AR, Aho I. Direct effects of
rate. Am J Physiol. 1979;236(2):F192–205. glucagon on renal hemodynamics and excretion of
192. Stokes JB. Potassium secretion by cortical collecting inorganic ions. Metabolism. 1967;16(4):358–73.
tubule: relation to sodium absorption, luminal sodium 210. Blachley JD, Crider BP, Johnson JH. Extrarenal
concentration, and transepithelial voltage. Am J potassium adaptation: role of skeletal muscle. Am J
Physiol. 1981;241(4):F395–402. Physiol. 1986;251(2 Pt 2):F313–8.
8 Physiology of the Developing Kidney: Potassium Homeostasis and Its Disorder 245

211. Hayslett JP, Binder HJ. Mechanism of potassium 229. Attali B, et al. A corticosteroid-induced gene
adaptation. Am J Physiol. 1982;243(2):F103–12. expressing an “IsK-like” K+ channel activity in
212. Alexander EA, Levinsky NG. An extrarenal mecha- Xenopus oocytes. Proc Natl Acad Sci U S A.
nism of potassium adaptation. J Clin Invest. 1995;92(13):6092–6.
1968;47(4):740–8. 230. Garty H, et al. A functional interaction between CHIF
213. Spat A, Hunyady L. Control of aldosterone secretion: and Na-K-ATPase: implication for regulation by
a model for convergence in cellular signaling path- FXYD proteins. Am J Physiol Renal Physiol.
ways. Physiol Rev. 2004;84(2):489–539. 2002;283(4):F607–15.
214. Young DB, et al. Multiplicative interaction between 231. Capurro C, et al. Cellular localization and regulation
angiotensin II and K concentration in stimulation of of CHIF in kidney and colon. Am J Physiol. 1996;271
aldosterone. Am J Physiol. 1984;247(3 Pt 1): (3 Pt 1):C753–62.
E328–35. 232. Wald H, et al. Aldosterone induction and epithelial
215. Engbretson BG, Stoner LC. Flow-dependent potas- distribution of CHIF. Am J Physiol. 1996;271(2 Pt 2):
sium secretion by rabbit cortical collecting tubule F322–9.
in vitro. Am J Physiol. 1987;253(5 Pt 2):F896–903. 233. Van Acker KJ, et al. Renin-angiotensin-aldosterone
216. Schwartz GJ, Burg MB. Mineralocorticoid effects on system in the healthy infant and child. Kidney Int.
cation transport by cortical collecting tubules in vitro. 1979;16(2):196–203.
Am J Physiol. 1978;235(6):F576–85. 234. Aperia A, et al. Sodium excretion in relation to
217. Palmer LG, Antonian L, Frindt G. Regulation of sodium intake and aldosterone excretion in newborn
apical K and Na channels and Na/K pumps in rat pre-term and full-term infants. Acta Paediatr Scand.
cortical collecting tubule by dietary K. J Gen Physiol. 1979;68(6):813–7.
1994;104(4):693–710. 235. Robillard JE, Nakamura KT, Lawton WJ. Effects
218. Masilamani S, et al. Aldosterone-mediated of aldosterone on urinary kallikrein and sodium
regulation of ENaC alpha, beta, and gamma subunit excretion during fetal life. Pediatr Res.
proteins in rat kidney. J Clin Invest. 1999;104(7): 1985;19(10):1048–52.
R19–23. 236. Stephenson G, et al. Ontogeny of renal mineralocor-
219. O’Neil RG, Hayhurst RA. Sodium-dependent modu- ticoid receptors and urinary electrolyte responses in
lation of the renal Na-K-ATPase: influence of miner- the rat. Am J Physiol. 1984;247(4 Pt 2):F665–71.
alocorticoids on the cortical collecting duct. J Membr 237. West ML, et al. New clinical approach to evaluate
Biol. 1985;85(2):169–79. disorders of potassium excretion. Miner Electrolyte
220. Pacha J, et al. Regulation of Na channels of the rat Metab. 1986;12(4):234–8.
cortical collecting tubule by aldosterone. J Gen 238. West ML, et al. Development of a test to evaluate the
Physiol. 1993;102(1):25–42. transtubular potassium concentration gradient in the
221. Summa V, et al. Short term effect of aldosterone on cortical collecting duct in vivo. Miner Electrolyte
Na, K-ATPase cell surface expression in kidney Metab. 1986;12(4):226–33.
collecting duct cells. J Biol Chem. 2001;276 239. Field MJ, Giebisch GJ. Hormonal control of renal
(50):47087–93. potassium excretion. Kidney Int. 1985;27(2):379–87.
222. Ikeda U, et al. Aldosterone-mediated regulation of 240. Rodriguez-Soriano J, Ubetagoyena M, Vallo
Na+, K+-ATPase gene expression in adult and A. Transtubular potassium concentration gradient: a
neonatal rat cardiocytes. J Biol Chem. 1991;266 useful test to estimate renal aldosterone bio-activity in
(18):12058–66. infants and children. Pediatr Nephrol. 1990;4
223. Garg LC, Knepper MA, Burg MB. Mineralocorticoid (2):105–10.
effects on Na-K-ATPase in individual nephron seg- 241. Malnic G, De Mello Aires M, Giebisch G. Potassium
ments. Am J Physiol. 1981;240(6):F536–44. transport across renal distal tubules during acid–base
224. Chen SY, et al. Epithelial sodium channel regulated disturbances. Am J Physiol. 1971;221(4):1192–208.
by aldosterone-induced protein sgk. Proc Natl Acad 242. Boudry JF, Stoner LC, Burg MB. Effect of acid lumen
Sci U S A. 1999;96(5):2514–9. pH on potassium transport in renal cortical collecting
225. Debonneville C, et al. Phosphorylation of Nedd4-2 by tubules. Am J Physiol. 1976;230(1):239–44.
Sgk1 regulates epithelial Na+ channel cell surface 243. Tabei K, et al. Potassium secretion is inhibited by
expression. EMBO J. 2001;20(24):7052–9. metabolic acidosis in rabbit cortical collecting ducts
226. Wulff P, et al. Impaired renal Na+ retention in vitro. Am J Physiol. 1995;268(3 Pt 2):F490–5.
in the sgk1-knockout mouse. J Clin Invest. 244. Beck FX, et al. The distribution of potassium, sodium
2002;110(9):1263–8. and chloride across the apical membrane of renal
227. Vallon V, et al. Role of Sgk1 in salt and potassium tubular cells: effect of acute metabolic alkalosis.
homeostasis. Am J Physiol Regul Integr Comp Pflugers Arch. 1988;411(3):259–67.
Physiol. 2005;288(1):R4–10. 245. Stone DK, et al. Mineralocorticoid modulation of
228. Huang DY, et al. Impaired regulation of renal K+ rabbit medullary collecting duct acidification. A
elimination in the sgk1-knockout mouse. J Am Soc sodium-independent effect. J Clin Invest.
Nephrol. 2004;15(4):885–91. 1983;72(1):77–83.
246 L.M. Satlin and D. Bockenhauer

246. Kone BC, Higham SC. A novel N-terminal splice potassium balance in severe chronic renal failure.
variant of the rat H+-K+-ATPase alpha2 subunit. Trans Assoc Am Physicians. 1967;80:207–16.
Cloning, functional expression, and renal adaptive 264. Sandle GI, et al. Enhanced rectal potassium secretion
response to chronic hypokalemia. J Biol Chem. in chronic renal insufficiency: evidence for large
1998;273(5):2543–52. intestinal potassium adaptation in man. Clin Sci
247. Moore-Ede MC, et al. Circadian variation in response (Lond). 1986;71(4):393–401.
to potassium infusion. Clin Pharmacol Ther. 265. Sandle GI, et al. Evidence for large intestinal control
1978;23(2):218–27. of potassium homoeostasis in uraemic patients under-
248. Moore-Ede MC, Herd JA. Renal electrolyte circadian going long-term dialysis. Clin Sci (Lond).
rhythms: independence from feeding and activity pat- 1987;73(3):247–52.
terns. Am J Physiol. 1977;232(2):F128–35. 266. Mathialahan T, et al. Enhanced large intestinal potas-
249. Gumz ML, et al. The circadian clock protein Period sium permeability in end-stage renal disease. J Pathol.
1 regulates expression of the renal epithelial sodium 2005;206(1):46–51.
channel in mice. J Clin Invest. 2009;119(8):2423–34. 267. Aizman R, Aizman O, Celsi G. Beta-adrenergic stim-
250. Zuber AM, et al. Molecular clock is involved in ulation of cellular K+ uptake in rat distal colon. Acta
predictive circadian adjustment of renal function. Physiol Scand. 1998;164(3):309–15.
Proc Natl Acad Sci U S A. 2009;106(38):16523–8. 268. Don BR, et al. Pseudohyperkalemia caused by fist
251. Rakova N, et al. Long-term space flight simulation clenching during phlebotomy. N Engl J Med.
reveals infradian rhythmicity in human Na(+) bal- 1990;322(18):1290–2.
ance. Cell Metab. 2013;17(1):125–31. 269. Stankovic AK, Smith S. Elevated serum potassium
252. Aizman RI, et al. Ontogeny of K+ transport in rat values: the role of preanalytic variables. Am J Clin
distal colon. Am J Physiol. 1996;271(2 Pt 1): Pathol. 2004;121(Suppl):S105–12.
G268–74. 270. Sevastos N, et al. Pseudohyperkalemia in serum: the
253. Foster ES, Hayslett JP, Binder HJ. Mechanism of phenomenon and its clinical magnitude. J Lab Clin
active potassium absorption and secretion in the rat Med. 2006;147(3):139–44.
colon. Am J Physiol. 1984;246(5 Pt 1):G611–7. 271. Sevastos N, et al. Pseudohyperkalemia in patients
254. Dawson DC. Ion channels and colonic salt transport. with increased cellular components of blood. Am J
Annu Rev Physiol. 1991;53:321–39. Med Sci. 2006;331(1):17–21.
255. Butterfield I, et al. Characterization of apical potas- 272. King MJ, Zanella A. Hereditary red cell membrane
sium channels induced in rat distal colon during disorders and laboratory diagnostic testing. Int J Lab
potassium adaptation. J Physiol. 1997;501 Hematol. 2013;35(3):237–43.
(Pt 3):537–47. 273. Montague BT, Ouellette JR, Buller GK. Retrospective
256. Pacha J, Popp M, Capek K. Corticosteroid regulation review of the frequency of ECG changes in
of Na+ and K+ transport in the rat distal colon during hyperkalemia. Clin J Am Soc Nephrol.
postnatal development. J Dev Physiol. 1988;10 2008;3(2):324–30.
(6):531–40. 274. Saxena K. Clinical features and management of poi-
257. Warth R, Bleich M. K+ channels and colonic func- soning due to potassium chloride. Med Toxicol
tion. Rev Physiol Biochem Pharmacol. Adverse Drug Exp. 1989;4(6):429–43.
2000;140:1–62. 275. Zettle RM, et al. Renal potassium handling during
258. Sausbier M, et al. Distal colonic K+ secretion states of low aldosterone bio-activity: a method to
occurs via BK channels. J Am Soc Nephrol. differentiate renal and non-renal causes. Am J
2006;17(5):1275–82. Nephrol. 1987;7(5):360–6.
259. Kunzelmann K, Mall M. Electrolyte transport in the 276. Dillon MJ, et al. Pseudohypoaldosteronism. Arch Dis
mammalian colon: mechanisms and implications for Child. 1980;55(6):427–34.
disease. Physiol Rev. 2002;82(1):245–89. 277. Hoorn EJ, et al. The calcineurin inhibitor tacrolimus
260. Rechkemmer G, Frizzell RA, Halm DR. Active potas- activates the renal sodium chloride cotransporter to
sium transport across guinea-pig distal colon: action cause hypertension. Nat Med. 2011;17(10):1304–9.
of secretagogues. J Physiol. 1996;493(Pt 2):485–502. 278. Bonny O, Rossier BC. Disturbances of Na/K balance:
261. Binder HJ, McGlone F, Sandle GI. Effects of cortico- pseudohypoaldosteronism revisited. J Am Soc
steroid hormones on the electrophysiology of rat Nephrol. 2002;13(9):2399–414.
distal colon: implications for Na+ and K+ transport. 279. Dellow EL, Unwin RJ, Honour JW. Pontefract cakes
J Physiol. 1989;410:425–41. can be bad for you: refractory hypertension and liquo-
262. Agarwal R, Afzalpurkar R, Fordtran JS. rice excess. Nephrol Dial Transplant. 1999;14
Pathophysiology of potassium absorption and secre- (1):218–20.
tion by the human intestine. Gastroenterology. 280. Seyberth HW, Schlingmann KP. Bartter- and
1994;107(2):548–71. Gitelman-like syndromes: salt-losing tubulopathies
263. Hayes Jr CP, McLeod ME, Robinson RR. An with loop or DCT defects. Pediatr Nephrol. 2011
extrarenal mechanism for the maintenance of Oct;26(10):1789–802
Physiology of the Developing Kidney:
Acid-Base Homeostasis and Its Disorders 9
Peter D. Yorgin, Elizabeth G. Ingulli, and Robert H. Mak

Contents Interpretation of Acid–Base Status


from Blood Gas Results . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248 General Approach to Evaluating
General Acid–Base Concepts . . . . . . . . . . . . . . . . . . . . . . 248 Acid–Base Status . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
pH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248 pH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
The Henderson–Hasselbalch Equation . . . . . . . . . . . . . . 248 Bicarbonate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 256
Buffering Systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249 Base Excess . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 258

Proximal Mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250 Metabolic and Respiratory Derangements . . . . . . . 260


Metabolic Acidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 260
NHE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250 Respiratory Acidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 261
Discriminating Between Acute and Chronic
V-ATPase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
Respiratory Acidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 262
Na+/K+-ATPase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251
Metabolic Alkalosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263
Carbonic Anhydrase (CA) . . . . . . . . . . . . . . . . . . . . . . . . . . . 252
Respiratory Alkalosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
Na+/HCO 3 Cotransporter (NBC) . . . . . . . . . . . . . . . . . . . . 252
Simple Acid–Base Disorders . . . . . . . . . . . . . . . . . . . . . . . . 265
Non-proximal Mechanisms . . . . . . . . . . . . . . . . . . . . . . . . 252 Mixed Acid–Base Disorders . . . . . . . . . . . . . . . . . . . . . . . . . 265
Loop of Henle and Thick Ascending Loop . . . . . . . 252 Nutrition and Acid–Base Balance . . . . . . . . . . . . . . . . . 267
Distal Convoluted Tubule . . . . . . . . . . . . . . . . . . . . . . . . . . 253 Acid–Base Balance in Neonates, Infants,
and Children . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 268
Ammonia Production and Transport . . . . . . . . . . . . . 254
Mechanisms of Growth Retardation in
Chronic Acidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 271
Effect of Acidosis on Bone and Calcium
Homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 272
P.D. Yorgin • R.H. Mak (*)
Acid–Base Balance in CKD . . . . . . . . . . . . . . . . . . . . . . . . 272
Pediatric Nephrology, University of California, San Diego,
CA, USA References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 274
Pediatric Nephrology Division, Rady Children’s Hospital,
San Diego, CA, USA
e-mail: romak@ucsd.edu
E.G. Ingulli
Department of Pediatrics, University of California,
San Diego, CA, USA
Division of Nephrology, Rady Children’s Hospital
San Diego, San Diego, CA, USA
Kidney Transplant Program, Rady Children’s Hospital,
San Diego, CA, USA

# Springer-Verlag Berlin Heidelberg 2016 247


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_8
248 P.D. Yorgin et al.

Introduction logarithmic scale to express H+ molality in a solu-


tion. The equation for pH is:
The body is highly dependent on acid–base control
by the kidneys, lungs, and buffer systems to pH ¼  log ½Hþ 
provide a cellular environment suitable for normal
health, growth, and development. The acid and A solution with a pH of 6 has 10 times the amount
alkali loads from ingesting food and fluid must be of H+ than a solution with a pH of 7. In water, the
managed so that the extracellular hydrogen pH of an acidic solution is less than 7 (H+ concen-
ion (H+) concentration is maintained within a very tration = 107 mmol/L), and a pH greater than
narrow range. There are serious consequences 7 is alkaline. In a healthy individual, the extracel-
from acid–base perturbations. Patients with severe lular fluid pH is maintained within a narrow range,
acidemia, high blood levels of H+, may have pH 7.38–7.42 (H+ concentration = 107.38–7.42 M).
problems with hyperkalemia, increased susceptibil- Serious health consequences are associated with
ity to cardiac dysrhythmias, osteopenia, recurrent abnormal blood pH values.
nephrolithiasis, skeletal muscle atrophy, and
growth retardation in children. Conversely, patients
with severe alkalemia, low blood H+ concentration, The Henderson--Hasselbalch Equation
may experience arteriolar constriction, refractory
dysrhythmias, hypoventilation, hypokalemia, The pH of blood can be directly measured, using a
decreased ionized calcium, paresthesias, and even pH probe (and the Nernst equation), by photo-
coma. The following information will serve as chemical sensors (optodes) [1] and semiconductor
practical assistance to physicians who manage sensor [2] methods, or can be calculated using a
acid–base problems in children. modified version of the Henderson–Hasselbalch
equation [3] which describes the relationship
between carbonic acid and bicarbonate:
General Acid–Base Concepts
H2 CO3 $ Hþ þ HCO3 
An acid is a compound, which donates a H+, often
The equation can be modified so that the con-
referred to as a proton, to a base, which is a proton
centration of H+ can be determined by knowing
acceptor. Protons react easily and rapidly with
both the HCO3 and H2CO3 concentrations.
other molecules in serum and particularly with
water (H2O). There are strong and weak acids.
Strong acids, like hydrogen chloride and sulfuric log½Hþ  ¼ pH ¼ pK þ log ð½HCO3  =½H2 CO3 Þ
acid, are good at donating hydrogen ions, and they
ionize fully when dissolved in water. Weak acids, The pK, in this equation, represents the pH
like carbonic acid, do not ionize fully when value at which HCO3 and H2CO3 are found in
dissolved in water. Solutions of weak acids and equal concentrations and represents the pH at
salts of their conjugate bases form buffer solu- which there is the greatest amount of buffering
tions. An acid is considered to be dissociated, capacity. The pK of the CO2 – HCO3 buffering
after the H+ has been donated. An alkali is a base system is 6.1. At a pH of 7.40, carbonic acid
that is soluble in water and donates hydroxide ions concentrations are very low and cannot be mea-
(OH). sured easily. In most equations, the carbonic acid
value is replaced by a solubility coefficient multi-
plied by the partial pressure of carbon dioxide
pH (PaCO2), which can be easily measured. pH can
then be defined:
pH provides a convenient means of expressing the
concentration of H+ in water. pH utilizes a pH ¼ pKa þ logð½HCO3  =½ð0:0308ÞðCO2 ÞÞ
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 249

Buffering Systems With this reaction, a strong base is exchanged for a


weak base, causing only a minor shift toward an
CO2/Bicarbonate Buffering System alkaline pH.
Buffers, found within cells and in blood, quickly The phosphate buffering system has a pKa of
limit the change in extracellular H+ concentrations 6.8, which means that there are similar amounts of
by combining with an acid or a base. An H2PO4 and HPO4 at the pH of 7.40. Therefore
acid–base buffer is usually composed of two or the phosphate buffering system has its best buff-
more chemical compounds. ering capacity in the normal blood pH range. Yet,
The carbonic acid + bicarbonate extracellular the concentration of H2PO4 and HPO4 is much
buffering system has two buffering molecules: less than that of bicarbonate system and therefore
carbonic acid (H2CO3) and bicarbonate (HCO3). contributes less buffering capacity.
H+ and bicarbonate can combine to form carbonic
acid. Carbonic acid, in a reversible process, disso- Intracellular Buffering
ciates to yield water and carbon dioxide: Cells and calcium salts in the bone contribute to
the buffering of extracellular fluid. Intracellular
Hþ þ HCO3  $ H2 CO3 $ H2 O þ CO2 proteins including hemoglobin, imidazole, and
histidine act as potent buffers and perhaps account
If an acid is added to the carbonic acid + bicar- for as much as three-quarters of all chemical buff-
bonate buffering system, more carbonic acid is ering in the body [4]. Lysosomes and mitochon-
generated as shown in the following reaction: dria may also play a role in intracellular buffering
of H+. Intracellular H+ concentrations are primar-
Hþ þ HCO3  $ H2 CO3 ily affected by carbon dioxide concentrations
which rapidly diffuse through the cell membrane
If a base is added to the carbonic acid + bicarbon- to affect intracellular pH [5]. Bicarbonate, which
ate buffering system, more bicarbonate is gener- can move into the cell via an electrogenic
ated as shown in the following reaction: Na–HCO3 cotransport mechanism, also influ-
ences intracellular pH [6].
OH þ H2 CO3 $ H2 O þ HCO3 
Pulmonary Regulation
The pulmonary compensatory response to acute
Phosphate and Protein Buffering
metabolic acidosis or alkalosis is swift. If a strong
Systems
acid is added to the blood, the pH begins to drop as
The phosphate buffering system functions like the
bicarbonate is consumed and PaCO2 begins to
carbonic acid + bicarbonate buffering system.
rise. Blood pH cannot directly influence the cen-
Two compounds, dihydrogen phosphate
tral respiratory center due to the blood–brain bar-
(H2PO4) and hydrogen phosphate (HPO4), act
rier. Instead, as PaCO2 increases, CO2 carbonic
as buffers. When a strong acid is added, the fol-
acid and H+ levels increase in the cerebral spinal
lowing reaction occurs:
fluid. Hydrogen ions directly stimulate the central
respiratory center causing ventilation to increase.
HCl þ Na2 HPO4 ! NaH2 PO4 þ NaCl Additional ventilatory stimuli are supplied by
peripheral chemoreceptors which respond to the
In this reaction, a strong acid, hydrochloric acid, is rising H+ concentrations. Because the lungs rep-
converted into a weak acid, NaH2PO4, thereby resent an open system by which CO2 can be
only causing a minor change in the pH. If a strong rapidly disposed, the blood pH can be rapidly
base is added to the buffer system, the following corrected to 7.40 by decreasing the PaCO2 value
reaction occurs: to less than 40 mmHg.
When a strong base is added to the blood, the
NaOH þ NaH2 PO4 ! Na2 HPO4 þ H2 0 pH rises rapidly and carbonic acid and PaCO2
250 P.D. Yorgin et al.

levels decrease. Ventilatory drive decreases in glutamine to bicarbonate and NH3. The bicarbon-
response to decreased serum and cerebrospinal ate moves out the distal tubule cell into the extra-
fluid H+ levels, causing an increase in PaCO2. cellular fluid by a chloride–bicarbonate
However, the effectiveness of hypoventilation as exchanger.
a compensatory response is limited, as patients
will not hypoventilate to the point where they
become hypoxic. Proximal Mechanisms

Renal Regulation Even though the proximal tubule of the nephron is


The kidneys play a major role in acid–base responsible for reabsorbing approximately 80 %
homeostasis through reabsorption of filtered of the filtered bicarbonate, there is no evidence of
bicarbonate, secretion of hydrogen ions, and direct bicarbonate reabsorption [7]. Bicarbonate
ammonia (NH3) excretion. The proximal tubule reabsorption in the proximal tubule involves the
reabsorbs 80 % of the filtered bicarbonate, the integrated function of multiple exchanger pro-
thick ascending limb of the nephron reabsorbs teins. The process of bicarbonate reabsorption
15 %, and the distal nephron reabsorbs the begins with H+ secretion by both the Na+/H+
remaining 5 %. Very little of the filtered HCO3 exchangers (NHE) (SLC9A3) and H+-ATPase
is excreted in the urine. (V-ATPase a2 isoform). Most of the H+ secreted
The second function of the kidney is to secrete by the proximal tubule is used to reclaim bicar-
hydrogen ions, in order to cope with dietary or bonate. The remaining secreted H+ will be com-
endogenous metabolic acids. These acids come bined with phosphate as titratable acid (Fig. 1) [4].
from dietary carbohydrate and fat metabolism,
which yields large quantities of CO2 and carbonic
acid. Dietary and endogenous acids amount to NHE
about 1 mEq/kg body weight per day in adults
ingesting a typical Western diet. Oxidation of Sodium–hydrogen ion exchangers (NHE) are
dibasic and sulfur-containing amino acids gener- members of a large monovalent cation–proton
ates small quantities of hydrogen ions. Hydrogen antiporter family. NHE isoforms 2, 3, and 8 are
ions secreted by the distal tubule react with expressed along the apical membrane and extrude
(1) dihydrogen phosphate (H2PO4) and hydro- hydrogen ions utilizing a high inward sodium
gen phosphate (HPO4) to form phosphoric acid gradient [8]. NHE2 (SLC9A2), which is
and (2) NH3 to form ammonium NH4+. The term expressed from the proximal tubule to the distal
titratable acid (TA) refers to acid excreted that has nephron, is thought to have a minor role in urinary
been bound to urinary buffers. It equals the acidification. NHE3 (SLC9A3) is expressed in the
amount of acid (H+) that is added to the tubular proximal tubule and thick ascending limb (TAL).
fluid by the nephron and is a function of both urine NHE3 generates most of the urinary H+ needed
pH and buffering capacity. Titratable acids for the reabsorption of bicarbonate in the proximal
(TA) found in the urine include phosphoric and tubule and TAL. NHE8 (SLC9A8) has been iden-
sulfuric acid, but not ammonium. tified in the proximal tubule of younger animals,
The production of new bicarbonate, which is but its role during early development is not
not filtered, by the kidney is quantitated by urinary known. NHE1 (SLC9A1) and NHE4 (SLC9A4),
net acid excretion (NAE), calculated as: which localize to the basolateral membrane, may
mediate NH4+ reabsorption in the TAL, prior to
 þ  its final secretion in the distal tubule/collecting
NAE ¼ NH4 þ TA  HCO3 
duct. Murine studies have suggested that
two-thirds of the bicarbonate reabsorption in the
Urinary NH3 excretion produces new bicarbonate proximal tubule is achieved through the NHE
in the distal tubule cell by the metabolism of [9–12].
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 251

Fig. 1 Bicarbonate reabsorption in the proximal tubule. accelerated by cytoplasmic CA II. H2CO3 rapidly dissoci-
Proximal tubule HCO3 reabsorption involves the inte- ates to H+ and HCO3, thereby replenishing the secreted
grated function of multiple proteins. H+ ions are secreted cytosolic H+. Cytosolic HCO3 exits across the basolateral
by both the Na+/H+ exchanger NHE3 and H+-ATPase and plasma membrane primarily by the Na+ (HCO3)
titrate luminal HCO3 to H2CO3. Luminal H2CO3 dehy- cotransporter NBCe1. Basolateral CA II and CA IV facil-
dration to H2O and CO2 is accelerated by luminal carbonic itate HCO3 transport (Weiner DI, Verlander JW. Renal
anhydrase (CA) activity mediated by CA IV. CO2 enters acidification mechanisms. Brenner & Rector’s The Kidney,
the cell via aquaporin-1 (AQP1) and, most likely, via 9th Edition, 2012, Chapter 9, pp 293–325. Reproduced
diffusive movement, where its hydration to H2CO3 is with permission from Ref. [4])

V-ATPase Na+/K+-ATPase

The remaining one-third of bicarbonate Sodium can move passively into the proximal
reabsorption is achieved through V-ATPase a2 iso- tubular cell facilitated by a concentration gradient
form which is similar to V-ATPase B1 isoform, the generated within the cell. The sodium concentra-
distal nephron [13]. The H+-ATPase [9–12, 14] is tion gradient in the tubular cell is generated by the
an apically located, multi-subunit vacuolar type basolateral Na+/K+-ATPase (ATP1A1). This is an
[15–17]. There is a maturational increase in both active process, which extrudes three sodium
NHE activity [18–22] and basolateral Na+/K+- ions in exchange for two potassium ions.
ATPase activity [23, 24] resulting in an increase in The sodium concentration inside the proximal
proximal tubule acidification during development. tubule is usually low, around 19 mM [25].
252 P.D. Yorgin et al.

The Na+/K+-ATPase pump indirectly provides the to the basolateral membrane of α-intercalated cells
energy that allows most of the transport proteins to in the collecting duct. mRNA levels of SLC26A7
translocate filtered solutes in the proximal tubule. increase in response to water deprivation.
The sodium–bicarbonate cotransporter 1,
NBC1 (SLC4A7), is an electrogenic sodium–
Carbonic Anhydrase (CA) bicarbonate transporter located on the basolateral
membrane of the proximal tubule [28]. NBC1 is
H+ in the urinary lumen reacts with HCO3 to responsible for transporting three bicarbonate
form carbonic acid (H2CO3). The rate of luminal molecules and one sodium molecule into the
dehydration of H2CO3 to CO2 and H2O is dramat- blood [29]. NBC1 appears to be the dominant
ically increased by the luminal carbonic pathway for HCO3 reabsorption in the S1 and
anhydrase isoenzyme type IV (CA IV), a member S2 segments of the proximal tubule, but a
of zinc metalloenzyme family. This zinc molecule basolateral Cl-HCO3 exchanger plays a role in
enhances H+ transfer to and from the active site by the S3 segment [30–32].
weakening the hydrogen–oxygen bond. Luminal
H2O and CO2 move across the apical plasma
membrane via aquaporin (AQP)-1. Cytosolic Non-proximal Mechanisms
CO2 is then hydrated in the presence of carbonic
anhydrase (CA) II, forming carbonic acid, which The distal nephron is responsible for net acid
then rapidly dissociates to H+ and HCO3, excretion. As in the proximal tubule, processes
thereby replenishing the secreted cytosolic H+26. are at work to reabsorb the remaining filtered
Currently, there are 15 known CA isoenzymes bicarbonate. In addition, protons are actively
which enhance H+/HCO3 transport [26]. CA II transported to the tubular lumen where they com-
accounts for 95 % of the CA activity in the kidney bine with NH3 and HPO42 to form H2PO4 and
and resides primarily within the cytosol. CA IV NH4+ resulting in urinary acidification.
and CA XII account for the remaining 5 % and are
membrane associated. CA II interacts with a num-
ber of transporters including AE1, AE2, and AE3 Loop of Henle and Thick
and proximal tubule NBCe1, which facilitate Ascending Loop
movement of HCO3 [27]. Basolateral CA II
and CA IV also facilitate HCO3 transport. The TAL of the loop of Henle contributes to
acid–base homeostasis through its roles in
reabsorbing approximately 15–20 % of the fil-
Na+/HCO3 Cotransporter (NBC) tered bicarbonate load and in contributing to the
development of the medullary interstitial ammo-
In general, there are two main groups of nia gradient that is critical for collecting
bicarbonate transporters: chloride–bicarbonate duct ammonia secretion. H+ is secreted by
exchangers (anion exchange transporter) and apical Na+/H+ exchange activity and vacuolar
sodium–bicarbonate cotransporter (NBC). Cyto- H+-ATPase. Quantitatively, apical Na+/H+
solic HCO3 exits across the basolateral plasma exchange activity is the predominant apical H+
membrane primarily by the Na cotransporter mechanism; vascular H+-ATPase is also present.
NBCe1 (gene: SLC4A4) in a 3 HCO3 to 1 Na+ Two Na+/H+ exchanger isoforms are present in the
ratio. The chloride–bicarbonate exchangers, TAL, NHE2 (SLC9A2), and NHE3 (SLC9A3).
SLC26A6 and SLC26A7, transport oxalate, sul- Cytoplasmic CA II catalyzes CO2 hydration to
fate, chloride, and bicarbonate out of tubule cells, form H2CO3, which dissociates to H+ and
across the basal lamina. SLC26A6 is distributed HCO3, thereby recycling the H+ secreted across
widely in the proximal tubule, TAL, macula densa, the apical plasma membrane. Cytosolic HCO3
and distal convoluted tubules. SLC26A7 localizes exists via a basolateral Cl/ HCO3 exchanger
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 253

Fig. 2 Intercalated cell subtypes in the distal nephron. The transport, including H+–adenosine triphosphatase
distal nephron – that is, the connecting segment, initial (ATPase), anion exchanger 1 (AE1), pendrin, Rh B glyco-
collecting tubule, cortical collecting duct (CCD), outer protein (Rhbg), and Rh C glycoprotein (Rhcg) (Weiner DI,
medullary collecting duct (OMCD), and inner medullary Verlander JW. Renal acidification mechanisms. Brenner &
collecting duct (IMCD) – has multiple distinct cell types. Rector’s The Kidney, 9th Edition, 2012, Chapter 9, pp
Three intercalated cell types can be distinguished based on 293–325. Modified from an original drawing by of
differential expression in different plasma membrane Ki-Hwan Han, M.D., Ph.D., Ewha Womans University,
domains of several proteins involved in renal acid–base Seoul, Korea. Reproduced with permission from Ref. [4])

(AE1) and possibly AE2, via an electroneutral contains both α- and β-intercalated cells, while
sodium–bicarbonate cotransporter (NBCn1, the OMCD contains mostly α-intercalated cells
SLC4A7), and possibly via basolateral Cl chan- (Figs. 2 and 3) [4].
nels. An apical K+-dependent HCO3 transporter The α-intercalated cell actively transports H+
mediates coupled K+ and HCO3 transport and into the urine via a vacuolar H+-ATPase and to a
functions in parallel with apical Na+/H+ exchange lesser extent a H+/K+-ATPase on the luminal sur-
and, because of a cytoplasmic to luminal K+ gra- face [34]. Intracellular CA II is required to gener-
dient, may secrete HCO3. NBCn1 is expressed ate intracellular H+ for active transport. An apical
in the basolateral membrane in the TAL and in the Na+/H+ exchanger (NHE2) also secretes H+ into
apical membranes of the outer (OMCD) and inner the tubule [35, 36]. Proton secretion in the
(IMCD) medullary collecting duct intercalated collecting duct serves to reabsorb the remaining
cells. Both metabolic acidosis and hypokalemia HCO3– that has evaded earlier transport
increase TAL NBCn1 expression. mechanisms of the nephron. In addition, secreted
H+ combines with HPO42 to form H2PO4 and
NH3 to trap NH4+. HCO3 is transported from
Distal Convoluted Tubule the cell to the interstitium via a basolateral
Cl/HCO3 anion exchanger (AE1). Other
Urine acidification occurs mainly in the collecting exchangers such as AE2, AE4, and SLC26A7
duct [33]. Two major cell types are found inter- (pendrin) have been identified on the basolateral
spersed in the collecting duct: the principal cell membrane of selective cells within the IMCD or
and the intercalated cell. The principal cell is OMCD cells contributing to HCO3 exchange
primarily involved with Na+ reabsorption and K+ [37–41].
secretion and plays a little role in acid–base bal- The β-intercalated cell is opposite in polarity to
ance. The intercalated cell is primarily involved in the α-intercalated cell. The function is to actively
acid–base transport and regulation. There are two secrete HCO3 into the urine coupled to Cl
types of intercalated cells that display different reabsorption but independent of Na+. HCO3 is
functions. The α-intercalated cells secrete pro- moved from the lumen into the cell through
tons, while the β-intercalated cells secrete bicar- pendrin (SLC26A4), a Cl/HCO3 exchanger
bonate. The cortical collecting duct (CCD) on the apical membrane [42].
254 P.D. Yorgin et al.

Fig. 3 Bicarbonate reabsorption by the type A interca- H2CO3. Cytosolic HCO3 exits across the basolateral
lated cell. The type A intercalated cell is present from the plasma membrane via the kidney isoform of anion
connecting segment through the initial inner medullary exchanger 1, kAE1. Cl that enters via kAE1 recycles via
collecting duct (IMCD). Two families of H+ transporters, a basolateral Cl channel. K+ that enters via apical H+-K+-
H+–adenosine triphosphatase (ATPase), and H+-K+- ATPase can either recycle via an apical Ba+-sensitive K+
ATPase are present in the apical plasma membrane. channel or be reabsorbed via a basolateral Ba+-sensitive K+
Secreted H+ ions titrate luminal HCO3 – to form H2CO3 – channel. A basolateral Na+/H+ exchanger is present but
which dehydrates to H2O and CO2. Luminal carbonic does not contribute to bicarbonate reabsorption and is not
anhydrase activity, most likely mediated by carbonic shown (Weiner DI, Verlander JW. Renal acidification
anhydrase IV (CA IV), is variably present in the collecting mechanisms. Brenner & Rector’s The Kidney, 9th Edition,
duct. Cytosolic H+ and HCO3 are formed from CA 2012, Chapter 9, pp 293–325. Reproduced with permission
II-accelerated hydration of CO2 and rapid dissociation of from Ref. [4])

Ammonia Production and Transport IMCD, all have the capability to synthesize
NH3, with glutamine being the primary meta-
NH3 is produced by almost all renal epithelial bolic substrate [43]. Phosphate-dependent gluta-
cells. The proximal tubule is the primary site minase (PDG) is involved in this process in each
for physiologically relevant ammoniagenesis of these segments [44]. The proximal tubule
(Fig. 4) [4]. The glomeruli proximal tubule seg- accounts for 60–70 % of the total renal NH3
ments, descending thin limb of the loop of Henle, production under basal conditions and at least
medullary and cortical TAL of the loop of Henle, 70–80 % of production in response to metabolic
distal convoluted tubule, CCD, OMCD, and acidosis [43].
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 255

Fig. 4 Summary of renal ammonia metabolism. The prox- 20 % of final urinary ammonia; the remaining approxi-
imal tubule produces ammonia, as NH4+, from glutamine. mately 80 % is secreted in the collecting duct through
NH4+ is then secreted preferentially into the luminal fluid, parallel NH3 and H+ transport. Numbers in red indicate
primarily by Na+/H+ exchanger isoform 3 (NHE3). It is the proportion of total urinary ammonia present at the
then reabsorbed by the thick ascending limb of the loop of indicated sites under baseline conditions (Weiner DI,
Henle (TAL), primarily by Na+/K+-2Cl cotransporter Verlander JW. Renal acidification mechanisms. Brenner
type 2 (NKCC2). This results in the delivery of ammonia & Rector’s The Kidney, 9th Edition, 2012, Chapter 9, pp
to the distal nephron, which accounts for approximately 293–325. Reproduced with permission from Ref. [4])

Multiple conditions simulate renal ammonia- the thin descending limb of the loop of Henle,
genesis. Both acute and chronic metabolic acidoses which results in countercurrent amplification of
stimulate ammoniagenesis [45] and appear to do so medullary interstitial ammonia concentration.
by stimulating the PDG pathway. NH3 produced in NH3 recycling predominantly involves NH3 trans-
the proximal tubule is secreted preferentially into port, with a smaller component of NH3
the tubule lumen, although there is some transport transport [47].
across the basolateral membrane. The proximal NH3 movement across collecting duct cell api-
tubule also can reabsorb luminal NH3; this appears cal and basolateral membrane appears to involve
to occur primarily in the late proximal tubule [46]. both diffusive and transporter-mediated NH3
The apical Na+/K+-2Cl cotransporter NKCC2 transport. The Rh glycoprotein Rhcg plays a crit-
mediates the majority of NH3 reabsorption. ical role in renal NH3 excretion. Rhcg is an NH3-
Some of the NH3 absorbed by the medullary specific transporter with no identifiable affinity for
TAL of the loop of Henle undergoes recycling into solutes other than NH3.
256 P.D. Yorgin et al.

Interpretation of Acid–Base Status bicarbonate will increase or decrease to compen-


from Blood Gas Results sate, thereby normalizing the pH. When a primary
respiratory or metabolic process is so severe that it
General Approach to Evaluating overwhelms the compensatory systems or the
Acid–Base Status compensatory response is inadequate to the per-
turbation, the blood pH will not be normal.
To effectively assess acid–base status, the clini-
cian is greatly assisted by reviewing simultaneous Example
blood chemistry and blood gas results. The fol- A neonate with respiratory distress has the follow-
lowing steps are most helpful in yielding a ing blood gas: pH: 7.16, PaCO2: 70 mmHg,
diagnosis: HCO3: 26 mmol/L. What is the primary
disturbance?
1. Assess the serum pH as it generally indicates
whether a patient is acidemic (pH < 7.38) or Answer In this particular case, the patient is
alkalemic (pH > 7.42). acidemic. The PaCO2 is very high due to a pri-
2. Assess the PaCO2 and serum total CO2/HCO3 mary respiratory acidosis. Note that the HCO3 is
values relative to the pH to determine the pri- slightly above normal so it is not the cause of the
mary cause of the pH disturbance. acidemia.
3. Calculate the expected respiratory or renal
compensation. If memorization of the compen-
sation rules seems overwhelming, the modified PaCO2
version of the Henderson–Hasselbalch equa- The partial pressure of carbon dioxide (PaCO2) in
tion can be used to determine the appropriate the blood (normal range 35–35 mmHg) has pro-
respiratory or renal response when one found effects on blood pH. The arterial PaCO2
assumes a normal pH. will vary inversely with alveolar ventilation in a
4. Calculate the anion gap, delta anion gap, and linear manner:
delta ratio to distinguish the cause for meta-
bolic acidosis and to determine if there is an pa CO2  K  V’CO2 =V’A
additional acid–base disorder.
where V’CO2 is the volume of carbon dioxide
produced by the body’s metabolism per minute
pH (l/min) and V’A is the alveolar ventilation (l/min).
Thus, assuming a constant metabolic produc-
The first step in analyzing a blood gas is to deter- tion of CO2, a reduction in alveolar ventilation by
mine whether the patient is acidemic or alklemic. half will lead to a doubling of PaCO2 from
The normal pH of extracellular fluids is 40 mmHg to 80 mmHg causing a respiratory
7.38–7.42. Acidemia is present whenever the acidosis. In the context of a low blood pH, high
blood pH is lower than 7.38 and will be attributed PaCO2 values typically point to a primary respi-
to an increase in PaCO2 or decrease in HCO3. ratory acidosis.
Any uncompensated decrease in PaCO2 or
increase in HCO3 such that the blood pH is
higher than 7.42 is characterized as an alkalemia. Bicarbonate
When there is a perturbation in the blood pH, the
etiology is either respiratory or metabolic. Serum bicarbonate (HCO3) or total CO2 values
Any acute change in the H+ concentration in are routinely measured using analytical clinical
the blood causes a defense of the pH by compen- laboratory or blood gas equipment. The actual
satory mechanisms. For example, if there is a bicarbonate, standard bicarbonate, and base
perturbation in PaCO2, then the serum excess are then computed from the pH and
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 257

Fig. 5 Scales for pH, Total-CO2 mMol/l plasma.


PaCO2, base excess of 60
HCO3− mEq/l plasma.
whole blood of different 60
hemoglobin concentrations, Base Excess
plasma bicarbonate, plasma 50 mEq/l blood or plasma.
CO2 by Siggaard-Andersen 50 +30
(Strnad Z. [Numerical
calculation of basic
+25 pH
indicators of blood acid- 40
40 8.0
base balance using an
equilibration method]. Vet +20
35 35 7.9
Med 1986;31:557–64.
Reproduced with
permission from Ref. [48]) 30 +15 7.8
30

+10 7.7
25 25
0 +5 7.6

5 7.5
20
20 −5
7.4

7.3
−10
15
7.2
15
7.1
−15
7.0

10 6.9
25
9 20
−20 6.8
10
8 15
6.7
9 10
7
00 ml.

6.6
8
lobin g/1

6 −25 5

7
Hemog

6
4
0
−30

PaCO2 using the Siggaard-Andersen nomogram acid (H2CO3), dissolved CO2, carbonate, and
(Fig. 5) [48]. Some laboratories will report serum CO2 bound to amino acids. Total CO2 can be
total CO2 values instead of HCO3. The total CO2 assayed by adding a strong acid to the blood.
value includes bicarbonate (HCO3), carbonic Because 95 % of the total CO2 value is HCO3,
258 P.D. Yorgin et al.

the total CO2 value is usually 2 mmol/L higher the pH of the blood. Normal values range from 2
than the calculated HCO3 value. Serum HCO3 to +2 mEq/L. Base excess can be determined by
levels can be estimated from total CO2 values. plotting the values on the Siggaard-Andersen
Serum bicarbonate and total CO2 can be deter- nomogram (Fig. 5) or by calculating the formula
mined from known pH and PaCO2 values using where the base excess is based on PaCO2 and pH
the Henderson–Hasselbalch equation. or PaCO2 and HCO3:
Serum total CO2 and HCO3 values change in
response to blood CO2 levels due, in part, to the H+ 
Base excess ¼ 0:9287 ðHCO3  24:4Þ
accepting capacity of hemoglobin and other pro-
þ ð14:83 ðpH  7:4ÞÞ
teins. When CO2 is bubbled through a bicarbonate
solution without hemoglobin, the bicarbonate con-
centration does not change. Therefore, hemoglobin Positive base excess numbers are seen when
plays a role in mediating bicarbonate response to there is a metabolic alkalosis, whereas negative
PaCO2. If sodium bicarbonate is infused into the numbers indicate the presence of a metabolic aci-
blood, PaCO2 values increase due to the carbon dosis. The base deficit can be used to calculate the
dioxide + water $ carbonic acid $ hydrogen + amount of bicarbonate (or other base) needed to
bicarbonate buffer system. normalize the pH. Since the volume of distribu-
tion of bicarbonate is 0.4–0.5 [50, 51], the formula
Example calculating the amount of base that is required to
A 17-year-old girl with renal tubular acidosis has normalize the pH of the blood is as follows:
a serum bicarbonate of 6 mmol/L, a pH of 7.10,
and a PaCO2 of 17.5 mmHg. After intravenous Base required ¼ ðBase deficitÞ  ðWeight in kgÞ
fluids and 100 mEq of sodium bicarbonate were  0:4  0:5
infused, a repeat blood gas shows a bicarbonate
value of 11 mmol/L, PaCO2 of 29, and a pH of
7.17. Why is the PaCO2 rising? Example
A 12-year-old male with chronic renal failure
Answer As sodium bicarbonate is infused, the (estimated GFR = 25.3 mL/min/1.73 m [2]) aspi-
PaCO2 rises because of a decrease in respiratory rates after having a hypertension-induced seizure
drive from the rising pH and due to the conversion and is now intubated and on a ventilator. His
of H+ and HCO3 to H2O and CO2. blood gas shows a pH of 7.04, PaCO2 of
39 mmHg, and a HCO3 of 12 mmol/L. His
In children and adults, the normal serum hemoglobin is 10.1 g/dL. The base excess is
bicarbonate value is 24 mmol/L (range 22–29 16.5 mEq/L. How much sodium bicarbonate
mmol/L). In newborns the values are significantly per kilogram would be needed to normalize the
lower due to an expanded extracellular volume, a HCO3?
decreased renal tubule ammonia production,
a lower bicarbonate reabsorption threshold, and Answer The amount of bicarbonate per kilogram
a limited ability to excrete hydrogen ions needed to correct the metabolic acidosis is deter-
[49]. Mean serum bicarbonate values are lower mined by multiplying the base deficit by the bicar-
in preterm infants (18–20 mmol/L) than those bonate volume of distribution (0.4–0.5). The
born at term (20–22 mmol/L). patient would need 6.6–8.3 mEq per kilogram of
sodium bicarbonate to achieve a serum total CO2
of 24 mmol/L.
Base Excess
As with the serum anion gap, serum albumin
Base excess or the base deficit is characterized by concentrations impact base excess calculations
the amount of base that is required to normalize [52]. If the serum albumin is low, the base excess
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 259

Cations Anions

Chloride
Sodium

Bicarbonate
ANION GAP Albumin
Potassium
Other Proteins
Calcium

Magnesium Amino acids

Fig. 6 Serum anion gap. Due to the demands of electro- making the anion gap greater than normal (Andersen
chemical neutrality, the concentration of positive and neg- OS. Blood acid-base alignment nomogram. Scales for
ative ions in serum must be equal. The serum sodium value pH, pCO2 base excess of whole blood of different hemo-
is greater than that of chloride and bicarbonate combined. globin concentrations, plasma bicarbonate, and plasma
The normal ion gap, 12  4 mmol/L, is composed of total-CO2. Scand J Clin Lab Invest 1963;15:211–7.
albumin, phosphates, and amino acids. If the anion gap is Reproduced with permission from Ref. [53])
elevated, bicarbonate has been replaced by other anions,

will be more positive than the reported value, as cations. Under normal conditions, the anion gap
demonstrated in this equation: is predominantly constituted of negative charges
on serum proteins including albumin [54, 55]. The
Baseexcesscorrection ¼0:25 anion gap falls by 2.5 mmol/L for every 1 g/L
 ð4:2  serumalbuming=dLÞ reduction in serum albumin [56]. A low anion gap
can be due to hypoalbuminemia, hypercalcemia,
hyperkalemia, hypermagnesemia, or lithium
Serum Anion Gap ingestion.
The serum anion gap is useful in determining the Some patients have more than one metabolic
source of a metabolic acidosis. Electrochemical derangement affecting the serum HCO3 value.
neutrality (e.g., the sum of all the cations equals The delta anion gap assesses the change in the
the sum of the anions) insures that there are equal anion gap relative to a normal anion gap of
concentrations of positive and negative ions in 12 mmol/L. The delta anion gap is calculated
body fluids (Fig. 6) [53]. The equation for the using the following equation:
serum anion gap is:
Delta anion gap ¼ ðcurrent anion gap  12Þ
þ
Anion gap ¼ Unmeasured cations þ Na
After solving for the delta anion gap, proceed to
¼ Unmeasured anions þ HCO3 and Cl
solve the delta gap which is of use to determine if
the anion gap is sufficient to explain the entire
Rearranged equation looks like this: metabolic acidosis:

Anion gap ¼ Naþ  ðHCO3 and Cl Þ Delta Gap ¼ delta anion gap
¼ Unmeasured anions  Unmeasured cations  delta HCO3 ð24 mmol  current HCO3 Þ

The normal anion gap is 12  4 mmol/L. Patients A significantly positive delta gap (more than
who have high anion gaps may have increased 6 mmol/L positive) is associated with a concurrent
unmeasured anions or decreased unmeasured metabolic alkalosis. Conversely, if significantly
260 P.D. Yorgin et al.

negative delta gap (more than 6 mmol/L nega- (3) serum bicarbonate levels are decreased by
tive), then a hyperchloremic metabolic acidosis dilution with a non-bicarbonate-containing solu-
is also present. tion [58]. The compensatory response to both
Some clinicians have found that a delta anion acute and chronic metabolic acidoses is respira-
gap ratio calculation is useful. tory alkalosis.
Delta anion gap ratio: delta anion gap/delta Any evaluation of any metabolic acidosis
HCO3 (24 mmol/L – current HCO3 mmol/L). should include a determination of the serum
Ratios less than 0.4 are seen with hyperchloremic anion gap because it can be used to determine
normal anion gap acidosis; 0.4–1 are common the cause of the metabolic acidosis [59].
with high anion gap and normal anion gap acido- Most patients with a metabolic acidosis either
sis; 1–2 are seen with a high anion gap acidosis, have an increased or normal anion gap. If, for
and >2 are found when there is a high anion gap example, hydrochloric acid is infused into the
acidosis and a metabolic alkalosis or a preexisting blood, then there is a milliequivalent per
compensated respiratory acidosis. milliequivalent replacement of bicarbonate for
chloride yielding a normal anion gap acidosis.
Example The same is true for intestinal or kidney losses of
A 3-year-old male with cerebral palsy who had an HCO3. However, if HCO3 is replaced by an
exacerbation of his respiratory disease was unmeasured anion, like lactic acid, the anion gap
brought to the emergency department. His blood will increase. A list of conditions which cause an
gas showed a pH of 7.14, PaCO2 of 50 mmHg, increased anion gap is shown in Table 1. Increases
and a HCO3 of 18 mmol/L. His serum sodium in the anion gap induce a respiratory compensa-
was 144 mEq/L, chloride 102 mEq/L, and serum tion; for every 1 mmol/L rise in serum anion gap,
total CO2 16 mmol/L. Calculate the delta the PaCO2 should decrease by 1 mmHg.
anion gap. Most of the acute change in serum bicarbonate
is repaired by intracellular buffering processes
Solution The anion gap is 26 mmol/L and the [60]. Hemoglobin, phosphorous, protein, and the
delta anion gap is 14 mmol/L. The delta gap bone [61, 62] all contribute to buffering H+.
is + 6, consistent with a concurrent metabolic Serum chloride levels rise in response to the
alkalosis. The delta anion gap is 2.33, indicating decrease in HCO3 levels.
the patient has at least two factors influencing Intravenous sodium bicarbonate infusions might
his metabolic acidosis – either a metabolic alka- seem to be appropriate therapy for all forms of
losis or a previously compensated respiratory aci- metabolic acidosis. Sodium bicarbonate treatment
dosis. The respiratory compensation is not is appropriately used in the treatment of a
appropriate given the severity of metabolic hyperchloremic metabolic acidosis caused by con-
acidosis. ditions including renal tubular acidosis, diarrhea,
ureterosigmoidostomy, and amino acid or chole-
styramine infusions, but not in the treatment of
critically ill patients with a high anion gap metabolic
Metabolic and Respiratory acidosis [4, 64, 65]. In addition to sodium bicarbon-
Derangements ate, sodium acetate (in intravenous solutions), oral
sodium citrate, and potassium citrate may be used to
Metabolic Acidosis correct a persistent hyperchloremic acidosis.
Since the injectable solution of sodium bicar-
There are three mechanisms that can cause serum bonate has a sodium content of 1,000 mEq/L,
bicarbonate levels to fall, resulting in metabolic large doses can cause significant hypernatremia
acidosis: (1) acid is added to or increased in body [64]. Intravenous use of diluted sodium bicarbon-
fluids, (2) bicarbonate is lost (through the gastro- ate solution in concentrations of 140 mEq/L
intestinal tract [57] or by the kidneys), or in patients with normal kidney excretory
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 261

Table 1 Causes of an anion gap acidosis THAM ðmL of 0:3 mol=L solutionÞ
Toxins and drugs Unmeasured anion ¼ lean body weight ðkgÞ
Ethanol Lactic acid  base deficit ðmmol=LÞ:
Ethylene glycol Oxalic acid
Isoniazid toxicity Lactic acid Serum potassium values inversely correlate with
Methanol Formic acid serum bicarbonate values. Serum potassium con-
Paraldehyde Acetic acid centrations fall in response to the infusion of
Salicylates Lactic acid bicarbonate due to the movement of potassium
Isopropyl alcohol Oxalic acid into cells in exchange for hydrogen ions. A pH
Lactic acidosis Lactic acid increase of 0.1 leads to a decrease in serum potas-
Uremia Uric, oxalic, succinic, sium of 0.2–0.4 mEq/L. Conversely, for every
pimelic, adipic acid
decrease in blood pH of 0.1, the serum potassium
Amino acidopathies
rises by 0.6 mEq/L.
Maple syrup urine α-Ketoisocaproic,
disease α-keto-β-methylvaleric,
α-ketoisovaleric,
indoleacetic, acetoacetic, Respiratory Acidosis
β-hydroxybutyric acid
Isovaleric acidemia Isovaleric acid
Any process that interferes with ventilation can
Glutaric acidemia Glutaric, lactic, isobutyric,
isovaleric, α-methylbutyric cause respiratory acidosis. The causes of ventila-
acid tory failure include chronic obstructive pulmo-
Propionyl coenzyme Propionic, methylcitric nary disease, drugs, an extreme ventilation
A carboxylase deficiency propionylglycine, perfusion mismatch, an extensive infiltrative pro-
acetoacetic,
cess, exhaustion, neuromuscular disorders, or
β-hydroxypropionate,
β-hydroxybutyric acid excessive CO2 production.
Methylmalonic Methylmalonic acid The compensatory response to respiratory aci-
aciduria dosis is metabolic alkalosis. Hypercapnia stimu-
Defects in lates the secretion of hydrogen ions by the distal
carbohydrate tubule (Fig. 5) which lowers urine pH [66–68].
metabolism
Serum bicarbonate values also increase because
Diabetic ketoacidosis Acetoacetic,
β-hydroxybutyric acid bicarbonate secretion in the distal tubule is
Fructose-1,6- Lactic, pyruvic acid inhibited [69]. The higher bicarbonate concentra-
diphosphatase deficiency tion is maintained by enhanced kidney bicarbonate
Glucose-6- Lactic acid reabsorption in both proximal and distal tubules.
phosphatase deficiency As serum bicarbonate levels increase, the serum
Pyruvate carboxylase Lactic, pyruvic acid chloride levels must decrease. Hypercapnia leads
deficiency
to a decrease in proximal sodium chloride
Succinyl-CoA- Acetoacetic,
transferase deficiency β-hydroxybutyric acid reabsorption and induces chloruresis. The compen-
satory response of metabolic alkalosis, to chronic
hypercarbia, usually takes 1–2 days.
function rarely causes hypernatremia. In situa-
tions where a patient is acidemic but is Example
significantly hypernatremic, tris-hydroxymethyl A 16-year-old male with a pulmonary hemorrhage
aminomethane (THAM) can be used [63]. due to Wegener granulomatosis has a pH of
THAM has been safely used in patients with 7.32, a PaCO2 of 52 mmHg, and a HCO3 of
renal acidosis, salicylate or barbiturate intoxica- 26 mmol/L.
tion, and increased intracranial pressure associ-
ated with cerebral trauma [65]. THAM dosing is Diagnosis The elevated bicarbonate could not
based on the following formula: have caused the acidemia, so the principal cause
262 P.D. Yorgin et al.

of the acidemia is a respiratory acidosis. Yet, the disorders (Tables 2 and 3). These formulas can
renal compensatory process is either incomplete assist with the determination of the primary event
or has not had time to correct the pH to normal. and the subsequent compensatory process. In
most cases, the direction of the pH deviation sug-
If there were no compensatory process the gests the primary process.
recognition of the inciting problem would be
quite easy. Formulas have been derived that
describe the relationship between carbon dioxide Discriminating Between Acute
and bicarbonate in acute and chronic acid–base and Chronic Respiratory Acidosis

Acute respiratory acidosis is typically caused by


Table 2 Rules of acute respiratory compensation an abrupt decline in ventilation, which causes
blood PaCO2 to rise and pH to fall. A child with
Change Rule Example
acute respiratory acidosis frequently is hypoxic
"PaCO2 For every 1 mmHg PaCO2
increase in PaCO2, 40 ! 60 and presents with tachypnea, dyspnea and
the pH will decrease pH hyperpnea.
by 0.008 pH unit 7.40 ! 7.24
Compensation The HCO3 will HCO3 Example
to "PaCO2 increase by 0.1 24 ! 26 An arterial blood gas obtained from a child with
mmol/L for every
1 mmHg increase in known severe asthma, now presenting to the
PaCO2 emergency room, with status asthmaticus reveals
#PaCO2 For every 1 mmHg PaCO2 a low pH (7.16) with a high PaCO2 (74 mmHg)
decrease in PaCO2, 40 ! 20 and a slightly elevated serum bicarbonate
the pH will increase pH (27 mmol/L).
by 0.007 pH unit 7.40 ! 7.54
Compensation The HCO3 will HCO3
to #PaCO2 decrease by 0.25 24 ! 19
Diagnosis These results are consistent with a
mmol/L for every child who has impending respiratory
1 mmHg decrease failure. Most asthmatics have a mild respiratory
in PaCO2 alkalosis at the time of presentation. The elevated

Table 3 Rules of chronic acid–base compensation


Chronic change Rule Example
"PaCO2 For every 1 mmHg increase in PaCO2, the pH decreases by 0.0025 PaCO2 40 ! 60
pH unit pH 7.40 ! 7.35
Compensation for The HCO3 will increase by 0.4 mmol/L for every 1 mmHg increase HCO3 24 ! 28
"PaCO2 in PaCO2
#PaCO2 For every 1 mmHg decrease in PaCO2, the pH increases by 0.003 pH PaCO2 40 ! 20
unit pH 7.40 ! 7.46
Compensation for The HCO3 will decrease by 0.5 mmol/L for every 1 mmHg HCO3 24 ! 14
#PaCO2 decrease in PaCO2
"HCO3 For every 1 mmHg increase in HCO3, the pH increases by HCO3 24 ! 34
0.003–0.008 pH unit pH7.40 ! 7.43–48
Compensation for The PaCO2 will increase by 0.2–0.9 mmHg for every 1 mmHg PaCO2 40 ! 48
"HCO3 increase in HCO3
#HCO3 For every 1 mmHg decrease in HCO3, the pH decreases by 0.012 HCO3 24 ! 14
pH unit pH 7.40 ! 7.28
Compensation for The PaCO2 will decrease by 1.25 mmHg for every 1 mmHg PaCO2 40 ! 28
#HCO3 decrease in HCO3
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 263

bicarbonate is attributable to buffering by volume contraction where chloride is lost dispro-


intracellular buffering mechanisms. The increase portionately to bicarbonate, (2) loss of hydrogen
in serum bicarbonate by 1 mmol/L for ion, or (3) net addition of HCO3 to the extracel-
every 10 mmHg increase in PaCO2 [70] is lular space.
immediate. Chloride loss, without concurrent H+ loss, most
commonly occurs with gastrointestinal disease,
Renal buffering does not noticeably impact the chemical diuretic use [73], cystic fibrosis [74, 75],
pH until 12–24 h after the respiratory acidosis Bartter syndrome [76], or Gitelman syndrome
begins [71]. Treatment of the patient with acute [77, 78]. With intractable chloride depletion, the
respiratory acidosis involves rapid recognition kidney increases the reabsorption of bicarbonate by
and correction of the inciting cause coupled with increasing distal tubule H+ secretion. A metabolic
oxygen administration. Sodium bicarbonate, alkalosis can develop when chloride is lost in
which can cause a rise in PaCO2, should be used excess of sodium from the gastrointestinal tract.
only to preclude the serious cardiovascular effects Patients with acute diarrhea may have chloride
of acidosis. losses that range from 10 to 110 mEq per liter of
The basis for chronic respiratory acidosis is stool [79]. Chemical diuretics, including loop
typically a decrease in alveolar ventilation. (furosemide, bumetanide, ethacrynic acid) and thi-
Plasma CO2 values are elevated; however, unlike azide (including chlorothiazide, hydrochlorothia-
in the patient with acute respiratory acidosis, zide, and metolazone) diuretics, can cause a
effective renal compensation has occurred. There- profound loss of chloride by the kidney.
fore, serum pH values are only slightly below Bicarbonate values rise acutely with the use of
normal. For each increase in the PaCO2 of loop and thiazide diuretics due to at least four or
1 mmHg, the bicarbonate value increases by five mechanisms. First, contraction of the intra-
0.41 mmol/L [72]. vascular volume, without net loss of bicarbonate,
causes an increase in serum bicarbonate levels.
Example Second, increased salt delivery to the distal
A 3-month-old infant with bronchopulmonary tubule stimulates potassium and hydrogen ion secre-
dysplasia on diuretic therapy has the following tion independent of an aldosterone effect [80]; thus
venous blood gas and laboratory results: pH, contributing to higher serum bicarbonate values.
7.38; PaCO2, 68 mmHg; PO2, 53 mmHg; HCO3, Third, extravascular volume contraction causes
38 mmol/L; serum sodium, 136 mEq/L; serum the GFR to decrease. As a result, more sodium and
potassium, 2.9 mEq/L; serum total CO2, water are reabsorbed in the proximal tubule.
39 mmol/L; and serum chloride, 86 mEq/L. Because sodium delivery is decreased to the
collecting duct, the juxtaglomerular apparatus
Solution This blood gas obtained from a child secretes renin into the afferent arteriole. A renin-
with stable bronchopulmonary dysplasia is mediated increase of angiotensin II leads to an
remarkable for the normal to slightly low pH, a increase in aldosterone levels. Aldosterone
markedly elevated PaCO2, and a hypochloremic decreases the H+ concentration in serum by increas-
metabolic alkalosis. This gas shows an effective ing distal tubule H+ secretion via the H+/K+-ATPase
renal compensation for the chronic respiratory luminal pump. As plasma potassium levels begin to
acidosis. decrease due to diuretic mediated potassium deple-
tion, aldosterone levels decrease. This represents an
important feedback mechanism to prevent severe
Metabolic Alkalosis hypokalemia and metabolic alkalosis.
Fourth, profound hypokalemia due to potas-
Metabolic alkalosis, characterized by an increased sium depletion causes K+ to egress from somatic
concentration of serum bicarbonate, can be caused cells. With significant potassium depletion, potas-
by three major mechanisms: (1) intravascular sium moves out of the cell in order to maintain the
264 P.D. Yorgin et al.

ratio of intracellular and extracellular potassium, excrete bicarbonate and decrease urinary acidifi-
and hydrogen ions move into the cell maintain cation in response to an alkali load [81], the alka-
electrochemical neutrality. Since all cells have a losis that develops in response to exogenous alkali
K+–Na+ pump that keeps intracellular levels of is typically mild.
sodium low, sodium cannot move into the cell in There are three mechanisms that act to return the
exchange for the potassium leaving the cell. The pH toward 7.40 when a metabolic alkalosis
loss of H+ from the extracellular space into intra- develops. There are intracellular buffering mecha-
cellular space causes the rise in serum HCO3. nisms by which H+, derived from intracellular pro-
Finally, thiazide diuretic-induced inhibition of teins and phosphate, is released into the
NDBCE in the intercalated cells may also block extracellular space. Lactic acid levels also increase
the loss of HCO3 in the urine, contributing to the and provide additional H+ for buffering [50]. The
metabolic alkalosis. kidney has the capacity to excrete bicarbonate via
Treatment of the hypokalemic, hypochloremic β-intercalated cells in the distal tubule [82]. Com-
metabolic alkalosis caused by diuretics typically pensatory respiratory acidosis is the third process by
consists of potassium chloride or potassium- which the pH is returned toward normal. Decreased
sparing diuretics. Potassium chloride therapy is ventilatory drive, limited by hypoxia, increases
advantageous in that both potassium depletion PaCO2 values to approximately 55 mmHg. With
and chloride deficiency are corrected. Sodium respiratory compensation, the PaCO2 should
chloride therapy can be used to correct the chlo- increase by 0.25–1 times the bicarbonate change.
ride depletion, but its use is controversial. There are factors including volume depletion
and aldosterone and potassium depletion that
Example interfere with renal bicarbonate wasting, thus
A 4-month-old female with bronchopulmonary maintaining the alkalosis [83]. Volume depletion
dysplasia has chronic CO2 retention and is receiv- decreases the amount of glomerular filtrate deliv-
ing furosemide and metolazone therapy. Arterial ered to the proximal tubule; therefore, sodium and
blood gas results show a pH of 7.43, PaCO2 of bicarbonate reabsorptions are increased. Aldoste-
58 mmHg, PO2 of 68 mmHg, and HCO3 of rone increases distal tubule H+ secretion via the
36 mmol/L. Her serum sodium is 137 mEq/L, H+/K+-ATPase luminal pump [84, 85]. Potassium
serum potassium is 2.4 mEq/L, serum chloride is depletion increases the rate of bicarbonate
86 mEq/L, and serum total CO2 is 35 mmol/L. reabsorption [86], thus helping to sustain meta-
How would one describe this acid–base problem? bolic alkalosis.
Occasionally, metabolic alkalosis can occur in
Diagnosis In this particular case, the child is patients who experience a rapid resolution of their
slightly alkalemic due to an excessive compensa- chronic hypercapnia but cannot excrete bicarbon-
tory metabolic alkalosis in the context of chronic ate rapidly enough. Conversion of large amounts
respiratory acidosis. The metabolic alkalosis is of lactic acid due to shock can also cause a meta-
excessive because of the excessive diuretic- bolic acidosis [87].
induced urinary chloride loss. The effective treatment of a metabolic alkalosis
depends upon the recognition of chloride-
Hydrogen ion can be lost through gastrointes- responsive and chloride-resistant processes.
tinal or renal mechanisms. The secretion of Sodium or potassium chloride replacement will
hydrochloric acid in the stomach leaves behind a lead to rapid resolution of the chloride depletion-
cation and bicarbonate in the serum. If the H+ ion induced metabolic alkalosis [88]. Blocking the
is lost from the body (e.g., vomiting), there will be reabsorption of bicarbonate in the urine by inhi-
a net increase in serum bicarbonate. bition of carbonic anhydrase can be very effective.
In the administration of bicarbonate or citrate, The use of even a single dose of acetazolamide
acetate or lactate will cause a rise in serum HCO3 can lead to a rapid improvement of a metabolic
levels. Because the kidney has the capacity to alkalosis [89, 90].
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 265

Individuals who have sodium chloride- Table 4 Simple acid–base disorders


resistant metabolic alkalosis may have Type of disorder pH PaCO2 HCO3
hyperaldosteronism, pseudohyperaldosteronism Metabolic acidosis # # #
(Liddle’s syndrome, type 1 pseudohyperaldos- Metabolic alkalosis " " "
teronism), aldosterone receptor gain of function Acute respiratory acidosis # " "
mutation (type 2 pseudohyperaldosteronism), Chronic respiratory acidosis # " "
apparent mineralocorticoid excess (AME) or per- Acute respiratory alkalosis " # #
haps potassium depletion [91]. Patients with Chronic respiratory " # #
alkalosis
AME experience growth failure, hypertension,
and a chronic hypokalemic metabolic alkalosis From Schrier RW. Renal and Electrolyte Disorders,
ed. 3, Boston, Little Brown and Company, 1986, p. 146;
[92]. Excessive aldosterone activity can be with permission
blocked with spironolactone [78, 93, 94],
triamterene [95], or amiloride [94].
Although not commonly used, the intravenous
infusion of hydrochloric acid has also been used Simple Acid–Base Disorders
to correct acute and severe metabolic alkalosis
[96–98]. Acid–base problems fall into two broad catego-
ries: those with a single primary disorder coupled
with a compensatory response, a simple acid–base
Respiratory Alkalosis disorder, and those in which two or more primary
disorders occur together, a mixed acid–base dis-
Respiratory alkalosis is caused by a process in order. The type of acid–base disorder can be deter-
which the pH rises in response to a decreasing mined by evaluating the pH, PaCO2, and the
PaCO2. The PaCO2 falls when ventilatory losses HCO3. In a simple acid–base problem, the
of carbon dioxide exceed CO2 production. Venti- PaCO2 and the HCO3 always move in the same
lation can be increased due to central or peripheral direction (Table 4). Movement of the PaCO2 and
neural stimulation, mechanical ventilation, or vol- the HCO3 in opposite directions indicates a
untary effort. mixed acid–base disorder. Assessment of the
The compensatory mechanism is a metabolic appropriateness of the compensation using
acidosis. Buffering by H+ release from intracellu- Tables 2 and 3 and Fig. 7 can also be helpful in
lar sources constitutes the first defense against uncovering a mixed acid–base problem.
respiratory alkalosis [99]. Buffering is complete
in minutes and persists for at least 2 h [100]. Example
In response to acute respiratory alkalosis, the A 2-year-old male with hypochloremic metabolic
HCO3 decreases by 1–3 mmol/L for every alkalosis is diagnosed as having Bartter syn-
10 mmHg decrease in PaCO2. drome. His initial blood gas result showed a pH
The kidney compensates in response to respi- of 7.41, PaCO2 54 mmHg and a HCO3 of
ratory alkalosis by reducing the amount of new 33 mmol/L.
HCO3 generated and by excreting HCO3
[101]. The process of renal compensation occurs Diagnosis This is an example of simple
within 24–48 h [102]. The stimulus for the renal acid–base disorder with a primary metabolic alka-
compensatory mechanism is not pH but PaCO2 losis and a compensatory respiratory acidosis.
[103, 104]. In chronic respiratory alkalosis, the
plasma HCO3 is decreased by 2–5 mmol/L for
every 10 mmHg decrease in PaCO2. The only Mixed Acid–Base Disorders
means to treat a respiratory alkalosis is to correct
the underlying disorder responsible for causing A mixed acid–base disorder is a combination of
the disorder. two primary disorders [55]. Recognition of a
266 P.D. Yorgin et al.

Fig. 7 Acid–base Arterial blood [H+] (nmol/L)


nomogram (or map).
Shaded areas represent the 100 90 80 70 60 50 40 30 20
95 % confidence limits of 60
120 110 100 90 80 70 60 50 40
normal respiratory and 56
metabolic compensations
for primary acid–base 52
disturbances. Data falling 35
48
outside the shaded areas METABOLIC
denote a mixed disorder if a 44
Arterial plasma [HCO3−] (mmol/L)
ALKALOSIS
CHRONIC 30
laboratory error is not RESPIRATORY
present (Dubose 40 ACIDOSIS
TD. Disorders of acid–base 25
36
balance. In The Kidney 8th
ed, Brenner B (ed.), 32
Philadelphia, Elsevier ACUTE 20
Saunders 2007;505–546. 28 RESPIRATORY
ACIDOSIS
Reproduced with 24 NORMAL
permission from Ref. [105]) ACUTE
15
20 RESPIRATORY
ALKALOSIS
16 CHRONIC
10
RESPIRATORY
12 ALKALOSIS
METABOLIC
8 ACIDOSIS
Pco2(mm Hg)
4

0
7.0 7.1 7.2 7.3 7.4 7.5 7.6 7.7 7.8
Arterial blood pH

mixed acid–base disorder is dependent on deter- respiratory acidosis. Note that the PaCO2 and
mining whether the compensatory process was HCO3 do not move in the same direction. With
adequate and appropriate. In mixed acid–base the high PaCO2, an increase in the total CO2
disorders, the blood gas result will fall outside of would be the normal compensatory response.
the predictive bands found in an acid–base nomo-
gram (Fig. 7). When there is a significant devia- The anion gap and delta anion gap can also
tion of the pH, one of the two primary disorders point to a mixed acid–base disorder.
blocks the compensation for the other
(Fig. 7) [105]. Example 2
A 13-month-old female presents with a 2-day
Example 1 history of non-bilious vomiting and diarrhea.
A 9-month-old male with renal tubular acidosis The child has vomited 10 times over the last
is unable to take his medication due to the respi- 36–48 h. Initial laboratory tests show a Na of
ratory distress associated with respiratory syncy- 132 mmol/L, chloride of 87 mmol/L, and total
tial virus. His admission arterial blood gas results CO2 of 18 mmol/L.
show a pH of 7.03, PaCO2 of 52 mmHg, PO2 of
67 mmHg, and HCO3 of 16 mmol/L. Diagnosis The anion gap is 27 mmol/L with a
delta gap of +11 mmol/L. This is an example of a
Solution This is an example of a mixed acid–base mixed acid–base disorder because the patient has
disorder with a metabolic acidosis and acute a metabolic acidosis but also has metabolic
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 267

alkalosis due to vomiting with loss of HCl. (1 mmol/L per 10 mmHg rise in PaCO2). More-
The metabolic alkalosis is not obvious due to the over, the blood gas value falls into the chronic
severity of the metabolic acidosis. Without the respiratory acidosis prediction band.
vomiting, the metabolic acidosis would have
been much worse.

Occasionally the amount of compensation is Nutrition and Acid–Base Balance


excessive given the clinical situation. For exam-
ple, children presenting with aspirin intoxication Dietary intake with daily generation of organic
will typically have PaCO2 values lower than acids (lactic acid, pyruvic acid, and acetic acid)
expected based upon the acidosis caused by the and excretion of bicarbonate in the stool result in
aspirin alone [106]. The excessive respiratory net daily acid production of approximately 1 mEq
alkalosis is attributable to the stimulatory effect hydrogen ions per kilogram body weight in adults
aspirin has upon ventilation. [107]. Dietary intake contributes to the majority of
Patients with chronic lung disease typically this daily acid production, with the latter two
have high PaCO2 values and an appropriate processes making a minimal contribution. With
compensatory metabolic alkalosis. The addition the development of agriculture and animal hus-
of diuretics, which are frequently used to bandry, our diets changed from net base or alkali
decrease alveolar interstitial edema, causes producing to net acid producing. This occurred as
serum HCO3 values to increase to levels greater net acid-producing animal foods and cereal grains
than expected on the basis of the respiratory replaced alkali-rich fruits and vegetables.
acidosis alone. The blood gas results are fre- The typical modern Western diet produces a net
quently consistent with a simple metabolic alka- acid load or hydrogen ion in an adult of approxi-
losis. A patient’s history may be the only means mately 50–100 mEq per day [107, 108]. Vegetarian
by which the mixed acid–base disorder may be diets that consist of vegetables, fruits, and nuts gen-
suspected. erate net base, whereas nonvegetarian diets includ-
ing meat, seafood, and dairy products generate net
Example acid [109]. This load is markedly increased in
A 15-month-old female with bronchopulmonary infants who are solely fed with cow milk-based
dysplasia receives furosemide at a dose of infant formula, which produces a net acid load of
1 mg/kg/day. Her typical blood gases on 1/2 l of approximately 2 mEq/kg/day compared to about 0.8
nasal O2 are as follows: pH, 7.49; PaCO2, mEq/kg/day when fed with human milk [110]. The
50 mmHg; PaO2, 87 mmHg; and HCO3, dietary net acid load in children in the West is
40 mmol/L. roughly 15–80 mEq/day or roughly 1–3 mEq/kg/
day, data obtained from population-based studies,
Diagnosis Mixed acid–base disorder, chronic being higher in adolescents than younger children
respiratory acidosis, and compensatory metabolic and in males compared to females [111–113].
alkalosis + metabolic alkalosis due to a severe Metabolism of sulfur-containing amino acids
contraction alkalosis. The child develops a viral cysteine and methionine, cationic amino acids
pneumonia. The arterial blood gas is as follows: arginine and lysine, and phosphorus produces
pH, 7.35; PaCO2, 80 mmHg; PaO2, 62 mmHg; acid. These substances are found in high concen-
and HCO3, 41 mmol/L. If one were not familiar trations in animal proteins, cereals, nuts, and dairy
with the case, this would appear to be an acute products. Conversely, metabolism of anionic
respiratory acidosis with an incomplete compen- amino acids, aspartic acid, and glutamic acid,
satory metabolic alkalosis; however, with an found in wheat, soy protein, cow’s milk, vegeta-
increase of the PaCO2 value by 40 mmHg bles, and potassium- and magnesium-containing
(above the normal 40 mmHg), one would antici- foods, like fruits, vegetables, nuts, and dairy prod-
pate an acute HCO3 compensation of 4 mmol/L ucts, consumes hydrogen ion [113].
268 P.D. Yorgin et al.

Physiologically, two types of acid are impor- 24 mEq/L. The imbalance between increased acid
tant: carbonic acid, which is a volatile acid and load and decreased ability for net acid excretion
produced by the metabolism of dietary carbohy- results in increased susceptibility of neonates and
drates and fats, and non-carbonic acids, produced infants to the development of metabolic acidosis.
from the metabolism of proteins. Metabolism The acid load from diet and metabolism in infants
of dietary carbohydrates and fats results in gener- is approximately one hundred percent higher than
ation of carbon dioxide that can be excreted by that of adults, adjusted for body weight. The
alveolar ventilation. Approximately 15,000 mmol increased acid load is exaggerated in infants fed
of volatile acid is produced per day [112]. On the with cow’s milk formula. In small preterm babies,
other hand, renal urinary excretion eliminates renal acid excretion capacity is low compared to
excess non-carbonic or nonvolatile acids, mainly term newborns. In both preterm and term infants,
sulfuric acid (H2SO4) that is generated endoge- maximum renal net acid excretion improves rap-
nously or from dietary intake of sulfur-containing idly during the first few weeks of life (Fig. 8)
amino acids. [110]. Insipient late metabolic acidosis is common
The nonvolatile acid produced from metabo- in premature infants in the first year of life, and its
lism and dietary intake is balanced with renal net pathophysiology is multifactorial (Fig. 9) [110].
acid excretion, thus maintaining acid–base equi- The growing bones of neonates and children
librium. The normal healthy kidney easily handles results in production of additional acid as a result
dietary net acid, but in the setting of reduced renal of hydroxyapatite production for bone minerali-
function, excess dietary load provides a challenge zation. Approximately 0.92 mmol of protons is
for excretion. In fact, there is evidence that even released into the extracellular circulation for every
with normal aging, gradual reduction in glomeru- 1 mmol of calcium incorporated into the skeleton
lar filtration rate (GFR) results in metabolic aci- [116]. Broadly speaking, renal regulation of
dosis on a normal diet [114]. hydrogen ion balance involves (a) glomerular fil-
Goodman, Lemann, and Lennon demonstrated tration; (b) reclamation of filtered bicarbonate,
that despite minor daily fluctuations, the cumula- thus repleting body stores; and (c) excretion of
tive acid balance in normal individuals is approx- hydrogen ion as titratable acid or ammonium,
imately zero [115]. Typical acid production in which excretes the nonvolatile acid from metabo-
adults, estimated from extrapolation of urinary lism, dietary intake, and, in children, bone growth.
sulfate and organic anion, is equal to urinary Net urinary acid excretion consists of the sum of
acid output. With chronic renal disease (CKD), titratable acid (hydrogen ion buffered by phos-
serum CO2 content reaches the acidotic level over phate and sulfate) plus ammonium.
a period of 6 days after withdrawal of bicarbonate Since the beginning of the twentieth century, it
therapy. Net acid production exceeds was observed that urine pH is higher in preterm
compromised or reduced net acid excretion, and term neonates and gradually decreases after
resulting in a positive balance of 21–30 mEq of several weeks. When preterm and term infants fed
acid per day [115]. Withdrawing the alkaline ther- human milk or formula were challenged with
apy of a patient with CKD may lead to metabolic acute or chronic acid loading using ammonium
acidosis resulting primarily from hydrogen ion chloride or calcium chloride, to determine maxi-
retention. mal net renal acid excretion, the results revealed
lower net acid excretion in preterm compared to
term infants. Furthermore, there was an increase
Acid–Base Balance in Neonates, in maximum net renal acid excretion during the
Infants, and Children first few weeks in term and preterm infants
(Fig. 8).
Newborns and infants have a lower serum bicar- Animal studies point to decreased activity and
bonate of about 20 mEq/L compared to older immaturity of almost every transporter involved
children and adults, whose serum bicarbonate is in bicarbonate reabsorption and acid secretion as
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 269

Fig. 8 Maximum renal Maximum renal net


excretion of net acid and acid excretion
ammonium in preterm and 9.0
term infants on nutrition
with human milk or (mmol/kg/d) Cow’s milk / Formula NAE
formulas (Kalhoff H, Manz 7.5 NH4
F. Nutrition, acid-base
Human milk NAE
status and growth in early
NH4
childhood. Eur J Nutr 6.0
2001;40:221–30.
Reproduced with
permission from Ref. [110]) 4.5

3.0

1.5

0
30 35 40 45 50 65 70
Mean developmental age (weeks)

Nutrition

“Inner milieu”
of the body

Ventilation

Growth
Incipient late Weight gain
metabolic
acidosis (ILMA)
Na-retention
Aldosterone Ca,P-retention
Renal acid excretion N-assimilation
capacity Arg. -vasopressin

Genetic
features

Urinary excretion

Fig. 9 Pathophysiological mechanisms in premature in early childhood. Eur J Nutr 2001;40:221–30.


infants with incipient late metabolic acidosis (ILMA) Reproduced with permission from Ref. [110])
(Kalhoff H, Manz F. Nutrition, acid-base status and growth

causes for the reduced ability of the neonatal potential mechanisms. Less bicarbonate reclama-
kidney to excrete acid. While it may not be possi- tion occurs in the proximal convoluted tubule of
ble to study these mechanisms in humans, animal infants compared to adults. Whereas in adults,
studies have provided substantial insights into approximately 85 % of filtered bicarbonate is
270 P.D. Yorgin et al.

Fig. 10 The apical protein expression of the Na+/H+ and glucocorticoids that occur during that time
exchanger during postnatal development. As shown, (Twombley K, Gattineni J, Bobulescu IA, Dw
NHE8 is highly expressed in the neonate, while NHE3 is arakanath V, Baum M. Effect of metabolic acidosis on
the predominant Na+/H+ exchanger in the adult proximal neonatal proximal tubule acidification. Am J Physiol
tubule. The isoform change occurs at the time of weaning Regul Integr Comp Physiol 2010;299:R1360-R68.
and is likely mediated by the increase in thyroid hormone Reproduced with permission from Ref. [118])

reabsorbed proximally, only 65 % of the bicar- but barely detectable in adults, as shown in Fig. 10
bonate is reabsorbed proximally in infants [110]. [118]. NHE8 was found to have significantly dif-
The Na+/H+ exchanger (NHE) is directly or ferent properties than NHE3 in terms of its regu-
indirectly responsible for most sodium bicarbon- lation and its sensitivity to amiloride analogues,
ate and NaCl reabsorption by the adult proximal which inhibit NHEs [119]. Metabolic acidosis in
tubule [117]. There are 10 NHE isoforms, but neonates in vivo and in cells expressing NHE8
NHE3 is the predominant isoform on the apical in vitro increase NHE8 activity consistent with
membrane of the adult proximal tubule [12]. The this isoform playing an important role in neonatal
abundance of NHE3 is far less on the brush border proximal tubule acidification.
of the neonate compared to the adult proximal Animal studies show that the lower neonatal
tubule [118]. However, unlike the adult proximal bicarbonate transport is due to lower activities of
tubule where all of NHE activity could be attrib- all the transporters involved in proximal bicarbon-
uted to NHE3, there is a discordance between ate reabsorption, that is, the apical Na+/H+
NHE activity and NHE3 mRNA and protein antiporter, apical H+-ATPase, the basolateral
abundance in the neonate. There is far greater Na+/3HCO3 transporter, and the basolateral
NHE activity than could be explained by NHE3 Na+/K+-ATPase [19, 20, 120, 121]. The activity
alone in the neonatal proximal tubule [22]. NHE8 of all these transporters reached adult levels by the
was found to be this developmental NHE, which age of 6–7 weeks in these studies. One of the
is highly expressed in neonatal proximal tubules major hormones that stimulate the development
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 271

of bicarbonate transport is glucocorticoid. Neo- at higher risk for developing metabolic acidosis.
nates are relatively glucocorticoid deficient during This limited capacity for acid excretion, in con-
the first weeks of life. There is a developmental junction with increased acid load from the diet and
increase in glucocorticoids which precedes the bone formation in growing children, is an impor-
increase in bicarbonate transport. Indeed, if glu- tant factor in acid–base homeostasis.
cocorticoids are administered to pregnant animals
prior to giving birth, the neonates have proximal
tubular bicarbonate transport rates which are com- Mechanisms of Growth Retardation in
parable to those in adults [20]. Thyroid hormone Chronic Acidosis
also affects the development of bicarbonate trans-
port but plays a much less important role. Poor growth occurs in many chronic disorders
The limited ability for urinary acidification such as chronic kidney disease (CKD), cystic
may in part be due to carbonic anhydrase II defi- fibrosis, rheumatologic conditions, and inflamma-
ciency in immature renal tubules. This isoform of tory bowel disease. Growth retardation is a fre-
carbonic anhydrase is found in the cytoplasm of quent, long-recognized complication of chronic
proximal and distal tubular cells. Karashima metabolic acidosis [128].
et al. found that carbonic anhydrase II levels Studies in acidotic children with classic renal
increased from 14 % of adult levels in 1-week- tubular acidosis have shown a blunted release of
old rats to 40 % at age 3 weeks and 97 % at age growth hormone in response to provocative
7 weeks [122]. Carbonic anhydrase IV is the iso- stimuli. The growth retardation associated with
form that is membrane bound on the luminal acidosis can be reversed by systemic bicarbonate
surface of tubular cells. There is decreased activity therapy [129]. The release of growth hormone
of carbonic anhydrase IV in neonatal rabbit kid- under acidotic conditions has been studied [130].
neys compared with adults [123]. Exploration in rats with normal anion gap acidosis
However, human studies provided contradic- from ingestion of ammonium chloride demon-
tory data. Studies of kidney tissue from human strated aberrant growth hormone (GH) secretion.
embryos showed positive staining for carbonic Significant inhibition of pulsatile GH secretion
anhydrase in proximal and distal tubules from was present in the acidotic rats such that both the
12 to 15 weeks gestation. The catalytic activity amplitude of the GH secretory pulse and the area
and the amount of carbonic anhydrase increased under the curve were significantly smaller than in
as gestational age increased from 19 to 26 weeks, controls. These reductions in pulse amplitude and
being comparable to those of adult renal cortex at area were correlated to decreased growth (weight)
26 weeks [124]. Thus, in humans, carbonic in the acidotic rats [130]. Evaluation of tibial
anhydrase may not play a major role in the matu- epiphyseal growth plate insulin growth factor-I
rational changes associated with renal acid–base (IGF-I) gene expression in acidotic and control
homeostasis. rats revealed that IGF-I messenger RNA abun-
α-Intercalated and β-intercalated cells in the dance was lower in the acidotic growth plates.
collecting duct are responsible for distal urinary IGF-I peptide was predominantly localized to the
acidification. There are fewer intercalated cells in hypertrophic zone of chondrocytes and was
the rabbit neonatal kidney and the activity of the weakly detectable in the proliferative zone in
apical H+-ATPase is reduced [125]. Additionally, both the acidotic and control rats’ growth plates.
the capacity for ammonium excretion is reduced Anthropomorphic measurements demonstrated
in newborn animals [126, 127]. Thus, the ability that acidotic rats grew less than did control rats
for distal urinary acidification by excreting hydro- in both length and weight, and these physical
gen ions is limited in newborns. measurements were reflected in the size of the
Therefore, maturational deficiencies in almost tibial epiphyseal growth plates being significantly
every transporter involved in acid–base homeo- smaller in the acidotic rats compared with the
stasis throughout the renal tubule put the neonate control group [131]. Taken together, these
272 P.D. Yorgin et al.

observations suggest that metabolic acidosis evidenced in the study by Goodman et al., in which
reduces IGF-I message abundance and induces few adult patients with RTA had normal
resistance to IGF-I peptide action within the tibial GFR [115].
epiphyseal growth plate. The use of GH to stimu- Patients with CKD with concomitant meta-
late growth in normal anion gap acidosis has been bolic acidosis and hyperparathyroidism are at an
ineffective experimentally, despite enhancement increased risk for skeletal demineralization.
of IGF-I- and IGF-binding protein immunoreac- Krieger et al. studied the effect of acidosis with
tivity within the stem cell chondrocyte zone of the and without parathyroid hormone on calvariae
tibial epiphyseal growth plate [131]. [133]. They found that acidosis and parathyroid
hormone (PTH) independently stimulated cal-
cium efflux from the bone, inhibited osteoblastic
Effect of Acidosis on Bone and Calcium collagen synthesis, and stimulated osteoclastic
Homeostasis beta-glucuronidase secretion. Furthermore, the
effects of acidosis and PTH on bone resorption
The renal response to acidosis, which consists of were additive.
H+ excretion, is relatively slow and takes days.
The initial response to acidosis is buffering by
extracellular bicarbonate and by cellular and Acid–Base Balance in CKD
bone buffers. This role of the skeleton as buffer
occurs at the expense of bone mineral content CKD is characterized by a progressive deteriora-
resulting in loss of calcium and phosphate tion of kidney function eventually leading to
[132]. Bone sodium and potassium are lost in end-stage renal disease necessitating either dialy-
exchange for hydrogen ion, and carbonate con- sis or renal transplantation. As renal function dete-
sumed as a buffer. Positive acid and negative riorates due to a progressive loss in nephron mass,
calcium balance (due to hypercalciuria) occurs in the ability of the kidney to excrete the daily acid
healthy individuals made chronically acidotic by load produced as either hydrogen ions or ammo-
ammonium chloride loading [133, 134]. A similar nium deteriorates. Endogenous net acid produc-
profile is found in patients with renal tubular aci- tion in infants and children is variable and has
dosis. Correction of the metabolic acidosis with been estimated to be between 1 and –3 mEq/kg/
bicarbonate therapy results in return of acid bal- day [141]. In adolescents, endogenous net acid
ance to zero and less negative calcium balance production is similar to that of adults, 1 mEq/kg/
[135]. Furthermore, bicarbonate administration day [142].
corrects bone mineral loss in patients with acido- Metabolic acidosis is a common complication
sis [136, 137]. of CKD. In the initial stages of CKD, the inability
Acutely, bone resorption is primarily due to to excrete hydrogen ions is compensated by an
physicochemical mineral dissolution, while cell- increase in ammonium excretion, and balance is
mediated mechanisms predominate after 24 h maintained [143]. However, as nephrons are lost
[132]. Chronic metabolic acidosis stimulates cal- and GFR falls, ammonium excretion cannot keep
cium efflux from the bone due to increased osteo- up and acidosis ensues [143–149]. Metabolic aci-
clastic bone resorption and decreased osteoblastic dosis usually appears when GFR falls below
activity. The negative calcium balance observed in 25 mL/min/1.73 m2 [144, 146, 150]. However, it
acidotic patients is due to calcium mobilization can appear earlier in the course of CKD, particu-
from the bone, resultant hypercalciuria [138, larly if additional defects in tubular acid secretion
139], and lack of concomitant increase in intestinal are present or damage to the collecting duct is
calcium absorption [136, 140]. There is net loss of present [149, 151].
body calcium. The hypercalciuria predisposes to Even in the late stages of CKD, the severity of
osteoporosis, nephrocalcinosis and nephrolithiasis. the acidosis is usually mild to moderate (serum
The latter renal effects result in renal impairment as bicarbonate 12–23 mEq/L) [144, 152, 153].
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 273

The reasons for the variability in the onset and result in chronic inflammation in the kidney and
severity of metabolic acidosis are not well under- progressive renal fibrosis [163].
stood but may reflect differences in tubular func- Treatment of children with acidosis and CKD is
tion. An additional contributing factor may be primarily with bicarbonate therapy. KDIGO guide-
differences in dietary animal protein intake, a lines recommend alkali therapy to be used to main-
major contributor to the endogenous net acid pro- tain serum bicarbonate levels within the normal
duction. Controlled studies have suggested that the range [164]. Sodium bicarbonate or sodium citrate
loss of the buffering ability of the bone may be an is typically used at doses of 0.5–1 mEq/kg/day.
important factor leading to acidosis [115, 140, Citrate is rapidly metabolized to bicarbonate in
154]. Uribarri et al [155] have looked at adults on the liver and thus should be avoided in patients
dialysis and not found that to be the case. Thus the with liver failure. In patients on maintenance dial-
role of the bone in playing a major role in acidosis ysis, the option to increase the bicarbonate concen-
in ESRD patients remains in dispute. tration in the dialysate is an option. Correction of
The anion gap in CKD patients with the meta- the acidosis in children with CKD can often result
bolic acidosis can be elevated or normal in improved appetite and growth.
(normochloremic versus hyperchloremic) Successful renal transplantation has resulted in
[143]. It had been suggested that a normal anion reduced mortality and improvement in quality of
gap pattern characterizes early CKD, but as GFR life for patients with ESRD on dialysis. Although
declines anion gap increases [149]. However, sev- renal transplantation may restore GFR, patients
eral studies have shown that a normal anion gap may demonstrate overt or subclinical metabolic
pattern can occur at any stage of CKD [149, acidosis. Several mechanisms may account for
153]. Patients with significant tubular damage metabolic acidosis after kidney transplantation
and hyporeninemic hypoaldosteronism are more [165]. Reduced nephron mass being the most obvi-
likely to have normal anion gap levels [156]. The ous. Patients have low serum bicarbonate levels
accumulation of anions such as phosphate, sul- and impaired renal acid excretion. In a study of
fate, urate, and hippurate is usually exacerbated over 800 adults posttransplant, approximately
by a diet high in animal proteins and reduced in 60 % of patients had low serum bicarbonate levels
diets rich in fruit (high in citrate) [157, 158]. [166]. Serum bicarbonate levels strongly correlated
Despite its lack of severity, persistent acidosis with GFR. However, low bicarbonate levels were
can have an adverse impact on cellular function also observed in patients where GFR was near
and contribute to increased morbidity and mortal- normal. The authors found an association between
ity in this patient population [143, 159, 160]. serum bicarbonate levels with higher serum phos-
Symptoms resulting from metabolic acidosis are phate and PTH concentrations and lower serum
more prominent when GFR drops below 25 % of calcium levels. They postulate that the low bicar-
normal [161]. These patients are in negative nitro- bonate levels induce calcium excretion, reduce
gen balance and experience (a) increased catabo- vitamin D synthesis, and stimulation of PTH
lism of muscle protein resulting in muscle release. The relationship between posttransplant
wasting; (b) metabolic bone disease with acidosis and disturbances in mineral metabolism
increased bone resorption, osteopenia, and wors- needs further investigation. Other known causes of
ening hyperparathyroidism; (c) depletion of the metabolic acidosis posttransplant include the
body’s buffering systems; (d) impaired myocar- development of renal tubular acidosis most often
dial function; (e) endocrine abnormalities such as caused by calcineurin inhibitor use (cyclosporine
resistance to growth hormone and impaired glu- and tacrolimus). It is postulated that calcineurin
coneogenesis; (f) hypertriglyceridemia; and inhibitors impair regulation of tubular transport
(g) systemic inflammation [162]. Metabolic aci- proteins involved in renal acid handling on a
dosis is postulated to activate the complement molecular level. It is also speculated that a defect
system and the renin–angiotensin system and in proximal tubule ammonia synthesis due to insu-
increase production of endothelin-1, all of which lin resistance is at play.
274 P.D. Yorgin et al.

References 18. Shah M, Quigley R, Baum M. Neonatal rabbit prox-


imal tubule basolateral membrane Na+/H+ antiporter
1. Mahutte CK. On-line arterial blood gas analysis and Cl-/base exchange. Am J Physiol. 1999;276:
with optodes: current status. Clin Biochem. 1998;31: R1792–7.
119–30. 19. Baum M. Developmental changes in rabbit
2. J-L H. High performance GdTixOy electrolyte- juxtamedullary proximal convoluted tubule acidifica-
insulator-semiconductor pH sensor and biosensor. tion. Pediatr Res. 1992;31:411–4.
Intl J Electrochem Sci. 2013;8:606–20. 20. Baum M, Quigley R. Maturation of proximal tubular
3. Story DA. Bench-to-bedside review: a brief history of acidification. Pediatr Nephrol. 1993;7:785–91.
clinical acid-base. Crit Care. 2004;8:253–8. 21. Schwartz GJ, Evan AP. Development of solute trans-
4. Weiner DI, Verlander JW. Renal acidification port in rabbit proximal tubule. I. HCO-3 and glucose
mechanisms. In: Brenner & Rector’s the kidney, 9th absorption. Am J Physiol. 1983;245:F382–90.
ed. Philadelphia: Elsevier/Saunders, Ch 9 2012. 22. Shah M, Gupta N, Dwarakanath V, Moe OW, Baum M.
p. 293–325. Ontogeny of Na+/H+ antiporter activity in rat proximal
5. Levraut J, Giunti C, Ciebiera JP, et al. Initial effect of convoluted tubules. Pediatr Res. 2000;48:206–10.
sodium bicarbonate on intracellular pH depends on 23. Fukuda Y, Aperia A. Differentiation of Na + -K+
the extracellular nonbicarbonate buffering capacity. pump in rat proximal tubule is modulated by
Crit Care Med. 2001;29:1033–9. Na + -H+ exchanger. Am J Physiol. 1988;255:
6. Burton RF. The roles of intracellular buffers and bone F552–7.
mineral in the regulation of acid-base balance in 24. Larsson SH, Rane S, Fukuda Y, Aperia A, Lechene
mammals. Comp Biochem Physiol Comp Physiol. C. Changes in Na influx precede post-natal increase in
1992;102:425–32. Na, K-ATPase activity in rat renal proximal tubular
7. Rector Jr FC, Carter NW, Seldin DW. The mechanism cells. Acta Physiol Scand. 1990;138:99–100.
of bicarbonate reabsorption in the proximal and 25. Wong PS, Johns EJ. The action of angiotensin II on
distal tubules of the kidney. J Clin Invest. the intracellular sodium content of suspensions of rat
1965;44:278–90. proximal tubules. J Physiol. 1996;497(Pt 1):219–27.
8. Bobulescu IA, Moe OW. Na+/H+ exchangers in renal 26. Purkerson JM, Schwartz GJ. The role of carbonic
regulation of acid-base balance. Semin Nephrol. anhydrases in renal physiology. Kidney Int.
2006;26:334–44. 2007;71:103–15.
9. Ledoussal C, Lorenz JN, Nieman ML, Soleimani M, 27. Pushkin A, Abuladze N, Gross E, et al. Molecular
Schultheis PJ, Shull GE. Renal salt wasting in mice mechanism of kNBC1-carbonic anhydrase II interac-
lacking NHE3 Na+/H+ exchanger but not in mice tion in proximal tubule cells. J Physiol.
lacking NHE2. Am J Physiol. 2001;281:F718–27. 2004;559:55–65.
10. Lorenz JN, Schultheis PJ, Traynor T, Shull GE, 28. Gross E, Kurtz I. Structural determinants and signif-
Schnermann J. Micropuncture analysis of single- icance of regulation of electrogenic Na(+)-HCO(3)(-)
nephron function in NHE3-deficient mice. Am J cotransporter stoichiometry. Am J Physiol. 2002;283:
Physiol. 1999;277:F447–53. F876–87.
11. Nakamura S, Amlal H, Schultheis PJ, Galla JH, Shull 29. Soleimani M, Grassi SM, Aronson PS. Stoichiometry
GE, Soleimani M. HCO-3 reabsorption in renal of Na + -HCO-3 cotransport in basolateral membrane
collecting duct of NHE-3-deficient mouse: a compen- vesicles isolated from rabbit renal cortex. J Clin
satory response. Am J Physiol. 1999;276:F914–21. Invest. 1987;79:1276–80.
12. Schultheis PJ, Clarke LL, Meneton P, et al. Renal and 30. Kondo Y, Fromter E. Axial heterogeneity of sodium-
intestinal absorptive defects in mice lacking the bicarbonate cotransport in proximal straight tubule of
NHE3 Na+/H+ exchanger. Nat Genet. rabbit kidney. Pflugers Arch. 1987;410:481–6.
1998;19:282–5. 31. Kondo Y, Fromter E. Evidence of chloride/bicarbon-
13. Kinne-Saffran E, Beauwens R, Kinne R. An ate exchange mediating bicarbonate efflux from S3
ATP-driven proton pump in brush-border membranes segments of rabbit renal proximal tubule. Pflugers
from rat renal cortex. J Membr Biol. 1982;64:67–76. Arch. 1990;415:726–33.
14. Wang T, Yang CL, Abbiati T, et al. Mechanism of 32. Seki G, Fromter E. The chloride/base exchanger in the
proximal tubule bicarbonate absorption in NHE3 null basolateral cell membrane of rabbit renal proximal
mice. Am J Physiol. 1999;277:F298–302. tubule S3 segment requires bicarbonate to operate.
15. Gluck SL, Lee BS, Wang SP, Underhill D, Nemoto J, Pflugers Arch. 1990;417:37–41.
Holliday LS. Plasma membrane V-ATPases in proton- 33. Steinmetz PR. Cellular organization of urinary acidi-
transporting cells of the mammalian kidney and fication. Am J Physiol. 1986;251:F173–87.
osteoclast. Acta Physiol Scand. 1998;643:203–12. 34. Breton S, Brown D. New insights into the regulation
16. Nakhoul NL, Hamm LL. Vacuolar H(+)-ATPase in of V-ATPase-dependent proton secretion. Am J
the kidney. J Nephrol. 2002;15 Suppl 5:S22–31. Physiol. 2007;292:F1–10.
17. Stone DK, Crider BP, Xie XS. Structural properties of 35. Wang T, Hropot M, Aronson PS, Giebisch G. Role of
vacuolar proton pumps. Kidney Int. 1990;38:649–53. NHE isoforms in mediating bicarbonate reabsorption
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 275

along the nephron. Am J Physiol. 2001;281: 51. Gunn VL, Barone MA. Johns hopkins hospital. Chil-
F1117–22. dren’s medical and surgical center. The Harriet Lane
36. Wang T, Malnic G, Giebisch G, Chan YL. Renal handbook : a manual for pediatric house officers. 16th
bicarbonate reabsorption in the rat. IV. Bicarbonate ed. Philadelphia: Mosby; 2002.
transport mechanisms in the early and late distal 52. Story DA, Morimatsu H, Bellomo R. Strong ions,
tubule. J Clin Invest. 1993;91:2776–84. weak acids and base excess: a simplified Fencl-
37. Stuart-Tilley AK, Shmukler BE, Brown D, Alper Stewart approach to clinical acid-base disorders. Br
SL. Immunolocalization and tissue-specific splicing J Anaesth. 2004;92:54–60.
of AE2 anion exchanger in mouse kidney. J Am Soc 53. Andersen OS. Blood acid-base alignment nomogram.
Nephrol. 1998;9:946–59. Scales for pH, pCO2 base excess of whole blood of
38. Alper SL, Stuart-Tilley AK, Biemesderfer D, different hemoglobin concentrations, plasma bicar-
Shmukler BE, Brown D. Immunolocalization of bonate, and plasma total-CO2. Scand J Clin Lab
AE2 anion exchanger in rat kidney. Am J Physiol. Invest. 1963;15:211–7.
1997;273:F601–14. 54. Emmett M, Narins RG. Clinical use of the anion gap.
39. Ko SB, Luo X, Hager H, et al. AE4 is a DIDS- Medicine (Baltimore). 1977;56:38–54.
sensitive Cl(-)/HCO(-)(3) exchanger in the 55. Kraut JA, Madias NE. Approach to patients with acid-
basolateral membrane of the renal CCD and the base disorders. Respir Care. 2001;46:392–403.
SMG duct. Am J Physiol Cell Physiol. 2002;283: 56. Figge J, Jabor A, Kazda A, Fencl V. Anion gap
C1206–18. and hypoalbuminemia. Crit Care Med. 1998;26:
40. Xu J, Worrell RT, Li HC, et al. Chloride/ 1807–10.
bicarbonate exchanger SLC26A7 is localized in 57. Weizman Z, Houri S, Ben-Ezer GD. Type of acidosis
endosomes in medullary collecting duct cells and is and clinical outcome in infantile gastroenteritis.
targeted to the basolateral membrane in hypertonicity J Pediatr Gastroenterol Nutr. 1992;14:187–91.
and potassium depletion. J Am Soc Nephrol. 58. Garella S, Chang BS, Kahn SI. Dilution acidosis and
2006;17:956–67. contraction alkalosis: review of a concept. Kidney Int.
41. Petrovic S, Barone S, Xu J, et al. SLC26A7: a 1975;8:279–83.
basolateral Cl-/HCO3- exchanger specific to interca- 59. Moe OW, Fuster D. Clinical acid-base pathophysiol-
lated cells of the outer medullary collecting duct. Am ogy: disorders of plasma anion gap. Best Pract Res
J Physiol. 2004;286:F161–9. Clin Endocrinol Metab. 2003;17:559–74.
42. McKinney TD, Burg MB. Bicarbonate absorption by 60. Schwartz WB, Orning KJ, Porter R. The internal
rabbit cortical collecting tubules in vitro. Am J distribution of hydrogen ions with varying degrees
Physiol. 1978;234:F141–5. of metabolic acidosis. J Clin Invest. 1957;36:373–82.
43. Good DW, Burg MB. Ammonia production by indi- 61. Burnell JM. Changes in bone sodium and carbonate in
vidual segments of the rat nephron. J Clin Invest. metabolic acidosis and alkalosis in the dog. J Clin
1984;73:602–10. Invest. 1971;50:327–31.
44. Curthoys NP, Lowry OH. The distribution of gluta- 62. Kurtzman NA. Relationship of extracellular volume
minase isoenzymes in the various structures of the and CO2 tension to renal bicarbonate reabsorption.
nephron in normal, acidotic, and alkalotic rat kidney. Am J Physiol. 1970;219:1299–304.
J Biol Chem. 1973;248:162–8. 63. Hoste EA, Colpaert K, Vanholder RC, et al. Sodium
45. Tannen RL, Sahai A. Biochemical pathways and bicarbonate versus THAM in ICU patients with mild
modulators of renal ammoniagenesis. Miner Electro- metabolic acidosis. J Nephrol. 2005;18:303–7.
lyte Metab. 1990;16:249–58. 64. White BC, Tintinalli JE. Effects of sodium bicarbon-
46. Hamm LL, Simon EE. Ammonia transport in the ate administration during cardiopulmonary resuscita-
proximal tubule in vivo. Am J Kidney Dis. tion. J Am Collage Emerg Phys. 1977;6:187–90.
1989;14:253–7. 65. Nahas GG, Sutin KM, Fermon C, et al. Guidelines for
47. Flessner MF, Mejia R, Knepper MA. Ammonium and the treatment of acidaemia with THAM. Drugs.
bicarbonate transport in isolated perfused rodent 1998;55:191–224.
long-loop thin descending limbs. Am J Physiol. 66. Jacobson HR. Medullary collecting duct acidification.
1993;264:F388–96. Effects of potassium, HCO3 concentration, and
48. Strnad Z. Numerical calculation of basic indicators of pCO2. J Clin Invest. 1984;74:2107–14.
blood acid-base balance using an equilibration 67. Schwartz GJ. Na+-dependent H+ efflux from proxi-
method. Vet Med. 1986;31:557–64. mal tubule: evidence for reversible Na+-H+
49. Edelmann CM, Soriano JR, Boichis H, Gruskin AB, exchange. Am J Physiol. 1981;241:F380–5.
Acosta MI. Renal bicarbonate reabsorption and 68. Madias NE, Adrogue HJ. Cross-talk between two
hydrogen ion excretion in normal infants. J Clin organs: how the kidney responds to disruption of
Invest. 1967;46:1309–17. acid-base balance by the lung. Nephron Physiol.
50. Swan RC, Axelrod DR, Seip M, Pitts RF. Distribution 2003;93:p61–6.
of sodium bicarbonate infused into nephrectomized 69. Schwartz GJ, Al-Awqati Q. Carbon dioxide causes
dogs. J Clin Invest. 1955;34:1795–801. exocytosis of vesicles containing H+ pumps in
276 P.D. Yorgin et al.

isolated perfused proximal and collecting tubules. J 87. Korosi A, Kahn T, Kalb T, Uribarri J. Marked
Clin Invest. 1985;75:1638–44. hyperlactatemia associated with severe alkalemia in
70. Brackett Jr NC, Cohen JJ, Schwartz WB. Carbon a patient with thrombotic thrombocytopenic purpura.
dioxide titration curve of normal man. Effect of Am J Kidney Dis. 2000;36:E6.
increasing degrees of acute hypercapnia on acid- 88. Jacobson HR, Seldin DW. On the generation, main-
base equilibrium. N Engl J Med. 1965;272:6–12. tenance, and correction of metabolic alkalosis. Am J
71. Schwartz WB, Cohen JJ. The nature of the renal Physiol. 1983;245:F425–32.
response to chronic disorders of acid-base equilib- 89. Mazur JE, Devlin JW, Peters MJ, Jankowski MA,
rium. Am J Med. 1978;64:417–28. Iannuzzi MC, Zarowitz BJ. Single versus multiple
72. Krapf R, Beeler I, Hertner D, Hulter HN. Chronic doses of acetazolamide for metabolic alkalosis in
respiratory alkalosis. The effect of sustained hyper- critically ill medical patients: a randomized, double-
ventilation on renal regulation of acid-base equilib- blind trial. Crit Care Med. 1999;27:1257–61.
rium. N Engl J Med. 1991;324:1394–401. 90. Marik PE, Kussman BD, Lipman J, Kraus P.
73. Wong HR, Chundu KR. Metabolic alkalosis in chil- Acetazolamide in the treatment of metabolic alkalosis
dren undergoing cardiac surgery. Crit Care Med. in critically ill patients. Heart Lung. 1991;20:455–9.
1993;21:884–7. 91. Amlal H, Habo K, Soleimani M. Potassium depriva-
74. Mauri S, Pedroli G, Rudeberg A, Laux-End R, tion upregulates expression of renal basolateral Na
Monotti R, Bianchetti MG. Acute metabolic alkalosis (+)-HCO(3)(-) cotransporter (NBC-1). Am J Physiol
in cystic fibrosis: prospective study and review of the Renal Physiol. 2000;279:F532–43.
literature. Miner Electrolyte Metab. 1997;23:33–7. 92. Dave-Sharma S, Wilson RC, Harbison MD,
75. Fustik S, Pop-Jordanova N, Slaveska N, Koceva S, et al. Examination of genotype and phenotype rela-
Efremov G. Metabolic alkalosis with hypoelec- tionships in 14 patients with apparent mineralocorti-
trolytemia in infants with cystic fibrosis. Pediatr Int. coid excess. J Clin Endocrinol Metab. 1998;83:
2002;44:289–92. 2244–54.
76. Hebert SC. Bartter syndrome. Curr Opin Nephrol 93. Colussi G, Rombola G, De Ferrari ME, Macaluso M,
Hypertens. 2003;12:527–32. Minetti L. Correction of hypokalemia with
77. Naesens M, Steels P, Verberckmoes R, Vanrenterghem antialdosterone therapy in Gitelman’s syndrome. Am
Y, Kuypers D. Bartter’s and Gitelman’s syndromes: J Nephrol. 1994;14:127–35.
from gene to clinic. Nephron Physiol. 2004;96:p65–78. 94. Ramsay LE, Hettiarachchi J, Fraser R, Morton JJ.
78. Schmidt H, Kabesch M, Schwarz HP, Kiess Amiloride, spironolactone, and potassium chloride
W. Clinical, biochemical and molecular genetic data in thiazide-treated hypertensive patients. Clin
in five children with Gitelman’s syndrome. Horm Pharmacol Ther. 1980;27:533–43.
Metab Res. 2001;33:354–7. 95. Vania A, Tucciarone L, Mazzeo D, Capodaglio PF,
79. Perez GO, Oster JR, Rogers A. Acid-base distur- Cugini P. Liddle’s syndrome: a 14-year follow-up of
bances in gastrointestinal disease. Dig Dis Sci. the youngest diagnosed case. Pediatr Nephrol.
1987;32:1033–43. 1997;11:7–11.
80. Bosch JP, Goldstein MH, Levitt MF, Kahn T. Effect of 96. Korkmaz A, Yildirim E, Aras N, Ercan
chronic furosemide administration on hydrogen and F. Hydrochloric acid for treating metabolic alkalosis.
sodium excretion in the dog. Am J Physiol. 1977;232: Jpn J Surg. 1989;19:519–23.
F397–404. 97. McLaughlin ML, Kassirer JP. Rational treatment of
81. van Buren M, Rabelink TJ, van Rijn HJ, Koomans acid-base disorders. Drugs. 1990;39:841–55.
HA. Effects of acute NaCl, KCl and KHCO3 loads on 98. Nasimi A, Cardona J, Berthier M, Oriot D.
renal electrolyte excretion in humans. Clin Sci Hydrochloric acid infusion for treatment of severe
(Lond). 1992;83:567–74. metabolic alkalosis in a neonate. Clin Pediatr
82. Schwartz GJ. Physiology and molecular biology of (Phila). 1996;35:271–2.
renal carbonic anhydrase. J Nephrol. 2002;15 Suppl 99. Giebisch G, Berger L, Pitts RF. The extrarenal
5:S61–74. response to acute acid-base disturbances of respira-
83. Khanna A, Kurtzman NA. Metabolic alkalosis. tory origin. J Clin Invest. 1955;34:231–45.
Respir Care. 2001;46:354–65. 100. Arbus GS, Herbert LA, Levesque PR, Etsten BE,
84. Arruda JA, Kurtzman NA. Mechanisms and classifi- Schwartz WB. Characterization and clinical
cation of deranged distal urinary acidification. Am J application of the “significance band” for acute
Physiol. 1980;239:F515–23. respiratory alkalosis. N Engl J Med. 1969;280:
85. Wagner CA, Kovacikova J, Stehberger PA, Winter C, 117–23.
Benabbas C, Mohebbi N. Renal acid-base transport: 101. Gledhill N, Beirne GJ, Dempsey JA. Renal response
old and new players. Nephron Physiol. 2006;103: to short-term hypocapnia in man. Kidney Int.
p1–6. 1975;8:376–84.
86. Roberts KE, Randall HT, Sanders HL, Hood 102. Gennari FJ, Goldstein MB, Schwartz WB. The nature
M. Effects of potassium on renal tubular reabsorption of the renal adaptation to chronic hypocapnia. J Clin
of bicarbonate. J Clin Invest. 1955;34:666–72. Invest. 1972;51:1722–30.
9 Physiology of the Developing Kidney: Acid-Base Homeostasis and Its Disorders 277

103. Cohen JJ, Madias NE, Wolf CJ, Schwartz 119. Joseph C, Twombley K, Gattineni J, Zhang Q,
WB. Regulation of acid-base equilibrium in chronic Dwarakanath V, Baum M. Acid increases NHE8 sur-
hypocapnia. Evidence that the response of the kidney face expression and activity in NRK cells. Am J
is not geared to the defense of extracellular (H+). Physiol Renal Physiol. 2012;302:16.
J Clin Invest. 1976;57:1483–9. 120. Baum M. Neonatal rabbit juxtamedullary proximal
104. Schwartz WB, Brackett Jr NC, Cohen JJ. The convoluted tubule acidification. J Clin Invest.
response of extracellular hydrogen ion concentration 1990;85:499–506.
to graded degrees of chronic hypercapnia: the physi- 121. Schwartz GJ, Evan AP. Development of solute trans-
ologic limits of the defense of Ph. J Clin Invest. port in rabbit proximal tubule. III. Na-K-ATPase
1965;44:291–301. activity. Am J Physiol. 1984;246:F845–52.
105. DuBose T. Acid-base disorders. In: Brenner BM, 122. Karashima S, Hattori S, Ushijima T, Furuse A,
editor. The kidney. 7th ed. Philadelphia: Saunders/ Nakazato H, Matsuda I. Developmental changes in
Elsevier; 2004. p. 2870. carbonic anhydrase II in the rat kidney. Pediatr
106. Temple AR. Pathophysiology of aspirin overdosage Nephrol. 1998;12:263–8.
toxicity, with implications for management. Pediat- 123. Winkler CA, Kittelberger AM, Watkins RH,
rics. 1978;62:873–6. Maniscalco WM, Schwartz GJ. Maturation of car-
107. Rennke H DB. Acid-base physiology and metabolic bonic anhydrase IV expression in rabbit kidney. Am
alkalosis. In: Renal pathophysiology: the essentials. J Physiol Renal Physiol. 2001;280:F895–903.
2nd ed. Boston: Lippincott Williams & Wilkins 2007. 124. Schwartz GJ, Olson J, Kittelberger AM,
p. 127–156. Matsumoto T, Waheed A, Sly WS. Postnatal devel-
108. Vormann J, Remer T. Dietary, metabolic, physiologic, opment of carbonic anhydrase IV expression in rabbit
and disease-related aspects of acid-base balance: fore- kidney. Am J Physiol. 1999;276:F510–20.
word to the contributions of the second International 125. Lonnerholm G, Wistrand PJ. Carbonic anhydrase
acid-base symposium. J Nutr. 2008;138:413S–4. in the human fetal kidney. Pediatr Res. 1983;17:
109. Remer T, Manz F. Estimation of the renal net acid 390–7.
excretion by adults consuming diets containing vari- 126. Goldstein L. Renal ammonia and acid excretion in
able amounts of protein. Am J Clin Nutr. infant rats. Am J Physiol. 1970;218:1394–8.
1994;59:1356–61. 127. Matsumoto T, Fejes-Toth G, Schwartz GJ. Postnatal
110. Kalhoff H, Manz F. Nutrition, acid-base status and differentiation of rabbit collecting duct intercalated
growth in early childhood. Eur J Nutr. cells. Pediatr Res. 1996;39:1–12.
2001;40:221–30. 128. Benyajati S, Goldstein L. Renal glutaminase adapta-
111. Alexy U, Kersting M, Remer T. Potential renal acid tion and ammonia excretion in infant rats. Am J
load in the diet of children and adolescents: impact of Physiol. 1975;228:693–8.
food groups, age and time trends. Public Health Nutr. 129. Chan JC. Acid-base disorders and the kidney. Adv
2008;11:300–6. Pediatr. 1983;30:401–71.
112. Prynne CJ, Ginty F, Paul AA, et al. Dietary acid-base 130. McSherry E, Morris Jr RC. Attainment and mainte-
balance and intake of bone-related nutrients in Cam- nance of normal stature with alkali therapy in infants
bridge teenagers. Eur J Clin Nutr. 2004;58:1462–71. and children with classic renal tubular acidosis. J Clin
113. Remer T, Dimitriou T, Manz F. Dietary potential renal Invest. 1978;61:509–27.
acid load and renal net acid excretion in healthy, free- 131. Challa A, Krieg Jr RJ, Thabet MA, Veldhuis JD, Chan
living children and adolescents. Am J Clin Nutr. JC. Metabolic acidosis inhibits growth hormone
2003;77:1255–60. secretion in rats: mechanism of growth retardation.
114. Frassetto LA, Morris Jr RC, Sebastian A. Effect of age Am J Physiol. 1993;265:E547–53.
on blood acid-base composition in adult humans: role 132. Hanna JDCA, Chan JCM, Han VKM. Insulin-like
of age-related renal functional decline. Am J Physiol. growth factor-1 gene expression in the tibial epiphy-
1996;271:F1114–22. seal growth plate of the acidotic and with nutritional
115. Goodman AD, Lemann Jr J, Lennon EJ, Relman limited rats. Pediatr Res. 1995;37:363A.
AS. Production, excretion, and net balance of fixed 133. Krieger NS, Frick KK, Bushinsky DA. Mechanism of
acid in patients with renal acidosis. J Clin Invest. acid-induced bone resorption. Curr Opin Nephrol
1965;44:495–506. Hypertens. 2004;13:423–36.
116. Manz F, Kalhoff H, Remer T. Renal acid excretion in 134. Lemann Jr J, Lennon EJ, Goodman AD, Litzow JR,
early infancy. Pediatr Nephrol. 1997;11:231–43. Relman AS. The net balance of acid in subjects given
117. Rector Jr FC. Sodium, bicarbonate, and chloride large loads of acid or alkali. J Clin Invest.
absorption by the proximal tubule. Am J Physiol. 1965;44:507–17.
1983;244:F461–71. 135. Lemann Jr J, Litzow JR, Lennon EJ. The effects of
118. Twombley K, Gattineni J, Bobulescu IA, chronic acid loads in normal man: further evidence for
Dwarakanath V, Baum M. Effect of metabolic acido- the participation of bone mineral in the defense
sis on neonatal proximal tubule acidification. Am J against chronic metabolic acidosis. J Clin Invest.
Physiol Regul Integr Comp Physiol. 2010;299:18. 1966;45:1608–14.
278 P.D. Yorgin et al.

136. Litzow JR, Lemann Jr J, Lennon EJ. The effect of aldosterone deficiency in hyperkalemic patients with
treatment of acidosis on calcium balance in patients chronic renal insufficiency. Kidney Int. 1980;17:
with chronic azotemic renal disease. J Clin Invest. 89–101.
1967;46:280–6. 152. Jr E. Hydrogen ion turnover in health and disease.
137. Cochran M, Wilkinson R. Effect of correction of Ann Intern Med. 1962;57:660–84.
metabolic acidosis on bone mineralisation rates in 153. Wallia R, Greenberg A, Piraino B, Mitro R, Puschett
patients with renal osteomalacia. Nephron. JB. Serum electrolyte patterns in end-stage renal dis-
1975;15:98–110. ease. Am J Kidney Dis. 1986;8:98–104.
138. Bleich HL, Moore MJ, Lemann Jr J, Adams ND, Gray 154. Kraut JA. Disturbances of acid-base balance and bone
RW. Urinary calcium excretion in human beings. disease in end-stage renal disease. Semin Dial.
N Engl J Med. 1979;301:535–41. 2000;13:261–6.
139. Lefebvre A, de Vernejoul MC, Gueris J, Goldfarb B, 155. Uribarri J, Zia M, Mahmood J, Marcus RA, Oh
Graulet AM, Morieux C. Optimal correction of aci- MS. Acid production in chronic hemodialysis
dosis changes progression of dialysis osteodystrophy. patients. J Am Soc Nephrol. 1998;9:114–20.
Kidney Int. 1989;36:1112–8. 156. Sebastian A, Schambelan M, Lindenfeld S, Morris Jr
140. Lemann Jr J, Bushinsky DA, Hamm LL. Bone buff- RC. Amelioration of metabolic acidosis with
ering of acid and base in humans. Am J Physiol Renal fludrocortisone therapy in hyporeninemic hypoaldos-
Physiol. 2003;285:F811–32. teronism. N Engl J Med. 1977;297:576–83.
141. Rodriguez-Soriano J, Vallo A. Renal tubular acidosis. 157. Kurtz I, Maher T, Hulter HN, Schambelan M,
Pediatr Nephrol. 1990;4:268–75. Sebastian A. Effect of diet on plasma acid-base com-
142. Halperin ML, Jungas RL. Metabolic production and position in normal humans. Kidney Int. 1983;24:
renal disposal of hydrogen ions. Kidney Int. 670–80.
1983;24:709–13. 158. Uribarri J, Levin NW, Delmez J, et al. Association of
143. Warnock DG. Uremic acidosis. Kidney Int. acidosis and nutritional parameters in hemodialysis
1988;34:278–87. patients. Am J Kidney Dis. 1999;34:493–9.
144. Hakim RM, Lazarus JM. Biochemical parameters in 159. Kopple JD, Kalantar-Zadeh K, Mehrotra R. Risks of
chronic renal failure. Am J Kidney Dis. 1988;11: chronic metabolic acidosis in patients with chronic
238–47. kidney disease. Kidney Int Suppl. 2005;95:S21–7.
145. Bailey JL. Metabolic acidosis: an unrecognized cause 160. Kovesdy CP, Anderson JE, Kalantar-Zadeh
of morbidity in the patient with chronic kidney dis- K. Association of serum bicarbonate levels with mor-
ease. Kidney Int Suppl. 2005;96:S15–23. tality in patients with non-dialysis-dependent CKD.
146. Kraut JA, Kurtz I. Metabolic acidosis of CKD: diag- Nephrol Dial Transplant. 2009;24:1232–7.
nosis, clinical characteristics, and treatment. Am J 161. Oh MS, Uribarri J, Weinstein J, et al. What unique
Kidney Dis. 2005;45:978–93. acid-base considerations exist in dialysis patients?
147. Uribarri J, Douyon H, Oh MS. A re-evaluation of the Semin Dial. 2004;17:351–64.
urinary parameters of acid production and excretion 162. Kovacic V, Roguljic L, Kovacic V. Metabolic acidosis
in patients with chronic renal acidosis. Kidney Int. of chronically hemodialyzed patients. Am J Nephrol.
1995;47:624–7. 2003;23:158–64.
148. Welbourne T, Weber M, Bank N. The effect of gluta- 163. Frassetto LA, Hsu CY. Metabolic acidosis and pro-
mine administration on urinary ammonium excretion gression of chronic kidney disease. J Am Soc
in normal subjects and patients with renal disease. Nephrol. 2009;20(9):1869–70. doi:10.1681/
J Clin Invest. 1972;51:1852–60. ASN.2009070710. Epub 2009 Aug 20.
149. Widmer B, Gerhardt RE, Harrington JT, Cohen 164. KDIGO. Chapter 3: Management of progression and
JJ. Serum electrolyte and acid base composition. complications of CKD. Kidney Int Suppl.
The influence of graded degrees of chronic renal 2013;3:73–90.
failure. Arch Intern Med. 1979;139:1099–102. 165. Ambuhl PM. Posttransplant metabolic acidosis: a
150. Hsu CY, Chertow GM. Elevations of serum phospho- neglected factor in renal transplantation? Curr Opin
rus and potassium in mild to moderate chronic renal Nephrol Hypertens. 2007;16:379–87.
insufficiency. Nephrol Dial Transplant. 2002;17: 166. Yakupoglu HY, Corsenca A, Wahl P, Wuthrich RP,
1419–25. Ambuhl PM. Posttransplant acidosis and associated
151. Schambelan M, Sebastian A, Biglieri EG. Prevalence, disorders of mineral metabolism in patients with a
pathogenesis, and functional significance of renal graft. Transplantation. 2007;84:1151–7.
Bone Developmental Physiology
10
MH Lafage-Proust

Contents Introduction
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 279
Ossification is a key stage of bone development
Embryonic Bone Development . . . . . . . . . . . . . . . . . . . . 279
and growth. This complex process combines
Ossification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 280 cell–cell contacts, cell proliferation and apoptosis,
Intramembranous Ossification . . . . . . . . . . . . . . . . . . . . . . . 282
cell migration and differentiation, vasculogenesis,
Endochondral Ossification . . . . . . . . . . . . . . . . . . . . . . . . . . . 282
Periosteum . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286 matrix synthesis, and mineralization. The com-
plete ossification of the skeleton is a very long
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286
process that begins as early as the first weeks of
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287 fetal development and ends at the end of growth,
some 20 years later. The local molecular control of
bone growth and development is summarized.
Hormonal regulation and the effects of genetic
mutations are beyond the scope of this review.

Embryonic Bone Development

Bones derive from the mesoderm embryonic


layer. The axial skeleton is formed from the
sclerotome compartment of the somites and the
limbs originate from the lateral plate mesoderm.
The skull arises from neural crest-derived mesen-
chymal cells. The site and the shape of each bone
are determined early during embryonic develop-
ment patterning, which is under tight genetic con-
trol. Briefly, the spine patterning follows the
anterior–posterior axis and results from the for-
mation of segmented mesodermal symmetric
structures, the somites, which develop succes-
M. Lafage-Proust (*)
sively, according sequential bursts of gene expres-
INSERM U 1059, Université de Lyon, Saint-Etienne,
France sion. These waves, which are rhythmically
e-mail: lafagemh@univ-st-etienne.fr activated, act as a molecular oscillator called the
# Springer-Verlag Berlin Heidelberg 2016 279
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_9
280 M. Lafage-Proust

Fig. 1 Control of limb bud


development: (a) Limb
patterning and growth along
the anterior/posterior and
proximal/distal axes and
illustration of feed-back
loops between FGFS and
Shh signaling pathways.
AER apical ectodermal
ridge, ZPA zone of
polarizing activity, BMP
bone morphogenetic
protein, FGF fibroblast
growth factor, Shh sonic
hedgehog, Grem1 gremlin
1. See text for explanations.
(b) Limb patterning and
growth along the ventral/
dorsal axis, illustration of the
Wnt7a/BMP signaling loop

“segmentation clock” ([1]). They involve the hedgehog (Shh) [2]. Finally, the remaining
Notch, Wnt/β-catenin and fibroblast growth factor non-AER ectoderm region of the limb bud con-
(FGF) signaling pathways, which exert specific trols limb development along the DV axis mainly
perimeters of influence. Vertebrae and ribs will via Wnt and BMP signaling control. Briefly, BMP
develop from the sclerotome, at the ventral part activates the expression of En1, an homeotic gene,
of the somites. in the ventral ectoderm [3], which leads to the
Limb patterning follows an even more com- suppression of Wnt7a expression in this region.
plex scheme according to three axes: the limb is Thus, expression of Wnt7a is restricted to the
elongated along a proximal–distal axis (PD: from dorsal ectoderm, where it induces, in the underly-
the shoulder to the last phalanx of the digits), it ing mesoderm, expression of Lmx1b, another
flattens along a dorsal–ventral axis (DV: from the homeobox transcription factor that establishes
back of the hand to the palm), and it becomes the dorsal identity (Fig. 1) [4]. Conversely,
asymmetric along an anterior–posterior axis (AP: BMPs may act directly in an En-1-independent
from the thumb to the little finger; Fig. 1). After manner via inhibition of Lmx1b expression. The
mesenchymal cell condensation at the presump- coordination of the three controlling zones is
tive site of the limb, a bud outgrowth takes place, mediated by inter-connected feed-back loops,
whose tip consists of the apical ectodermal ridge which lead to harmonious limb development.
(AER). The AER is a key region that controls the FGFs expressed in the AER, control Shh expres-
limb PD elongation via the balance of two major sion in the ZPA, which in turn, regulates FGFs.
signaling pathways: Wnt/beta cateninsignaling is Shh is also able to antagonize the inhibitory action
a positive regulator, while BMP signaling is an of BMP on AER FGF expression, via Gremlin
inhibitor of limb elongation. In addition, many 1, and in turn, high FGF levels in AER inhibit
members of the FGF family interact at this level, Gremlin1 [5].
especially via an FGF10–FGF8 feedback loop.
The zone of polarizing activity (ZPA), which is
located at the posterior part of the limb bud, close Ossification
to the AER, controls the AP hand patterning. The
early events are mediated via the combination of Bone can develop from two different mecha-
transcription factors, namely Alx 4, Gli 3, Twist nisms: endochondral ossification or
1, and dHAND, followed by the action of sonic intramembranous ossification. The term
10 Bone Developmental Physiology 281

Fig. 2 Fetal human woven bone. (a) Goldner’s trichrome observed under polarized light, same section as in (a),
stained section. Magnification 100, green mineralized illustrating the woven aspect of primary bone. (c) Adult
bone, pink osteoid tissue. (b) Bone histological sections bone with lamellar collagen
282 M. Lafage-Proust

Fig. 3 Schematic illustration of endochondral bone ossification

ossification is used because it results from the mineralize progressively. Some osteoblasts get bur-
transformation of a pre-existing mesenchymal ied in the matrix and differentiate into osteocytes.
template, which is cartilage in the case of endo- The ossification progresses in a centrifugal way, the
chondral ossification and a condensation of most proliferating and immature cells being at the
non-cartilaginous mesenchymal cells in the case periphery. The matrix deposited at this early step is
of membranous ossification. The skull, the jaw, not well organized. When observed under polar-
and a large part of the scapula undergo membra- ized light, the collagen fibers appear woven
nous ossification. Skeletal ossification results (Fig. 2). Later, it is resorbed by osteoclasts and
from the successive appearance of ossification lamellar collagen is synthesized by a second set
centers, which take place sequentially in each of osteoblasts. At the skull, the specialized struc-
bone-to-be, according to a pattern that is tightly tures called “sutures,” which connect the various
regulated in time and space. cranial bones, ossify more progressively during
growth. Morphogenesis and phenotypic preserva-
tion of the sutures are regulated by complex tissue
Intramembranous Ossification interactions, especially with the underlying dura
mater, thus allowing the harmonious development
The mesenchymal cells proliferate and condense of both the skull and the brain.
into nodules. Some cells differentiate into osteo-
blast precursors and then into osteoblasts. Others
become endothelial precursor cells and then endo- Endochondral Ossification
thelial cells, which migrate and form capillaries.
The matrix synthesized by osteoblasts, The ossification process summarized here takes a
called”osteoid”, which surrounds the cells, long bone as an example (Fig. 3).
10 Bone Developmental Physiology 283

The Cartilage Template • The resting zone is adjacent to the trabeculae of


The first step consists of the differentiation of the epiphysis, and contains a thin layer of
condensed mesenchymal cells into chondrocytes small, non-oriented, reserve chondrocytes.
and the formation of a cartilage anlage surrounded • The proliferation zone consists of stacks of flat
by perichondrium [6]. Chondrocytes proliferate oval cycling chondrocytes organized in longi-
and synthesize a matrix mainly comprising type tudinal parallel rows.
II collagen. Then, the deeper chondrocytes • The zone of hypertrophy comprises round and
located at the middle of the future diaphysis stop large chondrocytes within individual lacunae.
proliferating and become pre-hypertrophic. These This step is preceded by a phase where
cells then undergo hypertrophy, increasing their pre-hypertrophic chondrocytes exit the cell
volume and secreting extracellular matrix cycle. In the lowest layer, the walls of the
containing type X collagen, which will eventually lacunae are mineralized and some
mineralize. Meanwhile, the periosteal cells, which chondrocytes exhibit signs of apoptosis
surround the zone of hypertrophic chondrocytes, • The zone of ossification contains osteoclasts
differentiate into osteoblasts, leading to the for- that resorb the mineralized cartilage matrix.
mation of a hollow bony cylinder called the “peri- Their precursors are recruited at this specific
osteal collar.” Then, the hypertrophic site via trafficking through the walls of the
chondrocytes facing the bone collar undergo apo- numerous arch-shaped vessels that develop at
ptosis, which leads to the collapse of the cartilage contact with the hypertrophic zone. Then, a
matrix surrounding them, leaving a cavity with first set of osteoblasts differentiate and deposit
few intact septa. woven bone on the top of the remnants of
calcified cartilage matrix, using them as scaf-
Formation of the Primary Ossification folds. These trabeculae directly “hanging”
Center below the hypertrophic zone form “the primary
Sprouting blood vessels preceded by osteoclasts spongiosa.” They undergo remodeling [10,
(multinucleated bone-resorbing cells) penetrate 11], including a first step of resorption by oste-
the periosteal collar and invade the underlying oclasts, which remove the woven bone
mineralized cartilage, allowing the colonization followed by a second set of osteoblasts that
of the cavity by osteoblast precursors [7], which replace it with lamellar bone, leading to the
differentiate into osteoblasts and rapidly deposit formation of the “secondary spongiosa.”
woven type I collagen matrix, constituting the
primary ossification center (Fig. 4a) [8]. Ossifica- Regulation of Cell Proliferation
tion extends from the center of the diaphysis and Differentiation
toward the epiphyses as vascularization develops During bone elongation each zone “feeds” the
and the mineralized cartilage matrix is progres- adjacent one according to a complex regulation
sively replaced by bone. According to a similar that allows harmonious bone growth [12]. This
mechanism, secondary centers of ossification are progression is tightly regulated by a number of
formed at each epiphysis. Vascular invasion is interacting signaling pathways that balance chon-
stimulated by VEGF expression by hypertrophic drocyte proliferation and hypertrophy [13, 14].
chondrocytes owing to activation of the Hif-1 Briefly, Indian hedgehog (Ihh) is synthesized
alpha signaling pathway [9]. by the prehypertrophic chondrocytes. It diffuses
toward the proliferating chondrocytes and main-
The Growth Plate Allows Bone tains them in a proliferating state by stimulating
Elongation PTHrP expression [15]. PTHrP promotes chon-
The growth plates are located between the diaph- drocyte cycling and inhibits hypertrophy
ysis and the epiphyses of long bones. The growth [16]. This stimulus stops when chondrocytes
plate allows bone elongation until the end of progress and leave the area of PTHrP
growth. It comprises several zones (Fig. 4): diffusion. Ihh also modulates hypertrophy in a
284 M. Lafage-Proust

Fig. 4 (continued)
10 Bone Developmental Physiology 285

Fig. 4 Growth plate organization. (a) Schematic illustra- Magnification 250, toluidine blue staining, cartilage is
tion of the growth plate. (b, c) Growth plate section in a stained dark purple. (d) Tartrate-resistant acid phosphatase
growing rat. (b) Magnification 100, Goldner’s trichrome- (TRAP) histo-enzymological staining of osteoclasts at the
stained histological sections. Note the green (i.e., mineral- junction between the hypertrophic zone and the primary
ized) walls of the lacunae of hypertrophic chondrocytes. (c) spongiosa, aniline blue counter-staining, 400

PTHrP-independent manner [17]. The Wnt bone vascularization as well as osteoclast and
canonical and noncanonical signaling pathways osteoblast recruitment to the growth plate
interact with the Ihh/PTHrP-negative feedback [22]. FGF effects may also be down-regulated
and contribute to the regulation of the pace by local specific inhibitors such as Sef, which is
of chondrocyte proliferation, survival, and hyper- expressed in the periosteum and at the
trophy [18]. The spatial organization of the pro- osteochondral junction in the growth plate of neo-
liferation zone into columns is dependent on natal mice [23]. Basically, BMP and FGF signal-
the noncanonical frizzled signaling pathway ing pathways exert opposite effects in the growth
[19]. BMP signaling modulates Ihh expression, plate. For instance, it was recently shown that the
chondrocyte proliferation and maturation. It has deletion of BMP type I receptor a (BMPR1a) in
a dual action on hypertrophic chondrocytes, FGFR3 knock-out mice rescued the bone over-
whereby it slows down hypertrophy and promotes growth phenotype by reducing chondrocyte dif-
terminal maturation [20]. The major role of FGFs ferentiation, because FGFR3 activation induces
in bone growth was revealed 20 years ago when the proteasome degradation of Bmpr1a [24].
Shiang et al. [21] reported that mutations in the The master genes for chondrocyte and osteoblast
FGFR3 transmembrane domain were responsible differentiation are Sox9 [25] and Runx2 [26, 27]
for achondroplasia, one of the most common respectively. These transcription factors are essen-
forms of dwarfism caused by genetic mutations. tial molecular switches for the commitment of mes-
FGFR3 signaling inhibits chondrocyte prolifera- enchymal cells toward the cartilage and bone-
tion and hypertrophy. FGFR1 and FGFR2 are also forming cell lineages. They also play a major role
involved in the regulation of bone growth. FGF18 in the differentiation and the maintenance of their
is the major ligand in a number of cell types, phenotype. Therefore, they are necessary targets of
including hypertrophic chondrocytes and the above-mentioned signaling pathways. Basi-
perichondrial cells. It promotes both proliferation cally, activation of Wnt signaling leads to enhanced
and modulates initiation of chondrocyte hypertro- ossification and suppressed chondrocyte formation,
phy at early stages of chondrogenesis, mediates whereas genetic inactivation of beta-catenin, the
286 M. Lafage-Proust

main factor involved in intracellular canonical Wnt Periosteum


signal transduction, causes ectopic formation of
chondrocytes at the expense of osteoblast differen- The periosteum is composed of two distinct
tiation during both intramembranous and endo- layers (Fig. 5): an outer fibrous layer and an
chondral ossification [28]. Sox9 is phosphorylated inner proliferative cambium layer in contact
and activated by PTHrP and it antagonizes the Wnt with the underlying cortical bone. The outer
canonical signaling pathway by promoting beta- fibrous layer consists of fibroblasts and thick
catenin proteasome degradation [29]. Runx2 exerts collagen fibers. The inner layer is highly
complex effects according to the cells where it is vascularized and contains mesenchymal stem
activated, namely chondrocytes or perichondrial cells that can differentiate into osteoblasts and
cells. Runx2 deficiency in mice decreases chondro- chondrocytes. During growth, periosteal bone
cyte Ihh expression and retards chondrocyte hyper- apposition increases the outer perimeter of the
trophy [30]. In contrast, in perichondrial cells, bone, while resorption occurs at the endocortical
Runx2 activation (under the inhibitory control of surface, leading to harmonious radial expansion
Twist-1) leads to FGF18 release and inhibition of of both the bone cortex and the marrow cavity
chondrocyte maturation [31]. At the level of the [35]. However, progressive cortex thickening
primary and secondary spongiosae, the mecha- occurs, due to an excess of outer formation over
nisms of osteoblast differentiation are not inner resorption, and is associated with bone
completely understood. During long bone develop- tissue displacement away from the neutral axis
ment, all the populations of osteoblasts within peri- of the shaft, thus optimizing bone mechanical
osteum and osteoblasts at the endosteal and resistance to load [36]. Periosteal apposition is
trabecular bone surfaces within the marrow are continuous in the growing skeleton, characteris-
derived from the embryonic perichondrium. Differ- tic of bone modeling [37]. Studies in various
entiation of osteoblast precursors, chondrocyte apo- mouse strains showed that cortical bone geome-
ptosis, and the development of vessels at contact try (size and shape), which is partly determined
with the growth plate are functionally [32] and by periosteum activity, as seen above, is geneti-
spatially coupled. Skeletal stem cells borne by the cally controlled by different, although
vessel walls may differentiate into osteoblasts. overlapping, groups of polymorphic loci [38]. It
Recently, Kusumbe et al. [33] described two sub- is highly dependent on mechanical load. The
types of bone vessels according to their high (H) or biology of the periosteum has been less fre-
low (L) level of CD31 and endomucin expression. quently studied than that of longitudinal growth.
In young animals, H vessels contain more prolifer- Recently, it was shown that periosteal cells
ating endothelial cells. They are abundant in the express periostin, an extracellular matrix protein,
primary spongiosa and run along the cortical end- also expressed by cortical osteocytes. This
osteum. H vessel walls bear more osteoprogenitors expression is stimulated by load and PTH
and express higher levels of HIF-1α. Their number [39]. BMP2 expression in osteoblasts controls
decreases with age. Inhibition or activation of the periosteal vascularization [40].
HIF-1α signaling pathway in endothelial cells only,
modulates the number of perivascular osteopro-
genitors. In addition, Zhou et al. used mice in
which activation of gene promoters specific to Conclusion
hypertrophic or nonhypertrophic chondrocytes
(i.e., type X collagen and aggrecan respectively) Our knowledge of bone development and growth
could be observed and allowed cell lineage track- biology is not complete. Indeed, the physiology
ing. They showed that both chondrocytes before of endochondral ossification has been exten-
initial ossification and growth plate chondrocytes sively studied over the past 20 years at the molec-
before or after birth could transdifferentiate into ular level, which has brought major insights into
osteoblasts [34]. the pathophysiology of a number of skeletal
10 Bone Developmental Physiology 287

Fig. 5 Periosteum. (a) Schematic illustration of the periosteum during growth. (b, c) Goldner’s trichrome-stained bone
histological sections of periosteum in growing humans, green mineralized bone, pink osteoid tissue. (b) 100, (c) 250

diseases that affect the pediatric population.


Furthermore, this information has been some- References
how “recycled” in the research on osteoarthritis
1. Dequéant ML, Glynn E, Gaudenz K, Wahl M, Chen J,
(OA), which is a major public health concern, Mushegian A, Pourquié O. A complex oscillating net-
since some of the events are recapitulated during work of signaling genes underlies the mouse segmen-
OA progression at the osteochondral junction. In tation clock. Science. 2006;314(5805):1595–8.
2. Johnson RL, Laufer E, Riddle RD, Tabin C. Ectopic
contrast, the role of the periosteum and the pre-
expression of Sonic hedgehog alters dorsal-ventral pat-
cise molecular mechanisms that control its func- terning of somites. Cell. 1994;79(7):1165–73.
tions remain to be elucidated. Similarly, it is only 3. Pizette S, Abate-Shen C, Niswander L. BMP controls
recently that major breakthroughs have been proximodistal outgrowth, via induction of the apical
ectodermal ridge, and dorsoventral patterning in the
reported in bone vessel heterogeneity and its
vertebrate limb. Development. 2001;128(22):4463–74.
specific roles.
288 M. Lafage-Proust

4. Chen H, Johnson RL. Interactions between dorsal- 19. Li Y, Dudley AT. Non canonical frizzled signaling
ventral patterning genes lmx1b, engrailed-1 and regulates cell polarity of growth plate chondrocytes.
wnt-7a in the vertebrate limb. Int J Dev Biol. Development. 2009;136:1083–92.
2002;46(7):937–41. 20. Yoon BS, Pogue R, Ovchinnikov DA, Yoshii I,
5. Khokha MK, Hsu D, Brunet LJ, Dionne MS, Harland Mishina Y, Behringer RR, Lyons KM. BMPs regulate
RM. Gremlin is the BMP antagonist required for main- multiple aspects of growth-plate chondrogenesis
tenance of Shh and Fgf signals during limb patterning. through opposing actions on FGF pathways. Develop-
Nat Genet. 2003;34(3):303–7. ment. 2006;133(23):4667–78.
6. Gilbert SF. Developmental biology. 6th ed. Sunder- 21. Shiang R, Thompson LM, Zhu YZ, Church DM,
land: Sinauer Associates; 2000. Fielder TJ, Bocian M, Winokur ST, Wasmuth JJ. Muta-
7. Maes C, Kobayashi T, Selig MK, et al. Osteoblast tions in the transmembrane domain of FGFR3 cause
precursors, but not mature osteoblasts, move into the most common genetic form of dwarfism, achondro-
developing and fractured bones along with invading plasia. Cell. 1994;78(2):335–42.
blood vessels. Dev Cell. 2010;19:329–44. 22. Liu Z, Lavine KJ, Hung IH, Ornitz DM. FGF18 is
8. Colnot CI, Helms JA. A molecular analysis of matrix required for early chondrocyte proliferation, hypertro-
remodeling and angiogenesis during long bone devel- phy and vascular invasion of the growth plate. Dev
opment. Mech Dev. 2001;100:245–50. Biol. 2007;302(1):80–91.
9. Schipani E, Ryan HE, Didrickson S, Kobayashi T, 23. He Q, Yang X, Gong Y, Kovalenko D, Canalis E,
Knight M, Johnson RS. Hypoxia in cartilage: Rosen CJ, Friesel RE. Deficiency of Sef is associated
HIF-1alpha is essential for chondrocyte growth arrest with increased postnatal cortical bone mass by regulat-
and survival. Genes Dev. 2001;15(21):2865–76. ing Runx2 activity. J Bone Miner Res. 2014;29(5):
10. Parfitt AM. Targeted and nontargeted bone 1217–31.
remodeling: relationship to basic multicellular unit 24. Qi H, Jin M, Duan Y, Du X, Zhang Y, Ren F, Wang Y,
origination and progression. Bone. 2002;30(1):5. Tian Q, Wang X, Wang Q, Zhu Y, Xie Y, Liu C, Cao X,
11. Parfitt AM. Osteonal and hemi-osteonal remodeling: Mishina Y, Chen D, Deng CX, Chang Z, Chen
the spatial and temporal framework for signal traffic in L. FGFR3 induces degradation of BMP type I receptor
adult human bone. J Cell Biochem. 1994;55(3): to regulate skeletal development. Biochim Biophys
273–86. Acta. 2014;1843(7):1237–47.
12. Yang Y. Skeletal morphogenesis and embryonic devel- 25. Bi W, Deng JM, Zhang Z, Behringer RR, de
opment. In: Rosen CJ, editor. Primer on the metabolic Crombrugghe B. Sox9 is required for cartilage forma-
bone diseases and disorders of mineral metabolism. 8th tion. Nat Genet. 1999;22(1):85–9.
ed. Ames: Wiley-Blackwell; 2013. p. 3–15. 26. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G.
13. Kronenberg HM. Developmental regulation of the Osf2/Cbfa1: a transcriptional activator of osteoblast
growth plate. Nature. 2003;423:332–6. differentiation. Cell. 1997;89(5):747–54.
14. Minina E, Kreschel C, Naski MC, Ornitz DM, 27. Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K,
Vortkamp A. Interaction of FGF, Ihh/Pthlh, and BMP Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M,
signaling integrates chondrocyte proliferation and Sato M, Okamoto R, Kitamura Y, Yoshiki S, Kishimoto
hypertrophic differentiation. Dev Cell. 2002;3(3): S. Targeted disruption of Cbfa1 results in a complete
439–49. lack of bone formation owing to maturational arrest of
15. Karp SJ, Schipani E, St-Jacques B, Hunzelman J, osteoblasts. Cell. 1997;89(5):755–64.
Kronenberg H, McMahon AP. Indian hedgehog coor- 28. Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/
dinates endochondral bone growth and morphogenesis beta-catenin signaling in mesenchymal progenitors
via parathyroid hormone related-protein-dependent controls osteoblast and chondrocyte differentiation
and -independent pathways. Development. 2000;127: during vertebrate skeletogenesis. Dev Cell. 2005;8(5):
543–8. 739–50.
16. Lanske B, Karaplis AC, Lee K, Luz A, Vortkamp A, 29. Akiyama H, Lyons JP, Mori-Akiyama Y, Yang X,
Pirro A, Karperien M, Defize LH, Ho C, Mulligan RC, Zhang R, Zhang Z, Deng JM, Taketo MM,
Abou-Samra AB, Juppner H, Segre GV, Kronenberg Nakamura T, Behringer RR, McCrea PD, de
HM. PTH/PTHrP receptor in early development and Crombrugghe B. Interactions between Sox9 and beta-
Indian hedgehog-regulated bone growth. Science. catenin control chondrocyte differentiation. Genes
1996;273(5275):663–6. Dev. 2004;18(9):1072–87.
17. Mak KK, Kronenberg HM, Chuang PT, Mackem S, 30. Kim IS, Otto F, Zabel B, Mundlos S. Regulation of
Yang Y. Indian hedgehog signals independently of chondrocyte differentiation by Cbfa1. Mech Dev.
PTHrP to promote chondrocyte hypertrophy. Develop- 1999;80(2):159–70.
ment. 2008;135(11):1947–56. 31. Hinoi E, Bialek P, Chen YT, Rached MT, Groner Y,
18. Yang Y, Topol L, Lee H, Wu J. Wnt5a and Wnt5b Behringer RR, Ornitz DM, Karsenty G. Runx2 inhibits
exhibit distinct activities in coordinating chondrocyte chondrocyte proliferation and hypertrophy through
proliferation and differentiation. Development. its expression in the perichondrium. Genes Dev.
2003;130(5):1003–15. 2006;20(21):2937–42.
10 Bone Developmental Physiology 289

32. Gerber HP, Vu TH, Ryan AM, Kowalski J, Werb Z, 38. Volkman SK, Galecki AT, Burke DT, et al. Quantitative
Ferrara N. VEGF couples hypertrophic cartilage trait loci for femoral size and shape in a enetically
remodeling, ossification and angiogenesis during endo- heterogenous mouse population. J Bone Miner Res.
chondral bone formation. Nat Med. 1999;5(6):623–8. 2003;18(8):1497–505.
33. Kusumbe A, Ramasamy S, Adams RH. Coupling of 39. Bonnet N, Conway SJ, Ferrari SL. Regulation of
angiogenesis and osteogenesis by a specific vessel beta catenin signaling and parathyroid hormone ana-
subtype in bone. Nature. 2014;507:325–8. bolic effects in bone by the matricellular protein
34. Zhou X, von der Mark K, Henry S, Norton W, periostin. Proc Natl Acad Sci U S A. 2012;109(37):
Adams H, de Crombrugghe B. Chondrocytes transdif- 15048–53.
ferentiate into osteoblasts in endochondral bone during 40. Yang W, Guo D, Harris MA, Cui Y, Gluhak-Heinrich J,
development, postnatal growth and fracture healing in Wu J, Chen XD, Skinner C, Nyman JS, Edwards JR,
mice. PLoS Genet. 2014;10(12):e1004820. Mundy GR, Lichtler A, Kream BE, Rowe DW,
35. Seeman E. Periosteal bone formation – a neglected Kalajzic I, David V, Quarles DL, Villareal D, Scott G,
determinant of bone strength. N Engl J Med. Ray M, Liu S, Martin JF, Mishina Y, Harris
2003;349:320–3. S. Bmp2 in osteoblasts of periosteum and trabecular
36. Rauch F. Bone growth in length and width: the Yin and bone links bone formation to vascularization and
Yang of bone stability. J Musculoskelet Neuronal Inter- mesenchymal stem cells. J Cell Sci. 2013;126(Pt 18):
act. 2005;5(3):194–201. 4085–98.
37. Parfitt AM, Travers R, Rauch F, Glorieux
FH. Structural and cellular changes during bone
growth in healthy children. Bone. 2000;27(4):487–94.
Physiology of the Developing Kidney:
Disorders and Therapy of Calcium 11
and Phosphorous Homeostasis

€ppner
Amita Sharma, Rajesh V. Thakker, and Harald Ju

Contents Jansen’s Metaphyseal Chondrodysplasia (JMC) . . . . 298


Williams Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 298
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292 Infantile Hypercalcemia: CYP24A1 Deficiency . . . . 300
Regulators of Calcium and Phosphate Hypocalcemia and Hyperphosphatemia Due
Homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292 to Reduced Parathyroid Hormone Activity . . . . . . 300
Parathyroid Hormone (PTH) and PTH-Related Hypoparathyroidism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 300
Peptide (PTHrP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292 Abnormalities at the Calcium-Sensing Receptor
Vitamin D and Its Metabolites . . . . . . . . . . . . . . . . . . . . . . . 293 (CaSR) and the Downstream Signaling
Fibroblast Growth Factor 23 (FGF23) and Molecules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 307
Other Proteins with Phosphaturic Properties . . . . . . . . 294 Pseudohypoparathyroidism (PHP) . . . . . . . . . . . . . . . . . . 308
Hypercalcemia and Hypophosphatemia Due Blomstrand’s Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 310
to Increased Parathyroid Hormone Secretion . . . 295 Hyperphosphatemic Disorders with Reduced
Parathyroid Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 295 Secretion of Biologically Active FGF23 . . . . . . . . . . . 311
Hyperparathyroidism in Chronic Kidney Hyperphosphatemic Familial Tumoral
Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296 Calcinosis (HFTC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311
Non-syndromic Isolated Hyperparathyroidism . . . . . . 296 Hyperostosis with Hyperphosphatemia (HHS) . . . . . . 312
Hypercalcemic Disorders Without Elevated Hypophosphatemic Disorders . . . . . . . . . . . . . . . . . . . . . 312
Parathyroid Hormone Secretion . . . . . . . . . . . . . . . . . . 296 Hypophosphatemic Disorders with Increased
Disorders of the Calcium-Sensing Receptor (CaSR) FGF23 Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 312
and the Downstream Signaling Molecules . . . . . . . . . . 296 Hypophosphatemic Disorders with Normal or
Suppressed FGF23 Activity . . . . . . . . . . . . . . . . . . . . . . . . . 315
Other Hypophosphatemic Disorders . . . . . . . . . . . . . . . . 317
A. Sharma (*) Acute and Chronic Treatment of Calcium
Department of Pediatrics, Pediatric Nephrology Unit, and Phosphorus Disorders . . . . . . . . . . . . . . . . . . . . . . . . . 317
Massachusetts General Hospital and Harvard Medical Hypercalcemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317
School, Boston, MA, USA Hypocalcemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 318
e-mail: asharma5@mgh.harvard.edu Hyperphosphatemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 319
R.V. Thakker Hypophosphatemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320
Radcliffe Department of Medicine, Academic Endocrine Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321
Unit, University of Oxford, OCDEM (Oxford Centre for
Diabetes, Endocrinology and Metabolism), The Churchill References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321
Hospital Headington, Oxford, UK
e-mail: rajesh.thakker@ndm.ox.ac.uk
H. J€uppner
Departments of Medicine and Pediatrics, Endocrine Unit
and Pediatric Nephrology Unit, Massachusetts General
Hospital and Harvard Medical School, Boston, MA, USA
e-mail: jueppner@helix.mgh.harvard.edu

# Springer-Verlag Berlin Heidelberg 2016 291


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_10
292 A. Sharma et al.

Introduction Regulators of Calcium and Phosphate


Homeostasis
The regulation of calcium and phosphate homeo-
stasis involves several different hormones that Parathyroid Hormone (PTH) and
act on the kidney, intestine, and bone. The most PTH-Related Peptide (PTHrP)
important calcium-regulating peptide hormone is
parathyroid hormone (PTH). Its production and The mature secreted form of PTH peptide com-
secretion by the parathyroid glands increases prises 84 amino acids, which is derived from a
in response to a decrease in the extracellular longer prepropeptide (for review, see Ref. [10]).
calcium concentration. PTH increases (1) the PTH gene transcription (as well as PTH peptide
1α-hydroxylase activity in the proximal renal secretion) is regulated primarily by the extracel-
tubules to promote production of the biologically lular concentration of calcium and through a vita-
active 1,25-dihydroxyvitamin D (1,25(OH)2D) min D response element upstream of the
from its precursor 25-hydroxyvitamin D, thereby transcription start site [11, 12]. Phosphate affects
enhancing intestinal absorption of calcium (and also parathyroid gland activity, independent of
phosphorus); (2) it stimulates bone resorption, concomitant changes in ambient calcium concen-
thus releasing calcium and (phosphorus); (3) it tration, parathyroid gland activity; however, these
enhances calcium reabsorption in the distal renal effects take several hours and may result from
tubules; and (4) it promotes urinary phosphate secondary changes in hormone biosynthesis
excretion. These phosphaturic actions of PTH rather than secretion [13–15].
occur within minutes by reducing the expression PTH secretion by the parathyroid glands is
of two sodium-dependent phosphate regulated through the calcium-sensing receptor
cotransporters, NPT2a and NPT2c, in the proxi- (CaSR), which recognizes remarkably small per-
mal convoluted tubules. The long-term regula- turbations in calcium and responds quickly by
tion of phosphate homeostasis involves altering PTH secretion. This G protein-coupled
fibroblast growth factor 23 (FGF23), a more receptor is stimulated by extracellular calcium
recently discovered hormone made by osteocytes and requires coupling to two closely related G
and probably osteoblasts. Like PTH, FGF23 proteins, namely, Gαq and Gα11, to activate the
reduces the expression of NPT2a and NPT2c, Ca+2/IP3/PKC signaling pathway. Besides being
but the time courses for the effects of both hor- expressed in the chief cells of the parathyroid
mones are very different [1–3]. Furthermore, in glands, the CaSR is expressed in the kidneys and
contrast to the stimulatory actions of PTH on in several other tissues, albeit at lower abundance,
1α-hydroxylase, FGF23 reduces the expression which are not directly involved in the regulation
of this enzyme in the proximal renal tubules, of calcium homeostasis. In addition to regulating
and it enhances 24-hydroxylase, leading to two PTH secretion, the CaSR plays an important role
different mechanisms to a reduction in serum in regulating aquaporin trafficking in the
1, 25(OH)2D levels [4–8]. kidneys [16].
This chapter will review the physiological and The PTH-related peptide (PTHrP also known
biochemical mechanisms underlying calcium and as PTHrH, PTH-related hormone), which was
phosphate homeostasis and the genetic basis of first isolated from tumors that cause the humoral
rare inherited disorders that provided important hypercalcemia of malignancy, shares significant
novel insights into the regulation of these amino acid sequence homology with PTH
minerals [9]. [17–19]. PTH and PTHrP, although distinct
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 293

Multiple differentiation of growth plate chondrocytes


tissues [22, 27]. A closely related receptor, the PTH2
receptor, shares more than 50 % homology with
PTHrP PTH
the PTH/PTHrP receptor [28]. Its primary ligand,
Ca++ TIP39 (tuberoinfundibular peptide of 39 residues),
is involved in nociception and reproduction
Cartilage
Breast [29–31].
Bone
Teeth Kidney
Pancreas
Skin Regulation of
other tissues ? calcium and phosphate
Vitamin D and Its Metabolites
Fig. 1 Parathyroid hormone (PTH) mediates its endocrine
actions through the PTH/PTHrP receptor expressed in the Most of the vitamin D (D2 and D3) in healthy
kidney and the bone to regulate mineral ion homeostasis individuals is derived from the precursor
and bone metabolism. In numerous other tissues, this G 7-dehydrocholestrol through exposure to ultravi-
protein-coupled receptor mediates the paracrine actions of
olet light. It is made in the skin and is stored in
PTHrP; particularly important is its role in the growth plate
muscle or fat [32, 33]. Presumably through the
enzyme CYP2R1 [34, 35], the prohormone
undergoes hydroxylation in the liver to form the
products of different genes, exhibit considerable 25-hydroxyvitamin D metabolite (25(OH)D). In
functional and structural similarities, including the proximal convoluted tubular cells of the kid-
equivalent positions of the boundaries between ney, 25(OH)D is further hydroxylated by the
some of the coding exons and of the adjacent enzyme 25-hydroxyvitamin D-1α-hydroxylase
introns; both may have evolved from a shared (1α-hydroxylase; CYP27B) to yield the biologi-
ancestral gene [10, 20]. PTHrP is a larger, more cally active metabolite 1,25 dihydroxyvitamin D
complex protein than PTH that functions as an (1,25(OH)2D) [32, 33]. The expression of
autocrine/paracrine rather than an endocrine fac- CYP27B is regulated by extracellular concentra-
tor with a little influence on calcium homeostasis tions of ionized calcium, inorganic phosphate,
except when secreted in large concentrations PTH, and FGF23 [36]. 1,25(OH)2D binds in tar-
[21]. Its most prominent functions involve the get organs (e.g., the intestine, bones, kidneys,
regulation of chondrocyte proliferation and dif- and parathyroids) to the intracellular vitamin D
ferentiation, and consequently bone elongation receptor (VDR) [37] and thereby activates the
and growth [22]. It also affects mammary gland transcription of genes in the bone, kidney, and
development and function [23, 24]. enterocytes that help increase gut absorption of
PTH and PTHrP both mediate their actions calcium, reduce urinary calcium losses, and
through a common G protein-coupled receptor, increase bone resorption, thereby ensuring ade-
the PTH/PTHrP receptor [25, 26] (Fig. 1). In the quate extracellular concentration of calcium and
kidney and bone, it mediates the endocrine actions phosphate [33, 37]. Homozygous or compound
of PTH. However, the most abundant expression heterozygous mutations in CYP27B, CYP2R1,
of the PTH/PTHrP receptor occurs in or VDR lead to hypocalcemia, which increases
chondrocytes of the metaphyseal growth plate PTH secretion, thus enhancing urinary phosphate
where it mediates the autocrine/paracrine actions excretion, which leads to the development of rick-
of PTHrP, i.e., it delays the hypertrophic ets/osteomalacia [32–35].
294 A. Sharma et al.

~15 kDa ~8 kDa


1 25 176-9 251

FAM20c-dependent
phosphorylation

N-term.
RXTR… SAEDD..
C-term.

Cleavage by Furin-
like enzyme

Fig. 2 The primary structure of FGF23. The C-terminal like protease. Mutations at this site make FGF23 resistant
and the N-terminal portions of FGF23 are connected by an to the cleavage by a furin-like protease, thereby stabilizing
“RXXR” motif, which is the recognition site for a furin- the intact bioactive FGF23 molecule

Fibroblast Growth Factor 23 (FGF23) identified by reverse transcriptase (RT)-PCR in the


and Other Proteins with Phosphaturic heart, liver, thymus, small intestine, and brain [38,
Properties 43]. The most abundant expression of FGF23
occurs in the bone cells, particularly in osteocytes,
Fibroblast growth factor 23 (FGF23), which is which represent the largest population of bone cells
probably the most important phosphate-regulating [44–47]. Its regulation remains incompletely under-
hormone, belongs to a large family of structurally stood, but mice that are null for DMP1, ENPP1, or
related proteins. FGF23 was discovered through Fam20C show an increased expression of FGF23,
two independent approaches, namely, positional suggesting that each of these proteins are negative
cloning to define the molecular cause of autoso- regulators of FGF23 expression and/or activity
mal dominant hypophosphatemic rickets (ADHR) [48–50]. Consistent with these findings, humans
[38] and sequence analysis of cDNAs derived with inactivating mutations in any of these three
from rare mesenchymal tumors that cause tumor- genes show increased urinary phosphate excretion
induced osteomalacia (TIO) [6, 39]. The FGF23 leading to hypophosphatemia [48, 51–57].
gene consists of 3 exons that encode a 251-amino In the presence of αKlotho, a protein associ-
acid precursor protein comprising a hydrophobic ated with longevity [58, 59], FGF23 binds with
leader sequence (residues 1–24), thus allowing its high affinity to FGFR1 [59, 60] (Fig. 3). Consis-
secretion into the blood circulation (Fig. 2). The tent with a role of Klotho in the regulation of
Golgi casein kinase Fam20C phosphorylates phosphate homeostasis, mice that are “null” for
Ser180 of FGF23, which reduces O-glycosylation Klotho develop severe hyperphosphatemia due to
of Thr178 by the polypeptide N-acetylgalactosami- diminished urinary phosphate excretion. These
nyltransferase 3 (GALNT3), thus making the hor- animals furthermore show elevated 1, 25(OH)2D
mone prone to proteolytic cleavage between levels, i.e., findings that are similar to those
Arg179 and Ser180 by the subtilisin-like proprotein observed in the FGF23-null mice [4, 45,
convertase SPC2 [40, 41]. Only full-length 46]. Klotho-null mice have dramatically elevated
FGF23 has phosphaturic activity [7], yet FGF23 levels [60, 61].
C-terminal fragments appear to have some biolog- FGF23-null mice (FGF23/) develop
ical activity since they can antagonize the actions hyperphosphatemia and elevated serum 1,25
of the intact hormone [42] (Fig. 2). (OH)2D concentration, and they die prematurely,
FGF23 is most closely related to fibroblast secondary to renal failure because of glomerular
growth factors 21 and 19, but shows limited homol- capillary calcifications [45, 62, 63]. FGF23/
ogy also with other fibroblast growth factors [38, animals furthermore show reduced bone turnover,
43]. FGF23 mRNA could not be detected by North- an unexpected increase in osteoid, and diminished
ern blot analysis in normal tissues, but it has been osteoblast and osteoclast number and activity.
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 295

sFRP4
FGF23 FGF7
PTH

Klotho

Other R?
FGFR1c (co-receptor)
FGFR1c

Second messengers
Inhibition Stimulation of
??
of 1 α- 1α-hydroxylase
hydroxylase

NaPi-IIa NaPi-IIc NaPi-IIa NaPi-IIc

Fig. 3 Effect of FGF23 and PTH on the proximal tubular expression of NaPi-IIa and NaPi-IIc, whereas it simulates
cells. (a) FGF23 binds to the FGFR1c and Klotho complex 1α-hydroxylase activity. Thus, FGF23 only acts as stimu-
and inhibits 1α-hydroxylase. At the same time, this cascade late phosphaturia, and PTH acts both P and Ca homeostasis
inhibits the apical expression of NaPi-IIa and NaPi-IIc. (b) simultaneously
PTH binds to the PTHR(GPCR) and inhibits the

Animals overexpressing FGF23 transgenically several additional laboratory parameters is


show increased unmineralized osteoid because required for diagnostic work-up (Fig. 4).
of increased urinary phosphate excretion leading
to hypophosphatemia as well as significant wid-
ening of growth plates leading to deformities of Parathyroid Tumors
weight-bearing bones [45, 64].
In addition to FGF23, other cDNAs were over- Increased parathyroid gland activity can be seen in
represented in libraries derived from tumors that patients with multiple gland hyperplasia, adenomas,
cause oncogenic osteomalacia [6, 65, 66]. These and, rarely, parathyroid carcinoma. Two principal
included DMP1, MEPE, sFRP4, and FGF-7 and defects can lead to the development of parathyroid
were implicated in phosphate handling in vivo tumors: (1) a heterozygous mutation that enhances
and/or in vitro, suggesting that “phosphatonins” the activity of a gene (gain-of-function mutation),
other than FGF23 may be involved in the renal which is therefore referred to as a proto-oncogene,
regulation of phosphate homeostasis [66–68]. or (2) homozygous loss-of-function mutation in a
Their role needs to be elucidated more as the abla- tumor-suppressor gene, which is therefore referred
tion of sFRP4 in mice at least does not affect to as a recessive oncogene. Parathyroid tumors usu-
phosphate homeostasis [69]. ally occur as an isolated and sporadic
endocrinopathy or as part of inherited tumor syn-
dromes [70] such as the multiple endocrine neopla-
sias (MEN) or hereditary hyperparathyroidism with
Hypercalcemia and jaw tumors [71, 72]. Sporadic parathyroid tumors
Hypophosphatemia Due to Increased can be caused by single somatic mutations that lead
Parathyroid Hormone Secretion to the activation or overexpression of proto-
oncogenes, such as PRAD1 ( parathyroid adenoma
Hypercalcemia can be observed in several differ- 1) or RET (mutated in MEN2), or by mutations in
ent nonfamilial (sporadic) or familial disorders. tumor-suppressor genes – predicted to be located on
Besides physical examination and a careful several different chromosomes, for example, chro-
review of the family history, the evaluation of mosome 1p (RIZ1) and 11q3 (MEN1) – that allow
296 A. Sharma et al.

Hypercalcemia

Serum PTH↑ Serum PTH nl-↓

sP↓, uCa↑, ↓TRP sP↑, ↑TRP


sP↓-nl, uCa↑ sP↓-nl, uCa↓

Williams Syndrome Familial Benign Hypercalcemia (FBH)


Primary hyperparathyroidism CKD? McCune-Albright Syndrome FBH1: loss-of-function mutation in CaSR
Jansen’s disease: PTH/PTHrP FBH2: gain-of-function mutation in Gα11
receptor activation FBH3: activating mutation in AP2σ2
Sporadic (these three are associated with Autoimmune Hypocalciuric Hypercalcemia (AHH)
Familial developmental abnormalities)
Adenoma
Carcinoma Multiple endocrine neoplasia Type I Infantile hypercalcemia
Multiple endocrine neoplasia Type II
Hereditary hyperparathyroidism with jaw tumors
Neonatal severe hyperparathyroidism ( NSHPT)

Fig. 4 Flow diagram for the work-up of patients with hypercalcemia

for the clonal expansion of a single parathyroid cell presented with hypercalcemia and
and its progeny (see Ref. [73] for a detailed review hypophosphatemia, yet undetectable levels of
of this topic in adults). In children, parathyroid PTH in the circulation, was reported. A single
tumors are rare, with adenomas being the most parathyroid adenoma was identified that secreted
common abnormality [74]. a mutant PTH molecule, which was truncated
after amino acid residue 52, thus explaining why
different two-site immunometric PTH assays had
Hyperparathyroidism in Chronic been unable to detect elevated circulating levels of
Kidney Disease this hormone [85]. After surgical removal of the
adenoma, clinical symptoms and biochemical
Chronic overstimulation of the parathyroid glands abnormalities resolved.
frequently occurs in patients with chronic kidney
disease (CKD), which may lead to the development
of hyperplasia transitioning to adenoma (tertiary Hypercalcemic Disorders Without
hyperparathyroidism) due to the clonal expansion Elevated Parathyroid Hormone
of one or several parathyroid cells [75], through a Secretion
process possibly involving the reduced expression
of different cyclin-dependent kinase inhibitors [76] Disorders of the Calcium-Sensing
and an abnormal WNT β-catenin signaling path- Receptor (CaSR) and the Downstream
way [77]. Increased levels of biologically active Signaling Molecules
circulating FGF23 [78–81] and decreased levels
of 1,25(OH)2D [5, 82–84] contribute to the devel- CaSR is a G protein-coupled receptor (GPCR)
opment of hyperparathyroidism. located on chromosome 3q21.1 [86] that couples
through Gαq and Gα11 to the Ca2+/IP3/PKC sig-
naling pathway. Three hypocalciuric hypercalce-
Non-syndromic Isolated mic disorders are caused by abnormalities in
Hyperparathyroidism calcium-sensing receptor (CaSR) function:
(1) familial benign hypercalcemia (FBH), also
PTH structural changes as the cause of hyperpara- referred to as familial hypocalciuric hypercalce-
thyroidism appear to be rare. One patient, who mia (FHH), (2) neonatal severe
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 297

hyperparathyroidism (NSHPT), and (3) autoim- compounded with a mutation (N34S) in the pan-
mune hypocalciuric hypercalcemia (AHH) caused creatic secretory trypsin inhibitor gene
by acquired autoantibodies against the CaSR. (SPINK1) [104].

Familial Hypocalciuric Hypercalcemia Neonatal Severe Hyperparathyroidism


(FHH): FHH1 (CaSR), FHH2 (Ga11), (NSHPT)
FHH3 (AP2s1) Patients with severe neonatal hyperparathyroid-
FHH is a genetically heterogeneous disorder ism usually have homozygous or compound het-
which may be inherited as an autosomal dominant erozygous CaSR mutations that are both
trait, although patients often have no family his- inactivating [87, 88, 90, 105–107]. Infants with
tory as they may have developed a de novo muta- NSHPT usually exhibit severe bone disease
tion. Affected patients are usually asymptomatic driven by markedly increased PTH, leading to
or have nonspecific symptoms such as fatigue, significant mortality, if left untreated [108,
weakness, painful joints, and headache, and the 109]. Some patients with sporadic neonatal hyper-
diagnosis is often only suspected after routine parathyroidism have been reported to carry de
biochemical screening shows elevated serum cal- novo heterozygous CaSR mutations [89], thereby
cium levels. suggesting the involvement of factors other than
Three variants of FHH have been defined at the the dosage of the mutant gene or dominant-
molecular level; these are (1) loss-of-function negative actions on the wild-type CaSR [105]. A
mutations in CaSR (FHH1) [87–92]; (2) loss-of- novel heterozygous de novo mutation (R551K)
function in Gα11, the signaling protein at the was shown recently to cause NSHPT, which grad-
CaSR (FHH2) [93, 94]; and (3) loss-of-function ually reverted to asymptomatic FBH without the
mutations in AP2σ1 (adapter protein-2 sigma need for surgical intervention [110].
subunit), a central component of clathrin-coated
vesicles (FHH 3) [95]. FHH2 and FHH3 families Autoimmune Hypocalciuric
were initially linked genetically to chromosome Hypercalcemia (AHH)
19p (FHH19p) and 19q13 [96] (FHHOK), respec- AHH is an acquired disorder with circulating anti-
tively [97, 98]. Heterozygous loss-of-function bodies directed against the extracellular domain of
CaSR mutations were found in approximately the CaSR. Some of these antibodies stimulate the
two-thirds of the affected members of investigated release of PTH when tested with dispersed human
FHH kindreds (FHH1) [87, 92, 99, 100]. CaSR parathyroid cells in vitro, probably by reducing the
mutations cluster around low-affinity calcium- activation of the CaSR by extracellular calcium
binding sites (that are similar to calsequestrin) [111]. AHH diagnosis should be considered for
containing aspartate- and glutamate-rich regions patients who have hyperparathyroidism in combi-
(codons 39–300) within the extracellular domain nation with other autoimmune disorders [111, 112]
of the receptor [101, 102] and show gene-dosage and lack mutations associated with FHH. AHH was
effect [87–92, 102]; for details of CaSR muta- initially described in four individuals from two
tions, visit http://www.casrdb.mcgill.ca. Another unrelated kindreds. Among them, three patients
heterozygous mutation in exon 4 of CaSR gene had antithyroid antibodies, and one had celiac
(F180C, TTC > TGC) leads to FHH phenotype sprue with antigliadin and anti-endomyseal anti-
only in vitamin D deficiency state. Vitamin D bodies [111]. Another patient described had an
deficiency probably impaired CaSR expression, IgG4 blocking autoantibody against CaSR.
thus leading to increased PTH secretion; consis- Regression of biochemical abnormalities and auto-
tent with this conclusion, vitamin D supplemen- immune dysregulation followed glucocorticoid
tation resulted in the normalization of PTH levels treatment [112]. An autoantibody isolated from a
[103]. Individuals in FHH families carrying the patient with AHH caused distinct conformational
CaSR mutation L137P have shown increased sus- changes in CaSR favoring coupling to Gα11 and
ceptibility for chronic pancreatitis, when uncoupling from Gi. These allosteric changes
298 A. Sharma et al.

codon 223 (His ! Arg), codon 410 (Thr ! Pro


or Arg), and codon 458 (Ile ! Arg or Lys). The
expression of these mutant receptors in suitable
cells resulted in constitutive agonist-independent
accumulation of cAMP, while the basal accumula-
tion of inositol phosphates was not measurably
increased. The H223R mutation is the most fre-
quent cause of JMC, while all other mutations were
found in only single patients [116–122]. Trans-
genic mice, in which the expression of a
PTH/PTHrP receptor carrying the H223R mutation
was targeted to the growth plate by the rat α1
(II) collagen promoter, showed a significant delay
in chondrocyte differentiation, supporting the con-
clusion that the defect in endochondral bone for-
mation in JMC patients is caused by the
constitutively active mutant receptor [123]. The
slowed differentiation of growth plate
chondrocytes was associated with an upregulation
Fig. 5 Patient with a severe form of Jansen’s metaphyseal of cyclin- and E2F-dependent gene expression,
chondrodysplasia. (From [114], with permission) indicating that the PTH/PTHrP receptor controls
the timing of cell cycle exit and the onset of differ-
occurred at a site in close proximity to the binding entiation of chondrocytes [124]. The Thr410Arg
site for calcimimetics [113]. mutation in PTH/PTHrP receptor leads to less pro-
nounced skeletal and laboratory abnormalities, i.e.,
only mild skeletal dysplasia and relatively normal
Jansen’s Metaphyseal stature and high-normal plasma calcium concentra-
Chondrodysplasia (JMC) tion associated with normal or suppressed serum
PTH levels and with hypercalciuria leading to
JMC is a rare autosomal dominant disease that is nephrolithiasis in two individuals [125]. In com-
characterized by short-limbed dwarfism caused by parison to PTH/PTHrP receptors with the
an abnormal regulation of chondrocyte prolifera- Thr410Pro mutation, the Thr410Arg mutation
tion and differentiation in the metaphyseal growth showed less pronounced agonist-independent
plate [114] (Figs. 5 and 6) that is usually associated cAMP accumulation in vitro [117, 126].
with severe hypercalcemia and hypophosphatemia,
despite normal or undetectable serum levels of
PTH or PTHrP [115]. JMC is caused by heterozy- Williams Syndrome
gous mutations in the PTH/PTHrP receptor that
lead to constitutive PTH- and PTHrP-independent Williams syndrome (WS) is a hemizygous contin-
receptor activation [116–118]. Since the uous deletion syndrome caused by the deletion of
PTH/PTHrP receptor is most abundantly expressed 25–30 genes on chromosome 7 q11.23. WS is
in the kidney, bone, and growth plates, these find- characterized by vascular stenosis, hypertension,
ings provided a likely explanation for the abnor- elfin-like facies, developmental delay, and typical
malities observed in mineral homeostasis and for behavioral features. Endocrine abnormalities
the associated defects in chondrocyte growth and including hypercalcemia, diabetes mellitus, and
differentiation. Several different heterozygous subclinical hypothyroidism might be present.
mutations in the PTH/PTHrP receptor have been About 20 % of affected children may present
identified in the severe form of JMC; these involve with congenital anomalies of the kidney and
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 299

Fig. 6 Radio- and photographs of patients with a relatively mild form of Jansen’s disease (From [125] with permission)

urinary tract, hypercalciuria, or dysfunctional an important diagnostic tool. Other deleted pro-
voiding [127]. teins, like LIM-kinase [131], the cytoplasmic
Of the deleted genes, elastin is the most impor- linker protein-115 (CYLN2), and the transcription
tant. The ablation of elastin gene in mice results in factors GTF2I and GTF2IRD1 [132], may con-
vascular abnormalities similar to those observed tribute to some of the distinct neurological and
in WS patients [128]. Homozygous deletion at the cognitive deficits. In contrast, NCF1 deletions are
elastin locus was found in over 90 % of patients protective against hypertension [133]. In addition,
with the classical Williams phenotype [129], and a genes flanking aneuploid genes, some of which
series of 235 WS patients revealed submicro- are located several megabases away from the
scopic deletions of the elastin gene in 96 % of deletion, may contribute to the observed pheno-
the investigated individuals [130], thus making it typic variation [134, 135].
300 A. Sharma et al.

The etiology of hypercalcemia is less clear. develop often severe hypercalcemia and
Although it is clinically not severe in most cases, hypercalciuria leading to nephrocalcinosis and
some affected individuals require therapeutic failure to thrive. Besides elevated plasma and
interventions with bisphosphonates [136]. The urinary calcium levels, 1,25(OH)2D levels are
calcitonin receptor gene, located on chromosome increased, which was shown in one child to dra-
7q21, is not included in the deletions identified in matically enhance intestinal calcium absorption
WS [137]. Increased calcium retention, sensitiv- [143], thereby suppressing circulating PTH con-
ity, or abnormal synthesis or degradation of 1,25 centrations through two mechanisms, i.e., ele-
(OH)2 vitamin D has been postulated to be causal vated plasma calcium and 1,25(OH)2 vitamin D
[138, 139]. Claudin 3 and 4 though deleted were concentration.
not found to be causative for hypercalcemia
[140]. The transient receptor potential cation
channel 3 (TRPC3) overexpressed in the intes- Hypocalcemia and
tines and kidneys of a WS patient has been Hyperphosphatemia Due to Reduced
shown to increase calcium absorption in the gut Parathyroid Hormone Activity
and reabsorption in the kidneys [141].
As for hypercalcemic disorders, patients who
present with hypocalcemia require careful
Infantile Hypercalcemia: CYP24A1 clinical evaluation, as well as the assessment of
Deficiency laboratory parameters in serum and urine, and a
detailed review of the family history [147]
Initially referred to as idiopathic infantile hyper- (Fig. 7).
calcemia (IIH), this disorder was shown to be
caused by homozygous or compound heterozy-
gous mutations in the gene encoding the Hypoparathyroidism
24-hydroxylase (CYP24A1), the enzyme that
metabolizes and thus inactivates the biologically Hypoparathyroidism comprises a heterogeneous
active 1,25(OH)2D [142–146]. Affected infants group of disorders presenting with hypocalcemia

Hypocalcemia

Serum PTH nl-↓ (sP↑; uCa nl-↑) Serum PTH ↑

Hypoparathyroidism
sP↓; uCa nl-↓ sP↑; uCa↓

Isolated Forms Complex syndromic forms


autosomal dominant DiGeorge syndrome ( TBX1) Vitamin D deficiency Pseudohypoparathyroidism (PHP)
autosomal recessive HDR (GATA3) (25 (OH ) ↓;1,25(OH) NL-↑)
X-linked Kenny-Caffey/Sanjad-Sakati syndrome ( TBCE; FAM111A)
Autoimmune polyglandular syndrome
VDDR type I
CaSR abnormalities
Genetic mutations in: (25 (OH ) NL;1,25(OH) ↓) Without AHO With AHO
autosomal dominant hypocalcemic hypercalciuria ( ADHH)
PTH PHP type Ib PHP type 1a
Bartter syndrome Type V
GCMB autoimmune acquired hypoparathyroidism (AH) VDDR type II PHP type 1c
PHP type II
GNA11 Mitochondrial disorders associated with hypoparathyroidism (25 (OH ) NL;1,25(OH) ↑)
Rare associations
congenital lymphoedema
Barakat syndrome

Fig. 7 Flow diagram for the work-up of patients with hypocalcemia


11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 301

of varying severity, inappropriately normal or Several different PTH gene mutations were
elevated urinary calcium excretion, and identified in patients with autosomal dominant or
hyperphosphatemia, yet normal or diminished autosomal recessive isolated hypoparathyroidism.
PTH levels. Hypoparathyroidism may be A single-base substitution (T ! C) resulted in the
nonfamilial (sporadic) or familial, and it may substitution of arginine (CGT) at position 18 in
occur as an isolated defect or as a component of the signal peptide for cysteine (TGT), thus
disorders with additional manifestations, such as disrupting the hydrophobic core of the signal
pluriglandular autoimmune disorder or various sequence, which is required by the secreted pro-
developmental abnormalities, e.g., DiGeorge syn- teins for efficient translocation across the endo-
drome (DGS). Familial isolated hypoparathyroid- plasmic reticulum [158]. Another single-point
ism can present anytime from early infancy well mutation changes cysteine to arginine at the
into adulthood. 8 position of the signal peptide, which causes
The main treatment options available for interference with the normal targeting and
patients with acute or chronic hypoparathyroid- processing of secretory proteins, including the
ism are calcium salts, vitamin D or vitamin D normal PTH precursor, suggesting that the mutant
analogues, and drugs that increase renal tubular gene product exerts a dominant-negative effect
reabsorption of calcium (i.e., thiazides). The para- in vitro by trapping the hormone intracellularly,
thyroid hormone-dependent renal production of predominantly in endoplasmic reticulum
1,25-dihydroxyvitamin D is deficient in all (ER) [159], thereby causing stress-induced cell
hypoparathyroid states. Consequently, therapy death [160]. In another family, a single-base sub-
with a vitamin D analogue is used to improve stitution (T ! C) involving codon 23 of exon
serum calcium levels. However, because of the 2 was detected, which resulted in the substitution
absence of functional PTH and the lack of of proline (CCG) for the normal serine (TCG) in
PTH-dependent calcium reabsorption in the distal the signal peptide [161]. This mutation at the 3
renal tubules, patients affected by hypoparathy- position of the preproPTH protein cleavage site
roidism frequently develop hypercalciuria and most likely disrupts cleavage of the mutant pre-
nephrolithiasis, and chronic kidney disease, even cursor molecule and prevents the efficient forma-
if attempts are made to maintain plasma calcium tion and secretion of PTH [161]. The affected
within the lower end of the normal range individuals of one other kindred with autosomal
[148]. Considerable efforts were therefore made recessive isolated hypoparathyroidism showed a
to treat patients with recombinant PTH(1-34) or single-base transition (G ! C) at position 1 of
PTH(1-84) [149–155]. Compared with twice- intron 2 of the gene encoding PTH. This mutation
daily delivery, pump delivery of PTH(1-34) resulted in the deletion of exon 2, which encodes
appears to provide the most promising modality the initiation codon and the signal peptide,
for improving calcium homeostasis and bone thereby causing parathyroid hormone deficiency
turnover in children with severe congenital hypo- [162]. A nonsense mutation involving codon
parathyroidism [156, 157]. 23 (Ser23Stop) was recently reported in a girl
with autosomal recessive isolated hypoparathy-
Parathyroid Hormone (PTH) Gene roidism [163], and a heterozygous T to C point
Abnormalities mutation was identified that eliminates the initia-
Preproparathyroid hormone (preproPTH), the tor methionine, thereby deleting the first six amino
PTH precursor, contains a 25-residue amino-ter- acids of the signal peptide [164].
minal signal sequence followed by a 6-residue
pro-specific peptide and the mature hormone. GCM2 Abnormalities
The hydrophobic core of the human preproPTH Glial cells missing 2 (GCM2), the mammalian
signal peptide is composed of 12 contiguous homolog of the Drosophila Gcm2 gene, encodes
uncharged amino acids (residues 5 to 16 of a 506-amino acid parathyroid-specific transcrip-
the signal peptide). tion factor, which contributes to the regulation of
302 A. Sharma et al.

PTH gene expression [165]. Mice that are homo- established by demonstrating an identical mito-
zygous for the deletion of GCM2 (the murine chondrial DNA sequence, inherited via the mater-
homolog) lack parathyroid glands, and these ani- nal lineage, in affected males from the two
mals develop hypocalcemia and hyperpho- families [178]. Affected males suffered from
sphatemia without a compensatory increase in infantile onset of epilepsy and hypocalcemia
PTHrP or 1,25(OH)2 vitamin D levels [166]. [179]. Studies utilizing X-linked polymorphic
GCM2-null mice revealed a small cluster of markers in these families localized the mutant
PTH-expressing cells under the thymic capsule; gene to chromosome Xq26-q27 [180].
however, the amount of PTH was insufficient to Characterization of 906 kb region on Xq27 by
prevent hypocalcemia [167]. combined analysis of single nucleotide polymor-
The gene encoding GCM2 is located on chromo- phisms and sequence-tagged site identified a
some 6p23–24, and isolated hypoparathyroidism 23–25 kb deletion, which did not contain genes.
can be caused by inactivating mutations on both However, DNA fiber-FISH and pulsed-field gel
parental alleles or by a dominant-negative heterozy- electrophoresis revealed an approximately 340 kb
gous mutation. The first GCM2 mutation, a large insertion that replaced the deleted fragment and a
homozygous intragenic deletion, was identified in molecular deletional-insertion that involved chro-
the proband of an extended kindred with an autoso- mosome 2p25 and Xq27 [181]. This complex
mal recessive form of isolated hypoparathyroidism deletion-insertion {del(X) (q27.1) inv ins (X;2)
[168]. Subsequently, homozygous mutations were (q27.1; p25.3)} is located 67 kb downstream of
identified as additional causes of hypoparathyroid- the SOX3 gene. The developing parathyroid tis-
ism; these affect amino acid residues 63 (G63S) sue of mouse embryos has strong SOX expres-
[169] or 47 (R47L) [170], as well as several other sion. Thus, this mutation may have a position
residues [171–174]. Two closely related heterozy- effect on SOX3 expression. These findings
gous GCM2 mutations were furthermore identified added SOX3 to the growing list of transcription
in two families in which hypoparathyroidism fol- factors, including GCM2, GATA3, TBX1,
lows an autosomal dominant mode of inheritance HOXA3, PAX1, and PAX9, that operate in para-
[175, 176]. Both mutations lead to a shift in the open thyroid development [181].
reading frame and the replacement of the putative
transactivation domain within the carboxyl-terminal Autoimmune Pluriglandular
region by unrelated amino acid sequence, and both Hypoparathyroidism
mutant proteins have a dominant-negative effect Autoimmune pluriglandular syndromes associ-
on the wild-type GCM2 protein. A similar mecha- ated with hypoparathyroidism can be subdivided
nism may apply to the Asn502His mutation, which into three subtypes: (1) APS1, (2) APS3 (which is
localizes to the nucleus and retains the ability to associated with thyroid autoimmunity), and
bind to the GCM-consensus DNA recognition (3) APS4 (which is associated with other autoim-
motif but shows impaired gene transactivation and mune disorders).
a dominant-negative effect on the wild-type APS1 or autoimmune polyendocrinopathy-
protein [173]. For a large number of patients with candidiasis-ectodermal dystrophy (APECED) is
isolated hypoparathyroidism, however, no disease- an autosomal recessive disease with a high inci-
causing mutation has yet been identified [171], dence in isolated subpopulations in Central and
making it likely that mutations in additional Eastern Finland [182], which usually becomes
as-of-yet unidentified genes can also cause manifest during childhood. Establishing the diag-
hypoparathyroidism. nosis requires the presence of at least two of three
major components: hypoparathyroidism, candidi-
X-Linked Recessive Hypoparathyroidism asis, and adrenal insufficiency; however, hypo-
X-linked recessive hypoparathyroidism has been parathyroidism may be the only manifestation of
reported in two related multigenerational kindreds the syndrome. APS1 is caused by mutations
[177]. The relatedness of these two kindreds was within the autoimmune regulator (AIRE) gene
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 303

located on chromosome 21 (21q22). As part of Thus, the AIRE-mediated dominant-negative


this syndrome, autoantibodies against NACHT effect may cause autoimmune predisposition to
leucine-rich repeat protein 5 (NALP5Abs) can phenotypes distinct from APS [196]. In addition,
be detected. Non-APS forms of hypoparathyroid- there is a downstream abnormality as activated
ism show an association with class I human leu- CD4+ T cells of APECED demonstrate a selective
kocyte antigen (HLA) allele A*2601 and with IL-22 defect [197].
class II HLA alleles DRB1*01 and APS1 patients with hypoparathyroidism can
DRB1*09 [183]. have antibodies directed against the CaSR,
Linkage studies of Finnish families mapped which leads to a decrease in PTH secretion and
the APECED gene to chromosome 21q22.3 thus hypocalcemia [198]. The NACHT leucine-
[184]. Further, positional cloning approaches led rich repeat protein 5 (NALP5), which is predom-
to the isolation AIRE (autoimmune regulator), inantly expressed in the cytoplasm of parathyroid
which encodes a 545-amino acid protein. Specific chief cells, is another antigen, autoantibodies
domains of AIRE protein indicate that it is against which can be responsible for some forms
involved in transcriptional processes, including of hypoparathyroidism. NALP5-specific autoan-
(i) the amino-terminal HSR domain, (ii) nuclear tibodies were detected in almost half of the
localization signal (NLS), (iii) a SAND domain, patients with APS1 and hypoparathyroidism, but
(iv) two plant homeodomain (PHD) type zinc were absent in all APS1 patients without hypo-
fingers, and (v) four LXXLL motifs [185]. To parathyroidism [199]. However, the pathological
date, more than 50 different APS1-causing muta- significance of these antibodies has been
tions have been established in affected patients. questioned [200]. Instead of seeking AIRE muta-
Though mutations are distributed throughout the tions, which is a cumbersome process,
coding region of the gene, most mutations are IgG-neutralizing autoantibodies against type I
located in the amino-terminal HSR domain. The interferon can be used as a diagnostic criterion
R257X mutation in SAND domain is the most [201] as high titers of these antibodies were
prevalent mutation among Finnish patients, found in 100 % of Finnish and Norwegian patients
accounting for 83 % of the disease alleles [185, with two prevalent AIRE truncations [202]. Fur-
186]. This particular mutation is also frequently thermore, rising titers of anti-cytokine antibodies
found in Central and Eastern European and North- may precede clinical presentation [203].
ern Italian populations, indicating an early intro-
duction of the mutation into Caucasian DiGeorge Syndrome
populations [186, 187]. Characteristic mutations DiGeorge syndrome (DGS) is associated with the
have been reported also for various other ethnic- absence or hypoplasia of the thymus and the para-
ities [188–192]. Recently, a novel mechanism of thyroid glands, cardiovascular anomalies, and cra-
AIRE dysregulation has been described in which niofacial dysmorphism [204]. Most DGS cases
miR-220b inhibits AIRE gene translation through that result from the deletion of the 250–3,000 kb
the 3’UTR [193]. critical region that contains approximately
AIRE has been shown to regulate the elimina- 30 genes [205, 206] are designated as DGS type
tion of organ-specific T cells in the thymus and 1 (DGS1) [207]. DGS2 is associated with the
thus failure to delete forbidden T cells [194] and to deletion locus on chromosome 10p [208]. Approx-
disrupt of the nuclear organization [195], thereby imately 17 % of patients with the phenotypic
resulting in failure to establish immunologic tol- features of DGS have no detectable genomic
erance. A unique G228W variant in the SAND deletion [209].
domain appears to inhibit wild-type AIRE from Hypocalcemia, considered one of the cardinal
reaching the sites of active transcription in med- features of DGS, is invariably due to hypopara-
ullary thymic epithelial cells. This resulted in the thyroidism as originally described by DiGeorge in
failure to delete T cells reactive against antigens 1965 and documented by aplasia or hypoplasia of
specific for the thymus, leading to autoimmunity. parathyroid glands at surgery or autopsy [210].
304 A. Sharma et al.

Hypocalcemia was noticed in 203/340 subjects bilateral, symmetrical, nonprogressive sensori-


from a large European cohort; the majority of neural deafness involving all frequencies. The
affected individuals were found to have transient renal abnormalities consisted mainly of bilateral
hypocalcemia [211]. Similar transient hypocalce- cysts that compressed the glomeruli and tubules,
mia occurs at birth due to the abrupt cessation of leading to renal impairment in some patients.
active maternal calcium supply in combination Cytogenetic abnormalities were not detected and
with birth stress. Increased demands on parathy- abnormalities of the PTH gene were excluded.
roid reserve resolve as stress dissipates. Symp- Deletion mapping studies in two unrelated
tomatic or latent hypocalcemia may also be HDR patients showed cytogenic abnormalities
precipitated by the stress of surgery, pregnancy, involving chromosome 10p14-10pter [229].
or puberty [212]. Point prevalence of hypocalce- These two patients suffered from hypoparathy-
mia outside the neonatal period was 30 % in roidism, deafness, and growth and mental retarda-
27 subjects of all ages [213]. EDTA infusion can tion; one patient also had a solitary dysplastic
be used to unmask latent hypocalcemia [214]. kidney with vesicoureteric reflux and a uterus
Treatment is aimed at keeping serum calcium bicornuis unicollis, and the other HDR patient
in low-normal range since the absence of PTH also had cartilaginous exostoses and was shown
leads to high or inappropriately normal urinary to have a complex reciprocal insertional translo-
excretion of calcium, which increases the risk of cation of chromosomes 10p and 8q. Neither of
renal calcification and nephrolithiasis. these patients had immunodeficiency or heart
The deleted region in DGS1 contains several defects, which are key features of DGS2 (see
genes [207, 215–217]. The ablation of several of above), and further studies defined two
these genes causes developmental abnormalities nonoverlapping regions; thus, the DGS2 region
of the pharyngeal arches in transgenic mice was located on 10p13-14 and HDR on 10p14-
[218–220]. Tbx1-ablated transgenic mice [220] 10pter. It is important to note that HDR patients
and humans with point mutations of TBX1 have with GATA3 haploinsufficiency do not have
the DGS1 phenotype [221]; mutations in this gene immune deficiency. Similarly, the facial
are therefore likely to cause DGS1 [222]. TBX1 is dysmorphism, growth, and developmental delay,
a DNA-binding transcriptional factor of the T-Box commonly seen in patients with larger 10p dele-
family with an important role in organogenesis tions, were absent in the HDR patients carrying
and pattern formation. Tbx1-null mutant mice GATA3 mutations, further indicating that these
(Tbx1/) had all the developmental anomalies features were likely due to other genes on 10p
of DGS1, while haploinsufficiency (Tbx1+/) was [230]. Deletion mapping studies in two other
associated with a milder phenotype. The spectrum HDR patients further defined a critical 200 kb
of DGS1 malformations is thus elicited in a dose- region that contained GATA3 [230], which
dependent manner [223], suggesting that the Tbx1 belongs to a family of zinc finger transcription
dose can be modulated by other modifying genes, factors that bind to the consensus DNA sequence
thus causing phenotypic variability [224–227]. and are involved in vertebrae embryonic develop-
ment. DNA sequence analysis in other HDR
Hypoparathyroidism, Deafness, patients identified mutations that resulted in a
and Renal Anomalies (HDR) Syndrome haploinsufficiency and loss of GATA3 function
The combined occurrence of hypoparathyroidism, [230–232]. GATA3 has 2 zinc fingers. The
deafness, and renal dysplasia (HDR) as an auto- C-terminal finger (ZnF2) binds DNA, while the
somal dominant trait was reported in one family in N-terminal finger (ZnF1) stabilizes this DNA
1992 [228]. Patients had asymptomatic hypocal- binding and interacts with other zinc finger pro-
cemia with undetectable or inappropriately nor- teins, such as the Friends of GATA (FOG)
mal serum concentrations of PTH and normal [233]. HDR-associated mutations involving
brisk increases in plasma cAMP in response to GATA3 ZnF2 or the adjacent basic amino acids
the infusion of PTH. The patients also had were found to result in a loss of DNA binding,
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 305

while those involving ZnF1 either lead to a loss of (2) MELAS syndrome, and (3) mitochondrial
interaction with FOG2 ZnFs or altered trifunctional protein deficiency syndrome
DNA-binding affinity [232]. These findings are (MTPDS). Both the KSS and MELAS syndromes
consistent with the proposed 3-dimensional have been reported to occur with insulin-
model of GATA3 ZnF1, which has separate dependent diabetes mellitus and hypoparathyroid-
DNA and protein binding surfaces [232, 234]. ism [242, 243]. A point mutation in the mitochon-
Electrophoretic mobility shift assays (EMSAs) drial gene tRNA leucine (UUR) has been reported
revealed three classes of GATA3 mutations: in one patient with the MELAS syndrome who
those that lead to a loss of DNA binding (over also suffered from hypoparathyroidism and dia-
90 % of all mutations), those that lead to a loss of betes mellitus [101]. Large deletions, consisting
the carboxyl-terminal zinc finger and result in of 6,741 and 6,903 base pairs and involving
reduced DNA-binding affinity, and those (e.g., >38 % of the mitochondrial genome, have been
Leu348Arg) that do not alter DNA binding or reported in other patients who suffered from
the affinity but likely induce conformational hypoparathyroidism and sensorineural deafness
change in GATA3 [235]. Furthermore, a hetero- [244]. Rearrangements and duplication of mito-
zygous mutation, 432insG, has been described, chondrial DNA have also been reported in KSS.
which introduces a premature stop codon at exon MTPDS is a disorder of fatty acid oxidation that is
4 (K302X), resulting in a loss of both zinc finger associated with peripheral neuropathy, pigmen-
domains of the GATA3 protein [236]. No muta- tary retinopathy, and acute fatty liver degeneration
tions were identified in patients with isolated in pregnant women who carry an affected fetus.
hypoparathyroidism, indicating that GATA3 Hypoparathyroidism has been observed with
abnormalities are more likely to result in two or trifunctional protein deficiency [245, 246]. The
more of the phenotypic features of the HDR syn- role of these mitochondrial mutations in the etiol-
drome and not in isolated disease affecting only ogy of hypoparathyroidism remains to be further
the regulation of calcium homeostasis [235]. The elucidated.
co-occurrence of the 22q11 deletion and the HDR
due to a de novo deletion in chromosome 10p14 Kenny-Caffey and Sanjad-Sakati
has been described in a Japanese boy with severe Syndrome
phenotype, including progressive renal failure and Hypoparathyroidism has been reported to occur in
severe intellectual disabilities [237]. over 50 % of patients with the Kenny-Caffey syn-
The HDR phenotype is consistent with the drome (KCS), which is associated with short stat-
expression pattern of GATA3 during human and ure, osteosclerosis, and cortical thickening of the
mouse embryogenesis in the developing kidney, long bones, delayed closure of the anterior fonta-
otic vesicle, and parathyroids. Homozygous abla- nel, basal ganglia calcification, nanophthalmos,
tion of GATA3 in mice leads to CNS defects, but dental anomalies, and hyperopia [247]. Parathy-
heterozygous GATA3-null animals were initially roid tissue could not be found in a detailed
reported to have no abnormalities [238]. However, postmortem examination of one patient [248],
further studies have revealed that these heterozy- suggesting that hypoparathyroidism may be due
gous mice have a progressive hearing loss associ- to an embryological defect of parathyroid devel-
ated with continuous morphological degeneration opment. KCS, namely, KCS type 1 (KCS1), was
of the cochlea [239, 240] and that they also have convincingly demonstrated in 16 affected children
hypoparathyroidism [241]. in 6 unrelated sibships, born to healthy consan-
guineous parents of Bedouin ancestry [249].
Mitochondrial Disorders Associated In eight consanguineous Kuwaiti kindreds, link-
with Hypoparathyroidism age to a locus in the 1q42–q43 region was found
Hypoparathyroidism has been reported to occur in for the autosomal recessive form of Kenny-Caffey
three disorders associated with mitochondrial dys- syndrome [250]. This disease has been
function:(1) Kearns-Sayre syndrome (KSS), observed almost exclusively in the Middle East.
306 A. Sharma et al.

The only available data about its prevalence is located this gene to chromosome 1q42–q43 [256].
from Saudi Arabia, where its frequency is esti- Sanjad-Sakati syndrome resembles the autosomal
mated between 1:40,000 and 1:100,000 live births recessive form of KCS with similar manifesta-
[247]. An analysis of DNA from 14 children with tions but lacking osteosclerosis. Eight Sanjad-
severe short stature of unknown etiology by whole Sakati families from Saudi Arabia were
exome sequencing identified one subject with genotyped with polymorphic short tandem repeat
Kenny-Caffey syndrome, highlighting the fact markers from the SSS/KCS critical region. A
that rare syndromic causes of short stature may maximum multipoint LOD score of 14.32 was
be under-recognized and underdiagnosed [251]. obtained at marker D1S2649, confirming linkage
KCS1 is caused by mutations of the tubulin- of Sanjad-Sakati syndrome to the same region as
specific chaperone (TBCE), whereas patients autosomal recessive Kenny-Caffey syndrome.
with KCS2 were recently shown to carry de novo Haplotype analysis refined the critical region to
mutations in FAM111A (family with sequence 2.6 cM and identified a rare haplotype present in
similarity 111A) [252]. There is close genotype- all the Sanjad-Sakati syndrome disease alleles,
phenotype correlation. indicative of a common founder. In addition to
Five individuals with KCS2 and five with more the assignment of the Sanjad-Sakati syndrome in
severe osteocraniostenosis (OCS) were found to Saudi families and of the Kenny-Caffey syndrome
have heterozygous mutations in FAM111A. This in Kuwaiti families to overlapping genetic inter-
protein comprises 611 amino acids with homol- vals, comparison of the haplotypes unexpectedly
ogy to trypsin-like peptidases. Molecular model- demonstrated that the diseases shared an identical
ing revealed that these mutations are close to the haplotype. This finding, combined with the clini-
outer surface rather than on the active site, thus cal similarity between the two syndromes, sug-
raising the possibility that decreased binding of gests that the two conditions are not only allelic
FAM111A by its inactivating partner results in but are also caused by the same ancestral mutation
increased and/or deregulated FAM111A activity, [257]. Molecular genetic investigations led to the
a gain-of-function effect observed even in a het- conclusion that mutations of the tubulin-specific
erozygous state [253]. FAM111A functions as a chaperone (TBCE) are associated with both syn-
host range restriction factor which plausibly plays dromes [258]. TBCE encodes one of several chap-
a role in embryonic morphogenesis and appears to erone proteins required for the proper folding of
be crucial for parathyroid gland formation and α-tubulin subunits and the formation of α-β tubu-
function [252]. Mother-to-daughter transmission lin heterodimers. In addition, deletion and trunca-
of KCS2 was shown to be associated with a recur- tion mutations in the gene encoding a tubulin-
rent dominant FAM111A mutation, namely, specific chaperone cofactor E (TBCE) have been
pArg569His [254]. shown to cause the hypoparathyroidism, mental
In the Sanjad-Sakati syndrome, hypoparathy- retardation, and facial dysmorphism (HRD) syn-
roidism is associated with severe prenatal and drome which is associated with extreme growth
postnatal growth failure and dysmorphic features, failure. However, cryptic translational initiation at
and this has been reported in 12 patients from each of three out-of-frame AUG codons upstream
Saudi Arabia [255]. The presenting complaint in of the genetic lesion can rescue a mutant HRD
all patients was hypocalcemic tetany or general- allele by producing a functional TBCE protein
ized convulsions, usually detected in the first few [259]. The defect in the tubulin folding and
days or weeks of life. Consanguinity was noted in assembly pathway also has grave consequences
the families of 11 of the 12 patients, the majority on growth and PMN functions [260, 261].
of which originated from the Western province of
Saudi Arabia. This syndrome, which is inherited Additional Familial Syndromes
as an autosomal recessive disorder, has also been Single familial syndromes in which hypoparathy-
identified in families of Bedouin origin, and roidism is a component have been reported. The
homozygosity and linkage disequilibrium studies inheritance of the disorder in some instances has
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 307

been established, and molecular genetic analysis after the cloning of the CaSR, investigators
of the PTH gene has revealed no abnormalities. showed linkage of ADHH to a locus on chromo-
Thus, an association of hypoparathyroidism, renal some 3q13 [267], i.e., the same locus as for the
insufficiency, and developmental delay has been gene encoding the CaSR. Shortly afterward, a
reported in one Asian family in whom autosomal heterozygous missense mutation, Q127A, was
recessive inheritance of the disorder was identified as the cause of ADHH in an unrelated
established [262]. An analysis of the PTH gene family [266]. The majority (>80 %) of CaSR
in this family revealed no abnormalities [262]. mutations that result in a functional gain are
The occurrence of hypoparathyroidism, nerve located within the extracellular domain [102,
deafness, and a steroid-resistant nephrosis leading 266–270], which is different from the findings in
to renal failure, which has been referred to as other disorders that are the result of activating
the Barakat syndrome [263], has been reported mutations in G protein-coupled receptors. A sec-
in four brothers from one family, and an associa- ond hotspot for mutations is transmembrane
tion of hypoparathyroidism with congenital domains 6 and 7 and the intervening third extra-
lymphoedema, nephropathy, mitral valve pro- cellular loop, a site which maintains the CaSR in
lapse, and brachytelephalangy has been observed an inactive conformation and also is critical for the
in two brothers from another family [264]. Molec- binding and/or action of allosteric modulators
ular genetic studies have not been reported from including calcimimetics of the phenylalkylamine
these two families. class to which Cinacalcet belongs [271]. In addi-
tion, two deletion mutations have been described.
Most ADHH patients are heterozygous for the
Abnormalities at the Calcium-Sensing activating mutation. In one family, a homozygous
Receptor (CaSR) and the Downstream mutation was described, but it was not associated
Signaling Molecules with a more severe phenotype [272], and although
there is a spectrum of phenotypic severity for a
Abnormalities at the CaSR are associated with given genotype, the symptoms present in affected
several different hypocalcemic disorders. These members of the same family tend to be similar.
include (1) autosomal dominant forms of hypo- Thus, one mutated allele may be enough to shift
calcemic hypercalciuria (ADHH), (2) Bartter syn- the set point to left, and the presence of second
drome type V (i.e., ADHH with a Bartter-like mutated allele makes no change in the phenotype
syndrome), and (3) autoimmune hypoparathy- perhaps due to the “dominant-positive” effect of
roidism (AH). the mutant receptor.
In addition to heterozygous gain-of-function
Autosomal Dominant Hypocalcemic mutations in the CaSR (ADHH1), ADHH cases
Hypercalciuria (ADHH) Due to CaSR were recently identified that are associated with
and Ga11 Mutations heterozygous mutations in GNA11 (ADHH2), the
ADHH, although rare, in index cases may com- gene encoding Gα11 [93, 273]. Gα11 is one of
prise a sizeable fraction of cases of idiopathic two heterotrimeric G proteins that couple the
hypoparathyroidism, perhaps representing as CaSR to the Ca2+/IP3/PKC signaling pathway in
many as one-third of such cases [265]. CaSR parathyroid cells [274]. Functional studies of the
mutations that result in a loss-of-function are asso- Gα11 mutation R60L revealed a significantly
ciated with familial benign (hypocalciuric) hyper- decreased EC50 and MAP kinase activity. Com-
calcemia [87–92, 102]. It was therefore postulated pared with subjects with CaSR mutations, patients
that gain-of-function mutations in CaSR lead to with GNA11 mutations lacked significant
hypocalcemia with hypercalciuria, and the inves- hypercalciuria and had normal serum magnesium
tigation of kindreds with autosomal dominant levels [274]. FHH3 is caused by loss-of-function
forms of hypocalcemia has indeed identified AP2σ1 mutations [95]; however, 19 patients
such CaSR mutations [102, 266–270]. Soon (including 6 familial cases) with ADHH, who
308 A. Sharma et al.

did not have CaSR or GNA11 mutations, were have autoantibodies to the extracellular domain
found not to have AP2σ1 mutations [275], indi- of the CaSR [111, 112, 284]. The CaSR autoanti-
cating that gain-of-function AP2σ1 mutations are bodies are not persistent [284]. The majority of the
not a cause of ADHH. patients who had CaSR autoantibodies were
females, a finding that is similar to that found in
Bartter Syndrome Type V other autoantibody-mediated diseases. Indeed a
Bartter syndrome is a heterozygous group of auto- few AH patients have also had features of auto-
somal recessive disorders of electrolyte homeosta- immune polyglandular syndrome type 1 (APS1).
sis characterized by hypokalemic alkalosis, renal These findings establish that the CaSR is an
salt wasting that may lead to hypotension, hyper- autoantigen in AH [111, 284]. CaSR autoanti-
reninemic hyperaldosteronism, increased urinary bodies bind to the CaSR ECD and have, in some
prostaglandin excretion, and hypercalciuria with cases, been shown to enhance receptor function,
nephrocalcinosis [276, 277]. Mutations of several thereby reducing PTH secretion. Thus, hypopara-
ion transporters and channels have been associated thyroidism associated with activating CaSR auto-
with Bartter syndrome, and five types are now antibodies results from activated parathyroid
recognized [277]. The CaSR-related cases of CaSR function and not from immune-mediated
Bartter’s syndrome identified to date have been destruction of the parathyroid glands and is of
inherited in an autosomal dominant manner, unlike varying severity [284].
other subtypes that are inherited as autosomal
recessive traits.
Bartter syndrome type V is due to activating Pseudohypoparathyroidism (PHP)
mutations of the CaSR. Activating mutations of
the CaSR gene in three patients involving L125P, The term pseudohypoparathyroidism (PHP) was
C131W, and A843E, which inhibited the activity first introduced to describe patients with hypocal-
of the ROMK channel, provided the missing link cemia and hyperphosphatemia due to PTH resis-
that explains why some activating mutations of tance rather than PTH deficiency [285]. Affected
CaSR can cause the Bartter syndrome phenotype individuals show partial or complete resistance to
[278, 279]. Another recent mutation in monozy- biologically active exogenous PTH as demon-
gotic twins involving the K29E in the ECD of the strated by a lack of increase in urinary cyclic
CaSR described mild hypokalemia, minimal aldo- AMP and urinary phosphate excretion; this con-
sterone and renin production, and absent alkalosis dition is now referred to as PHP type I
but notable hypocalcemia [280]. The K29E muta- [286–288]. If associated with other endocrine
tion is an activating mutation of the CaSR [281] deficiencies and characteristic physical stigmata,
and buttresses previous observations that the phe- now collectively termed Albright’s hereditary
notype of Bartter syndrome is variable and not osteodystrophy (AHO), the condition is referred
directly related to the in vitro potency of the to as PHP type Ia. This latter syndrome is caused
known genetic changes associated with this syn- by heterozygous inactivating mutations within
drome [282]. CaSR mutations causing type V exons 1–13 of GNAS located on chromosome
Bartter syndrome have been shown to increase 20q13.3, which encode the stimulatory G protein
ER to cytosol calcium gradient, thus explaining (Gsα) (for review, see [288, 289]). These muta-
the higher sensitivity of CaSR gain-of-function tions were shown to lead to an approximately
variants to external calcium [283]. 50 % reduction in Gsα activity/protein in readily
accessible tissues, like erythrocytes and fibro-
Autoimmune Acquired blasts, and explain, at least partially, the resistance
Hypoparathyroidism (AH) toward PTH and other hormones that mediate
Twenty percent of patients, who had acquired their actions through G protein-coupled receptors
hypoparathyroidism (AH) in association with [286–288]. A similar reduction in Gsα activity/
autoimmune hypothyroidism, were found to protein is also found in patients with
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 309

pseudopseudohypoparathyroidism (PPHP), who that a somatic mutation is required in addition to


often show the same physical appearance as indi- heterozygosity at the GNAS locus [306].
viduals with PHP-Ia, but lack endocrine abnor- Mutations in the GNAS gene encoding Gsα
malities [286, 290–295]. However, contrary to have not been detected in patients with PHP type
previous reports, recent studies have shown that Ib (PHP-Ib), a disorder in which affected individ-
not all AHO features are observed in PPHP uals show PTH-resistant hypocalcemia and
patients. For example, obesity, which was thought hyperphosphatemia, but usually lack develop-
to be a hallmark of PHP-Ia and PPHP, was not mental defects. Initially it was therefore thought
evident in a large number of PPHP patients [296]. that this variant of PHP is caused by PTH/PTHrP
Patients affected by PHP-Ia or PPHP are typi- receptor mutations; however, mutations in its
cally found within the same kindred, but not gene and mRNA could not be identified
within the same sibship, and hormonal resistance [307–310]. Furthermore, it was shown that there
is parentally imprinted, i.e., PHP-Ia occurs only if is an increased incidence of TSH resistance in
the genetic defect is located on the maternal allele, PHP-Ib patients [261, 287, 311, 312] and that
while PPHP occurs only if the defect is located on some individuals affected by this disease variant
the paternal allele [297, 298]. Observations con- show some shortening of the fourth metacarpals,
sistent with these findings in humans were made suggesting some overlap between the develop-
in mice that are heterozygous for the ablation of mental features of PHP-Ia and PHP-Ib [313, 314].
exons 1 or 2 of the Gnas locus [299–301]. Animals A genome-wide search to identify the location
that inherited the mutant allele from a female of the “PHP-Ib gene” mapped the PHP-Ib locus to
showed much reduced Gsα protein in the renal chromosome 20q13.3, which contains the GNAS
cortex and decreased plasma calcium concentra- locus [315], and it was furthermore shown that the
tion due to resistance toward PTH. In contrast, genetic defect is parentally imprinted, i.e., it is
offspring that had obtained the mutant allele inherited in the same mode as the PTH-resistant
from a male showed no evidence for abnormali- hypocalcemia in kindreds with PHP-Ia and/or
ties in the regulation of mineral ion homeostasis. PPHP [297, 298]. Patients affected by PHP-Ib
These findings in mice with ablation of Gnas exon furthermore show a loss of methylation on the
1 or 2 and in humans with inactivating mutations maternal allele, which is usually restricted to
in GNAS exons 1–13 indicate that certain tissues, GNAS exon A/B [316, 317]. In most families
such as the proximal renal tubules, express Gsα with the autosomal dominant form of PHP-Ib
predominantly from the maternal allele; expres- with parental imprinting (AD-PHP-Ib), affected
sion from the paternal allele is silenced through individuals and healthy carriers were shown to
some unknown mechanisms. These data provided carry a 3 kb deletion located with the syntaxin
a reasonable explanation for the finding that het- 16 (STX16) gene about 220 kb upstream of exon
erozygous GNAS mutations result in a dominant A/B [318–321] (Fig. 8). Other AD-PHP-Ib kin-
phenotype with regard to the mineral ion abnor- dreds, in which the affected members show a loss
malities, if inherited through females. of A/B methylation alone, revealed either a 4.4 kb
Progressive osseous heteroplasia (POH) is deletion within STX16 overlapping with the 3 kb
caused also by heterozygous inactivating muta- deletion by 1,286 bp [322] or a 24.6 kb deletion
tions in the GNAS exons encoding Gsα comprising most STX16 exons [323]. Methylation
[302–305]. Interestingly, POH became apparent changes at GNAS exon A/B alone were further-
predominantly when a Gsα mutation was inherited more observed in the affected members of a fam-
from a male. Furthermore, the majority of muta- ily with a 18.9 kb deletion comprising exon
tions occurred in GNAS exons 2–13, thus impli- NESP55 and a large part of AS intron 4 [324]. In
cating XLαs rather than Gsα in the pathogenesis of affected individuals with either mutation, the dele-
POH. Recent evidence, however, indicated that tion is always found on the maternal allele, while
the abnormal bone formation process followed a it occurs on the paternal allele in unaffected
dermomyotomal distribution pattern, suggesting healthy carriers.
310 A. Sharma et al.

P11.23

P11.21

q11.21
q11.22
q11.23

q13.12

q13.13

q13.31
q13.32
q13.33
P12.3

P12.2

P12.1

q13.2
P13

q12
4.2–kb
18.9–kb
24.6–kb
4.0/4.7–kb
Gsα exons
4.4–kb
3.0
3.0–kb

1 1a 23 456 7 8 5 NESP554 3 21 XL A/B 1 2 3 N1 4 5 6 7 13

STX16 P + – – –
Control
M – + + +

3.0/4.4/24.6/18.9–kb del.
P + – – –
M – + + –
P + – – –
4.0/4.2/4.7–kb del.
M +/– – – –

Fig. 8 Genomic structure of PHP-Ib locus to chromosome mechanisms related to the defect of GNAS are described
20q13.3. This locus contains GNAS, encoding in the text
the stimulatory G protein (Gsα). The pathophysiological

The affected members of two families with deletions affect signaling through the PTH/PTHrP
broader methylation changes within the GNAS receptor in the proximal renal tubules, but not in
locus were shown to carry two distinct deletions most other tissues. Mice lacking the murine
on the maternal allele that remove GNAS exon homolog of exon A/B were recently shown to
NESP55 and the antisense exons 3 and 4 [325], have biallelic and thus increased Gsα transcription
a third deletion removes only antisense exons [329] (Fig. 8). Loss of exon A/B methylation on
3 and 4 [326]. Although indistinguishable broad the maternal allele allowing active transcription
methylation changes were observed in most from the promoters of both parental alleles and
patients with sporadic PHP-Ib, no deletions or this transcript therefore seems to have a prominent
point mutations have yet been identified in these role in suppressing Gsα expression.
individuals [327]. In fact, only patients with pater-
nal uniparental isodisomy involving either the
long arm of chromosome 20 (patUPD20q) or the Blomstrand’s Disease
entire chromosome (patUPD20) have provided a
molecular explanation for this variant of PHP-Ib Blomstrand’s chondrodysplasia is an autosomal
[328]. Taken together, these findings suggest that recessive human disorder characterized by early
several different deletions upstream or within the lethality, dramatically advanced bone maturation,
GNAS locus or patUPD20q lead to indistinguish- and accelerated chondrocyte differentiation [330].
able clinical and laboratory findings. However, it Affected infants are typically born to consanguin-
remains uncertain how the deletion affecting eous healthy parents (only in one instance did
STX16 results in a loss of exon A/B methylation unrelated healthy parents have two affected off-
alone, while the deletion of GNAS exon NESP spring) [331–335] and show pronounced
results in a broader loss of methylation. Further- hyperdensity of the entire skeleton (Fig. 9) [336]
more, it remains uncertain how the different and markedly advanced ossification, and
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 311

with Blomstrand’s disease. Two of these frameshift


mutations secondary to a homozygous single
nucleotide deletion in exon EL2 [339] and 27 bp
insertion between exon M4 and EL2 led to a trun-
cated receptor protein [340]. The other defect, a
nonsense mutation at residue 104, also resulted in
the same receptor protein defect [341]. Affected
infants show abnormalities in other organs, includ-
ing secondary hyperplasia of the parathyroid
glands (presumably due to hypocalcemia), besides
the described skeletal defects. In addition, analysis
of fetuses with Blomstrand’s disease revealed
abnormal breast development and tooth impaction,
highlighting the involvement of the PTH/PTHrP
receptor in the normal development of breast and
tooth [342].
Fig. 9 Radiological findings in a patient with
Blomstrand’s disease. (From [333], with permission from
Loshkajian with permission). Note the markedly advanced
ossification of all skeletal elements, and extremely short Hyperphosphatemic Disorders
limbs, despite comparatively normal size and shape of with Reduced Secretion of Biologically
hands and feet. In addition, notice that the clavicles are Active FGF23
relatively long and abnormally shaped

Hyperphosphatemic Familial Tumoral


particularly the long bones are extremely short and Calcinosis (HFTC)
poorly modeled. PTH/PTHrP receptor mutations
that impair its functional properties were identified HFTC needs to be differentiated from sporadic
as the most likely cause of Blomstrand’s disease. and secondary TC. It is a rare autosomal recessive
One of these defects is caused by a nucleotide disorder [343], which can be severe, sometimes
exchange in exon M5 of the maternal PTH/PTHrP fatal, and is typically characterized by hyperpho-
receptor allele, which introduces a novel splice sphatemia and often massive calcium deposits in
acceptor site and thus leads to the synthesis of a skin and subcutaneous tissues; in some patients,
receptor mutant that does not mediate, despite however, only few minor abnormalities are noted
seemingly normal cell surface expression, the [344]. The biochemical hallmark of tumoral cal-
actions of PTH or PTHrP; the patient’s paternal cinosis is hyperphosphatemia caused by loss-of-
PTH/PTHrP receptor allele is, for yet unknown function mutations in genes relevant to the pro-
reasons, only poorly expressed [336]. In a second duction of an intact bioactive form of FGF23, i.e.,
patient with Blomstrand’s disease, the product of a mutations in (1) FGF23 [40], (2) GALNT3 [345],
consanguineous marriage, a nucleotide exchange or resistance to the end-organ effects of FGF23
was identified that changes proline at position bioactivity (αKlotho) [346]. Homozygous or
132 to leucine [337, 338]. The resulting compound heterozygous mutations in GALNT3,
PTH/PTHrP receptor mutant despite reasonable which encodes a glycosyltransferase responsible
cell surface expression had severely impaired bind- for initiating mucin-type O-glycosylation, appear
ing to radiolabeled PTH and PTHrP analogues, to occur more frequently. Furthermore, the disor-
resulting in greatly reduced agonist-stimulated der can be caused by loss-of-function mutations in
cAMP accumulation and no measurable inositol FGF23. Mutations in either gene lead to indistin-
phosphate response. Additional loss-of-function guishable changes in FGF23, namely, low serum
mutations of the PTH/PTHrP receptor have intact FGF23, but significantly elevated
recently been identified in three unrelated patients C-terminal FGF23 concentrations [347, 348].
312 A. Sharma et al.

In contrast, HFTC caused by mutations in αKL is approaches has helped to elucidate some of the
characterized by profound increases of both forms mechanisms underlying these disorders of hered-
of FGF23. itary hypophosphatemic rickets (Table 1).

Hyperostosis with Hyperphosphatemia Hypophosphatemic Disorders


(HHS) with Increased FGF23 Activity

HHS is an autosomal recessive disorder, which Autosomal Dominant


shares several clinical and metabolic features Hypophosphatemic Rickets (ADHR)
with HFTC. The combination of hyperostosis Autosomal dominant hypophosphatemic rickets
with hyperphosphatemia was first described in is characterized by FGF23 excess due to “activat-
1970 [349, 350]. Most cases appear to be spo- ing” mutations in the FGF23 gene. Genetic link-
radic, but consanguineous parents were described age studies mapped the ADHR locus to
for some patients, implying that the disease can be chromosome 12p13.3 [355] and defined a
recessive. All affected individuals in two 1.5 Mb critical region that contained 12 genes.
unrelated Arab-Israeli HHS families were found Positional cloning in affected members of four
to harbor a previously described splice site muta- unrelated families identified three different mis-
tion in GALNT3, indicating that HFTC and HHS sense mutations involving a new member of the
are allelic variants [351]. Four novel GALNT3 fibroblast growth factor (FGF) family [38].
gene mutations were found in additional unrelated The C-terminal and the N-terminal portions of
subjects with considerable phenotypic overlap, FGF23 are connected by an “RXXR” motif,
again underscoring the fact that these two disor- which is the recognition site for a furin-like pro-
ders are variants of the same disease [352]. The tease. At this site, FGF23 can be cleaved and thus
molecular reasons for the enhanced skeletal inactivated. Missense mutations that replace the
involvement in HHS are currently unknown, but arginine (R) residue at either position 176 or
could potentially be due to modifier gene 179 with glutamine (Q) or tryptophan
interactions. (W) disrupt the cleavage site, thus making
FGF23 resistant to enzymatic cleavage (Fig. 2),
thereby stabilizing the intact bioactive FGF23
Hypophosphatemic Disorders molecule [6, 39, 48, 356, 357]. Iron deficiency
stimulates FGF23 expression and leads mice
The different forms of hypophosphatemia repre- engineered to carry one of the ADHR mutations
sent the most common cause of hereditary rickets, to hypophosphatemia [358], and similar observa-
which can be divided into two main groups tions were made in human with ADHR [359].
according to the predominant metabolic abnor-
mality [353, 354]. In the first group, Oncogenic Osteomalacia (OOM)
hypophosphatemia is the result of a renal tubular OOM (also referred to as tumor-induced osteoma-
defect, which may consist of either a single (iso- lacia (TIO)) is a rare disorder characterized by
lated) defect in renal phosphate handling, as it hypophosphatemia, phosphaturia, low circulating
occurs in the X-linked hypophosphatemia levels of 1,25-dihydroxyvitamin D, and osteoma-
(XLH), or of multiple tubular defects as encoun- lacia that develops in previously unaffected indi-
tered in Fanconi syndrome. In the second group, viduals [360]. Similarities between OOM,
vitamin D metabolism is abnormal, either because ADHR, and XLH suggested that they may
of a defect in the 1α-hydroxylase enzyme or involve the same phosphate-regulating pathway,
because of defects in the 1,25-dihydroxyvitamin and it is important to note that OOM tumors do
D3 receptor (VDR) leading to end-organ resis- express PHEX [361, 362], which is mutated in
tance. The application of molecular genetics XLH (see below). The possibility that FGF23,
11

Table 1 Biochemical findings in several inherited hypo- and hyperphosphatemic disorders and underlying genetic defects
TRP or Serum
FGF-23 TmP/GFR 1,25(OH)2D PTH calcium Urinary calcium Mutant gene
Hypophosphatemic disorders
FGF23 dependent
XLH Increased/ Low Low/ Normal/ Normal Normal PHEX
inappropriately inappropriately increased
normal normal
ADHR Increased/ Low Low/ Normal Normal Normal FGF23
inappropriately inappropriately
normal normal
ARHP Increased/ Low Low/ Normal Normal Normal DMP1, ENPP1, FAM20C
inappropriately inappropriately
normal normal
Hypophosphatemic disorders
FGF23 independent
HHRH Low/normal Low High Low Normal/ High NPT2c
increased
Renal Fanconi syndrome Low/normal Low High Low Normal/ High NPT2a
increased
Hyperphosphatemic disorders
Tumoral calcinosis Intact: low High Normal-high Low Normal/ Increased FGF23 or GALNT3
C-terminal: very increased (glycosyltransferase)
high
Extremely high High Normal-high Elevated Normal/ Increased Klotho
(intact and increased
C-terminal)
Isolated hypoparathyroidism Normal-increased Elevated Low-normal Low- Low Increased or Calcium-sensing receptor, PTH,
normal inappropriately GCM2, GNA11, and unknown genetic
Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . .

normal defects
Pseudohypoparathyroidism Normal-increased Elevated Low-normal High Low Low PHP-Ia: GNAS exons encoding Gsα
type Ia (PHP-Ia) or Ib PHP-Ib: microdeletions within or
(PHP-Ib) upstream of GNAS
313
314 A. Sharma et al.

which is mutated in ADHR, may also be FGF23 alone [45, 46], indicating that FGF23
expressed in OOM tumors and that FGF23 may resides genetically downstream of PHEX. Fur-
be a secreted protein was therefore explored [39, thermore, recent studies have shown that Hyp
65, 356, 363]. Indeed, OOM tumors were found mice normalize their plasma phosphate concen-
by Northern blot analysis to contain high levels of tration and heal their rachitic changes, when
FGF23 mRNA and protein. Consistent with this injected with inactivating antibodies to FGF23,
finding, FGF23 plasma concentrations can be indicating that FGF23 is indeed the phosphaturic
increased considerably in OOM patients, until principle in XLH [373]. XLH patients exhibited a
successful tumor removal [356, 357, 364, 365]. similar response to a single injection of anti-
Tumors responsible for oncogenic osteomala- FGF23 antibodies [374].
cia produce two molecular forms of FGF23 (~32
and ~12 kDa), and both variants were also Autosomal Recessive
observed when assessing conditioned medium Hypophosphatemia (ARHP)
from different cell lines expressing full-length There are different forms of autosomal recessive
wild-type FGF23 [39]. When conditioned hypophosphatemia (ARHP) characterized by renal
medium from cells expressing R176Q FGF23 or phosphate wasting. ARHP can be caused by muta-
R179Q FGF23 was investigated by Western blot tions in DMP1 (Dentin matrix protein 1), which
analysis, only the larger protein band was belongs to the family of integrin-binding ligand
observed [7, 8, 366]. These findings showed that N-linked glycoproteins (SIBLING proteins) that
the ADHR mutations impair FGF23 degradation, are secreted phosphoproteins involved in bone
thus enhancing and/or prolonging its biological mineralization [375]. DMP1 mutations lead to
activity. In addition to FGF23, these tumors have increased FGF23 expression and thus increased
also been shown to express other factors, includ- urinary phosphate excretion and defective osteo-
ing DMP1, sFRP4, and MEPE [367]. cyte maturation [376, 377]. ARHP was first
reported in consanguineous kindreds, suggesting
X-Linked Hypophosphatemia (XLH) an autosomal recessive form of hypophosphatemia
Like ADHR, patients with XLH present with [378–380]. Patients affected by ARHP have
hypophosphatemia, hyperphosphaturia, low cir- FGF23 levels that are either elevated or inappro-
culating 1,25-dihydroxyvitamin D levels, and priately normal for the level of serum phosphorous
osteomalacia. XLH is caused by inactivating [48, 51]. Of the several different DMP1 mutations
mutations in PHEX, a gene located on Xp22.1 identified thus far, one mutation alters the transla-
[368, 369], which shows significant amino acid tion initiation codon (M1V), two mutations are
sequence homology to the M13 family of zinc located in different intron-exon boundaries, and
metallopeptidases. In the past, substrates for three are frameshift mutations within exon 6. All
PHEX considered were FGF23, matrix extracel- mutations appear to be inactivating, suggesting that
lular phosphoglycoprotein (MEPE) [68, 370], and the loss of DMP1 causes increased FGF23 produc-
secreted frizzled-related protein 4 (sFRP4) [65, tion leading to hypophosphatemia. Accordingly,
67]. Among these proteins, FGF23 levels were Dmp1-null mice show severe defects in dentine,
found to be elevated in about two-thirds of XLH bone, and cartilage, as well as hypophosphatemia
patients [78, 261, 356] and more than tenfold in and osteomalacia [381, 382]. Furthermore, FGF23
Hyp mice, the murine homolog of XLH [63, 371]. levels in osteocytes and in serum are drastically
Although PHEX-dependent cleavage of FGF23 elevated in these animals [48]. Given the
was observed in one study in vitro [372], these established importance of DMP1 in osteoblast
findings were not confirmed by others [40]. How- function, loss of DMP1 actions in osteoblasts and
ever, genetic ablation of FGF23 in male Hyp extracellular matrix may also contribute to the phe-
mice, i.e., animals that are null for FGF23 and notype of patients with ARHR. Consistent with
PHEX, led to blood phosphate levels that are this hypothesis, a high-calcium/high-phosphate
indistinguishable from those in mice lacking diet which is capable to rescue osteomalacia in
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 315

VDR-null mice does not seem to prevent bone and Fanconi syndrome and hypophosphatemic
dentine mineralization defect in Dmp1-null mice rickets [388].
[383]. Other forms of ARHP are caused by muta- These findings are similar to those observed
tion in the gene encoding ENPP1 gene in mice with homozygous ablation of Npt2a
(ectonucleotide pyrophosphatase/phosphodiester- (Npt2a/) [389]. Due to hypophosphatemia,
ase), which catalyzes phosphoester cleavage of Npt2a/ mice show an appropriate elevation in
adenosine triphosphate, thereby generating pyro- the serum levels of 1,25-dihydroxyvitamin D, lead-
phosphate, a mineralization inhibitor. All forms of ing to hypercalcemia, hypercalciuria and decreased
hypophosphatemia are often associated with ossi- serum parathyroid hormone levels, and increased
fication of the posterior spinal ligaments, and serum alkaline phosphatase activity.
ENPP1 mutations can also be associated with cal-
cifications of the vascular endothelium, as Hereditary Hypophosphatemic Rickets
observed in GACI (generalized arterial calcifica- with Hypercalciuria (HHRH)
tion of infancy) [51, 54, 384]. Homozygous or compound heterozygous mutations
in the sodium phosphate cotransporter NPT2c
(SLC34A3) are the cause of HHRH, an autosomal
Hypophosphatemic Disorders recessive disorder affecting renal tubular phosphate
with Normal or Suppressed FGF23 reabsorption, leading to hypophosphatemia,
Activity increased 1,25(OH)2 vitamin D levels, and
hypercalciuria, which lead in a considerable number
Nephrolithiasis and Osteoporosis of patients to nephrolithiasis [390–393]; even indi-
Associated with Hypophosphatemia viduals with heterozygous NPT2c mutations can
Two different heterozygous mutations (A48P and show laboratory abnormalities and nephrolithiasis.
V147M) in NPT2a, the gene encoding a sodium- Long-term phosphate supplementation as the sole
dependent phosphate transporter, have been therapy leads, with the exception of persistently
reported in patients with urolithiasis or osteoporo- decreased TmP/GFR, to reversal of the clinical and
sis and persistent idiopathic hypophosphatemia biochemical abnormalities [390]. Treatment with
due to decreased tubular phosphate reabsorption calcitriol is contraindicated since it can further
[385]. When expressed in Xenopus laevis oocytes, increase urinary calcium excretion.
the mutant NPT2a showed impaired function and,
when co-injected, dominant-negative properties. Vitamin D-Dependent Rickets (VDDR)
However, these in vitro findings were not con- Patients with VDDR type I (pseudo-vitamin
firmed in another study using oocytes and OK D-resistant rickets) show clinical and laboratory
cells, raising the concern that the identified findings that are similar to patients with vitamin
NPT2a mutation alone cannot explain the findings D-deficient rickets but do not respond to treatment
in the described patients [386]. On the other hand, with 25(OH) vitamin D; instead treatment with
additional heterozygous NPT2a variations 1,25(OH)2 vitamin D is required [394]. Clarifica-
(in-frame deletion or missense change) have tion of the abnormal vitamin D metabolism [395]
recently been identified upon analyzing a large led to the recognition of two different forms of
cohort of hypercalciuric stone-forming kindreds; rickets: VDDR type I (defect in the 1-
however, these genetic variations do not seem to α-hydroxylation) and vitamin D-dependent rick-
cause functional abnormalities [387]. Homozygous ets type II (VDDR type II) (end-organ resistance
in-frame duplication of 21 bp in SLC34A1 to 1,25(OH)2D).
resulting in complete loss of function of the mutant
NPT2a, one of two sodium-dependent phosphate 1-Alpha Hydroxylase Deficiency
cotransporters expressed in the proximal renal (VDDR Type I)
tubules, was identified in two siblings from a con- Patients affected by VDDR type I (autosomal
sanguineous family with autosomal recessive recessive) show almost all the clinical and
316 A. Sharma et al.

biochemical features of vitamin D-deficient rick- analogous to the classical steroid hormones
ets except that vitamin D intake is usually ade- [415]. The interactions between 1,25(OH)2 vita-
quate. The permanent teeth show marked enamel min D and its intracellular receptor have been
hypoplasia unlike in XLH [396]. The serum con- studied using cultured skin fibroblasts from con-
centration of 1,25(OH)2 vitamin D is low in trol subjects and VDDR type II patients [416,
untreated patients [397] and in vitamin 417]. Several defects were identified, including
D-sufficient patients with normal or elevated absent receptors, a decreased number of receptors
serum 25(OH)D3 levels, which provided the first with normal affinity, a normal receptor-hormone
hint that the renal 1α-hydroxylase is deficient binding but a subsequent failure to translocate the
[398–400]; molecular genetic studies later con- hormone to the nucleus, and a post-receptor
firmed this conclusion. Indeed, genetic linkage defect, in which normal receptors are present,
studies in affected French-Canadian families but there is a deficiency in the induction of the
mapped VDDR type I to a region on chromosome 25-OHD-24 hydroxylase enzyme in response to
12q13.3 [401], which is the location of the gene 1,25(OH)2 vitamin D. Thus, the heterogeneity
encoding the 25(OH)D 1α-hydroxylase suggested from clinical observations in VDDR
(CYP27B). DNA sequence analysis of patients type II patients could be demonstrated at the cel-
affected by VDDR type 1 identified more than lular level with various combinations of defective
20 different mutations of CYP27B [402–406] in receptor-hormone binding and expression. Vita-
26 kindreds. All patients with VDDR type 1 were min D receptor (VDR) is an intracellular protein,
found to carry homozygous or compound hetero- which has a molecular weight of 60,000 Daltons.
zygous mutations, while the obligate carriers were The binding site for 1,25(OH)2 vitamin D resides
heterozygous for the mutant allele. Mutations that in the C-terminal part of the protein, while the
confer partial enzyme activity in vitro were found N-terminal part of the molecule possesses the
in the two patents with mild laboratory abnormal- DNA-binding domain [418]. This hormone-
ities, suggesting that such mutations contribute to receptor complex binds to a DNA region, which
the phenotypic variation observed in patients with is located upstream of the promoter of genes
1α-hydroxylase deficiency [407]. encoding calcium-binding proteins and other
proteins.
End-Organ Resistance (VDR Mutations, The availability of cDNAs encoding the avian
VDDR Type II) and human VDR [419] helped to clarify the
Vitamin D-dependent rickets type II is an autoso- molecular basis of VDDR type II [420]. Nucleo-
mal recessive disorder caused by end-organ resis- tide sequence analysis of genomic DNA revealed
tance to 1,25(OH)2 vitamin D [408–410]. that the human VDR gene consists of nine exons;
Alopecia, ectodermal defects like oligodontia, exons 2 and 3 encode the DNA-binding domain,
and epidermal cysts might be present. The disease while exons 7, 8, and 9 encode the vitamin
varies in its clinical and biochemical manifesta- D-binding domain. The gene is located on chro-
tions, which suggest heterogeneity in the under- mosome 12q12–q14 in man [401], i.e., in a region
lying molecular defects [410]. Most of the patients that comprises the gene encoding the 1-
have early-onset rickets, but the first reported α-hydroxylase. Mutational analysis of the VDR
patient was a 22-year-old woman who had skele- gene in VDDR type II patients demonstrated the
tal pain for 7 years [408], and another patient presence of nonsense and missense mutations
presented at the age of 50 years following affecting different parts of the receptor. The
5 years of symptoms [411]. Alopecia in some expression of these mutations in COS-1 monkey
cases is associated with poor therapeutic response kidney cells demonstrated that these mutations
to the 1α-(OH) vitamin D [412, 413]. result in a reduction or a loss of VDR function
1,25(OH)2 vitamin D actions are mediated by similar to the heterogeneous effects observed in
an intracellular receptor that binds DNA and con- cultured fibroblasts from VDDR type II patients.
centrates the hormone in the nucleus [414], Furthermore, null mutant, i.e., “knockout” mice
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 317

for VDR produced by targeted gene disruption cause increased endogenous FGF23 from bone,
[421, 422], was found to have features consistent thus causing hypophosphatemia and osteomalacia
with those observed in patients with VDDR type [442]. Furthermore, fibrous dysplasia can be asso-
II. The severity of resistance to 1,25(OH)2 vitamin ciated with increased urinary phosphate excretion.
D is variable, and some patients have improved This disorder is caused by heterozygous activating,
following therapy with very large doses of vita- post-zygotic mutations in exon 8 of GNAS, the gene
min D [423] or 1,25(OH)2 vitamin D encoding the alpha-subunit of the stimulatory G
[424–426]. However, currently most patients are protein (Gsα) [443, 444]. These mutations lead in
treated with long-term nocturnal intravenous cal- the dysplastic regions to a cAMP-dependent
cium infusions followed by oral calcium supple- increase in FGF23 production by osteoblasts/oste-
mentation [427, 428]; cinacalcet to reduce PTH ocytes and fibrous cells [44, 445].
secretion has been tried with some success [429].

Acute and Chronic Treatment


Other Hypophosphatemic Disorders of Calcium and Phosphorus Disorders

There are several other genetic disorders associated Goals of acute and chronic therapy are dependent
with hypophosphatemia and often with other upon the specific clinical setting in which the dis-
defects in proximal tubular function. These include turbance occurs, and management decisions usu-
Dent’s disease, an X-linked recessive disorder, ally require individual attention to the underlying
which is caused by mutations in CLCN5 encoding pathophysiology of each patient’s underlying dis-
the chloride/proton antiporter, chloride channel order. Broad principles for correcting calcium and
CLC-5 [430, 431], and Lowe syndrome (oculocer- phosphorus abnormalities are discussed.
ebrorenal syndrome) [432], another X-linked reces-
sive disorder that is caused by mutations in OCRL1
[433, 434]. Other rare diseases that can be associ- Hypercalcemia
ated with severe hypophosphatemia are the
Fanconi-Bickel syndrome, which is caused by Acute Intervention
homozygous or compound heterozygous mutations Hypercalcemia is the biochemical hallmark of
in GLUT2 [435, 436], a glucose transporter local- increased PTH or 1,25(OH)2 vitamin D levels,
ized on the basolateral membrane of the proximal less frequently in patients with fat necrosis or malig-
renal tubules. The hallmarks of this disorder are nancies. When severe it can be life-threatening and
severe failure to thrive, hepatomegaly, and proximal requires urgent medical intervention. If cardiac and
renal tubular dysfunction with massive glycosuria, renal function is normal, increasing sodium clear-
fasting hypoglycemia, and postprandial hypergly- ance, maximizing glomerular filtration rate, and
cemia, in addition to variable phosphaturia, acido- correcting dehydration increase calcium excretion.
sis, and aminoaciduria. Other hypophosphatemic Other available pharmacological options are calci-
disorders include osteoglophonic dysplasia (OGD) tonin, bisphosphonates, and cinacalcet. Calcitonin
[437], an autosomal dominant disorder, which was causes rapid reduction in serum calcium level by
shown to be caused by different heterozygous mis- inhibiting bone resorption and enhancing urinary
sense mutations in the FGFR1 [438, 439], and calcium excretion, but its use is limited by the
linear nevus sebaceous syndrome (LNSS), also requirement for frequent dosing and tachyphylaxis.
known as epidermal nevus syndrome (ENS) or Bisphosphonates inhibit osteoclast activity. Typi-
Schimmelpenning-Feuerstein-Mims syndrome, in cally, a clinical response is seen within 1–4 days,
which elevated FGF23 was observed [440, with nadir in calcium observed by 4–7 days. Cau-
441]. Somatic activating mutations of HRAS or tion is required, because acute renal failure and
NRAS (mitogen-activated protein kinase) associ- osteonecrosis of the jaw can be encountered as
ated with giant epidermal or melanocytic nevi rare side effects. Cinacalcet is an allosteric activator
318 A. Sharma et al.

of CaSR (calcimimetic) that increases the receptor’s Hypercalcemia associated with AHH may
affinity for calcium, leading to a reduction in PTH improve following glucocorticoid administration
synthesis and an increase in renal calcium excretion in some, but not all, patients [118].
because of reduced distal tubular calcium
reabsorption. Peritoneal or hemodialysis with low
calcium concentration in the dialysis fluid might be Hypocalcemia
required in severe cases. Corticosteroids to reduce
intestinal calcium absorption are not always A decrease in ionized calcium is frequently a
effective. manifestation of defective vitamin D homeostasis
or inadequate PTH secretion or action. The
Chronic Intervention PTH-dependent renal production of 1,25-
Chronic treatment is guided by the underlying dis- dihydroxyvitamin D is deficient in all
order. Primary hyperparathyroidism (PHPT), e.g., hypoparathyroid states.
parathyroid tumors, NSPHT, and MEN, respond
well to surgical removal of the parathyroid glands. Acute Intervention
No effective medical therapy for the long-term Symptomatic hypocalcemia or severe asymptom-
management of PHPT is currently approved for atic hypocalcemia is a medical emergency and
children. In some NSHPT patients, pamidronate often requires intravenous 10 % calcium gluco-
[446] has been used to manage life-threatening nate or calcium chloride infusion. Concomitant
hypercalcemia and skeletal demineralization prior hypomagnesemia and hyperkalemia, if present,
to parathyroidectomy. Intraoperative PTH mea- should be corrected. The transition from acute
surements have been shown to be effective for management of hypocalcemia to chronic mainte-
monitoring whether parathyroidectomy is com- nance is highly dependent on the individual clin-
plete [447]. Cinacalcet has been used as well, ical situation. It is often possible to begin oral
but with equivocal results, since CaSR mutations supplementation within a few days of intravenous
can disrupt interaction with this drug. Heterozy- therapy with a few days of overlap. Some children
gous loss-of-function mutations of the CaSR with abnormal parathyroid development, e.g.,
(FHH) usually cause mild-to-moderate hypercalce- DGS, maintain normal calcium when healthy,
mia that is not driven by excess PTH. FHH is a but have decreased parathyroid reserve and there-
benign disorder characteristically associated with fore develop symptomatic hypocalcemia when
hypocalciuria, which unlike PHPT requires no stressed, for example, due to surgery or infections,
treatment except reassurance. Parathyroidectomy or when normal intestinal calcium intake is
is not indicated. reduced because of diarrhea or vomiting.
Hypercalcemia driven by increased intestinal
calcium absorption, e.g., Williams syndrome, Chronic Intervention
usually responds to a low-calcium diet with elim- The main treatment options available for patients
ination of vitamin D. Low-calcium milk (Locasol) with acute or chronic hypoparathyroidism are cal-
and protection from the sunlight can be useful cium salts, vitamin D or vitamin D analogues, and
[148]. Corticosteroids may be a useful adjunct to drugs that increase renal tubular reabsorption of
lower plasma calcium levels by decreasing calcium (i.e., thiazides). The selection of the dose
absorption from the gut. Steroids can be usually and type of the vitamin D analogue varies with the
discontinued after a few days when dietary underlying condition and the response to therapy.
changes have shown to be effective. Treatment Calcitriol is the preferred metabolite as it is consis-
of infantile hypercalcemia consists of calcium- tently effective at nontoxic doses and has a short
and vitamin D-restricted diets, steroids, and half-life, thus allowing rapid correction should
bisphosphonates, if necessary. Cellulose phos- hypercalcemia develop. Monitoring of therapy is
phate has also been used to decrease calcium essential to avoid hypercalcemia, hypercalciuria,
absorption. and nephrocalcinosis. Furthermore, recombinant
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 319

PTH has been used in some cases of severe hypo- Bartter syndrome is characterized by increased
parathyroidism. Compared with twice-daily deliv- prostaglandin E2 production, which stimulates the
ery, pump delivery of PTH(1-34) appears to provide 1α-hydroxylase activity, thereby increasing 1,25
the most promising modality for improving calcium (OH)2D levels. Indomethacin (or other COX2
homeostasis and bone turnover in these children inhibitors) decreases prostaglandin levels, which
[154]. The absence of functional PTH and the furthermore restricts intestinal calcium and vita-
resulting lack of PTH-dependent calcium min D absorption from the diet, thus helping to
reabsorption in the distal renal tubules predispose correct hypercalciuria and nephrocalcinosis.
to hypercalciuria, nephrolithiasis, and chronic kid- However, indomethacin itself possesses serious
ney disease. Maintaining serum calcium at the lower nephrotoxicity; appropriate dose in each patient
end of the normal range avoids clinical symptoms should be considered.
and minimizes the risk of hypercalciuria. This is Long-term therapy of hypocalcemia due to
exactly important, and if the hypocalcemia is not pseudohypoparathyroidism (PHP) is similar to
symptomatic, maintenance of lower serum Ca level other forms of hypoparathyroidism, namely, oral
is recommended. Hypercalciuria, if it occurs, may calcium and calcitriol. However, PHP patients are
respond to oral hydrochlorothiazide. Monitoring for at little risk of developing increased urinary cal-
these complications at regular intervals is the key to cium excretion, unless medications are not suffi-
avoid complications. ciently reduced after the skeleton is fully
Hypocalcemia due to vitamin D deficiency is mineralized after pubertal development. Treat-
treated with cholecalciferol or related analogues. ment with calcitriol can thus be usually more
It is important to ensure an adequate calcium aggressive than that of other forms of hypocalce-
supply with the vitamin D replacement, as rapid mia, and normocalcemia and normal or close-to-
mineralization of the skeleton may follow initial normal PTH should be achieved. Patients with
treatment, potentially causing a precipitous drop PHP-Ia (and PHP-Ib) should furthermore be rou-
in serum calcium (hungry bone syndrome). Treat- tinely screened and eventually treated for any
ment of VDDR I (1α-hydroxylase deficiency) associated endocrinopathy, in particular, hypothy-
requires lifelong replacement with calcitriol or roidism. In addition, careful physical examination
alphacalcidiol, thus bypassing the 1- and, when necessary, specific psychological
α-hydroxylation step. Some patients with VDDR investigations should be performed annually to
II (end-organ resistance to 1,25(OH2) vitamin D) detect and follow up the presence/evolution of
may respond to very high doses of calcitriol or specific AHO features (heterotopic ossifications,
alphacalcidiol, but most pediatric patients require cognitive delay, brachydactyly). Increasing evi-
regular intravenous calcium infusions or high dence suggests that children should be screened
doses of oral calcium. with appropriate provocative tests for GH defi-
Hypocalcemia due to calcium-sensing receptor ciency [448]; note that PHP-Ia patients often
(CaSR) mutations is due to increased CaSR sen- show “normal” growth rates early in life, but
sitivity (ADHH). These patients therefore have fuse their growth plates prematurely, making it
serum PTH levels that are in the low-normal necessarily to start treatment with rhGH as soon
range, which is different from patients with hypo- as possible. Finally, there are no specific treat-
parathyroidism, who often have undetectable ments for the various manifestations of AHO.
serum PTH levels or patients with pseudohypo-
parathyroidism, who typically have major PTH
elevations. Treatment with active metabolites of Hyperphosphatemia
vitamin D to correct the hypocalcemia can result
in marked hypercalciuria, nephrocalcinosis, Acute Intervention
nephrolithiasis, and renal impairment, which Acute hyperphosphatemia usually does not cause
may be only partially reversible after cessation symptoms unless reciprocal reduction in serum
of the vitamin D treatment. calcium leads to hypocalcemia. In the setting of
320 A. Sharma et al.

normal kidney function, hyperphosphatemia is in 4–5 daily doses; high doses of phosphate are
usually self-limited because of the capacity of associated with diarrhea. Other adjunctive therapy
kidney to excrete the phosphorus load. In the is guided by the underlying disorder. Furthermore,
setting of markedly compromised renal function, anti-FGF23 antibodies may become available for
dialysis can be required, in addition to oral phos- FGF23-dependent hypophosphatemic disorders.
phate binders. In patients with oncogenic osteomalacia
(OOM), complete resection of the FGF23-
Chronic Intervention secreting tumor leads to rapid correction of the
Chronic hyperphosphatemia management is biochemical abnormalities. Somatostatin and
dependent on the underlying disorder. Acetazol- cinacalcet has been tried with some success if
amide has been used to treat hyperphosphatemia tumor resection is incomplete. Anti-FGF23 anti-
and tumor-like extraosseous calcifications due to bodies are likely to ameliorate potentially harmful
inactivating mutations in FGF23 [449] or effects of long-standing phosphorus and vitamin
GALNT3 in patients with FTC. This treatment D therapy before the tumor is found and resected.
induces mild metabolic acidosis, thus improving The primary goal for the treatment or X-linked
calcium-phosphate complex solubility. In severe hypophosphatemia (XLH) and other FGF23-
cases, regular hemodialysis has been prescribed dependent forms of hypophosphatemia is to cor-
with reasonable success. rect or minimize rachitic changes and aim for an
Secondary hyperparathyroidism seen in acceptable height velocity; in addition, meticu-
chronic kidney disease is driven by hyperpho- lous dental care is advised because of the high
sphatemia, reduced 1,25(OH)2D production and incidence of dental abscesses. Serum alkaline
thus hypocalcemia, and possibly elevated FGF23 phosphatase is the most useful laboratory bio-
levels. The cornerstone of management is dietary marker to gauge the success of therapy, in addition
phosphate restriction, use of phosphate binders, to radiographs. The current therapy consists of the
and dialysis to aid with phosphate removal. oral administration of phosphate and calcitriol.
Cinacalcet has been shown in EVOLVE trial to Cinacalcet and calcitonin have been tried with
decrease bone turnover and tissue fibrosis some success as adjunctive therapies. Anti-
[450]. Parathyroidectomy is often reserved for FGF23 antibodies were recently shown to normal-
ESRD patients, who have profound PTH eleva- ize blood phosphate levels in adult XLH patients
tions and thus increased bone resorption. [374]. Treatment with recombinant human growth
hormone (rhGH) is controversial given the lack of
adequately controlled trials. Lower limb correc-
Hypophosphatemia tive osteotomies or minimally invasive
hemiepiphysiodesis are needed for severe bony
Acute Intervention deformities in older children. For all patients
Acute severe hypophosphatemia particularly with inherited forms of hypophosphatemia, iron
presenting as hemolysis, rhabdomyolysis, flaccid stores should be replenished, if deficient.
paralysis, or cardiac dysfunction is a medical Hereditary hypophosphatemic rickets with
emergency that should be corrected promptly hypercalciuria (HHRH) is a hypophosphatemic
with intravenous sodium- or potassium- disorder characterized by normal or suppressed
phosphate, as appropriate. Cumulative phosphate FGF23 activity and thus elevated 1,25(OH)2 vita-
deficit cannot be accurately predicted from serum min D levels. This disorder is therefore managed
levels. Once the levels are above 1.5 mg/dL, tran- differently from the hypophosphatemia such as
sition to oral replacement should be attempted. XLH, ARHP, or ADHR. Oral phosphate supple-
ments are effective. Calcitriol is contraindicated,
Chronic Intervention as 1,25(OH)2 vitamin D levels are elevated.
The cornerstone of management is oral phospho- Fanconi-Bickel syndrome is a disorder of gen-
rus replacement, which needs to be administered eralized proximal tubular dysfunction that is
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 321

caused by GLUT2 mutations. Treatment aims at phosphate homeostasis. Endocrinology. 2004;


correcting acidosis, treating the hypophosphatemic 145(7):3087–94.
6. Shimada T, Mizutani S, Muto T, Yoneya T, Hino R,
rickets, and using fructose-based diets to circum- Takeda S, et al. Cloning and characterization of
vent hypoglycemia. Long-term prognosis is FGF23 as a causative factor of tumor-induced osteo-
unknown, but there is a risk of atherosclerotic malacia. Proc Natl Acad Sci USA. 2001;98(11):
vascular disease, repeated fractures from general- 6500–5.
7. Shimada T, Muto T, Urakawa I, Yoneya T,
ized osteopenia, and markedly short stature. Yamazaki Y, Okawa K, et al. Mutant FGF-23 respon-
sible for autosomal dominant hypophosphatemic
rickets is resistant to proteolytic cleavage and causes
Concluding Remarks hypophosphatemia in vivo. Endocrinology.
2002;143:3179–82.
8. Bai XY, Miao D, Goltzman D, Karaplis AC. The
Remarkable advances have been made in identify- autosomal dominant hypophosphatemic rickets
ing key proteins that are involved, either directly or R176Q mutation in fibroblast growth factor 23 resists
indirectly, in the regulation of calcium and phos- proteolytic cleavage and enhances in vivo biological
potency. J Biol Chem. 2003;278(11):9843–9.
phate homeostasis, the hormones that are involved 9. Jueppner H, Thakker RV. In: Pollack M, editor. The
in these mechanisms, and receptors that mediate Kidney. Philadelphia: WB Saunders; 2008.
these hormonal actions in the different target tissues. p. 311–45.
Although most of these conditions are rare, their 10. Gardella TJ, J€uppner H, Brown EM, Kronenberg HM,
Potts Jr JT. Parathyroid hormone and parathyroid
molecular definition resulted in the identification of hormone-related peptide in the regulation of calcium
several proteins that are involved in the normal homeostasis and bone development. In: DeGroot LJ,
regulation of mineral ion homeostasis and/or bone Jameson JL, editors. Endocrinology, vol. 1. 6th
development. These advances have added to ed. Philadelphia: W.B. Saunders; 2010. p. 1040–73.
11. Okazaki T, Igarashi T, Kronenberg HM. 5'-Flanking
targeted therapy for some of these disorders. region of the parathyroid hormone gene mediates
negative regulation by 1,25(OH)2 vitamin D3. J Biol
Chem. 1989;263:2203–8.
12. Demay MB, Kiernan MS, DeLuca HF, Kronenberg
References HM. Sequences in the human parathyroid hormone
gene that bind the 1,25- dihydroxyvitamin D3 recep-
1. Segawa H, Kawakami E, Kaneko I, Kuwahata M, tor and mediate transcriptional repression in response
Ito M, Kusano K, et al. Effect of hydrolysis-resistant to 1,25-dihydroxyvitamin D3. Proc Natl Acad Sci
FGF23-R179Q on dietary phosphate regulation of the USA. 1992;89(17):8097–101.
renal type-II Na/Pi transporter. Pflugers Arch. 13. Naveh-Many T, Rahaminov R, Livini N, Silver
2003;446(5):585–92. J. Parathyroid cell proliferation in normal and chronic
2. Segawa H, Yamanaka S, Onitsuka A, Tomoe Y, renal failure in rats. The effects of calcium, phosphate,
Kuwahata M, Ito M, et al. Parathyroid hormone- and vitamin D. J Clin Invest. 1995;96:1786–93.
dependent endocytosis of renal type IIc Na-Pi 14. Almaden Y, Canalejo A, Hernandez A, Ballesteros E,
cotransporter. Am J Physiol Renal Physiol. Garcia-Navarro S, Torres A, et al. Direct effect of
2007;292(1):F395–403. phosphorus on PTH secretion from whole rat para-
3. Miyamoto K, Ito M, Tatsumi S, Kuwahata M, Segawa thyroid glands in vitro. J Bone Miner Res Off J Am
H. New aspect of renal phosphate reabsorption: the Soc Bone Miner Res. 1996;11(7):970–6.
type IIc sodium-dependent phosphate transporter. Am 15. Slatopolsky E, Finch J, Denda M, Ritter C, Zhong M,
J Nephrol. 2007;27(5):503–15. Dusso A, et al. Phosphorus restriction prevents para-
4. Shimada T, Urakawa I, Yamazaki Y, Hasegawa H, thyroid gland growth. High phosphorus directly stim-
Hino R, Yoneya T, et al. FGF-23 transgenic mice ulates PTH secretion in vitro. J Clin Invest.
demonstrate hypophosphatemic rickets with reduced 1996;97:2534–40.
expression of sodium phosphate cotransporter type 16. Hebert SC. Extracellular calcium-sensing receptor:
IIa. Biochem Biophys Res Commun. 2004;314(2): implications for calcium and magnesium handling in
409–14. the kidney. Kidney Int. 1996;50(6):2129–39.
5. Larsson T, Marsell R, Schipani E, Ohlsson C, 17. Suva LJ, Winslow GA, Wettenhall RE, Hammonds
Ljunggren O, Tenenhouse HS, et al. Transgenic RG, Moseley JM, Diefenbach-Jagger H, et al. A para-
mice expressing fibroblast growth factor 23 under thyroid hormone-related protein implicated in malig-
the control of the alpha1(I) collagen promoter exhibit nant hypercalcemia: cloning and expression. Science.
growth retardation, osteomalacia, and disturbed 1987;237:893–6.
322 A. Sharma et al.

18. Mangin M, Webb AC, Dreyer BE, Posillico JT, 31. Coutellier L, Usdin TB. Enhanced long-term fear
Ikeda K, Weir EC, et al. Identification of a cDNA memory and increased anxiety and depression-like
encoding a parathyroid hormone-like peptide from a behavior after exposure to an aversive event in mice
human tumor associated with humoral hypercalcemia lacking TIP39 signaling. Behav Brain Res. 2011;
of malignancy. Proc Natl Acad Sci U S A. 222(1):265–9.
1988;85:597–601. 32. Whyte MP. Rickets and osteomalacia. In: Wass J,
19. Strewler GJ, Stern PH, Jacobs JW, Eveloff J, Klein Shalet S, editors. Oxford textbook of endocrinology,
RF, Leung SC, et al. Parathyroid hormone-like pro- Oxford: Oxford University Press; 2002. p. 697–715.
tein from human renal carcinoma cells. Structural 33. Liberman U, Marx SJ. Vitamin D and other
and functional homology with parathyroid hormone. calciferols. In: Beaudlt A, Sly W, Valle D, editors.
J Clin Invest. 1987;80:1803–7. The metabolic and molecular bases of inherited
20. Broadus AE, Stewart AF. Parathyroid hormone- disease. 8th ed. New York: McGraw-Hill; 2001.
related protein: structure, processing, and physiolog- p. 4223–40.
ical actions. In: Bilezikian JP, Levine MA, Marcus R, 34. Cheng JB, Levine MA, Bell NH, Mangelsdorf DJ,
editors. The parathyroids basic and clinical concepts. Russell DW. Genetic evidence that the human
New York: Raven; 1994. p. 259–94. CYP2R1 enzyme is a key vitamin D
21. Strewler GJ. Mechanisms of disease: the physiology 25-hydroxylase. Proc Natl Acad Sci U S A.
of parathyroid hormone-related protein. New Engl J 2004;101(20):7711–5.
Med. 2000;342:177–85. 35. Al Mutair AN, Nasrat GH, Russell DW. Mutation of
22. Kronenberg H. Developmental regulation of the the CYP2R1 vitamin D 25-hydroxylase in a Saudi
growth plate. Nature. 2003;423:332–6. Arabian family with severe vitamin D deficiency.
23. VanHouten J, Dann P, McGeoch G, Brown EM, J Clin Endocrinol Metab. 2012;97(10):E2022–5.
Krapcho K, Neville M, et al. The calcium-sensing 36. White KE, Larsson TE, Econs MJ. The roles of spe-
receptor regulates mammary gland parathyroid cific genes implicated as circulating factors involved
hormone-related protein production and calcium in normal and disordered phosphate homeostasis:
transport. J Clin Invest. 2004;113(4):598–608. frizzled related protein-4, matrix extracellular
24. Hiremath M, Wysolmerski J. Parathyroid hormone- phosphoglycoprotein, and fibroblast growth factor
related protein specifies the mammary mesenchyme 23. Endocr Rev. 2006;27(3):221–41.
and regulates embryonic mammary development. 37. Haussler MR, Haussler CA, Jurutka PW, Thompson
J Mammary Gland Biol Neoplasia. 2013;18(2): PD, Hsieh JC, Remus LS, et al. The vitamin D hor-
171–7. mone and its nuclear receptor: molecular actions and
25. J€uppner H, Abou-Samra AB, Freeman MW, Kong disease states. J Endocrinol. 1997;154(Suppl):
XF, Schipani E, Richards J, et al. A G protein-linked S57–73.
receptor for parathyroid hormone and parathyroid 38. ADHR Consortium T, White KE, Evans WE,
hormone-related peptide. Science. 1991;254:1024–6. O’Riordan JLH, Speer MC, Econs MJ, Lorenz-
26. Abou-Samra AB, J€ uppner H, Force T, Freeman MW, Depiereux B, Grabowski M, Meittinger T, Strom
Kong XF, Schipani E, et al. Expression cloning of a TM. Autosomal dominant hypophosphataemic rick-
common receptor for parathyroid hormone and para- ets is associated with mutations in FGF23. Nat Genet.
thyroid hormone-related peptide from rat osteoblast- 2000;26:345–8.
like cells: a single receptor stimulates intracellular 39. White KE, Jonsson KB, Carn G, Hampson G, Spector
accumulation of both cAMP and inositol triphos- TD, Mannstadt M, et al. The autosomal dominant
phates and increases intracellular free calcium. Proc hypophosphatemic rickets (ADHR) gene is a secreted
Natl Acad Sci U S A. 1992;89:2732–6. polypeptide overexpressed by tumors that cause
27. Lee K, Deeds JD, Segre GV. Expression of parathy- phosphate wasting. J Clin Endocrinol Metabol.
roid hormone-related peptide and its receptor messen- 2001;86(2):497–500.
ger ribonucleic acid during fetal development of rats. 40. Benet-Pages A, Lorenz-Depiereux B, Zischka H,
Endocrinology. 1995;136:453–63. White K, Econs M, Strom T. FGF23 is processed by
28. Usdin TB, Gruber C, Bonner TI. Identification and proprotein convertases but not by PHEX. Bone.
functional expression of a receptor selectively recog- 2004;35:455–62.
nizing parathyroid hormone, the PTH2 receptor. 41. Tagliabracci VS, Engel JL, Wiley SE, Xiao J,
J Biol Chem. 1995;270:15455–8. Gonzalez DJ, Nidumanda Appaiah H, et al. Dynamic
29. Usdin TB, Hoare SRJ, Wang T, Mezey E, Kowalak regulation of FGF23 by Fam20C phosphorylation,
JA. Tip39: a new neuropeptide and PTH2-receptor GalNAc-T3 glycosylation, and furin proteolysis.
agonist from hypothalamus. Nat Neurosci. Proc Natl Acad Sci U S A. 2014;111(15):5520–5.
1999;2:941–3. 42. Goetz R, Nakada Y, Hu MC, Kurosu H, Wang L,
30. Dimitrov EL, Petrus E, Usdin TB. Tuberoin- Nakatani T, et al. Isolated C-terminal tail of FGF23
fundibular peptide of 39 residues (TIP39) signaling alleviates hypophosphatemia by inhibiting FGF23-
modulates acute and tonic nociception. Exp Neurol. FGFR-Klotho complex formation. Proc Natl Acad
2010;226(1):68–83. Sci U S A. 2010;107:407–12.
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 323

43. Shimada T, Yoneya T, Hino R, Takeuchi Y, calcification of infancy and pseudoxanthoma


Fukumoto S, Yamashita T, editors. Transgenic mice elasticum can be caused by mutations in either
expressing fibroblast growth factor 23 (FGF23) ENPP1 or ABCC6. Am J Hum Genet. 2012;90(1):
demonstrate hypophosphatemia with low serum 25–39.
1,25-dihydroxyvitamin D [1,25(OH)2D] and rickets/ 56. Ishikawa HO, Xu A, Ogura E, Manning G, Irvine
osteomalacia. Twenty-third Annual Meeting of the KD. The Raine syndrome protein FAM20C is a
American Society for Bone and Mineral Research, Golgi kinase that phosphorylates bio-mineralization
Phoenix; 2001; J Bone Mineral Res proteins. PLoS One. 2012;7(8):e42988.
44. Riminucci M, Collins M, Fedarko N, Cherman N, 57. Rafaelsen SH, Raeder H, Fagerheim AK,
Corsi A, White K, et al. FGF-23 in fibrous dysplasia Knappskog P, Carpenter TO, Johansson S, et al.
of bone and its relationship to renal phosphate Exome sequencing reveals FAM20c mutations asso-
wasting. J Clin Invest. 2003;112:683–92. ciated with fibroblast growth factor 23-related
45. Sitara D, Razzaque MS, Hesse M, Yoganathan S, hypophosphatemia, dental anomalies, and ectopic
Taguchi T, Erben RG, et al. Homozygous ablation of calcification. J Bone Miner Res Off J Am Soc Bone
fibroblast growth factor-23 results in hyperpho- Miner Res. 2013;28(6):1378–85.
sphatemia and impaired skeletogenesis, and reverses 58. Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H,
hypophosphatemia in Phex-deficient mice. Matrix Suga T, Utsugi T, et al. Mutation of the mouse klotho
Biol. 2004;23:421–32. gene leads to a syndrome resembling ageing. Nature.
46. Liu S, Zhou J, Tang W, Jiang X, Rowe DW, Quarles 1997;390(6655):45–51.
LD. Pathogenic role of Fgf23 in Hyp mice. Am J 59. Kuro-o M. Klotho as a regulator of fibroblast
Physiol Endocrinol Metab. 2006;291(1):E38–49. growth factor signaling and phosphate/calcium
47. Bonewald LF. The amazing osteocyte. J Bone Miner metabolism. Curr Opin Nephrol Hypertens.
Res Off J Am Soc Bone Miner Res. 2011;26(2): 2006;15(4):437–41.
229–38. 60. Urakawa I, Yamazaki Y, Shimada T, Iijima K,
48. Feng JQ, Ward LM, Liu S, Lu Y, Xie Y, Yuan B, Hasegawa H, Okawa K, et al. Klotho converts canon-
et al. Loss of DMP1 causes rickets and osteomalacia ical FGF receptor into a specific receptor for FGF23.
and identifies a role for osteocytes in mineral metab- Nature. 2006;444:770–4.
olism. Nat Genet. 2006;38(11):1310–5. 61. Ohnishi M, Nakatani T, Lanske B, Razzaque
49. Wang X, Wang S, Li C, Gao T, Liu Y, Rangiani A, MS. Reversal of mineral ion homeostasis and soft-
et al. Inactivation of a novel FGF23 regulator, tissue calcification of klotho knockout mice by dele-
FAM20C, leads to hypophosphatemic rickets in tion of vitamin D 1alpha-hydroxylase. Kidney Int.
mice. PLoS Genet. 2012;8(5):e1002708. 2009;75(11):1166–72.
50. Mackenzie NC, Zhu D, Milne EM, Van’t Hof R, 62. Shimada T, Kakitani M, Yamazaki Y, Hasegawa H,
Martin A, Darryl Quarles L, et al. Altered bone devel- Takeuchi Y, Fujita T, et al. Targeted ablation of Fgf23
opment and an increase in FGF-23 expression in demonstrates an essential physiological role of
Enpp1(/) mice. PLoS One. 2012;7(2):e32177. FGF23 in phosphate and vitamin D metabolism.
51. Lorenz-Depiereux B, Bastepe M, Benet-Pages A, J Clin Invest. 2004;113(4):561–8.
Amyere M, Wagenstaller J, Muller-Barth U, 63. Liu S, Brown T, Zhou J, Xiao Z, Awad H, Guilak F,
et al. DMP1 mutations in autosomal recessive et al. Role of matrix extracellular phosphogly-
hypophosphatemia implicate a bone matrix protein coprotein in the pathogenesis of X-linked
in the regulation of phosphate homeostasis. Nat hypophosphatemia. J Am Soc Nephrol. 2005;16:
Genet. 2006;38(11):1248–50. 1645–53.
52. Mäkitie O, Pereira RC, Kaitila I, Turan S, Bastepe M, 64. Shimada T, Yamazaki Y, Takahashi M, Hasegawa H,
Laine T, et al. Long-term clinical outcome and carrier Urakawa I, Oshima T, et al. Vitamin D receptor-
phenotype in autosomal recessive hypophosphatemia independent FGF23 actions in regulating phosphate
caused by a novel DMP1 mutation. J Bone Miner Res. and vitamin D metabolism. Am J Physiol Renal
2010;25:2165–174. Physiol. 2005;289(5):F1088–95.
53. Turan S, Aydin C, Bereket A, Akcay T, Guran T, 65. Jan De Beur S, Finnegan R, Vassiliadis J, Cook B,
Yaralioglu BA, et al. Identification of a novel dentin Barberio D, Estes S, et al. Tumors associated with
matrix protein-1 (DMP-1) mutation and dental anom- oncogenic osteomalacia express genes important in
alies in a kindred with autosomal recessive bone and mineral metabolism. J Bone Miner Res Off J
hypophosphatemia. Bone. 2010;46(2):402–9. Am Soc Bone Miner Res. 2002;17:1102–10.
54. Lorenz-Depiereux B, Schnabel D, Tiosano D, 66. Carpenter TO, Ellis BK, Insogna KL, Philbrick WM,
Hausler G, Strom TM. Loss-of-function ENPP1 muta- Sterpka J, Shimkets R. Fibroblast growth factor 7: an
tions cause both generalized arterial calcification of inhibitor of phosphate transport derived from onco-
infancy and autosomal-recessive hypophosphatemic genic osteomalacia-causing tumors. J Clin Endocrinol
rickets. Am J Hum Genet. 2010;86(2):267–72. Metab. 2005;90(2):1012–20.
55. Nitschke Y, Baujat G, Botschen U, Wittkampf T, du 67. Berndt T, Craig T, Bowe A, Vassiliadis J, Reczek D,
Moulin M, Stella J, et al. Generalized arterial Finnegan R, et al. Secreted frizzled-related protein 4 is
324 A. Sharma et al.

a potent tumor-derived phosphaturic agent. J Clin and biologically active. J Clin Endocrinol Metab.
Invest. 2003;112:785–94. 2010;95(2):578–85.
68. Rowe P, Kumagai Y, Gutierrez G, Garrett I, 81. Smith ER, Cai MM, McMahon LP, Holt SG. Biolog-
Blacher R, Rosen D, et al. MEPE has the properties ical variability of plasma intact and C-terminal FGF23
of an osteoblastic phosphatonin and minhibin. Bone. measurements. J Clin Endocrinol Metab. 2012;97(9):
2004;34:303–19. 3357–65.
69. Christov M, Koren S, Yuan Q, Baron R, Lanske 82. Larsson T, Zahradnik R, Lavigne J, Ljunggren Ö,
B. Genetic ablation of sfrp4 in mice does not affect J€
uppner H, Jonsson K. Immunohistochemical detec-
serum phosphate homeostasis. Endocrinology. tion of FGF-23 protein in tumors that cause oncogenic
2012;152(5):2031–6. osteomalacia. Eur J Endocrinol. 2003;148:269–76.
70. Thakker RV. Molecular genetics of parathyroid 83. Gutierrez O, Isakova T, Rhee E, Shah A, Holmes J,
disease. Curr Opin Endocrinol Diabetes Obes. Collerone G, et al. Fibroblast growth factor-23 miti-
1996;3:521–8. gates hyperphosphatemia but accentuates calcitriol
71. Szabo J, Heath B, Hill VM, Jackson CE, Zarbo RJ, deficiency in chronic kidney disease. J Am Soc
Mallette LE, et al. Hereditary hyperparathyroidism- Nephrol. 2005;16(7):2205–15.
jaw tumor syndrome: the endocrine tumor gene 84. Portale A, Wolf M, J€ uppner H, Cayetano S, Kumar J,
HRPT2 maps to chromosome 1q21-q31. Am J Hum Wesseling-Perry K, et al. Disordered FGF23 and
Genet. 1995;56(4):944–50. mineral metabolism in the chronic kidney disease in
72. Carpten JD, Robbins CM, Villablanca A, Forsberg L, children (CKiD) cohort. Clin J Am Soc Nephrol.
Presciuttini S, Bailey-Wilson J, et al. HRPT2, 2014;9:344–53.
encoding parafibromin, is mutated in 85. Au AY, McDonald K, Gill A, Sywak M, Diamond T,
hyperparathyroidism-jaw tumor syndrome. Nat Conigrave AD, et al. PTH mutation with primary
Genet. 2002;32(4):676–80. hyperparathyroidism and undetectable intact PTH. N
73. Thakker RV, Newey PJ, Walls GV, Bilezikian J, Engl J Med. 2008;359(11):1184–6.
Dralle H, Ebeling PR, et al. Clinical practice 86. Janicic N, Soliman E, Pausova Z, Seldin MF,
guidelines for multiple endocrine neoplasia type Riviere M, Szpirer J, et al. Mapping of the calcium-
1 (MEN1). J Clin Endocrinol Metab. 2012;97(9): sensing receptor gene (CASR) to human chromosome
2990–3011. 3q13.3-21 by fluorescence in situ hybridization, and
74. Hsu SC, Levine MA. Primary hyperparathyroidism in localization to rat chromosome 11 and mouse chro-
children and adolescents: the Johns Hopkins Chil- mosome 16. Mamm Genome Off J Int Mamm
dren’s Center experience 1984–2001. J Bone Miner Genome Soc. 1995;6(11):798–801.
Res Off J Am Soc Bone Miner Res. 2002;17 Suppl 2: 87. Pollak MR, Brown EM, WuChou YH, Hebert SC,
N44–50. Marx SJ, Steinmann B, et al. Mutations in the
75. Arnold A, Brown MF, Urena P, Gaz RD, Sarfati E, human Ca2+-sensing receptor gene cause familial
Drueke TB. Monoclonality of parathyroid tumors in hypocalciuric hypercalcemia and neonatal severe
chronic renal failure and in primary parathyroid hyperparathyroidism. Cell. 1993;75:1297–303.
hyperplasia. J Clin Invest. 1995;95(5):2047–53. 88. Chou YH, Pollak MR, Brandi ML, Toss G,
76. Buchwald PC, Akerstrom G, Westin G. Reduced Arnqvist H, Atkinson AB, et al. Mutations in the
p18INK4c, p21CIP1/WAF1 and p27KIP1 mRNA human Ca(2+)-sensing-receptor gene that cause
levels in tumours of primary and secondary hyper- familial hypocalciuric hypercalcemia. Am J Hum
parathyroidism. Clin Endocrinol (Oxf). 2004;60(3): Genet. 1995;56(5):1075–9.
389–93. 89. Pearce S, Trump D, Wooding C, Besser G, Chew S,
77. Bjorklund P, Åkerstrom G, Westin G. Accumulation Grant D, et al. Calcium-sensing receptor mutations in
of nonphosphorylated beta-catenin and c-myc in pri- familial benign hypercalcaemia and neonatal hyper-
mary and uremic secondary hyperparathyroid tumors. parathyroidism. J Clin Invest. 1995;96:2683–92.
J Clin Endocrinol Metab. 2007;92(1):338–44. 90. Janicic N, Pausova Z, Cole DE, Hendy GN. Insertion
78. Jonsson KB, Zahradnik R, Larsson T, White KE, of an Alu sequence in the Ca(2+)-sensing receptor
Sugimoto T, Imanishi Y, et al. Fibroblast growth gene in familial hypocalciuric hypercalcemia and
factor 23 in oncogenic osteomalacia and X-linked neonatal severe hyperparathyroidism. Am J Hum
hypophosphatemia. New Engl J Med. 2003;348(17): Genet. 1995;56(4):880–6.
1656–63. 91. Aida K, Koishi S, Inoue M, Nakazato M, Tawata M,
79. Gutierrez OM, Mannstadt M, Isakova T, Rauh-Hain Onaya T. Familial hypocalciuric hypercalcemia asso-
JA, Tamez H, Shah A, et al. Fibroblast growth factor ciated with mutation in the human Ca(2+)-sensing
23 and mortality among patients undergoing hemodi- receptor gene. J Clin Endocrinol Metab. 1995;80(9):
alysis. N Engl J Med. 2008;359(6):584–92. 2594–8.
80. Shimada T, Urakawa I, Isakova T, Yamazaki Y, 92. Heath III H, Odelberg S, Jackson CE, Teh BT,
Epstein M, Wesseling-Perry K, et al. Circulating Hayward N, Larsson C, et al. Clustered inactivating
fibroblast growth factor 23 in patients with end-stage mutations and benign polymorphisms of the calcium
renal disease treated by peritoneal dialysis is intact receptor gene in familial benign hypocalciuric
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 325

hypercalcemia suggest receptor functional domains. J for chronic pancreatitis? Scand J Gastroenterol.
Clin Endocrinol Metab. 1996;81:1312–7. 2006;41(3):343–8.
93. Nesbit MA, Hannan FM, Howles SA, Babinsky VN, 105. Pollak MR, Chou YH, Marx SJ, Steinmann B, Cole
Head RA, Cranston T, et al. Mutations affecting DE, Brandi ML, et al. Familial hypocalciuric hyper-
G-protein subunit alpha11 in hypercalcemia and calcemia and neonatal severe hyperparathyroidism.
hypocalcemia. N Engl J Med. 2013;368(26): Effects of mutant gene dosage on phenotype. J Clin
2476–86. Invest. 1994;93(3):1108–12.
94. Mannstadt M, Harris M, Bravenboer B, Chitturi S, 106. Bai M, Pearce SH, Kifor O, Trivedi S, Stauffer UG,
Dreijerink KM, Lambright DG, et al. Germline muta- Thakker RV, et al. In vivo and in vitro characterization
tions affecting Galpha11 in hypoparathyroidism. of neonatal hyperparathyroidism resulting from a de
N Engl J Med. 2013;368(26):2532–4. novo, heterozygous mutation in the Ca2+sensing
95. Nesbit MA, Hannan FM, Howles SA, Reed AA, receptor gene: normal maternal calcium homeostasis
Cranston T, Thakker CE, et al. Mutations in AP2S1 as a cause of secondary hyperparathyroidism in famil-
cause familial hypocalciuric hypercalcemia type ial benign hypocalciuric hypercalcemia. J Clin Invest.
3. Nat Genet. 2013;45(1):93–7. 1997;99(1):88–96.
96. Lloyd SE, Pannett AA, Dixon PH, Whyte MP, 107. Ward BK, Cameron FJ, Magno AL, McDonnell CM,
Thakker RV. Localization of familial benign Stuckey BG, Ratajczak T. A novel homozygous dele-
hypercalcemia, Oklahoma variant (FBHOk), to chro- tion in the calcium-sensing receptor ligand-binding
mosome 19q13. Am J Hum Genet. 1999;64(1): domain associated with neonatal severe hyperpara-
189–95. thyroidism. J Pediat Endocrinol Metab. 2006;19(1):
97. McMurtry CT, Schranck FW, Walkenhorst DA, 93–100.
Murphy WA, Kocher DB, Teitelbaum SL, et al. 108. Marx SJ, Fraser D, Rapoport A. Familial
Significant developmental elevation in serum para- hypocalciuric hypercalcemia. Mild expression of the
thyroid hormone levels in a large kindred with famil- gene in heterozygotes and severe expression in homo-
ial benign (hypocalciuric) hypercalcemia. Am J Med. zygotes. Am J Med. 1985;78(1):15–22.
1992;93(3):247–58. 109. Cole D, Forsythe CR, Dooley JM, Grantmyre EB,
98. Trump D, Whyte MP, Wooding C, Pang JT, Pearce Salisbury SR. Primary neonatal hyperparathyroidism:
SH, Kocher DB, et al. Linkage studies in a kindred a devastating neurodevelopmental disorder if left
from Oklahoma, with familial benign (hypocalciuric) untreated. J Craniofac Genet Dev Biol. 1990;10(2):
hypercalcaemia (FBH) and developmental elevations 205–14.
in serum parathyroid hormone levels, indicate a third 110. Toke J, Czirjak G, Patocs A, Enyedi B, Gergics P,
locus for FBH. Hum Genet. 1995;96(2):183–7. Csakvary V, et al. Neonatal severe hyperparathyroid-
99. Pearce SH, Bai M, Quinn SJ, Kifor O, Brown EM, ism associated with a novel de novo heterozygous
Thakker RV. Functional characterization of calcium- R551K inactivating mutation and a heterozygous
sensing receptor mutations expressed in human A986S polymorphism of the calcium-sensing recep-
embryonic kidney cells. J Clin Invest. 1996;98(8): tor gene. Clin Endocrinol (Oxf). 2007;67(3):385–92.
1860–6. 111. Kifor O, Moore Jr FD, Delaney M, Garber J, Hendy
100. Bai M, Quinn S, Trivedi S, Kifor O, Pearce SH, Pollak GN, Butters R, et al. A syndrome of hypocalciuric
MR, et al. Expression and characterization of hypercalcemia caused by autoantibodies directed at
inactivating and activating mutations in the human the calcium-sensing receptor. J Clin Endocrinol
Ca2+-sensing receptor. J Biol Chem. 1996;271(32): Metab. 2003;88(1):60–72.
19537–45. 112. Pallais J, Kifor O, Chen Y, Slovik D, Brown E.
101. Morten KJ, Cooper JM, Brown GK, Lake BD, Acquired hypocalciuric hypercalcemia due to auto-
Pike D, Poulton J. A new point mutation associated antibodies against the calcium-sensing receptor.
with mitochondrial encephalomyopathy. Hum Mol N Engl J Med. 2004;351:362–9.
Genet. 1993;2(12):2081–7. 113. Makita N, Sato J, Manaka K, Shoji Y, Oishi A,
102. Brown EM, MacLeod RJ. Extracellular calcium sens- Hashimoto M, et al. An acquired hypocalciuric hyper-
ing and extracellular calcium signaling. Physiol Rev. calcemia autoantibody induces allosteric transition
2001;81(1):239–97. among active human Ca-sensing receptor conforma-
103. Zajickova K, Vrbikova J, Canaff L, Pawelek PD, tions. Proc Natl Acad Sci U S A. 2007;104(13):
Goltzman D, Hendy GN. Identification and functional 5443–8.
characterization of a novel mutation in the calcium- 114. Frame B, Poznanski AK. Conditions that may be
sensing receptor gene in familial hypocalciuric hyper- confused with rickets. In: DeLuca HF, Anast AS,
calcemia: modulation of clinical severity by vitamin editor. Pediatric diseases related to calcium.
D status. J Clin Endocrinol Metab. 2007;92(7): New York: Elsevier; 1980. p. 269–89.
2616–23. 115. J€
uppner H, Schipani E, Silve C. Jansen’s metaphyseal
104. Felderbauer P, Klein W, Bulut K, Ansorge N, chondrodysplasia and Blomstrand’s lethal
Dekomien G, Werner I, et al. Mutations in the chondrodysplasia: two genetic disorders caused by
calcium-sensing receptor: a new genetic risk factor PTH/PTHrP receptor mutations. In: Bilezikian J,
326 A. Sharma et al.

Raisz L, Rodan G, editors. Principles of bone biology, 127. Pober BR. Williams-Beuren syndrome. N Engl J
vol. 2. San Diego: Academic; 2002. p. 1117–35. Med. 2010;362(3):239–52.
116. Schipani E, Kruse K, J€ uppner H. A constitutively 128. Li D, Brooke B, Davis E, Mecham R, Sorensen L,
active mutant PTH-PTHrP receptor in Jansen-type Boak B, et al. Elastin is an essential determinant of
metaphyseal chondrodysplasia. Science. 1995;268: arterial morphogenesis. Nature. 1998;393:276–80.
98–100. 129. Ewart AK, Morris CA, Atkinson DL, Jin W,
117. Schipani E, Langman CB, Parfitt AM, Jensen GS, Sternes K, Spallone P, et al. Hemizygosity at the
Kikuchi S, Kooh SW, et al. Constitutively activated elastin locus in a developmental disorder. Williams
receptors for parathyroid hormone and parathyroid syndrome. Nat Genet. 1993;5:11–6.
hormone-related peptide in Jansen’s metaphyseal 130. Lowery MC, Morris CA, Ewart A, Brothman LJ, Zhu
chondrodysplasia. N Engl J Med. 1996;335:708–14. XL, Leonard CO, et al. Strong correlation of elastin
118. Schipani E, Langman CB, Hunzelman J, LeMerrer M, deletions, detected by FISH, with Williams syn-
Loke KY, Dillon MJ, et al. A novel PTH/PTHrP drome: evaluation of 235 patients. Am J Hum
receptor mutation in Jansen’s metaphyseal Genet. 1995;57(1):49–53.
chondrodysplasia. J Clin Endocrinol Metab. 131. Tassabehji M, Metcalfe K, Fergusson WD,
1999;84:3052–7. Carette MJ, Dore JK, Donnai D, et al. LIM-kinase
119. Minagawa M, Arakawa K, Minamitani K, Yasuda T, deleted in Williams syndrome. Nat Genet. 1996;13
Niimi H. Jansen-type metaphyseal chondrodysplasia: (3):272–3.
analysis of PTH/PTH-related protein receptor mes- 132. van Hagen JM, van der Geest JN, van der Giessen RS,
senger RNA by the reverse transcription-polymerase Lagers-van Haselen GC, Eussen HJ, Gille JJ,
chain method. Endocr J. 1997;44:493–9. et al. Contribution of CYLN2 and GTF2IRD1 to
120. Brown WW, J€uppner H, Langman CB, Price H, Far- neurological and cognitive symptoms in Williams
row EG, White KE, et al. Hypophosphatemia with syndrome. Neurobiol Dis. 2007;26(1):112–24.
elevations in serum fibroblast growth factor 23 in a 133. Del Campo M, Antonell A, Magano LF, Munoz FJ,
child with Jansen’s metaphyseal chondrodysplasia. Flores R, Bayes M, et al. Hemizygosity at the NCF1
J Clin Endocrinol Metab. 2009;94(1):17–20. gene in patients with Williams-Beuren syndrome
121. Onuchic L, Ferraz-de-Souza B, Mendonca BB, decreases their risk of hypertension. Am J Hum
Correa PH, Martin RM. Potential effects of Genet. 2006;78(4):533–42.
alendronate on fibroblast growth factor 23 levels and 134. Edelmann L, Prosnitz A, Pardo S, Bhatt J, Cohen N,
effective control of hypercalciuria in an adult with Lauriat T, et al. An atypical deletion of the Williams-
Jansen’s metaphyseal chondrodysplasia. J Clin Beuren syndrome interval implicates genes associated
Endocrinol Metab. 2012;97(4):1098–103. with defective visuospatial processing and autism.
122. Savoldi G, Izzi C, Signorelli M, Bondioni MP, J Med Genet. 2007;44(2):136–43.
Romani C, Lanzi G, et al. Prenatal presentation and 135. Merla G, Howald C, Henrichsen CN, Lyle R, Wyss C,
postnatal evolution of a patient with Jansen Zabot MT, et al. Submicroscopic deletion in patients
metaphyseal dysplasia with a novel missense muta- with Williams-Beuren syndrome influences expres-
tion in PTH1R. Am J Med Genet A. 2013;161(10): sion levels of the nonhemizygous flanking genes.
2614–9. Am J Hum Genet. 2006;79(2):332–41.
123. Schipani E, Lanske B, Hunzelman J, Kovacs CS, 136. Cagle AP, Waguespack SG, Buckingham BA,
Lee K, Pirro A, et al. Targeted expression of consti- Shankar RR, Dimeglio LA. Severe infantile hypercal-
tutively active PTH/PTHrP receptors delays endo- cemia associated with Williams syndrome success-
chondral bone formation and rescues PTHrP-less fully treated with intravenously administered
mice. Proc Natl Acad Sci U S A. 1997;94:13689–94. pamidronate. Pediatrics. 2004;114(4):1091–5.
124. Beier F, LuValle P. The cyclin D1 and cyclin a genes 137. Perez Jurado LA, Li X, Francke U. The human calci-
are targets of activated PTH/PTHrP receptors in tonin receptor gene (CALCR) at 7q21.3 is outside the
Jansen’s metaphyseal chondrodysplasia. Mol deletion associated with the Williams syndrome.
Endocrinol. 2002;16(9):2163–73. Cytogenet Cell Genet. 1995;70(3–4):246–9.
125. Bastepe M, Raas-Rothschild A, Silver J, Weissman I, 138. Garabedian M, Jacqz E, Guillozo H, Grimberg R,
J€uppner H, Gillis D. A form of Jansen’s metaphyseal Guillot M, Gagnadoux MF, et al. Elevated plasma
chondrodysplasia with limited metabolic and skeletal 1,25-dihydroxyvitamin D concentrations in infants
abnormalities is caused by a novel activating with hypercalcemia and an elfin facies. N Engl J
PTH/PTHrP receptor mutation. J Clin Endocrinol Med. 1985;312(15):948–52.
Metab. 2004;89:3595–600. 139. Taylor AB, Stern PH, Bell NH. Abnormal regulation
126. Schipani E, Jensen GS, Pincus J, Nissenson RA, of circulating 25-hydroxyvitamin D in the Williams
Gardella TJ, J€uppner H. Constitutive activation of syndrome. N Engl J Med. 1982;306(16):972–5.
the cAMP signaling pathway by parathyroid hormone 140. Lameris AL, Huybers S, Burke JR, Monnens LA,
(PTH)/PTH-related peptide (PTHrP) receptors Bindels RJ, Hoenderop JG. Involvement of claudin
mutated at the two loci for Jansen’s metaphyseal 3 and claudin 4 in idiopathic infantile
chondrodysplasia. Mol Endocrinol. 1997;11:851–8. hypercalcaemia: a novel hypothesis? Nephrol Dial
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 327

Transplant Off Publ Eur Dial Transplant Assoc Eur 153. Winer KK, Yanovski JA, Sarani B, Cutler Jr GB. A
Ren Assoc. 2010;25(11):3504–9. randomized, cross-over trial of once-daily versus
141. Letavernier E, Rodenas A, Guerrot D, Haymann JP. twice-daily parathyroid hormone 1–34 in treatment
Williams-Beuren syndrome hypercalcemia: is of hypoparathyroidism. J Clin Endocrinol Metab.
TRPC3 a novel mediator in calcium homeostasis? 1998;83(10):3480–6.
Pediatrics. 2012;129(6):e1626–30. 154. Winer KK, Zhang B, Shrader JA, Peterson D,
142. Schlingmann KP, Kaufmann M, Weber S, Irwin A, Smith M, Albert PS, et al. Synthetic human
Goos C, John U, et al. Mutations in CYP24A1 and parathyroid hormone 1–34 replacement therapy: a
idiopathic infantile hypercalcemia. N Engl J Med. randomized crossover trial comparing pump
2011;365:410–21. versus injections in the treatment of chronic hypo-
143. Dauber A, Nguyen TT, Sochett E, Cole DE, Horst R, parathyroidism. J Clin Endocrinol Metab. 2012;97
Abrams SA, et al. Genetic defect in CYP24A1, the (2):391–9.
vitamin D 24-hydroxylase gene, in a patient with 155. Mannstadt M, Clarke BL, Vokes T, Brandi ML,
severe infantile hypercalcemia. J Clin Endocrinol Ranganath L, Fraser WD, et al. Efficacy and safety
Metab. 2012;97(2):E268–74. of recombinant human parathyroid hormone (1–84) in
144. Fencl F, Blahova K, Schlingmann KP, Konrad M, hypoparathyroidism (REPLACE): a double-blind,
Seeman T. Severe hypercalcemic crisis in an infant placebo-controlled, randomised, phase 3 study.
with idiopathic infantile hypercalcemia caused Lancet Diabetes Endocrinol. 2013;1(4):275–83.
by mutation in CYP24A1 gene. Eur J Pediatr. 156. Winer KK, Fulton KA, Albert PS, Cutler GB, Jr.
2013;172(1):45–9. Effects of pump versus twice-daily injection delivery
145. Tebben PJ, Milliner DS, Horst RL, Harris PC, Singh of synthetic parathyroid hormone 1-34 in children
RJ, Wu Y, et al. Hypercalcemia, hypercalciuria, and with severe congenital hypoparathyroidism. J Pediatr.
elevated calcitriol concentrations with autosomal 2014;165:556–63.
dominant transmission due to CYP24A1 mutations: 157. Linglart A, Rothenbuhler A, Gueorgieva I, Lucchini
effects of ketoconazole therapy. J Clin Endocrinol P, Silve C, Bougneres P. Long-term results of contin-
Metab. 2012;97(3):E423–7. uous subcutaneous recombinant PTH (1–34) infusion
146. Castanet M, Mallet E, Kottler ML. Lightwood syn- in children with refractory hypoparathyroidism. J Clin
drome revisited with a novel mutation in CYP24 and Endocrinol Metab. 2011;96(11):3308–12.
vitamin D supplement recommendations. J Pediatr. 158. Arnold A, Horst SA, Gardella TJ, Baba H, Levine
2013;163(4):1208–10. MA, Kronenberg HM. Mutation of the signal peptide-
147. Zwermann O, Piepkorn B, Engelbach M, Beyer J, encoding region of the preproparathyroid hormone
Kann P. Abnormal pentagastrin response in a patient gene in familial isolated hypoparathyroidism. J Clin
with pseudohypoparathyroidism. Exp Clin Invest. 1990;86:1084–7.
Endocrinol Diabetes. 2002;110(2):86–91. 159. Karaplis AC, Lim SK, Baba H, Arnold A,
148. Mitchell DM, Regan S, Cooley MR, Lauter KB, Kronenberg HM. Inefficient membrane targeting,
Vrla MC, Becker CB, et al. Long-term follow-up of translocation, and proteolytic processing by signal
patients with hypoparathyroidism. J Clin Endocrinol peptidase of a mutant preproparathyroid hormone
Metab. 2012;97(12):4507–14. protein. J Biol Chem. 1995;270:1629–35.
149. Winer KK, Ko CW, Reynolds JC, Dowdy K, Keil M, 160. Datta R, Waheed A, Shah GN, Sly WS. Signal
Peterson D, et al. Long-term treatment of hypopara- sequence mutation in autosomal dominant form of
thyroidism: a randomized controlled study comparing hypoparathyroidism induces apoptosis that is
parathyroid hormone-(1–34) versus calcitriol and corrected by a chemical chaperone. Proc Natl Acad
calcium. J Clin Endocrinol Metab. 2003;88(9): Sci U S A. 2007;104(50):19989–94.
4214–20. 161. Sunthornthepvarakul T, Churesigaew S,
150. Winer KK, Sinaii N, Peterson D, Sainz Jr B, Cutler Jr Ngowngarmratana S. A novel mutation of the signal
GB. Effects of once versus twice-daily parathyroid peptide of the preproparathyroid hormone gene asso-
hormone 1–34 therapy in children with hypoparathy- ciated with autosomal recessive familial isolated
roidism. J Clin Endocrinol Metab. 2008;93(9): hypoparathyroidism. J Clin Endocrinol Metab.
3389–95. 1999;84(10):3792–6.
151. Winer KK, Sinaii N, Reynolds J, Peterson D, 162. Parkinson D, Thakker R. A donor splice site mutation
Dowdy K, Cutler Jr GB. Long-term treatment of in the parathyroid hormone gene is associated with
12 children with chronic hypoparathyroidism: a ran- autosomal recessive hypoparathyroidism. Nat Genet.
domized trial comparing synthetic human parathyroid 1992;1:149–53.
hormone 1–34 versus calcitriol and calcium. J Clin 163. Ertl DA, Stary S, Streubel B, Raimann A, Haeusler
Endocrinol Metab. 2010;95(6):2680–8. G. A novel homozygous mutation in the parathyroid
152. Winer KK, Yanovski JA, Cutler Jr GB. Synthetic hormone gene (PTH) in a girl with isolated hypopara-
human parathyroid hormone 1–34 vs calcitriol and thyroidism. Bone. 2012;51(3):629–32.
calcium in the treatment of hypoparathyroidism. 164. Tomar N, Gupta N, Goswami R. Calcium-sensing
JAMA. 1996;276(8):631–6. receptor autoantibodies and idiopathic
328 A. Sharma et al.

hypoparathyroidism. J Clin Endocrinol Metab. the calcium-sensing receptor gene: effect of a


2013;98(9):3884–91. dominant-negative GCM2 mutant associated with
165. Kawahara M, Iwasaki Y, Sakaguchi K, Taguchi T, autosomal dominant hypoparathyroidism. Hum
Nishiyama M, Nigawara T, et al. Involvement of Mutat. 2009;30(1):85–92.
GCMB in the transcriptional regulation of the 177. Peden VH. True idiopathic hypoparathyroidism as a
human parathyroid hormone gene in a parathyroid- sex-linked recessive trait. Am J Hum Genet.
derived cell line PT-r: effects of calcium and 1960;12:323–37.
1,25(OH)2D3. Bone. 2010;47(3):534–41. 178. Mumm S, Whyte MP, Thakker RV, Buetow KH,
166. G€unther T, Chen ZF, Kim J, Priemel M, Rueger JM, Schlessinger D. mtDNA analysis shows common
Amling M, et al. Genetic ablation of parathyroid ancestry in two kindreds with X-linked recessive
glands reveals another source of parathyroid hor- hypoparathyroidism and reveals a heteroplasmic
mone. Nature. 2000;406(6792):199–203. silent mutation. Am J Hum Genet. 1997;60(1):153–9.
167. Liu Z, Yu S, Manley NR. Gcm2 is required for the 179. Whyte M, Kim G, Kosanovich M. Absence of para-
differentiation and survival of parathyroid precursor thyroid tissue in sex-linked recessive hypoparathy-
cells in the parathyroid/thymus primordia. Dev Biol. roidism. J Paediatr. 1986;109:915.
2007;305(1):333–46. 180. Thakker RV, Davies KE, Whyte MP, Wooding C,
168. Ding C, Buckingham B, Levine MA. Familial iso- O’Riordan JL. Mapping the gene causing X-linked
lated hypoparathyroidism caused by a mutation in the recessive idiopathic hypoparathyroidism to Xq26-
gene for the transcription factor GCMB. J Clin Invest. Xq27 by linkage studies. J Clin Invest. 1990;86(1):
2001;108(8):1215–20. 40–5.
169. Thomee C, Schubert SW, Parma J, Le PQ, 181. Bowl M, Nesbit M, Harding B, Levy E, Jefferson A,
Hashemolhosseini S, Wegner M, et al. GCMB muta- Volpi E, et al. An interstitial deletion-insertion involv-
tion in familial isolated hypoparathyroidism with ing chromosomes 2p25.3 and Xq27.1, near SOX3,
residual secretion of parathyroid hormone. J Clin causes X-linked recessive hypoparathyroidism. J Clin
Endocrinol Metab. 2005;90(5):2487–92. Invest. 2005;115:2822–31.
170. Baumber L, Tufarelli C, Patel S, King P, Johnson CA, 182. Ahonen P. Autoimmune polyendocrinopathy–candi-
Maher ER, et al. Identification of a novel mutation dosis–ectodermal dystrophy (APECED): autosomal
disrupting the DNA binding activity of GCM2 in recessive inheritance. Clin Genet. 1985;27(6):
autosomal recessive familial isolated hypoparathy- 535–42.
roidism. J Med Genet. 2005;42(5):443–8. 183. Goswami R, Singh A, Gupta N, Rani R. Presence of
171. Maret A, Ding C, Kornfield SL, Levine MA. Analysis strong association of the major histocompatibility
of the GCM2 gene in isolated hypoparathyroidism: a complex (MHC) class I allele HLA-A*26:01 with
molecular and biochemical study. J Clin Endocrinol idiopathic hypoparathyroidism. J Clin Endocrinol
Metab. 2008;93(4):1426–32. Metab. 2012;97(9):E1820–4.
172. Bowl MR, Mirczuk SM, Grigorieva IV, Piret SE, 184. Aaltonen J, Bjorses P, Sandkuijl L, Perheentupa J,
Cranston T, Southam L, et al. Identification and char- Peltonen L. An autosomal locus causing autoimmune
acterization of novel parathyroid-specific transcrip- disease: autoimmune polyglandular disease type I
tion factor Glial Cells Missing Homolog B (GCMB) assigned to chromosome 21. Nat Genet. 1994;8(1):
mutations in eight families with autosomal recessive 83–7.
hypoparathyroidism. Hum Mol Genet. 2010;19(10): 185. Nagamine K, Peterson P, Scott HS, Kudoh J,
2028–38. Minoshima S, Heino M, et al. Positional cloning of
173. Mirczuk SM, Bowl MR, Nesbit MA, Cranston T, the APECED gene. Nat Genet. 1997;17(4):393–8.
Fratter C, Allgrove J, et al. A missense glial cells 186. Scott HS, Heino M, Peterson P, Mittaz L, Lalioti MD,
missing homolog B (GCMB) mutation, Asn502His, Betterle C, et al. Common mutations in autoimmune
causes autosomal dominant hypoparathyroidism. polyendocrinopathy-candidiasis-ectodermal dystro-
J Clin Endocrinol Metab. 2010;95(7):3512–6. phy patients of different origins. Mol Endocrinol.
174. Tomar N, Bora H, Singh R, Gupta N, Kaur P, 1998;12(8):1112–9.
Chauhan SS, et al. Presence and significance of a 187. Cihakova D, Trebusak K, Heino M, Fadeyev V,
R110W mutation in the DNA-binding domain of Tiulpakov A, Battelino T, et al. Novel AIRE muta-
GCM2 gene in patients with isolated hypoparathy- tions and P450 cytochrome autoantibodies in Central
roidism and their family members. Eur J Endocrinol. and Eastern European patients with APECED. Hum
2010;162(2):407–21. Mutat. 2001;18(3):225–32.
175. Mannstadt M, Bertrand G, Muresan M, Weryha G, 188. Wang CY, Davoodi-Semiromi A, Huang W,
Leheup B, Pulusani SR, et al. Dominant-negative Connor E, Shi JD, She JX. Characterization of muta-
GCMB mutations cause an autosomal dominant tions in patients with autoimmune polyglandular syn-
form of hypoparathyroidism. J Clin Endocrinol drome type 1 (APS1). Hum Genet. 1998;103(6):
Metab. 2008;93(9):3568–76. 681–5.
176. Canaff L, Zhou X, Mosesova I, Cole DE, Hendy 189. Heino M, Scott HS, Chen Q, Peterson P, Maebpaa U,
GN. Glial Cells Missing-2 (GCM2) transactivates Papasavvas MP, et al. Mutation analyses of North
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 329

American APS-1 patients. Hum Mutat. 1999;13(1): 202. Meager A, Visvalingam K, Peterson P, Moll K,
69–74. Murumagi A, Krohn K, et al. Anti-interferon autoan-
190. Pearce SH, Cheetham T, Imrie H, Vaidya B, Barnes tibodies in autoimmune polyendocrinopathy syn-
ND, Bilous RW, et al. A common and recurrent 13-bp drome type 1. PLoS Med. 2006;3(7):e289.
deletion in the autoimmune regulator gene in British 203. Wolff AS, Sarkadi AK, Marodi L, Karner J, Orlova E,
kindreds with autoimmune polyendocrinopathy type Oftedal BE, et al. Anti-cytokine autoantibodies pre-
1. Am J Hum Genet. 1998;63(6):1675–84. ceding onset of autoimmune polyendocrine syndrome
191. Bjorses P, Halonen M, Palvimo JJ, Kolmer M, type I features in early childhood. J Clin Immunol.
Aaltonen J, Ellonen P, et al. Mutations in the AIRE 2013;33(8):1341–8.
gene: effects on subcellular location and 204. Lammer EJ, Opitz JM. The DiGeorge anomaly as a
transactivation function of the autoimmune developmental field defect. Am J Med Genet Suppl.
polyendocrinopathy-candidiasis-ectodermal dystro- 1986;2:113–27.
phy protein. Am J Hum Genet. 2000;66(2):378–92. 205. Gong W, Emanuel BS, Collins J, Kim DH, Wang Z,
192. Rosatelli MC, Meloni A, Devoto M, Cao A, Scott HS, Chen F, et al. A transcription map of the DiGeorge
Peterson P, et al. A common mutation in Sardinian and velo-cardio-facial syndrome minimal critical
autoimmune polyendocrinopathy-candidiasis-ecto- region on 22q11. Hum Mol Genet. 1996;5(6):
dermal dystrophy patients. Hum Genet. 1998;103 789–800.
(4):428–34. 206. Scambler PJ. The 22q11 deletion syndromes. Hum
193. Matsuo T, Noguchi Y, Shindo M, Morita Y, Oda Y, Mol Genet. 2000;9(16):2421–6.
Yoshida E, et al. Regulation of human autoimmune 207. Scambler PJ, Carey AH, Wyse RK, Roach S,
regulator (AIRE) gene translation by miR-220b. Dumanski JP, Nordenskjold M, et al. Microdeletions
Gene. 2013;530(1):19–25. within 22q11 associated with sporadic and familial
194. Liston A, Lesage S, Wilson J, Peltonen L, Goodnow DiGeorge syndrome. Genomics. 1991;10(1):201–6.
CC. Aire regulates negative selection of organ- 208. Monaco G, Pignata C, Rossi E, Mascellaro O,
specific T cells. Nat Immunol. 2003;4(4):350–4. Cocozza S, Ciccimarra F. DiGeorge anomaly associ-
195. Meloni A, Incani F, Corda D, Cao A, Rosatelli ated with 10p deletion. Am J Med Genet. 1991;39(2):
MC. Role of PHD fingers and COOH-terminal 215–6.
30 amino acids in AIRE transactivation activity. Mol 209. Stoller JZ, Epstein JA. Identification of a
Immunol. 2008;45(3):805–9. novel nuclear localization signal in Tbx1 that is
196. Su MA, Giang K, Zumer K, Jiang H, Oven I, Rinn JL, deleted in DiGeorge syndrome patients harboring
et al. Mechanisms of an autoimmunity syndrome in the 1223delC mutation. Hum Mol Genet.
mice caused by a dominant mutation in Aire. J Clin 2005;14(7):885–92.
Invest. 2008;118(5):1712–26. 210. Conley ME, Beckwith JB, Mancer JF, Tenckhoff
197. Laakso SM, Kekalainen E, Heikkila N, L. The spectrum of the DiGeorge syndrome. J Pediatr.
Mannerstrom H, Kisand K, Peterson P, et al. In vivo 1979;94(6):883–90.
analysis of helper T cell responses in patients with 211. Ryan AK, Goodship JA, Wilson DI, Philip N, Levy A,
autoimmune polyendocrinopathy – candidiasis – Seidel H, et al. Spectrum of clinical features associ-
ectodermal dystrophy provides evidence in support ated with interstitial chromosome 22q11 deletions:
of an IL-22 defect. Autoimmunity. 2014;47:556–62. a European collaborative study. J Med Genet.
198. Gavalas NG, Kemp EH, Krohn KJ, Brown EM, Wat- 1997;34(10):798–804.
son PF, Weetman AP. The calcium-sensing receptor is 212. Cuneo BF, Langman CB, Ilbawi MN,
a target of autoantibodies in patients with autoim- Ramakrishnan V, Cutilletta A, Driscoll DA. Latent
mune polyendocrine syndrome type 1. J Clin hypoparathyroidism in children with conotruncal car-
Endocrinol Metab. 2007;92(6):2107–14. diac defects. Circulation. 1996;93(9):1702–8.
199. Alimohammadi M, Bjorklund P, Hallgren A, 213. Taylor SC, Morris G, Wilson D, Davies SJ, Gregory
Pontynen N, Szinnai G, Shikama N, et al. Autoim- JW. Hypoparathyroidism and 22q11 deletion syn-
mune polyendocrine syndrome type 1 and NALP5, drome. Arch Dis Child. 2003;88(6):520–2.
a parathyroid autoantigen. N Engl J Med. 214. Gidding SS, Minciotti AL, Langman CB. Unmasking
2008;358(10):1018–28. of hypoparathyroidism in familial partial DiGeorge
200. Kemp EH, Habibullah M, Kluger N, Ranki A, Sandhu syndrome by challenge with disodium edetate. N Engl
HK, Krohn KJ, et al. Prevalence and clinical associ- J Med. 1988;319(24):1589–91.
ations of calcium-sensing receptor and NALP5 215. Augusseau S, Jouk S, Jalbert P, Prieur M. DiGeorge
autoantibodies in Finnish APECED patients. J Clin syndrome and 22q11 rearrangements. Hum Genet.
Endocrinol Metab. 2014;99(3):1064–71. 1986;74(2):206.
201. Meloni A, Furcas M, Cetani F, Marcocci C, Falorni A, 216. Budarf ML, Collins J, Gong W, Roe B, Wang Z,
Perniola R, et al. Autoantibodies against type I Bailey LC, et al. Cloning a balanced translocation
interferons as an additional diagnostic criterion for associated with DiGeorge syndrome and identifica-
autoimmune polyendocrine syndrome type I. J Clin tion of a disrupted candidate gene. Nat Genet.
Endocrinol Metab. 2008;93(11):4389–97. 1995;10(3):269–78.
330 A. Sharma et al.

217. Yamagishi H, Garg V, Matsuoka R, Thomas T, 231. Muroya K, Hasegawa T, Ito Y, Nagai T, Isotani H,
Srivastava D. A molecular pathway revealing a Iwata Y, et al. GATA3 abnormalities and the pheno-
genetic basis for human cardiac and craniofacial typic spectrum of HDR syndrome. J Med Genet.
defects. Science. 1999;283(5405):1158–61. 2001;38(6):374–80.
218. Lindsay EA, Botta A, Jurecic V, Carattini-Rivera S, 232. Nesbit MA, Bowl MR, Harding B, Ali A, Ayala A,
Cheah YC, Rosenblatt HM, et al. Congenital heart Crowe C, et al. Characterization of GATA3 mutations
disease in mice deficient for the DiGeorge syndrome in the hypoparathyroidism, deafness, and renal dys-
region. Nature. 1999;401(6751):379–83. plasia (HDR) syndrome. J Biol Chem. 2004;279(21):
219. Magnaghi P, Roberts C, Lorain S, Lipinski M, 22624–34.
Scambler PJ. HIRA, a mammalian homologue of Sac- 233. Tsang AP, Visvader JE, Turner CA, Fujiwara Y, Yu C,
charomyces cerevisiae transcriptional co-repressors, Weiss MJ, et al. FOG, a multitype zinc finger protein,
interacts with Pax3. Nat Genet. 1998;20(1):74–7. acts as a cofactor for transcription factor GATA-1 in
220. Jerome LA, Papaioannou VE. DiGeorge syndrome erythroid and megakaryocytic differentiation. Cell.
phenotype in mice mutant for the T-box gene, Tbx1. 1997;90(1):109–19.
Nat Genet. 2001;27(3):286–91. 234. Dai YS, Markham BE. p300 Functions as a
221. Yagi H, Furutani Y, Hamada H, Sasaki T, Asakawa S, coactivator of transcription factor GATA-4. J Biol
Minoshima S, et al. Role of TBX1 in human Chem. 2001;276(40):37178–85.
del22q11.2 syndrome. Lancet. 2003;362(9393): 235. Ali A, Christie PT, Grigorieva IV, Harding B,
1366–73. Van Esch H, Ahmed SF, et al. Functional characteri-
222. Baldini A. DiGeorge’s syndrome: a gene at last. Lan- zation of GATA3 mutations causing the
cet. 2003;362(9393):1342–3. hypoparathyroidism-deafness-renal (HDR) dysplasia
223. Liao J, Kochilas L, Nowotschin S, Arnold JS, syndrome: insight into mechanisms of DNA binding
Aggarwal VS, Epstein JA, et al. Full spectrum of by the GATA3 transcription factor. Hum Mol Genet.
malformations in velo-cardio-facial syndrome/ 2007;16(3):265–75.
DiGeorge syndrome mouse models by altering Tbx1 236. Saito T, Fukumoto S, Ito N, Suzuki H, Igarashi T,
dosage. Hum Mol Genet. 2004;13(15):1577–85. Fujita T. A novel mutation in the GATA3 gene
224. Guris DL, Fantes J, Tara D, Druker BJ, Imamoto of a Japanese patient with PTH-deficient hypopara-
A. Mice lacking the homologue of the human thyroidism. J Bone Miner Metab. 2009;27(3):
22q11.2 gene CRKL phenocopy neurocristopathies 386–9.
of DiGeorge syndrome. Nat Genet. 2001;27(3): 237. Fukai R, Ochi N, Murakami A, Nakashima M,
293–8. Tsurusaki Y, Saitsu H, et al. Co-occurrence of 22q11
225. Paylor R, Glaser B, Mupo A, Ataliotis P, Spencer C, deletion syndrome and HDR syndrome. Am J Med
Sobotka A, et al. Tbx1 haploinsufficiency is linked to Genet A. 2013;161(10):2576–81.
behavioral disorders in mice and humans: implica- 238. Pandolfi PP, Roth ME, Karis A, Leonard MW,
tions for 22q11 deletion syndrome. Proc Natl Acad Dzierzak E, Grosveld FG, et al. Targeted disruption
Sci U S A. 2006;103(20):7729–34. of the GATA3 gene causes severe abnormalities in the
226. Gogos JA, Santha M, Takacs Z, Beck KD, Luine V, nervous system and in fetal liver haematopoiesis. Nat
Lucas LR, et al. The gene encoding proline dehydro- Genet. 1995;11(1):40–4.
genase modulates sensorimotor gating in mice. Nat 239. van der Wees J, van Looij MA, de Ruiter MM,
Genet. 1999;21(4):434–9. Elias H, van der Burg H, Liem SS, et al. Hearing
227. Maynard TM, Meechan DW, Dudevoir ML, loss following Gata3 haploinsufficiency is caused by
Gopalakrishna D, Peters AZ, Heindel CC, et al. cochlear disorder. Neurobiol Dis. 2004;16(1):
Mitochondrial localization and function of a subset 169–78.
of 22q11 deletion syndrome candidate genes. 240. van Looij MA, van der Burg H, van der Giessen RS,
Mol Cell Neurosci. 2008;39(3):439–51. de Ruiter MM, van der Wees J, van Doorninck JH,
228. Bilous R, Murty G, Parkinson D, Thakker R, et al. GATA3 haploinsufficiency causes a rapid dete-
Coulthard M, Burn J, et al. Autosomal dominant rioration of distortion product otoacoustic emissions
familial hypoparathyroidism, sensineural deafness (DPOAEs) in mice. Neurobiol Dis. 2005;20(3):
and renal dysplasia. N Engl J Med. 1992;327: 890–7.
1069–84. 241. Grigorieva IV, Mirczuk S, Gaynor KU, Nesbit MA,
229. Lichtner P, Konig R, Hasegawa T, Van Esch H, Grigorieva EF, Wei Q, et al. Gata3-deficient mice
Meitinger T, Schuffenhauer S. An HDR (hypopara- develop parathyroid abnormalities due to
thyroidism, deafness, renal dysplasia) syndrome dysregulation of the parathyroid-specific transcription
locus maps distal to the DiGeorge syndrome region factor Gcm2. J Clin Invest. 2010;120(6):2144–55.
on 10p13/14. J Med Genet. 2000;37(1):33–7. 242. Moraes CT, DiMauro S, Zeviani M, Lombes A,
230. Van Esch H, Groenen P, Nesbit MA, Schuffenhauer S, Shanske S, Miranda AF, et al. Mitochondrial DNA
Lichtner P, Vanderlinden G, et al. GATA3 haplo- deletions in progressive external ophthalmoplegia
insufficiency causes human HDR syndrome. Nature. and Kearns-Sayre syndrome. N Engl J Med.
2000;406(6794):419–22. 1989;320(20):1293–9.
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 331

243. Zupanc ML, Moraes CT, Shanske S, Langman CB, linkage-disequilibrium mapping of the syndrome of
Ciafaloni E, DiMauro S. Deletion of mitochondrial congenital hypoparathyroidism, growth and mental
DNA in patients with combined features of retardation, and dysmorphism to a 1-cM interval on
Kearns-Sayre and MELAS syndromes. Ann Neurol. chromosome 1q42-43. Am J Hum Genet. 1998;63(1):
1991;29(6):680–3. 163–9.
244. Isotani H, Fukumoto Y, Kawamura H, Furukawa K, 257. Diaz GA, Gelb BD, Ali F, Sakati N, Sanjad S, Meyer
Ohsawa N, Goto Y, et al. Hypoparathyroidism and BF, et al. Sanjad-Sakati and autosomal recessive
insulin-dependent diabetes mellitus in a patient with Kenny-Caffey syndromes are allelic: evidence for an
Kearns-Sayre syndrome harbouring a mitochondrial ancestral founder mutation and locus refinement. Am
DNA deletion. Clin Endocrinol (Oxf). 1996;45(5): J Med Genet. 1999;85(1):48–52.
637–41. 258. Parvari R, Hershkovitz E, Grossman N,
245. Dionisi-Vici C, Garavaglia B, Burlina AB, Bertini E, Gorodischer R, Loeys B, Zecic A, et al. Mutation of
Saponara I, Sabetta G, et al. Hypoparathyroidism TBCE causes hypoparathyroidism-retardation-
in mitochondrial trifunctional protein deficiency. dysmorphism and autosomal recessive Kenny-Caffey
J Pediatr. 1996;129(1):159–62. syndrome. Nat Genet. 2002;32(3):448–52.
246. Labarthe F, Benoist JF, Brivet M, Vianey-Saban C, 259. Tian G, Huang MC, Parvari R, Diaz GA, Cowan
Despert F, de Baulny HO. Partial hypoparathyroidism NJ. Cryptic out-of-frame translational initiation of
associated with mitochondrial trifunctional protein TBCE rescues tubulin formation in compound
deficiency. Eur J Pediatr. 2006;165(6):389–91. heterozygous HRD. Proc Natl Acad Sci U S A.
247. Franceschini P, Testa A, Bogetti G, Girardo E, 2006;103(36):13491–6.
Guala A, Lopez-Bell G, et al. Kenny-Caffey syn- 260. Hershkovitz E, Rozin I, Limony Y, Golan H,
drome in two sibs born to consanguineous parents: Hadad N, Gorodischer R, et al. Hypoparathyroidism,
evidence for an autosomal recessive variant. Am J retardation, and dysmorphism syndrome: impaired
Med Genet. 1992;42(1):112–6. early growth and increased susceptibility to severe
248. Boynton JR, Pheasant TR, Johnson BL, Levin DB, infections due to hyposplenism and impaired
Streeten BW. Ocular findings in Kenny’s syndrome. polymorphonuclear cell functions. Pediatr Res.
Arch Ophthalmol. 1979;97(5):896–900. 2007;62(4):505–9.
249. Tahseen K, Khan S, Uma R, Usha R, Al Ghanem 261. Weber T, Liu S, Indridason O, Quarles L. Serum
MM, Al Awadi SA, et al. Kenny-Caffey syndrome FGF23 levels in normal and disordered phosphorus
in six Bedouin sibships: autosomal recessive inheri- homeostasis. J Bone Miner Res. 2003;18:1227–34.
tance is confirmed. Am J Med Genet. 1997;69(2): 262. Parkinson DB, Shaw NJ, Himsworth RL, Thakker
126–32. RV. Parathyroid hormone gene analysis in autosomal
250. Diaz GA, Khan KT, Gelb BD. The autosomal reces- hypoparathyroidism using an intragenic
sive Kenny-Caffey syndrome locus maps to chromo- tetranucleotide (AAAT)n polymorphism. Hum
some 1q42-q43. Genomics. 1998;54(1):13–8. Genet. 1993;91(3):281–4.
251. Guo MH, Shen Y, Walvoord EC, Miller TC, Moon JE, 263. Barakat AY, D’Albora JB, Martin MM, Jose PA.
Hirschhorn JN, et al. Whole exome sequencing to Familial nephrosis, nerve deafness, and hypoparathy-
identify genetic causes of short stature. Horm Res roidism. J Pediatr. 1977;91(1):61–4.
Paediatr. 2014;82(1):44–52. 264. Dahlberg PJ, Borer WZ, Newcomer KL, Yutuc
252. Unger S, Gorna MW, Le Bechec A, Do Vale-Pereira S, WR. Autosomal or X-linked recessive syndrome of
Bedeschi MF, Geiberger S, et al. FAM111A mutations congenital lymphedema, hypoparathyroidism,
result in hypoparathyroidism and impaired skeletal nephropathy, prolapsing mitral valve, and brachytele-
development. Am J Hum Genet. 2013;92(6):990–5. phalangy. Am J Med Genet. 1983;16(1):99–104.
253. Fine DA, Rozenblatt-Rosen O, Padi M, Korkhin A, 265. Lienhardt A, Bai M, Lagarde JP, Rigaud M, Zhang Z,
James RL, Adelmant G, et al. Identification of Jiang Y, et al. Activating mutations of the calcium-
FAM111A as an SV40 host range restriction and sensing receptor: management of hypocalcemia.
adenovirus helper factor. PLoS Pathog. 2012;8(10): J Clin Endocrinol Metab. 2001;86(11):5313–23.
e1002949. 266. Pollak MR, Brown EM, Estep HL, McLaine PN,
254. Nikkel S, Ahmed A, Smith A, Marcadier J, Kifor O, Park J, et al. Autosomal dominant
Bulman D, Boycott K. Mother-to-daughter transmis- hypocalcaemia caused by a Ca2+-sensing receptor
sion of Kenny-Caffey syndrome associated with gene mutation. Nat Genet. 1994;8:303–7.
the recurrent, dominant FAM111A mutation p. 267. Finegold DN, Armitage MM, Galiani M, Matise TC,
Arg569His. Clin Genet. 2014;86:394–95. Pandian MR, Perry YM, et al. Preliminary localiza-
255. Sanjad SA, Sakati NA, Abu-Osba YK, Kaddoura R, tion of a gene for autosomal dominant hypoparathy-
Milner RD. A new syndrome of congenital hypopara- roidism to chromosome 3q13. Pediatr Res.
thyroidism, severe growth failure, and dysmorphic 1994;36(3):414–7.
features. Arch Dis Child. 1991;66(2):193–6. 268. Pearce SH, Williamson C, Kifor O, Bai M, Coulthard
256. Parvari R, Hershkovitz E, Kanis A, Gorodischer R, MG, Davies M, et al. A familial syndrome of hypo-
Shalitin S, Sheffield VC, et al. Homozygosity and calcemia with hypercalciuria due to mutations in the
332 A. Sharma et al.

calcium-sensing receptor. N Engl J Med. 1996;335 281. Hu J, Mora S, Weber G, Zamproni I, Proverbio MC,
(15):1115–22. Spiegel AM. Autosomal dominant hypocalcemia in
269. Baron J, Winer K, Yanovski J, Cunningham A, monozygotic twins caused by a de novo germline
Laue L, Zimmerman D, et al. Mutations in the mutation near the amino-terminus of the human cal-
Ca(2+)-sensing receptor gene cause autosomal cium receptor. J Bone Miner Res Off J Am Soc Bone
dominant and sporadic hypoparathyroidism. Hum Miner Res. 2004;19(4):578–86.
Mol Genet. 1996;5:601–6. 282. Bettinelli A, Vezzoli G, Colussi G, Bianchetti MG,
270. Okazaki R, Chikatsu N, Nakatsu M, Takeuchi Y, Sereni F, Casari G. Genotype-phenotype correlations
Ajima M, Miki J, et al. A novel activating mutation in normotensive patients with primary renal tubular
in calcium-sensing receptor gene associated with a hypokalemic metabolic alkalosis. J Nephrol.
family of autosomal dominant hypocalcemia. J Clin 1998;11(2):61–9.
Endocrinol Metab. 1999;84:363–6. 283. Ranieri M, Tamma G, Di Mise A, Vezzoli G,
271. Hu J, McLarnon SJ, Mora S, Jiang J, Thomas C, Soldati L, Svelto M, et al. Excessive signal transduc-
Jacobson KA, et al. A region in the seven- tion of gain-of-function variants of the calcium-
transmembrane domain of the human Ca2+ receptor sensing receptor (CaSR) are associated with increased
critical for response to Ca2+. J Biol Chem. ER to cytosol calcium gradient. PLoS One.
2005;280(6):5113–20. 2013;8(11):e79113.
272. Lienhardt A, Garabedian M, Bai M, Sinding C, 284. Li Y, Song YH, Rais N, Connor E, Schatz D, Muir A,
Zhang Z, Lagarde JP, et al. A large homozygous or et al. Autoantibodies to the extracellular domain of the
heterozygous in-frame deletion within the calcium- calcium sensing receptor in patients with acquired
sensing receptor’s carboxylterminal cytoplasmic tail hypoparathyroidism. J Clin Invest. 1996;97(4):
that causes autosomal dominant hypocalcemia. J Clin 910–4.
Endocrinol Metab. 2000;85(4):1695–702. 285. Albright F, Burnett CH, Smith PH, Parson W.
273. Hannan FM, Nesbit MA, Zhang C, Cranston T, Pseudohypoparathyroidism – an example of
Curley AJ, Harding B, et al. Identification of 70 “Seabright-Bantam syndrome”. Endocrinology.
calcium-sensing receptor mutations in hyper- and 1942;30:922–32.
hypo-calcaemic patients: evidence for clustering of 286. Weinstein LS. Albright hereditary osteodystrophy,
extracellular domain mutations at calcium-binding pseudohypoparathyroidism, and Gs deficiency. In:
sites. Hum Mol Genet. 2012;21(12):2768–78. Spiegel AM, editor. G proteins, receptors, and dis-
274. Li D, Opas EE, Tuluc F, Metzger DL, Hou C, ease. Totowa: Humana Press; 1998. p. 23–56.
Hakonarson H, et al. Autosomal dominant hypopara- 287. de Jan Beur S, Levine M. Pseudohypopar-
thyroidism caused by germline mutation in GNA11: athyroidism: clinical, biochemical, and molecular fea-
phenotypic and molecular characterization. J Clin tures. In: Bilezikian JP, Markus R, Levine MA,
Endocrinol Metab. 2014;99:E1774–83. editors. The parathyroids: basic and clinical concepts.
275. Rogers A, Nesbit MA, Hannan FM, Howles SA, New York: Academic; 2001. p. 807–25.
Gorvin CM, Cranston T, et al. Mutational analysis 288. Bastepe M, J€ uppner H. Pseudohypoparathyroidism,
of the adaptor protein 2 sigma subunit (AP2S1) Albright’s hereditary osteodystrophy, and progressive
gene: search for autosomal dominant hypocalcemia osseous heteroplasia: disorders caused by inactivating
type 3 (ADH3). J Clin Endocrinol Metab. 2014;99(7): GNAS mutations. In: DeGroot LJ, Jameson JL,
E1300–5. editors. Endocrinology, 1. 6th ed. Philadelphia:
276. Thakker RV. Molecular pathology of renal chloride W.B. Saunders; 2010. p. 1223–35.
channels in Dent’s disease and Bartter’s syndrome. 289. Bastepe M, J€ uppner H. Pseudohypoparathyroidism,
Exp Nephrol. 2000;8(6):351–60. Gsα, and the GNAS locus. BoneKEy. 2005;2:20–32.
277. Hebert SC. Bartter syndrome. Curr Opin Nephrol 290. Levine MA. Pseudohypoparathyroidism. In:
Hypertens. 2003;12(5):527–32. Bilezikian JP, Raisz LG, Rodan GA, editors. Princi-
278. Watanabe S, Fukumoto S, Chang H, Takeuchi Y, ples of bone biology. New York: Academic; 1996.
Hasegawa Y, Okazaki R, et al. Association between p. 853–76.
activating mutations of calcium-sensing receptor and 291. Schuster V, Eschenhagen T, Kruse K, Gierschik P,
Bartter’s syndrome. Lancet. 2002;360(9334):692–4. Kreth HW. Endocrine and molecular biological stud-
279. Vargas-Poussou R, Huang C, Hulin P, Houillier P, ies in a German family with Albright hereditary
Jeunemaitre X, Paillard M, et al. Functional charac- osteodystrophy. Eur J Pediatr. 1993;152:185–9.
terization of a calcium-sensing receptor mutation in 292. Miric A, Vechio JD, Levine MA. Heterogeneous
severe autosomal dominant hypocalcemia with a mutations in the gene encoding the α-subunit of the
Bartter-like syndrome. J Am Soc Nephrol. stimulatory G protein of adenylyl cyclase in Albright
2002;13(9):2259–66. hereditary osteodystrophy. J Clin Endocrinol Metab.
280. Vezzoli G, Arcidiacono T, Paloschi V, Terranegra A, 1993;76:1560–8.
Biasion R, Weber G, et al. Autosomal dominant 293. Weinstein LS, Gejman PV, Friedman E, Kadowaki T,
hypocalcemia with mild type 5 Bartter syndrome. Collins RM, Gershon ES, et al. Mutations of the
J Nephrol. 2006;19(4):525–8. Gs αsubunit gene in Albright hereditary
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 333

osteodystrophy detected by denaturing gradient progressive osseous heteroplasia (POH) and other
gel electrophoresis. Proc Natl Acad Sci U S A. forms of GNAS-based heterotopic ossification. Am J
1990;87:8287–90. Med Genet A. 2008;146A(14):1788–96.
294. Ahrens W, Hiort O, Staedt P, Kirschner T, 306. Cairns DM, Pignolo RJ, Uchimura T, Brennan TA,
Marschke C, Kruse K. Analysis of the GNAS1 gene Lindborg CM, Xu M, et al. Somitic disruption of
in Albright’s hereditary osteodystrophy. J Clin GNAS in chick embryos mimics progressive osseous
Endocrinol Metab. 2001;86(10):4630–4. heteroplasia. J Clin Invest. 2013;123(8):3624–33.
295. Linglart A, Carel JC, Garabedian M, Le T, Mallet E, 307. Schipani E, Weinstein LS, Bergwitz C, Iida-Klein A,
Kottler ML. GNAS1 lesions in pseudohypopar- Kong XF, Stuhrmann M, et al. Pseudohypopar-
athyroidism Ia and Ic: genotype phenotype relation- athyroidism type Ib is not caused by mutations in
ship and evidence of the maternal transmission of the the coding exons of the human parathyroid hormone
hormonal resistance. J Clin Endocrinol Metab. (PTH)/PTH-related peptide receptor gene. J Clin
2002;87(1):189–97. Endocrinol Metab. 1995;80:1611–21.
296. Long DN, McGuire S, Levine MA, Weinstein LS, 308. Bettoun JD, Minagawa M, Kwan MY, Lee HS,
Germain-Lee EL. Body mass index differences in Yasuda T, Hendy GN, et al. Cloning and characteri-
pseudohypoparathyroidism type 1a versus zation of the promoter regions of the human parathy-
pseudopseudohypoparathyroidism may implicate roid hormone (PTH)/PTH-related peptide receptor
paternal imprinting of Galpha(s) in the development gene: analysis of deoxyribonucleic acid from normal
of human obesity. J Clin Endocrinol Metab. subjects and patients with pseudohypoparathyroidism
2007;92(3):1073–9. type Ib. J Clin Endocrinol Metab. 1997;82:1031–40.
297. Davies AJ, Hughes HE. Imprinting in Albright’s 309. Suarez F, Lebrun JJ, Lecossier D, Escoubet B,
hereditary osteodystrophy. J Med Genet. Coureau C, Silve C. Expression and modulation of
1993;30:101–3. the parathyroid hormone (PTH)/PTH-related peptide
298. Wilson LC, Oude-Luttikhuis MEM, Clayton PT, receptor messenger ribonucleic acid in skin fibro-
Fraser WD, Trembath RC. Parental origin of Gsα blasts from patients with type Ib pseudohypopar-
gene mutations in Albright’s hereditary athyroidism. J Clin Endocrinol Metab. 1995;80:
osteodystrophy. J Med Genet. 1994;31:835–9. 965–70.
299. Germain-Lee EL, Schwindinger W, Crane JL, 310. Fukumoto S, Suzawa M, Takeuchi Y, Nakayama K,
Zewdu R, Zweifel LS, Wand G, et al. A mouse Kodama Y, Ogata E, et al. Absence of mutations in
model of Albright hereditary osteodystrophy gener- parathyroid hormone (PTH)/PTH-related protein
ated by targeted disruption of exon 1 of the Gnas receptor complementary deoxyribonucleic acid in
gene. Endocrinology. 2005;146(11):4697–709. patients with pseudohypoparathyroidism type Ib.
300. Yu S, Yu D, Lee E, Eckhaus M, Lee R, Corria Z, J Clin Endocrinol Metab. 1996;81:2554–8.
et al. Variable and tissue-specific hormone resistance 311. Bastepe M, Lane AH, J€ uppner H. Paternal uniparental
in heterotrimeric Gs protein alpha-subunit (Gsα) isodisomy of chromosome 20q (patUPD20q) – and
knockout mice is due to tissue-specific imprinting the resulting changes in GNAS1 methylation – as a
of the Gsα gene. Proc Natl Acad Sci U S A. plausible cause of pseudohypoparathyroidism. Am J
1998;95(15):8715–20. Hum Genet. 2001;68:1283–9.
301. Yu S, Gavrilova O, Chen H, Lee R, Liu J, Pacak K, 312. Bastepe M, J€ uppner H. Pseudohypoparathyroidism:
et al. Paternal versus maternal transmission of a stim- new insights into an old disease. In: Strewler GJ,
ulatory G-protein alpha subunit knockout produces editor. Endocrinology and Metabolism Clinics of
opposite effects on energy metabolism. J Clin Invest. North America: Hormones and disorders of mineral
2000;105:615–23. metabolism, vol. 29. Philadelphia: W. B. Saunders;
302. Eddy MC, De Jan Beur SM, Yandow SM, McAlister 2000. p. 569–89.
WH, Shore EM, Kaplan FS, et al. Deficiency of the 313. de Pérez Nanclares G, Fernández-Rebollo E, Santin I,
alpha-subunit of the stimulatory G protein and severe Garcia-Cuartero B, Gaztambide S, Menendez E,
extraskeletal ossification. J Bone Miner Res Off J Am et al. Epigenetic defects of GNAS in patients with
Soc Bone Miner Res. 2000;15(11):2074–83. pseudohypoparathyroidism and mild features of
303. Yeh GL, Mathur S, Wivel A, Li M, Gannon FH, Albright’s hereditary osteodystrophy. J Clin
Ulied A, et al. GNAS1 mutation and Cbfa1 Endocrinol Metab. 2007;92(6):2370–3.
misexpression in a child with severe congenital plate- 314. Unluturk U, Harmanci A, Babaoglu M, Yasar U,
like osteoma cutis. J Bone Miner Res Off J Am Soc Varli K, Bastepe M, et al. Molecular diagnosis and
Bone Miner Res. 2000;15(11):2063–73. clinical characterization of pseudohypopar-
304. Shore E, Ahn J, de Jan Beur S, Li M, Xu M, Gardner athyroidism type-Ib in a patient with mild Albright’s
RM, et al. Paternally-inherited inactivating mutations hereditary osteodystrophy-like features, epileptic sei-
of the GNAS1 gene in progressive osseous zures, and defective renal handling of uric acid. Am J
heteroplasia. N Engl J Med. 2002;346:99–106. Med Sci. 2008;336(1):84–90.
305. Adegbite NS, Xu M, Kaplan FS, Shore EM, Pignolo 315. J€
uppner H, Schipani E, Bastepe M, Cole DEC,
RJ. Diagnostic and mutational spectrum of Lawson ML, Mannstadt M, et al. The gene
334 A. Sharma et al.

responsible for pseudohypoparathyroidism type Ib is pseudohypoparathyroidism type Ib and biparental


paternally imprinted and maps in four unrelated kin- defects of GNAS methylation in cis. J Clin Endocrinol
dreds to chromosome 20q13.3. Proc Natl Acad Sci Metab. 2010;95:3993–4002.
U S A. 1998;95:11798–803. 327. Linglart A, Bastepe M, J€ uppner H. Similar clinical
316. Liu J, Litman D, Rosenberg M, Yu S, Biesecker L, and laboratory findings in patients with symptomatic
Weinstein L. A GNAS1 imprinting defect in autosomal dominant and sporadic pseudohypopar-
pseudohypoparathyroidism type IB. J Clin Invest. athyroidism type Ib despite different epigenetic
2000;106:1167–74. changes at the GNAS locus. Clin Endocrinol (Oxf).
317. Bastepe M, Pincus JE, Sugimoto T, Tojo K, 2007;67(6):822–31.
Kanatani M, Azuma Y, et al. Positional dissociation 328. Dixit A, Chandler KE, Lever M, Poole RL,
between the genetic mutation responsible for Bullman H, Mughal MZ, et al. Pseudohypopar-
pseudohypoparathyroidism type Ib and the associated athyroidism type 1b due to paternal uniparental
methylation defect at exon A/B: evidence for a long- disomy of chromosome 20q. J Clin Endocrinol
range regulatory element within the imprinted GNAS1 Metab. 2013;98(1):E103–8.
locus. Hum Mol Genet. 2001;10:1231–41. 329. Williamson CM, Ball ST, Nottingham WT, Skinner
318. Bastepe M, Fröhlich LF, Hendy GN, Indridason OS, JA, Plagge A, Turner MD, et al. A cis-acting control
Josse RG, Koshiyama H, et al. Autosomal dominant region is required exclusively for the tissue-specific
pseudohypoparathyroidism type Ib is associated with imprinting of Gnas. Nat Genet. 2004;36(8):894–9.
a heterozygous microdeletion that likely disrupts a 330. Blomstrand S, Claësson I, Säve-Söderbergh J. A case
putative imprinting control element of GNAS. J Clin of lethal congenital dwarfism with accelerated skele-
Invest. 2003;112(8):1255–63. tal maturation. Pediatr Radiol. 1985;15:141–3.
319. Laspa E, Bastepe M, J€ uppner H, Tsatsoulis 331. Young ID, Zuccollo JM, Broderick NJ. A lethal skel-
A. Phenotypic and molecular genetic aspects of etal dysplasia with generalised sclerosis and advanced
pseudohypoparathyroidism type Ib in a Greek kin- skeletal maturation: Blomstrand chondrodysplasia. J
dred: evidence for enhanced uric acid excretion due Med Genet. 1993;30:155–7.
to parathyroid hormone resistance. J Clin Endocrinol 332. Leroy JG, Keersmaeckers G, Coppens M, Dumon JE,
Metab. 2004;89:5942–7. Roels H. Blomstrand lethal chondrodysplasia. Am J
320. Mahmud FH, Linglart A, Bastepe M, J€ uppner H, Med Genet. 1996;63:84–9.
Lteif AN. Molecular diagnosis of pseudohypopar- 333. Loshkajian A, Roume J, Stanescu V, Delezoide AL,
athyroidism type Ib in a family with presumed parox- Stampf F, Maroteaux P. Familial Blomstrand
ysmal dyskinesia. Pediatrics. 2005;115(2):e242–4. Chondrodysplasia with advanced skeletal maturation:
321. Liu J, Nealon J, Weinstein L. Distinct patterns of further delineation. Am J Med Genet. 1997;71:283–8.
abnormal GNAS imprinting in familial and sporadic 334. den Hollander NS, van der Harten HJ, Vermeij-Keers-
pseudohypoparathyroidism type IB. Hum Mol Genet. C, Niermeijer MF, Wladimiroff JW. First-trimester
2005;14:95–102. diagnosis of Blomstrand lethal osteochondro-
322. Linglart A, Gensure RC, Olney RC, J€ uppner H, dysplasia. Am J Med Genet. 1997;73:345–50.
Bastepe M. A novel STX16 deletion in autosomal 335. Oostra RJ, Baljet B, Dijkstra PF, Hennekam RCM.
dominant pseudohypoparathyroidism type Ib Congenital anomalies in the teratological collection of
redefines the boundaries of a cis-acting imprinting museum Vrolik in Amsterdam, The Netherlands. II:
control element of GNAS. Am J Hum Genet. Skeletal dysplasia. Am J Med Genet. 1998;77:
2005;76:804–14. 116–34.
323. Elli FM, de Sanctis L, Peverelli E, Bordogna P, 336. Jobert AS, Zhang P, Couvineau A, Bonaventure J,
Pivetta B, Miolo G, et al. Autosomal dominant Roume J, Le Merrer M, et al. Absence of functional
pseudohypoparathyroidism type Ib: a novel inherited receptors for parathyroid hormone and parathyroid
deletion ablating STX16 causes loss of imprinting at hormone-related peptide in Blomstrand
the A/B DMR. J Clin Endocrinol Metab. 2014;99: chondrodysplasia. J Clin Invest. 1998;102(1):34–40.
E724–28. 337. Zhang P, Jobert AS, Couvineau A, Silve C. A homo-
324. Richard N, Abeguile G, Coudray N, Mittre H, zygous inactivating mutation in the parathyroid hor-
Gruchy N, Andrieux J, et al. A new deletion ablating mone/parathyroid hormone-related peptide receptor
NESP55 causes loss of maternal imprint of A/B causing Blomstrand chondrodysplasia. J Clin
GNAS and autosomal dominant pseudohypopar- Endocrinol Metab. 1998;83:3365–8.
athyroidism type Ib. J Clin Endocrinol Metab. 338. Karaplis AC, Bin He MT, Nguyen A, Young ID,
2012;97(5):E863–7. Semeraro D, Ozawa H, et al. Inactivating mutation
325. Bastepe M, J€uppner H. The GNAS locus and in the human parathyroid hormone receptor type
pseudohypoparathyroidism. Horm Res. 2005;63: 1 gene in Blomstrand chondrodysplasia. Endocrinol-
65–74. ogy. 1998;139:5255–8.
326. Chillambhi S, Turan S, Hwang DY, Chen HC, 339. Karperien MC, van der Harten HJ, van Schooten R,
J€uppner H, Bastepe M. Deletion of the noncoding Farih-Sips H, den Hollander NS, Kneppers ALJ,
GNAS antisense transcript causes et al. A frame-shift mutation in the type I parathyroid
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 335

hormone/parathyroid hormone-related peptide recep- processing of FGF-23. Philadelphia: American


tor causing Blomstrand lethal osteochondrodysplasia. Society of Nephrology; 2004.
J Clin Endocrinol Metab. 1999;84:3713–20. 352. Ichikawa S, Baujat G, Seyahi A, Garoufali AG, Imel
340. Karperien M, Sips H, Hvd H, Wijnaents L, EA, Padgett LR, et al. Clinical variability of familial
Kindblom L, Papapoulos S, et al. Novel mutations tumoral calcinosis caused by novel GALNT3 muta-
in the type I PTH/PTHrP receptor causing Blomstrand tions. Am J Med Genet A. 2010;152a(4):896–903.
lethal osteochondrodysplasia (abstract). J Bone Miner 353. Trump D, Thakker R. Inherited hypophosphataemic
Res. 2001;16 Suppl 1:S549. rickets. In: Thakker R, editor. Molecular genetics of
341. Hoogendam J, Farih-Sips H, Wynaendts LC, Lowik endocrine disorders. London: Chapman and Hall;
CW, Wit JM, Karperien M. Novel mutations in the 1997. p. 123–51.
parathyroid hormone (PTH)/PTH-related peptide 354. Glorieux F, Karsenty G, Thakker R. Metabolic bone
receptor type 1 causing Blomstrand osteochondro- disease in children. In: Avioli LV, Krane SM, editors.
dysplasia types I and II. J Clin Endocrinol Metab. Metabolic bone disease. 3rd ed. New York: Aca-
2007;92(3):1088–95. demic; 1997. p. 759–83.
342. Wysolmerski JJ, Cormier S, Philbrick W, Dann P, 355. Econs M, McEnery P, Lennon F, Speer M.
Zhang J, Roume J, et al. Absence of functional type Autosomal dominant hypophosphatemic rickets is
1 PTH/PTHrP receptors in humans is associated with linked to chromosome 12p13. J Clin Invest.
abnormal breast development and tooth impactation. 1997;100:2653–7.
J Clin Endocrinol Metab. 2001;86:1788–94. 356. Yamazaki Y, Okazaki R, Shibata M, Hasegawa Y,
343. Ichikawa S, Lyles KW, Econs MJ. A novel GALNT3 Satoh K, Tajima T, et al. Increased circulatory level
mutation in a pseudoautosomal dominant form of of biologically active full-length FGF-23 in patients
tumoral calcinosis: evidence that the disorder is with hypophosphatemic rickets/osteomalacia. J Clin
autosomal recessive. J Clin Endocrinol Metab. Endocrinol Metab. 2002;87:4957–60.
2005;90(4):2420–3. 357. Jonsson KB, Zahradnik R, Larsson T, White KE,
344. Ichikawa S, Imel EA, Sorenson AH, Severe R, Sugimoto T, Imanishi Y, et al. Fibroblast growth
Knudson P, Harris GJ, et al. Tumoral calcinosis factor 23 in oncogenic osteomalacia and X-linked
presenting with eyelid calcifications due to novel hypophosphatemia. N Engl J Med. 2003;348(17):
missense mutations in the glycosyl transferase 1656–63.
domain of the GALNT3 gene. J Clin Endocrinol 358. Farrow EG, Yu X, Summers LJ, Davis SI, Fleet JC,
Metab. 2006;91(11):4472–5. Allen MR, et al. Iron deficiency drives an autosomal
345. Topaz O, Shurman DL, Bergman R, Indelman M, dominant hypophosphatemic rickets (ADHR) pheno-
Ratajczak P, Mizrachi M, et al. Mutations in type in fibroblast growth factor-23 (Fgf23) knock-in
GALNT3, encoding a protein involved in O-linked mice. Proc Natl Acad Sci U S A. 2011;108(46):
glycosylation, cause familial tumoral calcinosis. Nat E1146–55.
Genet. 2004;36(6):579–81. 359. Imel EA, Peacock M, Gray AK, Padgett LR, Hui SL,
346. Ichikawa S, Imel EA, Kreiter ML, Yu X, Mackenzie Econs MJ. Iron modifies plasma FGF23 differently in
DS, Sorenson AH, et al. A homozygous missense autosomal dominant hypophosphatemic rickets
mutation in human KLOTHO causes severe tumoral and healthy humans. J Clin Endocrinol Metab.
calcinosis. J Clin Invest. 2007;117(9):2684–91. 2011;96(11):3541–9.
347. Larsson T, Davis S, Garringer H, Mooney S, 360. Drezner MK. Phosphorus homeostasis and related
Draman M, Cullen M, et al. Fibroblast growth disorders. In: Bilezikian JP, Raisz LG, Rodan GA,
factor-23 mutants causing familial tumoral calcinosis editors. Principles in bone biology, vol. 1. 2nd ed. -
are differentially processed. J Clin Endocrinol Metab. New York: Academic; 2002. p. 321–38.
2005;146:3883–91. 361. Lipman M, Panda D, Bennett H, Henderson J,
348. Araya K, Fukumoto S, Backenroth R, Takeuchi Y, Shane E, Shen Y, et al. Cloning of human PEX
Nakayama K, Ito N, et al. A novel mutation in fibro- cDNA. Expression, subcellular localization, and
blast growth factor 23 gene as a cause of tumoral endopeptidase activity. J Biol Chem. 1998;273:
calcinosis. J Clin Endocrinol Metab. 2005;90(10): 13729–37.
5523–7. 362. John M, Wickert H, Zaar K, Jonsson K, Grauer A,
349. Melhem R, Najjar S, Khachadurian A. Cortical hyper- Ruppersberger P, et al. A case of neuroendocrine
ostosis with hyperphosphatemia: a new syndrome? oncogenic osteomalacia associated with a PHEX
J Pediatr. 1970;77:986–90. and fibroblast growth factor-23 expressing sinusidal
350. Narchi H. Hyperostosis with hyperphosphatemia: evi- malignant schwannoma. Bone. 2001;29:393–402.
dence of familial occurrence and association with 363. Seufert J, Ebert K, M€ uller J, Eulert J, Hendrich C,
tumoral calcinosis. Pediatrics. 1997;99:745–8. Werner E, et al. Octreotide therapy for tumor-induced
351. Frishberg Y, Araya K, Rinat C, Topaz O, Yamazaki Y, osteomalacia. N Engl J Med. 2001;345:1883–8.
Feinstein Y, et al., editors. Hyperostosis- 364. Imel E, Peacock M, Pitukcheewanont P, Heller H,
hyperphosphatemia syndrome caused by mutations Ward L, Shulman D, et al. Sensitivity of fibroblast
in GALNT3 and associated with augmented growth factor 23 measurements in tumor-induced
336 A. Sharma et al.

osteomalacia. J Clin Endocrinol Metab. evolution and human disease. Nat Rev Genet.
2006;91:2055–61. 2007;8(10):762–75.
365. van Boekel G, Ruinemans-Koerts J, Joosten F, 378. Perry W, Stamp T. Hereditary hypophosphataemic
Dijkhuizen P, van Sorge A, de Boer H. Tumor pro- rickets with autosomal recessive inheritance and
ducing fibroblast growth factor 23 localized by severe osteosclerosis. A report of two cases. J Bone
two-staged venous sampling. Eur J Endocrinol. Joint Surg Br. 1978;60-B:430–4.
2008;158(3):431–7. 379. Scriver C, Reade T, Halal F, Costa T, Cole D. Auto-
366. White KE, Carn G, Lorenz-Depiereux B, Benet- somal hypophosphataemic bone disease responds to
Pages A, Strom TM, Econs MJ. Autosomal-dominant 1,25-(OH)2D3. Arch Dis Child. 1981;56:203–7.
hypophosphatemic rickets (ADHR) mutations stabi- 380. Bastepe M, Shlossberg H, Murdock H, J€ uppner H,
lize FGF-23. Kidney Int. 2001;60(6):2079–86. Rittmaster R. A lebanese family with osteosclerosis
367. Imanishi Y, Hashimoto J, Ando W, Kobayashi K, and hypophosphatemia. J Bone Miner Res. 1999;14
Ueda T, Nagata Y, et al. Matrix extracellular Suppl 1:S558.
phosphoglycoprotein is expressed in causative tumors 381. Ye L, Mishina Y, Chen D, Huang H, Dallas S,
of oncogenic osteomalacia. J Bone Miner Metab. Dallas M, et al. Dmp1-deficient mice display severe
2012;30(1):93–9. defects in cartilage formation responsible for a
368. Consortium TH. A gene (PEX) with homologies to chondrodysplasia-like phenotype. J Biol Chem.
endopeptidases is mutated in patients with X-linked 2005;280:6197–203.
hypophosphatemic rickets. The HYP Consortium. 382. Feng JQ, Scott G, Guo D, Jiang B, Harris M, Ward T,
Nat Genet. 1995;11:130–6. et al. Generation of a conditional null allele for Dmp1
369. Holm IA, Huang X, Kunkel LM. Mutational analysis in mouse. Genesis. 2008;46(2):87–91.
of the PEX gene in patients with X-linked 383. Liu S, Zhou J, Tang W, Menard R, Feng JQ,
hypophosphatemic rickets. Am J Hum Genet. Quarles LD. Pathogenic role of Fgf23 in Dmp1-null
1997;60(4):790–7. mice. Am J Physiol Endocrinol Metab. 2008;295(2):
370. Rowe P, de Zoysa P, Dong R, Wang H, White K, E254–61.
Econs M, et al. MEPE, a new gene expressed in 384. Levy-Litan V, Hershkovitz E, Avizov L, Leventhal N,
bone marrow and tumors causing osteomalacia. Bercovich D, Chalifa-Caspi V, et al. Autosomal-
Genomics. 2000;67:54–68. recessive hypophosphatemic rickets is associated
371. Aono Y, Shimada T, Yamazaki Y, Hino R, with an inactivation mutation in the ENPP1 gene.
Takeuchi M, Fujita T, et al., editors. The neutraliza- Am J Hum Genet. 2010;86(2):273–8.
tion of FGF-23 ameliorates hypophosphatemia and 385. Prié D, Huart V, Bakouh N, Planelles G, Dellis O,
rickets in Hyp mice. Meeting of the American Society Gérard B, et al. Nephrolithiasis and osteoporosis asso-
for Bone and Mineral Research, Minneapolis; 2003. ciated with hypophosphatemia caused by mutations
372. Bowe A, Finnegan R, Jan de Beur S, Cho J, Levine M, in the type 2a sodium-phosphate cotransporter.
Kumar R. FGF-23 inhibits renal tubular phosphate N Engl J Med. 2002;347:983–91.
transport and is a PHEX substrate. Biochem Biophys 386. Virkki L, Forster I, Hernando N, Biber J, Murer
Res Commun. 2001;284:977–81. H. Functional characterization of two naturally occur-
373. Yamazaki Y, Tamada T, Kasai N, Urakawa I, Aono Y, ring mutations in the human sodium-phosphate
Hasegawa H, et al. Anti-FGF23 neutralizing anti- cotransporter type IIa. J Bone Miner Res Off J Am
bodies show the physiological role and structural Soc Bone Miner Res. 2003;18:2135–41.
features of FGF23. J Bone Miner Res Off J Am Soc 387. Lapointe JY, Tessier J, Paquette Y, Wallendorff B,
Bone Miner Res. 2008;23(9):1509–18. Coady MJ, Pichette V, et al. NPT2a gene variation
374. Carpenter TO, Imel EA, Ruppe MD, Weber TJ, in calcium nephrolithiasis with renal phosphate leak.
Klausner MA, Wooddell MM, et al. Randomized Kidney Int. 2006;69(12):2261–7.
trial of the anti-FGF23 antibody KRN23 in X-linked 388. Magen D, Berger L, Coady MJ, Ilivitzki A,
hypophosphatemia. J Clin Invest. 2014;124(4): Militianu D, Tieder M, et al. A loss-of-function muta-
1587–97. tion in NaPi-IIa and renal Fanconi’s syndrome.
375. George A, Sabsay B, Simonian PA, Veis A. N Engl J Med. 2010;362(12):1102–9.
Characterization of a novel dentin matrix acidic 389. Beck L, Karaplis AC, Amizuka N, Hewson AS,
phosphoprotein. Implications for induction of Ozawa H, Tenenhouse HS. Targeted inactivation of
biomineralization. J Biol Chem. 1993;268(17): Ntp2 in mice leads to severe renal phosphate wasting,
12624–30. hypercalciuria, and skeletal abnormalities. Proc Natl
376. Narayanan K, Ramachandran A, Hao J, He G, Park Acad Sci U S A. 1998;95:5372–7.
KW, Cho M, et al. Dual functional roles of dentin 390. Tieder M, Modai D, Samuel R, Arie R, Halabe A,
matrix protein 1. Implications in biomineralization Bab I, et al. Hereditary hypophosphatemic rickets
and gene transcription by activation of intracellular with hypercalciuria. N Engl J Med. 1985;312:611–7.
Ca2+ store. J Biol Chem. 2003;278(19):17500–8. 391. Bergwitz C, Roslin NM, Tieder M, Loredo-Osti JC,
377. Chen JM, Cooper DN, Chuzhanova N, Ferec C, Bastepe M, Abu-Zahra H, et al. SLC34A3 mutations
Patrinos GP. Gene conversion: mechanisms, in patients with hereditary hypophosphatemic
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 337

rickets with hypercalciuria predict a key role for the gene in patients with pseudovitamin D-deficiency
sodium-phosphate cotransporter NaPi-IIc in rickets. N Engl J Med. 1998;338(10):653–61.
maintaining phosphate homeostasis. Am J Hum 405. Yoshida T, Monkawa T, Tenenhouse HS, Goodyer P,
Genet. 2006;78(2):179–92. Shinki T, Suda T, et al. Two novel 1alpha-hydroxylase
392. Lorenz-Depiereux B, Benet-Pages A, Eckstein G, mutations in French-Canadians with vitamin D
Tenenbaum-Rakover Y, Wagenstaller J, Tiosano D, dependency rickets type I1. Kidney Int. 1998;54(5):
et al. Hereditary hypophosphatemic rickets with 1437–43.
hypercalciuria is caused by mutations in the sodium- 406. Wang JT, Lin CJ, Burridge SM, Fu GK, Labuda M,
phosphate cotransporter gene SLC34A3. Am J Hum Portale AA, et al. Genetics of vitamin D 1alpha-
Genet. 2006;78(2):193–201. hydroxylase deficiency in 17 families. Am J Hum
393. Ichikawa S, Sorenson AH, Imel EA, Friedman NE, Genet. 1998;63(6):1694–702.
Gertner JM, Econs MJ. Intronic deletions in the 407. Wang X, Zhang MY, Miller WL, Portale AA. Novel
SLC34A3 gene cause hereditary hypophosphatemic gene mutations in patients with 1alpha-hydroxylase
rickets with hypercalciuria. J Clin Endocrinol Metab. deficiency that confer partial enzyme activity in vitro.
2006;91(10):4022–7. J Clin Endocrinol Metab. 2002;87(6):2424–30.
394. Prader A, Illig R, Heierli E. An unusual form of 408. Brooks MH, Bell NH, Love L, Stern PH, Orfei E,
primary vitamin D-resistant rickets with hypocalce- Queener SF, et al. Vitamin-D-dependent rickets
mia and autosomal-dominant hereditary transmission: type II. Resistance of target organs to 1,25-
hereditary pseudo-deficiency rickets. Helv Paediatr dihydroxyvitamin D. N Engl J Med. 1978;298(18):
Acta. 1961;16:452–68. 996–9.
395. Fraser D, Kooh SW, Kind HP, Holick MF, Tanaka Y, 409. Marx SJ, Spiegel AM, Brown EM, Gardner DG,
DeLuca HF. Pathogenesis of hereditary vitamin-D- Downs Jr RW, Attie M, et al. A familial syndrome
dependent rickets. An inborn error of vitamin D metab- of decrease in sensitivity to 1,25-dihydroxyvitamin
olism involving defective conversion of D. J Clin Endocrinol Metab. 1978;47(6):1303–10.
25-hydroxyvitamin D to 1 alpha,25-dihydroxyvitamin 410. Liberman UA, Samuel R, Halabe A, Kauli R,
D. N Engl J Med. 1973;289(16):817–22. Edelstein S, Weisman Y, et al. End-organ resistance
396. Scriver CR. Vitamin D, dependency. Pediatrics. to 1,25-dihydroxycholecalciferol. Lancet. 1980;1
1970;45(3):361–3. (8167):504–6.
397. Delvin EE, Glorieux FH, Marie PJ, Pettifor 411. Fujita T, Nomura M, Okajima S, Furuya H. Adult-
JM. Vitamin D dependency: replacement therapy onset vitamin D-resistant osteomalacia with the
with calcitriol? J Pediatr. 1981;99(1):26–34. unresponsiveness to parathyroid hormone. J Clin
398. Scriver CR, Reade TM, DeLuca HF, Hamstra AJ. Endocrinol Metab. 1980;50(5):927–31.
Serum 1,25-dihydroxyvitamin D levels in normal 412. Liberman UA, Eil C, Marx SJ. Resistance to Vitamin
subjects and in patients with hereditary rickets or D. In: Cohn DV, Fujita T, Potts Jr JT, Talmage RV,
bone disease. N Engl J Med. 1978;299(18):976–9. editors. Endocrine control of bone and calcium
399. DeLuca HF. Vitamin D metabolism and function. metabolism, International congress series 619, vol.
Arch Intern Med. 1978;138(Spec No):836–47. 8A. Amsterdam: Exerpta Medica; 1984. p. 32–40.
400. Chesney RW, Rosen JF, Hamstra AJ, DeLuca 413. Fraher LJ, Karmali R, Hinde FR, Hendy GN, Jani H,
HF. Serum 1,25-dihydroxyvitamin D levels in normal Nicholson L, et al. Vitamin D-dependent rickets type
children and in vitamin D disorders. Am J Dis Child. II: extreme end organ resistance to 1,25-dihydroxy
1980;134(2):135–9. vitamin D3 in a patient without alopecia. Eur J
401. Labuda M, Morgan K, Glorieux FH. Mapping auto- Pediatr. 1986;145(5):389–95.
somal recessive vitamin D dependency type I to chro- 414. Haussler MR, Manolagas SC, Deftos LJ. Evidence for
mosome 12q14 by linkage analysis. Am J Hum a 1,25-dihydroxyvitamin D3 receptor-like macromol-
Genet. 1990;47(1):28–36. ecule in rat pituitary. J Biol Chem. 1980;255(11):
402. St-Arnaud R, Messerlian S, Moir JM, Omdahl JL, 5007–10.
Glorieux FH. The 25-hydroxyvitamin D 1-alpha- 415. O’Malley BW. Steroid hormone action in eucaryotic
hydroxylase gene maps to the pseudovitamin cells. J Clin Invest. 1984;74(2):307–12.
D-deficiency rickets (PDDR) disease locus. J Bone 416. Eil C, Liberman UA, Rosen JF, Marx SJ. A cellular
Miner Res Off J Am Soc Bone Miner Res. defect in hereditary vitamin-D-dependent rickets type
1997;12(10):1552–9. II: defective nuclear uptake of 1,25-dihydroxyvitamin
403. Fu GK, Lin D, Zhang MY, Bikle DD, Shackleton CH, D in cultured skin fibroblasts. N Engl J Med.
Miller WL, et al. Cloning of human 1981;304(26):1588–91.
25-hydroxyvitamin D-1 alpha-hydroxylase and muta- 417. Eil C, Marx SJ. Nuclear uptake of 1,25-dihydroxy
tions causing vitamin D-dependent rickets type 1. [3H]cholecalciferol in dispersed fibroblasts cultured
Mol Endocrinol. 1997;11(13):1961–70. from normal human skin. Proc Natl Acad Sci U S A.
404. Kitanaka S, Takeyama K, Murayama A, Sato T, 1981;78(4):2562–6.
Okumura K, Nogami M, et al. Inactivating mutations 418. Haussler MR, Mangelsdorf DJ, Komm BS, et al.
in the 25-hydroxyvitamin D3 1alpha-hydroxylase Molecular biology of the vitamin D hormone.
338 A. Sharma et al.

In: Cohn DV, Martin TJ, Mennier PJ, editors. Calcium 431. Lloyd SE, Pearce SH, Fisher SE, Steinmeyer K,
regulation and bone metabolism: basic and clinic Schwappach B, Scheinman SJ, et al. A common
aspects, vol. 9. Amsterdam: Elsevier; 1987. molecular basis for three inherited kidney stone dis-
p. 465–74. eases. Nature. 1996;379(6564):445–9.
419. McDonnell DP, Mangelsdorf DJ, Pike JW, Haussler 432. Lowe CU, Terrey M, Mac LE. Organic-aciduria,
MR, O’Malley BW. Molecular cloning of comple- decreased renal ammonia production,
mentary DNA encoding the avian receptor for vitamin hydrophthalmos, and mental retardation; a clinical
D. Science. 1987;235(4793):1214–7. entity. AMA Am J Dis Child. 1952;83(2):164–84.
420. Hughes MR, Malloy PJ, Kieback DG, Kesterson RA, 433. Silver DN, Lewis RA, Nussbaum RL. Mapping the
Pike JW, Feldman D, et al. Point mutations in the Lowe oculocerebrorenal syndrome to Xq24-q26 by
human vitamin D receptor gene associated with hypo- use of restriction fragment length polymorphisms.
calcemic rickets. Science. 1988;242(4886):1702–5. J Clin Invest. 1987;79:282–5.
421. Yoshizawa T, Handa Y, Uematsu Y, Sekine K, 434. Leahey AM, Charnas LR, Nussbaum RL. Nonsense
Takeda S, Yoshihara Y, et al. Disruption of the vitamin mutations in the OCRL-1 gene in patients with the
D receptor (VDR) in the mouse. J Bone Miner Res. oculocerebrorenal syndrome of Lowe. Hum Mol
1996;11:S119. Genet. 1993;2(4):461–3.
422. Li YC, Amling M, Pirro AE, Priemel M, Meuse J, 435. Santer R, Schneppenheim R, Dombrowski A,
Baron R, et al. Normalization of mineral ion homeo- Gotze H, Steinmann B, Schaub J. Mutations in
stasis by dietary means prevents hyperparathyroid- GLUT2, the gene for the liver-type glucose trans-
ism, rickets, and osteomalacia, but not alopecia in porter, in patients with Fanconi-Bickel syndrome.
vitamin D receptor-ablated mice. Endocrinology. Nat Genet. 1997;17(3):324–6.
1998;139:4391–6. 436. Mannstadt M, Magen D, Segawa H, Stanley T,
423. Tsuchiya Y, Matsuo N, Cho H, Kumagai M, Sharma A, Sasaki S, et al. Fanconi-Bickel syndrome
Yasaka A, Suda T, et al. An unusual form of vitamin and autosomal recessive proximal tubulopathy with
D-dependent rickets in a child: alopecia and marked hypercalciuria (ARPTH) are allelic variants caused
end-organ hyposensitivity to biologically active vita- by GLUT2 mutations. J Clin Endocrinol Metab.
min D. J Clin Endocrinol Metab. 1980;51(4):685–90. 2012;97(10):E1978–86.
424. Rosen JF, Fleischman AR, Finberg L, Hamstra A, 437. Beighton P. Osteoglophonic dysplasia. J Med Genet.
DeLuca HF. Rickets with alopecia: an inborn error 1989;26(9):572–6.
of vitamin D metabolism. J Pediatr. 1979;94(5): 438. White K, Cabral J, Evans W, Ichikawa S, Davis S,
729–35. Ornitz D, et al. A missense mutation in FGFR1 causes
425. Liberman UA, Eil C, Marx SJ. Resistance to 1,25- a novel syndrome: craniofacial dysplasia with
dihydroxyvitamin D. Association with heterogeneous hypophosphatemia (CFDH). Am J Hum Genet.
defects in cultured skin fibroblasts. J Clin Invest. 2005;76:361–67.
1983;71(2):192–200. 439. Farrow EG, Davis SI, Mooney SD, Beighton P,
426. Castells G, Greig F, Fusi L, Finberg L, Yasmura A, Mascarenhas L, Gutierrez YR, et al. Extended muta-
Liberman U, et al. Vitamin D dependent rickets Type tional analyses of FGFR1 in osteoglophonic dyspla-
II (VDDRII) with alopecia. Treatment with mega sia. Am J Med Genet A. 2006;140(5):537–9.
doses of 1,25(OH)2D3 overcomes affinity defect in 440. Hoffman W, J€ uppner H, Deyoung B, O’dorisio M,
receptor for 1,25(OH)2D3. Pediatr Res. Given K. Elevated fibroblast growth factor-23 in
1984;18:1174. hypophosphatemic linear nevus sebaceous syndrome.
427. Balsan S, Garabédian M, Larchet M, Gorski AM, Am J Med Genet A. 2005;134:233–6.
Cournot G, Tau C, et al. Long-term nocturnal calcium 441. Heike C, Cunningham M, Steiner R, Wenkert D,
infusions can cure rickets and promote normal Hornung R, Gruss J, et al. Skeletal changes in epider-
mineralization in hereditary resistance to 1,25- mal nevus syndrome: does focal bone disease harbor
dihydroxyvitamin D. J Clin Invest. 1986;77(5): clues concerning pathogenesis? Am J Med Genet
1661–7. A. 2005;139:67–77.
428. Tiosano D, Hadad S, Chen Z, Nemirovsky A, 442. Lim YH, Ovejero D, Sugarman JS, Deklotz CM,
Gepstein V, Militianu D, et al. Calcium absorption, Maruri A, Eichenfield LF, et al. Multilineage somatic
kinetics, bone density, and bone structure in patients activating mutations in HRAS and NRAS cause
with hereditary vitamin D-resistant rickets. J Clin mosaic cutaneous and skeletal lesions, elevated
Endocrinol Metab. 2011;96(12):3701–9. FGF23 and hypophosphatemia. Hum Mol Genet.
429. Srivastava T, Alon US. Cinacalcet as adjunctive ther- 2014;23(2):397–407.
apy for hereditary 1,25-dihydroxyvitamin D-resistant 443. Weinstein LS, Shenker A, Gejman PV, Merino MJ,
rickets. J Bone Miner Res Off J Am Soc Bone Miner Friedman E, Spiegel AM. Activating mutations of
Res. 2013;28(5):992–6. the stimulatory G protein in the McCune-Albright
430. Scheinman SJ, Guay-Woodford LM, Thakker RV, syndrome. N Engl J Med. 1991;325:1688–95.
Warnock DG. Genetic disorders of renal electrolyte 444. Schwindinger W, Francomano C, Levine M. Identifi-
transport. N Engl J Med. 1999;340(15):1177–87. cation of a mutation in the gene encoding the alpha
11 Physiology of the Developing Kidney: Disorders and Therapy of Calcium and Phosphorous. . . 339

subunit of the stimulatory G protein of adenylyl 448. Germain-Lee EL, Groman J, Crane JL, de Beur SM J,
cyclase in McCune-Albright syndrome. Proc Natl Levine MA. Growth hormone deficiency in
Acad Sci U S A. 1992;89:5152–6. pseudohypoparathyroidism type 1a: another manifes-
445. Kobayashi K, Imanishi Y, Koshiyama H, Miyauchi A, tation of multihormone resistance. J Clin Endocrinol
Wakasa K, Kawata T, et al. Expression of FGF23 is Metab. 2003;88(9):4059–69.
correlated with serum phosphate level in isolated 449. Finer G, Price HE, Shore RM, White KE,
fibrous dysplasia. Life Sci. 2006;78(20):2295–301. Langman CB. Hyperphosphatemic familial tumoral
446. Waller S, Kurzawinski T, Spitz L, Thakker R, calcinosis: response to acetazolamide and postulated
Cranston T, Pearce S, et al. Neonatal severe hyper- mechanisms. Am J Med Genet A. 2014;164a(6):
parathyroidism: genotype/phenotype correlation and 1545–9.
the use of pamidronate as rescue therapy. Eur J 450. Parfrey PS, Chertow GM, Block GA, Correa-Rotter R,
Pediatr. 2004;163(10):589–94. Drueke TB, Floege J, et al. The clinical course of
447. Garcia-Garcia E, Dominguez-Pascual I, Requena- treated hyperparathyroidism among patients receiv-
Diaz M, Cabello-Laureano R, Fernandez-Pineda I, ing hemodialysis and the effect of cinacalcet:
Sanchez-Martin MJ. Intraoperative parathyroid hor- the EVOLVE trial. J Clin Endocrinol Metab.
mone monitoring in neonatal severe primary hyper- 2013;98(12):4834–44.
parathyroidism. Pediatrics. 2014;134(4):e1203–5.
Nutrition Management in Childhood
Kidney Disease: An Integrative and 12
Lifecourse Approach

Lauren Graf, Kimberly Reidy, and Frederick J. Kaskel

Contents Common Phosphate Additives Used By


the Food Industry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 342 Hypoallergenic Diet as a Treatment for Idiopathic
A Life Course Approach to the Renal Diet: Nephrotic Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 356
Integration with Developmental Physiology . . . . . . 342 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
A Life Course Approach to the Renal Diet: References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 358
Integration with Etiology of Renal Disease . . . . . . . 347
A Life Course Approach to the Renal Diet:
Integration with Prevention of Cardiovascular
Disease and Inflammation . . . . . . . . . . . . . . . . . . . . . . . . . 348
Effects of Diet on Inflammation and Heart Disease
and Immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 349
The Anti-Inflammatory Diet for Kidney Disease:
A Paradigm Shift . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 350
Fiber . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
Phosphorus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 351
Protein . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 353
Dietary Guidelines for CKD and ESRD . . . . . . . . . . 354
Guidelines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
Phosphate Preservatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355

L. Graf (*)
Children’s Hospital at Montefiore, Albert Einstein College
of Medicine, Bronx, NY, USA
e-mail: Lgraf@montefiore.org
K. Reidy • F.J. Kaskel
Division of Pediatric Nephrology, Children’s Hospital at
Montefiore, Albert Einstein College of Medicine, Bronx,
NY, USA
e-mail: Kreidy@montefiore.org;
Frederick.kaskel@einstein.yu.edu

# Springer-Verlag Berlin Heidelberg (outside the USA) 2016 341


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_11
342 L. Graf et al.

Introduction adult. A healthy child doubles their height in the


first 4 years of life and requires significant net
Infants, children, and adolescents with chronic kid- calcium and phosphorus absorption for bone
ney disease (CKD), end-stage renal disease accretion. Early childhood is a period of peak
(ESRD), and kidney disorders such as nephrotic growth velocity, with growth of 7–11 cm/year,
syndrome face unique nutritional challenges. The and over 50 % of adult height is attained in the
goals of this chapter are to outline the nutritional first 4 years of life. Thus, optimal nutritional sup-
problems associated with various forms of kidney port for infants and young children with CKD will
disease and describe interventions to correct them. have far-reaching consequences for ultimate adult
The approach to nutritional management varies height.
greatly based on the age of the child and type of There are several unique challenges to opti-
kidney disorder. The primary goals of nutrition in mize nutrition in infants with CKD. First, the
infants and young children with CKD are to pro- major source of calories and nutrients in the first
mote adequate weight gain, linear growth, and year of life is liquid. In addition, caloric intake is
hydration as well as maintain electrolyte homeo- the major determinant of growth in the first year of
stasis. For older children and teenagers with CKD life and outweighs the effect of growth hormone
and ESRD, the traditional nutritional goals have [1]. Thus, it is critical to optimize the caloric
focused on the management of potassium and intake of both infants and toddlers with CKD
phosphorus, fluid balance, and growth. However, [2, 3]. This often requires supplementation with
as our understanding of nutrition evolved, it is now either oral feeds or gastrostomy tube feeding (see
recognized that diet (quantity and quality of food) Table 1 for nutritional formulas and supplements
has far-reaching effects on overall health. Nutrition frequently used in kidney disease). Estimates of
is extremely important in modulating inflamma- caloric needs can be based on estimated energy
tion, immune function, and the gut microbiome, requirements for age (see Table 2) [4]. These
all of which play a major role in preventing chronic should be used as a guide and increased if inade-
diseases including cardiovascular disease (CVD) quate weight gain is achieved. These guidelines
and cancer. A large body of research over the past are also intended for chronic care, and different
decade has led to a paradigm shift in nutrition and guidelines may apply to children in the intensive
kidney disease. The recommendations for protein care unit, for example.
intake in the ESRD population as well as the most Maintenance of euvolemia and serum potas-
effective way to manage dietary phosphorus have sium is also particularly an issue for anuric
changed dramatically. In addition, in some cases of infants. Breast milk is the preferred source of
idiopathic nephrotic syndrome, diet therapy has nutrition for infants with or without chronic kid-
been effective as a treatment of the disease rather ney disease. Especially for infants with
than simply managing symptoms. The following hyperkalemia, breast milk potassium content is
sections will identify nutritional problems in vari- typically lower than commercially available for-
ous kidney disorders and describe the diet therapy mulas. Similac PM 60/40 ® can be used for infants
to best help manage them. who require formulas. In severe cases, potassium
content can be lowered further in formula or
breast milk by mixing sodium polystyrene with
A Life Course Approach to the Renal formula, allowing it to settle for 30–45 min, and
Diet: Integration with Developmental decanting the supernatant (leaving the sediment
Physiology behind) [5, 6]. Potassium is exchanged for
sodium, so monitoring for hypernatremia is advis-
The approach to nutrition in chronic kidney dis- able. It is preferable to avoid direct administration
ease is modified by developmental changes in of sodium polystyrene to infants as it has been
physiology. First, the nutritional needs of a grow- associated with necrotizing enterocolitis. To limit
ing infant or child are distinct from that of an fluid overload, anuric infants can be given higher
12

Table 1 Nutrition Formulas for Children with Renal Disease


Age range of Carbohydrates (g/L Na/K Ca/P mOsm/kg
use Manufacturer kcal/ml Protein (g/L) sources Fat sources sources) (mEq/L) (mg/L) water
Infant–toddler Similac Ross 0.67 16 (whey, caseinate) 38 g/L (soy, coconut) 69 (lactose) 7/15 380/ 250
PM products 190
60/40
Infant–toddler Amin- R&D 2 19 (free amino acids) 46 g/L (partially 365 (maltodextrin, <15/ – 700
Aid Laboratories hydrogenated soybean sucrose) <15
oil)
Infant–toddler MCT oil Multiple 7.7 – 14 g/15 mL –
Toddler–adult Suplena Ross product 1.8 44 (caseinates) 95 g/L (high oleic 199 (maltodextrin, 33/28 1,060/ 600
safflower and soy oil) sucrose, cornstarch) 700
Toddler–adult Renalcal Nestle 2 34 (essential L amino 82 g/L (MCT oil, 290 (maltodextrin, – – 600
Nutrition acids, select nonessential canola oil, corn oil, soy modified cornstarch)
amino acids, whey lecithin)
protein concentrate)
Toddler–adult Nepro Ross 1.8 81 (caseinates) 96 g/L (high oleic 167 (corn syrup, 46/27 1,060/ 585
with products safflower oil and soy sucrose, FOS) 700
Carb oil)
Steady
Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach
343
344 L. Graf et al.

Table 2 Equations to Estimate Energy Requirements for Children at Healthy Weights


Age Estimate energy requirement (EER) (kcal/day) = total energy expenditure + energy deposition
0–3 months EER = (89  weight (kg) – 50) + 175
4–6 months EER = (89  weight (kg) – 50) + 56
7–12 months EER = (89  weight (kg) – 50) + 22
13–35 months EER = (89  weight (kg) – 50) + 20
3–8 years Boys EER = 88.5 – 61.9  age (y) + PA  (26.7  weight (kg) + 903  height (m)) + 20
Girls EER = 125.3 – 30.8  age (y) + PA  (10  weight (kg) + 934  height (m)) + 20
9–18 years Boys EER = 88.5 – 61.9  age (y) + PA  (26.7  weight (kg) + 903  height (m)) + 25
Girls EER = 135.3 – 30.8  age (y) + PA  (10  weight (kg) + 934  height (m)) + 25
Physical Activity Coefficients for Determination of Energy Requirements in Children Ages 3–18 years (adapted from
KDOQI guidelines [4])
Gender Sedentary Low active Active Very active
Typical ADL + 30–60 min of ADL + ADL + 60 min of daily moderate activity
activities of daily moderate activity 60 min + an additional 60 min of vigorous activity
daily living (e.g., walking 5–7 km/h) daily or 120 min of moderate activity
(ADL) only moderate
activity
Boys 1.0 1.13 1.26 1.42
Girls 1.0 1.16 1.31 1.56
PA = physical activity

caloric feeding (up to 30 kcal/oz) by either pre- from infant formula due to the much higher calcium
paring more concentrated formula or adding either content in infant formulas (42–50 mg/100 mL
PM 60/40 ® or medium chain triacyglyceol (MCT) vs. 21–26 mg/100 mL in breast milk) [11]. There
oil to breast milk. is a transient drop in serum calcium after birth that
Nutritional management of CKD must also leads to increased parathyroid hormone secretion
account for the changes in normal physiology of (up to adult levels within 48 h), which in turn
mineral homeostasis, as it differs for infants, chil- stimulates synthesis of calcitriol (1,25-OH vitamin
dren, and adults. A prime example of this is the D) in the kidney [7]. By later infancy, intestinal
changes in calcium and phosphorus homeostasis. calcium absorption becomes dependent on
The developmental regulation of calcium and calcitriol. On average, 30–60 % of ingested calcium
phosphorus during infancy and childhood reflects is absorbed, but up to 90 % may be absorbed in
the need for net calcium and phosphorus preterm infants [11]. The recommended daily intake
reabsorption, for bone growth, and for minerali- of calcium varies with age (Table 3) (http://ods.od.
zation (Figs. 1 and 2). Before birth, the placenta nih.gov/factsheets/Calcium-HealthProfessional/).
actively transports up to 300 mg/day of calcium to In children, 30–350 mg of calcium/day is
the developing fetus, mostly during the third tri- deposited in growing bones [7, 12]. In compari-
mester [7]. This is dependent on parathyroid son, the average adult ingests 1,000 mg of calcium
hormone-related protein (PTHrp), likely secreted per day. Of this, 350 mg is absorbed, but 800 mg
by the placenta [8]. Thus, calcium and phosphorus gets excreted in the stool (650 not absorbed and
requirements in premature infants born before this 150 lost through gut cell sloughing and bile secre-
critical window for bone growth and mineraliza- tion), and 200 mg is excreted in the urine,
tion may be higher than in full-term newborns [9]. maintaining a net zero balance for homeostasis.
After birth, the intestine becomes the major ave- In the adult, 98% of the 7,500 mg filtered through
nue for calcium via passive absorption [7]. Although the kidney is reabsorbed. In the neonate calcium
calcium is best absorbed from breast milk [10], excretion in the urine is higher, especially in the
infants have greater net absorption of calcium premature infant [13, 14]. Despite this, there is net
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 345

Fig. 1 Developmental changes in calcium handling: a to the positive calcium balance. Premature infants and
positive calcium balance is required for bone growth in neonates have increased urinary calcium excretion, which
infants and children, whereas adults maintain a net zero decreases with age. Serum calcium levels may be higher in
calcium balance to maintain homeostasis. Increased intes- childhood than in adults
tinal absorption during infancy and childhood contributes

calcium accretion throughout childhood. Figure 1 nucleic acids and lipid membranes, and the pri-
illustrates the distinct age-related normal physio- mary source of cellular energy, ATP. In addition,
logic changes in calcium absorption, renal excre- phosphorylation (addition of phosphates to pro-
tion, serum calcium concentration, and bone teins) is key for the regulation of multiple cell
deposition. signaling pathways. Phosphorus excretion in the
Similar to calcium, phosphorus balance is reg- urine serves as a buffer and contributes to
ulated by intestinal absorption, distribution of acid–base homeostasis.
phosphorus between the extracellular and bone/ The average adult has an intake of 1,000–1,600
intracellular storage pool, and renal phosphate mg/day of phosphorus (mean 20 mg/kg/day), and
excretion (Fig. 2) [11]. In addition to being 80 % of this is absorbed in the intestine
required for bone mineralization, phosphorus is [11]. There is some secretion of phosphorus into
required for basic cellular function [15]. Inside the the gut as a component of bile (3 mg/kg/day), so
cell, phosphorus is predominantly in the form of net phosphorus absorption is approximately
phosphate anion. Phosphates are the most abun- 13 mg/kg/day. In the adult, approximately
dant intracellular anion, a major component of 3 mg/kg/day will enter the bone for bone
346 L. Graf et al.

Fig. 2 Developmental changes in phosphorus han- absorption and decreased renal excretion contribute to the
dling: the majority of phosphorus is intracellular (as a higher serum phosphorus levels and positive phosphorus
component of nucleic acids, ATP, or phospholipids) or balance in the infant. In the adult, renal excretion of
stored in the bone, and serum inorganic phosphorus repre- ingested phosphorus results in a net zero phosphorus bal-
sents <0.1 % of whole body phosphorus. Increased ance and maintains homeostasis

Table 3 Recommended intake of calcium (Adapted from tubule. Fibroblast growth factor 23 (FGF-23)
http://ods.od.nih.gov/factsheets/Calcium-HealthProfessional/) and parathyroid hormone regulate phosphorus
Age Recommended daily intake (mg/day) excretion in the adult. In the steady state, the
0–6 months 200 adult maintains net zero phosphorus balance and
6–12 months 260 all the ingested phosphorus is excreted in the
1–3 years 700 urine.
4–8 years 1,000 In the infant, a net positive phosphorus balance
8–18 years 1,300 is required for bone accretion and growth. Thus,
Adult 1,000–1,200 normal serum phosphate levels vary with age,
with the highest levels in infants (up to 8.5
mg/dL) which decrease with age (4.5 mg/dL) in
adults. Human milk has a concentration of 11–17
remodeling. Phosphorus is fully filtered by the mg/dL phosphorus, while standard cow milk for-
glomerulus and the tubular fluid concentration is mulas contain 28–39 mg/dL of phosphorus
the same as the adult. In the adult, 85 % of [11]. As with calcium, the recommended guide-
filtered phosphorus is reabsorbed in the proximal lines for phosphorus intake vary by age (Table 4)
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 347

Table 4 Recommended minimum phosphorus intake Table 5 Target Serum Calcium and Phosphorus Levels
(Adapted from http://www.nal.usda.gov/fnic/DRI/DRI_ By Age
Tables/recommended_intakes_individuals.pdf)
Calcium Phosphorus
Recommended minimum intake Age (mg/dL) (mg/dL)
Age (mg/day) 0–5 months 8.7–11.3 5.2–8.4
0–6 months 100 6–12 8.7–11.0 5.0–7.8
6–12 275 months
months 1–5 years 9.4–10.8 4.5–6.5
1–3 years 380 6–12 years 9.4–10.3 3.6–5.8
4–8 years 405 13–20 years 8.8–10.2 2.3–4.5
9–18 years 1,055
Adult 580

(http://www.nal.usda.gov/fnic/DRI/DRI_Tables/ A Life Course Approach to the Renal


recommended_intakes_individuals.pdf). How- Diet: Integration with Etiology of Renal
ever, the average diet likely greatly exceeds this Disease
recommended minimum intake [15].
Up to 90 % of ingested phosphorus is absorbed in The approach to nutrition is also modified by the
the newborn via passive and active transport in the etiology of chronic kidney disease. Congenital
intestine (predominantly jejunum) via paracellular abnormalities of the kidney and urinary tract
HPO4 transport and NaPi-IIb sodium-dependent (CAKUT) are the most common cause of chronic
phosphate transporters, respectively [11]. The new- and end-stage kidney disease in childhood. Dys-
born kidney tubules are relatively insensitive to plasia and obstructive uropathy often result in
parathyroid hormone, leading to decreased phos- polyuria, with concomitant salt and water
phorus excretion and higher serum phosphate levels wasting. Thus, infants and children with CKD
[16]. Newborn kidneys also express a NaPi trans- secondary to CAKUT require unique approach
porter isoform that also favors phosphorus to their nutritional management. Severely
reabsorption [16]. The role of FGF-23 in neonatal polyuric infants have daily fluid requirements
phosphate excretion is still under investigation [17]. that are 1.5–2 times that of a healthy infant.
The developmental differences in calcium/ Thus, for polyuric infants, it may be advisable
phosphorus homeostasis have significant implica- not to concentrate formulas to provide addi-
tions for management of the CKD-induced meta- tional free water. When this is insufficient, water
bolic bone disease in childhood. First, the boluses can be given either orally or via a
recommended target goals for calcium and phos- gastrostomy tube.
phorus are age based (Table 5). Impaired phos- Sodium accretion is required for normal
phorus excretion is an issue for the infant with growth. Sodium supplementation in infants with
CKD. Breast milk remains the optimal nutrition, salt wasting forms of CKD improves linear
again, due its lower content of phosphorus. Renal growth [19]. Sodium may be administered as
formulas (Table 1) also have lower phosphorus sodium bicarbonate (in patients with concomitant
content than typical infant formulas (albeit still acidosis) or with commercially available sodium
higher than breast milk). Calcium carbonate chloride solutions. Typically, sodium chloride
remains a commonly used phosphate binder in solutions need to be given via G-tube as they
infants. Calcium carbonate can be mixed with may not be tolerated orally. Close monitoring of
daytime and nighttime feeds. However, in infants serum sodium is advisable. Other, rarer renal dis-
with hypercalcemia, the newly available eases, such as nephropathic cystinosis with
sevelamer (Renvela ®) powder can be used as an Fanconi syndrome, often require electrolyte sup-
alternative [18]. Renvela ® is first reconstituted in plementation, including phosphorus, orally or via
water and then mixed with the overnight feeds. G-tube to support growth [20, 21].
348 L. Graf et al.

A Life Course Approach to the Renal Traditional cardiac risk factors are compounded
Diet: Integration with Prevention by those secondary to uremia. The largest contrib-
of Cardiovascular utors to vascular changes in children with ESRD
Disease and Inflammation are disturbances in mineral metabolism (high phos-
phorus, calcium–phosphorus product, and PTH)
Cardiovascular disease (CVD) is the leading [24]. Soft tissue calcification and coronary artery
cause of death in both children and adults with calcification (CAC) are evident in pediatric patients
chronic and end-stage kidney disease (ESRD). with advanced kidney disease. One study of young
Good nutrition plays a major role in preventing adults with childhood-onset CKD found coronary
and reversing CVD in ESRD. Children with CKD artery calcifications present in 92 % of patients
are at very high risk of developing premature [27]. Markers of inflammation associated with
CVD during young adulthood. Adults with a his- heart disease such as elevated C-reactive protein
tory of childhood-onset ESRD have significantly (CRP) and homocysteine are often elevated in
shorter life expectancies than their peers. Dialysis ESRD and remained high even after renal trans-
patients live 40–50 years less, and transplant plantation. Given the multitude of risk factors in
patients live 20–25 years less than age-matched children with kidney disease, the high prevalence
peers in the general population. In addition, young of early onset cardiovascular disease in this popu-
adults (25–34 years old) on maintenance dialysis lation is not surprising. An epidemiological report
die at rates 100 times higher of cardiovascular found that young adults on dialysis (ages 25–34)
causes than age- and race-matched peers in the have the same cardiovascular mortality risk as an
general population [22]. Renal transplantation average 80–85-year-old in the general
significantly reduces cardiovascular mortality population [28].
[23]. However, even in these patients, the mortal- Over the past 30 years, advances in medicine
ity rates are still 10 times higher than in the gen- have improved outcomes for children with CKD
eral pediatric population secondary to CVD and ESRD, with increasing numbers surviving
factors. into adulthood. Patients with CKD no longer die
The causes of early onset CVD in the pediatric of kidney disease but of heart disease [29]. How-
kidney disease population are multifactorial. The ever, little progress has been made in reducing risk
prevalence of both traditional and uremia-related factors for heart disease such as coronary artery
cardiovascular risk factors is extremely high in calcifications. This may be largely due to the fact
children with advanced kidney disease [24]. Tra- that little attention has been given to diet as a
ditional risk factors for heart disease include therapy.
hypertension, diabetes, dyslipidemia, obesity, A large body of evidence suggests that many
and insulin resistance. The Chronic Kidney Dis- causes of heart and blood vessel disease in chil-
ease in Children (CKiD) study, an observational dren can be greatly ameliorated with aggressive
cohort study of children aged 1–16 years with nutritional intervention. Mineral metabolism and
stages 2–4 CKD, provides valuable insight into fluid overload are directly related to nutrition. It is
the cardiac risk factors present in this population increasingly recognized that nutrition also plays
[25]. Hypertension was present in 54 % of sub- a major role in improving inflammation, hyper-
jects at enrollment. Even with antihypertensive tension, and metabolic acidosis in kidney disease
medications, 48 % of participants had poorly con- [30, 31].
trolled blood pressure. Dyslipidemia was present The goal in pediatric nephrology is to improve
in 45 % of children in the CKiD study and 15 % both quantity and quality of life. Pediatric
were obese [26]. Although less common, meta- nephrology has had great success increasing the
bolic disturbances are also found in children with survival rates of young children with kidney dis-
CKD. In the CKiD cohort, 19 % of subjects were ease. A new goal now is to help these patients
insulin resistant. These risk factors tend to worsen transition into adolescence and young adulthood
as CKD progresses. with much lower cardiovascular risk factors,
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 349

which would place them on a path toward a than 100 trillion microbial cells, which play a
healthier and more productive life. A greater significant role in immunity and inflammation.
focus on nutrition may be the most effective, Both diet and diseases including CKD and
risk-free strategy to help prevent early onset ESRD directly impact the quantity and type of
CVD in children with advanced kidney disease. microbes present in the gut [35]. This in turn
This requires careful attention to the overall qual- impacts inflammation and CVD risk. The compo-
ity of food and diet patterns rather than simply sition of bacteria in the gut can be manipulated by
focusing on individual nutrients. changes in diet.
A diet high in whole, plant-based foods (fiber)
has been shown to favor the growth of beneficial
Effects of Diet on Inflammation bacteria [30, 36]. High fiber intake results in short
and Heart Disease and Immunity chain fatty acid production by microbes in the gut,
which decrease the intestinal pH. This suppresses
The standard American diet (characterized by a the growth of harmful bacteria such as E. coli and
high intake of refined carbohydrates, simple sugars, other members of Enterobacteriaceae family.
red meat, and processed food and low intake of Studies have found that European children have
fiber, fruits, and vegetables) is a major cause of depleted levels of the beneficial bacteria
CVD, inflammation, hypertension, dyslipidemia, Bacteroidetes and higher levels of Enterobac-
and type 2 diabetes [32]. It is also well established teriaceae compared with rural African children,
that a high-fiber diet based on unprocessed, plant- which the authors believe is due to the lower
based foods that is low in simple sugar drastically fiber intake among Europeans [37]. Analysis of
reduces risk of CVD and in many cases is an the fecal microbiota of hemodialysis patients
effective way to reverse it [33]. High fiber intake revealed that the total number of enterobacteria
is associated with lower levels of inflammation and was approximately 100 times higher in subjects
reduced mortality in the general population. Recent with ESRD compared to healthy controls
studies suggest that a high-fiber diet may be even [38]. Hemodialysis patients also had lower num-
more effective in reducing inflammation in CKD bers of bifidobacteria and higher Clostridium
than in the general population. One large cohort perfringens than controls. The authors speculate
study analyzed data from the National Health and that the dysbiosis may be due to multiple factors
Nutrition Examination Survey (NHANES III) data- including uremia, frequent antibiotic use, as well
base to examine the association between dietary as decreased intake of fiber.
fiber intake and elevated CRP levels in CKD and The production of short chain fatty acids from
non-CKD populations [34]. Higher intake of die- fiber has many benefits that are protective to the
tary fiber was associated with lower CRP levels in heart and vascular system [30]. They inhibit cho-
both the CKD and non-CKD groups, but the effect lesterol production by the liver and increase the
was more dramatic in those with kidney disease. production of cholesterol-binding bile acids,
For every 10-g increase in daily dietary fiber, the which aids in the elimination of lipids. Short
odds of elevated CRP were decreased to 11 % in the chain fatty acids also improve insulin sensitivity
non-CKD group and 38 % in the CKD group. In and help quell uremic toxins.
subjects without kidney disease, dietary total fiber On the other hand, high protein intake pro-
intake was not significantly associated with motes the growth of harmful bacteria that induce
reduced mortality. However, in the CKD group, inflammation. In the general population, a high
dietary fiber was inversely associated with mortal- protein intake increases the activity of bacterial
ity. Therefore, a high dietary fiber intake is associ- enzymes that produce toxic metabolites that trig-
ated with reduced inflammation and mortality ger inflammation [30, 36]. The harmful effects of
in CKD. proteolytic fermentation appear to be exaggerated
Diet has an enormous influence on the gut in CKD and ESRD. The fermentation of protein
microbiome. The human gut is home to more by bacteria in the large intestine results in indoles
350 L. Graf et al.

and phenols including p-cresyl sulfate and was associated with a lower mortality risk over
indoxyl sulfate. These are gut-derived uremic time. One of the highlights of this study is that it
toxics that are absorbed into the blood and nor- focused on diet patterns rather than individual
mally cleared by the kidney. However, they are nutrients. The authors also drew attention to the
resistant to clearance by dialysis. Elevated levels need for food-based guidelines rather than micro-
of p-cresyl sulfate and indoxyl sulfate are found in nutrient guidelines. The current guidelines focus
CKD and ESRD and appear to accelerate the on micronutrients and macronutrients (sodium,
progression of kidney disease. Elevated levels of potassium, phosphorus, protein) with little focus
indoxyl sulfate are associated with vascular stiff- on actual food. There is a trend in nutritional
ness, aortic calcification, and increased cardiovas- research to focus more on dietary patterns rather
cular mortality in patients with kidney disease than individual nutrients of supplements. This
[39–41]. Evidence suggests that diet may play a comes with the recognition that many studies
significant role in controlling the level of these looking at nutrients in isolation yield misleading
uremic toxins in the blood even as CKD pro- results because the larger picture is often not taken
gresses. Serum concentrations of indoxyl sulfate into account. Nutrients are not eaten in isolation;
and p-cresyl vary in subjects with similar esti- rather they are consumed as part of a whole food
mated glomerular filtration rates (GFR), which is with a vast array of nutrients and antioxidants that
due in part to differences in fiber intake. Dietary act synergistically. We suggest that food-based
fiber suppresses colonic production of p-cresyl guidelines would make it easier for patients to
and indoxyl sulfate, whereas protein exacerbates visualize and understand, which may improve
it [30]. Therefore, maximizing intake of fiber from adherence.
whole foods as well as ensuring adequate but not
excessive protein intake will help keep inflamma-
tory uremic toxins at bay. The Anti-Inflammatory Diet for Kidney
High-fiber, plant-based diets are also more Disease: A Paradigm Shift
effective in keeping serum phosphorus within
the target range compared with animal protein- Over the past decade, advances have been made in
based diet. One crossover trial compared the our understanding of how food and nutrition
effects of a vegetarian diet versus a meat-based impact the health of patients with kidney disease.
diet on serum phosphorus levels [42]. Although The foundation of an anti-inflammatory diet for
both diets had equivalent amounts of phosphorus, kidney disease is plant-based foods
they found significantly lower serum phosphorus (low-potassium fruits, vegetables, unrefined
in the vegetarian group. This is due to decreased whole grains, nuts, and seeds) combined with
absorption from plant sources of phosphorus. smaller amounts of unprocessed lean protein
Plant foods naturally contain phytate, the storage such as turkey, chicken, or fish. The diet is also
form of phosphorus, which is not digestible to low in processed food, dairy, and refined carbo-
humans. Therefore, much of the phosphorus hydrates such as sugar-sweetened beverages,
from plant foods passes through undigested white-bread, sugary cereals, and deli meat. Avoid-
[43–45]. It is important to take into account the ance of processed foods containing phosphate
source (plants, meat, and processed food) rather additives should be particularly emphasized.
than simply the amount of phosphorus. The veg- Traditionally, the renal nutritional community
etarian group also had lower FGF-23 levels, has taken a reductionist approach – evaluating
suggesting decreased inflammation. food simply based on the number of calories,
A recent observational study of 3,972 adults milligrams of sodium, or potassium and phospho-
with CKD found that a diet high in processed rus content. The conventional nutritional goals in
meat, fried food, and sugary beverages was asso- children with kidney disease have been to pro-
ciated with higher mortality [46]. A plant-based mote adequate weight gain and linear growth,
diet high in fruits and vegetables as well as fish manage serum phosphorus and potassium levels,
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 351

and maintain fluid balance. However, little ESRD are often instructed to limit or avoid
emphasis was placed on the healthfulness of the many plant foods due to a high potassium or
diet as a whole or how nutrition influences risk of phosphorus content. However, there are many
developing cardiovascular disease. Moreover, plant foods that do not contain an excessive
some traditional nutritional advice given to amount of potassium or phosphorus. Furthermore,
patients, once thought to improve outcomes, much of the phosphorus in whole plant foods is
such as an emphasis on animal protein, may actu- poorly absorbed and can be tolerated in people
ally be detrimental. In addition, there are a number with advanced CKD without raising serum phos-
of foods that may appear appropriate for a renal phorus. The importance of fiber is not usually
diet because they are low in potassium and phos- discussed in traditional diet counseling for renal
phorus such as a lemon–lime soda and other patients. In fact many of the standard nutritional
sugar-sweetened beverages. However, these prod- recommendations given to patients with advanced
ucts are high in sugar and fructose, which may kidney disease lead to much lower fiber intakes. In
lead to inflammation and dyslipidemia and cases where obtaining adequate fiber through food
worsen hypertension [47–52]. As our understand- alone is difficult, fiber supplements such as
ing of nutrition has evolved, two additional goals Benefiber ® or Unifiber ® may be considered.
have emerged: to reduce inflammation in the body However, they should not serve as a replacement
and reduce heart disease risk. These new goals for healthy, wholesome food. Nutritional educa-
focus on prevention and should be included in tion for children and adolescents should empha-
nutritional counseling for patients with kidney size healthy, fiber-rich foods that can be
disease. incorporated into a renal diet.

Fiber Phosphorus

Fiber is a type of carbohydrate indigestible to It is well documented that limiting dietary phos-
humans and is found exclusively in plants. Fiber phorus is crucial for patients with advanced kid-
intake is a marker of the amount of plant foods in ney disease. Phosphorus restriction helps prevent
the diet. There are two main types of fiber: soluble elevations in serum phosphorus, which lead to
and insoluble. Soluble fiber dissolves in water and hyperparathyroidism, renal osteodystrophy, heart
slows the movement of food through the gastro- and blood vessel calcifications, and increased
intestinal tracts. Insoluble fiber does not dissolve mortality. Maintaining serum phosphorus in the
in water and provide bulking which can help target range is one of the most difficult goals to
prevent constipation. Both soluble and insoluble achieve for patients with advanced kidney dis-
fibers can be fermented in the large intestine into ease. Phosphorus often remains difficult to control
active by-products such as prebiotics. Prebiotics even with patients who are adherent with phos-
serve as food or fuel for the probiotics. Therefore, phate binders. There is increasing evidence that
adequate consumption of fiber is imperative to excessive phosphorus intake is harmful even to
maintain sufficient amounts of beneficial bacteria the general population without kidney disease.
in the gut. Despite the health benefits of dietary Recent research suggests that excess dietary phos-
fiber, more than 90 % of adults and children in the phorus is an arterial toxin [53]. One double-blind
general population fail to meet the minimum crossover study found that high phosphorus
recommended requirements [34]. The average intake impairs flow-mediated dilation and causes
intake is 17 g per day, which is well below the arteries to stiffen within 2 h of ingestion [54]. A
recommended intake of 25–30 g daily. In people post hoc analysis report of 4,127 subjects found
with advanced CKD, average dietary fiber intake that serum phosphorus levels at the high end of the
is even lower at 15 g per day [34]. This is not normal range (>3.5 mg/dL) are associated with
surprising given that patients with CKD and increased risk of heart failure and myocardial
352 L. Graf et al.

infarction [55]. A recent report using the Food sources of inorganic phosphorus are
NHANES III data of over 9,000 adults (ages mainly additives in processed food. Phosphate
20–80 years) found that high phosphorus intake additives are found in a wide variety of products
is associated with increased mortality [56]. including processed meats, cheeses, frozen foods,
Phosphorus is present in food in both organic soups, yogurts, cereals, nondairy milk, and sports
and inorganic forms. Organic phosphorus is natu- drinks. In contrast to organic phosphorus, the
rally present in both plant and animal foods and is absorption rate of phosphorus from food additives
bound to protein and other molecules naturally is very high at 80–100 % [43, 45]. Phosphate
found in protein-rich foods. Most animal protein additives in processed food are the largest obsta-
including meat, poultry, fish, and dairy are high in cles to achieving adequate serum phosphorus con-
phosphorus. High-protein plant foods such as trol in patients with advanced kidney disease.
nuts, seeds, legumes, some whole grains, as well Phosphates are added to processed food to extend
as cacao and chocolate are also high in phospho- shelf life as well as improve flavor and texture.
rus. Organic phosphorus is broken down in the gut Over the past two decades, the amount of phos-
and then absorbed into circulation as inorganic phate preservatives in a variety of food products
phosphorus. The gastrointestinal tract absorbs has risen dramatically. In the early 1990s, phos-
about 40–60 % of the phosphorus naturally pre- phate additives contributed about 500 mg per day
sent in animal foods. Plant-based foods have an to the standard Western diet. Today, phosphate
even lower absorption rate of only about 20–50 % additives alone contribute as much as 1,000 mg
[43]. Phosphorus from plant foods is mostly in the per day to a typical Western diet [45, 57, 58]. In
storage form of phytic acid or phytate. Humans do the United States, 11 different phosphate salts are
not contain the enzyme phytase, which is required allowed to be injected into meat and poultry to
to degrade phytate. Therefore, the bioavailability improve tenderness and flavor and extend shelf
of phosphorus from most plant foods is low. life [59]. This can increase phosphorus levels in
Within the plant kingdom, there are several factors the meat by as much as 70 %. This practice is
that alter phosphorus bioavailability. Yeast con- currently banned in Europe.
tains phytase, which breaks down some of the Avoiding phosphate additives is challenging
phytate in whole grains making the phosphorus given that they are ubiquitous in processed and
in leavened breads more readily absorbed than packaged food. Effectively limiting phosphate
flatbread, cereal, or other non-yeast-containing additives is doable for most patients but requires
grains [45]. The process of soaking and sprouting detailed nutritional education by a dietitian with
grains and nuts removes a significant portion of instructions to carefully read ingredient lists.
phytic acid. Sprouting a grain or nut involves Given the wide variability in phosphorus absorp-
soaking it in water for a period of hours. tion from different foods, understanding the
This moist environment begins the germination source of phosphorus is equally as important as
process and activates enzymes, including the amount present in a given food. Food com-
phytase, which is naturally present in some panies are not required by law to report the quan-
grains. Wheat and rye are particularly high in tity of phosphorus on the nutritional facts section
phytase. Thus, the soaking and sprouting process of the label. However, they are mandated to
renders the phosphorus more bioavailable. report the presence of phosphate additives in the
Sprouted grain breads and nuts are increasingly ingredient list. All packaged and processed foods
appearing on grocery store shelves and are being contain complete ingredient lists in descending
marketed as health foods due to the higher nutri- order or predominance. Given that phosphate
ent bioavailability. While these foods have health additives are the most readily absorbed form of
benefits for the general population, those with phosphorus, paying careful attention to the often
CKD and ESRD should avoid soaked and overlooked ingredient list is one of the most
sprouted grains and nuts due to the more efficient important tools to teach patients. Encouraging
phosphorus absorption. patients to minimize processed food and
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 353

make fresh food and low-potassium produce reduced the recommended protein intake for all
dietary staples will result in higher intake of ages and stages of CKD based on the most current
fiber, phytochemicals, and antioxidants. Limit- literature [4]. Protein recommendations had pre-
ing processed food will not only restrict the viously been much higher for dialysis patients
most efficiently absorbed form of phosphorus than age-matched healthy children. This was
but will simultaneously minimize other harmful based on the knowledge that small amounts of
ingredients often found in packaged food such as amino acids are lost through dialysis membranes
sugar, refined flour, and vegetable oil. Decreas- as well as the belief that dialysis induces catabo-
ing processed food will have a myriad of health lism. While both peritoneal and hemodialysis ses-
benefits such as decreasing inflammation, sions result in some protein loss, the amount is
improving lipid profile and blood pressure, as relatively small and can be easily regained
well as improved serum phosphorus control. through diet or supplementation. Studies of chil-
Exposure to a healthy, anti-inflammatory diet dren on peritoneal dialysis have found that protein
during childhood will set the foundation for losses decrease with age. Average protein losses
healthy nutrition and lifestyle habits as patients for infants on peritoneal dialysis are 0.28 g/kg per
grow, receive kidney transplants, and transition day [61]. During adolescence the average protein
into adulthood. losses decrease to less than 0.1 g/kg/day [61]. Pro-
tein losses in hemodialysis have not been studied
extensively in children. Research in adults has
Protein found that an average of 1–3 g of protein is lost
per hemodialysis session [62–65]. Children
Adequate protein intake is important to make would be expected to have slightly higher dialytic
enzymes and hormones, build and repair tissue, protein losses than adults. The updated KDOQI
and support linear growth. Advanced CKD is guidelines provide formulas for estimating protein
often accompanied by a decrease in both protein requirements for children based on the stage of
and overall calorie intake, particularly in young CKD, the type of renal replacement therapy, and
children and infants. Regular nutritional evalua- age. The formula takes into account the small
tions that include diet recalls as well as measure- amount of protein lost through different dialysis
ments of weight, linear growth, and growth modalities.
velocity are important determinants of nutritional The belief that dialysis induces catabolism
status. and that higher protein intake is required to
Correcting protein deficiency can usually be improve weight gain and linear growth is not
achieved easily through nutritional counseling supported by the literature. Several clinical trials
and in some cases through supplementation. Ade- have studied the effects of selectively increasing
quate but not excessive protein intake appears to protein intake by using amino acid-based dialysis
be the most beneficial for patients with CKD. solutions rather than glucose in children on
There does not appear to be any benefit from peritoneal dialysis. Protein intake beyond
restricting protein lower than the dietary reference recommended levels did not improve nutritional
intake (DRI) for age in terms of slowing the pro- status or linear growth but did result in accumu-
gression of kidney disease [4]. However, protein lation of nitrogenous waste [66–70]. Too high
intake significantly beyond recommended levels protein intakes appear to be harmful to patients
increases nitrogenous waste products and serum with advanced CKD in several ways. Excess
phosphorus and can have detrimental effects on protein has been found to worsen metabolic aci-
the bone [4, 60]. This is important to note because dosis, increases bone loss, and is associated with
it is easy to exceed protein needs especially when hyperphosphatemia. A DEXA study of 20 chil-
supplements or tube feedings are used as a pri- dren on peritoneal dialysis with a mean protein
mary source of nutrition. The most recent pediat- intake that was 144 % of the recommended die-
ric KDOQI nutritional guidelines from 2008 tary allowance (RDA) was inversely correlated
354 L. Graf et al.

Table 6 Protein recommendations for children with CKD stages 3–5 (Adapted from KDOQI Guidelines)
DRI Recommendations for Recommendations for CKD Recommended Recommended
(g/kg/ CKD stage 3 (g/kg/day) stages 4–5 (g/kg/day) for HD (g/kg/ for PD (g/kg/
Age day) 100–140 % DRI (100–120 % DRI) day) day)
0–6 1.5 1.5–2.1 1.5–1.8 1.6 1.8
months
7–12 1.2 1.2–1.7 1.2–1.5 1.3 1.5
months
1–3 1.05 1.05–1.5 1.05–1.2 1.15 1.3
years
4–13 0.95 0.95–1.35 0.95–1.15 1.05 1.1
years
14–18 0.85 0.85–1.2 0.85–1.05 0.95 1.0
years

with bone mineral density, lean body mass, and calories is as important as the quantity. The goal
serum bicarbonate level [60]. Studies in adults is to promote as many whole, plant-based foods
have found that diets rich in vegetables and fruit as possible and limit excess animal protein,
lower acid levels and reduce kidney injury refined sugar, processed food, and chemical
(slowing the progression of disease) and help additives.
improve blood pressure without inducing
hyperkalemia in patients with advanced kidney The Following Low-Potassium Vegetables
disease [31, 71, 72] (Table 6). and Fruit Can Be Consumed Regularly
on a Renal Diet

Dietary Guidelines for CKD and ESRD Low-Potassium Vegetables


Asparagus, broccoli, cabbage, carrots, cau-
liflower, celery, cucumbers, green beans, green
peas, eggplant, kale, lettuce, mushrooms,
onions, parsley, peppers, turnips, and water
chestnuts
Low-Potassium Fruit
Apple, applesauce, blackberries, blue-
berries, cherries, cranberries, lemons, limes,
grapes, peaches, pears, pineapple, plum, and
watermelon
Herbs and Spices: Enhance the Flavor of Food
and Are Low in Potassium and High in Anti-
oxidants and Phytochemicals
Allspice, basil, bay leaf, cardamom, chili
powder, cilantro, cinnamon, chives, coriander,
cumin, curry, garlic, ginger, fennel, horserad-
ish, mint, nutmeg, oregano, paprika, parsley,
pepper, peppermint, rosemary, saffron, sage,
thyme, turmeric, vanilla, and wasabi
As a child transitions to solid food, adequate
Healthiest Fats and Oils
calorie intake remains important. However, it is All oils are calorically dense and have
important to recognize that the quality of those 120 cal per tablespoon. Oils have a very low
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 355

mineral content so they are a good way to boost phosphates and other preservatives. Read the
calories without adding potassium or phos- ingredient list carefully. To add flavor to a
phorus. Healthy oils include olive oil, coconut plain whole grain cereal, add cinnamon,
oil, and flaxseed oil. Although coconut oil is fruit, a small amount of maple syrup or
high in saturated fat, research suggests it has a honey, chopped nuts, or seeds.
very different effect on the body compared 5. Avoid red and processed deli meat or limit to
with saturated fat from animal products [73]. special occasions. Limit animal protein to two
Coconut oil is high in lauric acid, which is a servings per day. Choose fresh meat such as
medium-chain triglyceride (MCT). MCTs are turkey, chicken, beef, fish, or eggs. If con-
easily digestible, have a favorable impact on suming meat, choose grass fed, organic
blood cholesterol, and contain antimicrobial when possible.
properties. Coconut oil also contains a variety 6. Beans and legumes can be consumed once a
of protective phytochemicals including day. Serving size should be limited to about
phenolic acid [73]. It is advisable to limit or ½ cup due to the high potassium content
avoid adding vegetable oils such as soybean of most beans. Chickpeas and black beans
oil, corn oil, and sunflower oil as they are contain the lowest amount of potassium.
high in omega-6 fats, which are inflammatory Avoid adzuki beans as they are very high in
when consumed in excess [74]. Trans fat (par- potassium.
tially hydrogenated oils) found in some mar- 7. Avoid or limit cow’s milk and cheese to one
garines and shortening should be avoided serving daily. Try a plant-based milk such as
because they are linked to inflammation almond milk or rice milk. Choose a brand that
and CVD. does not contain phosphate additives. Hemp
milk and soy milk should be limited as they
are high in phosphorus.
Guidelines 8. Carefully read ingredient lists on all packaged
food including nondairy milk, meat, cereal,
1. Avoid sugar-sweetened beverages including bread, crackers, etc. Avoid products with
natural fruit juice, fruit drinks, iced tea, and phosphate additives, partially hydrogenated
soda regardless of the amount of potassium or oils (trans fat), and high-fructose corn syrup.
phosphorus. The high sugar content can 9. Choose real food snacks when possible such
increase blood pressure and promote inflam- as fruit, nuts or nut butter, granola bar, vege-
mation in the body. Instead, drink water or tables, or whole grain cracker with hummus.
water with lemon, lime, or mint slices. Try 10. To add additional calories, try mixing extra
seltzer or natural herbal tea. Do not introduce olive oil into pasta, rice, or other whole grain
sugary beverages to young children. or vegetable dishes. Coconut oil is a good
2. Aim to eat low-potassium vegetables and fruit addition to smoothies or breakfasts such as
with every meal. Expose young children to a oats or cream of wheat cereal. Whole food
variety of vegetables and fruit regularly. sources of fat, calories, and protein such as
Repeated exposure helps develop and train peanut butter or almond butter can also be a
the palate. good way to boost calorie intake, but they do
3. Choose unrefined whole grains such as brown come with some extra potassium and phos-
rice, barley, buckwheat, oats, quinoa, and phorus as well.
100 % whole grain breads. Avoid sprouted
grains as the phosphorus is more readily
absorbed. Phosphate Preservatives
4. Be cautious with boxed cereal as many are not
whole grain and contain significant amounts Phosphate additives sneak into food products and
of added sugar and trans fat as well as are listed on the ingredient list in many forms. The
356 L. Graf et al.

examples below are not a complete list of all the


phosphate additives but give an idea of how to
identify them on a package. Food companies are
currently not required to report the quantity of
phosphorus present in a food on the nutritional
facts panel. However, they are required to report
any phosphate additives in the product, and this
will appear in the ingredient list. Given that phos-
phate preservatives are the most readily absorbed
form of phosphorus, it is important for patients to
be taught to read the ingredient list (Fig. 3,
Table 7).

Common Phosphate Additives Used By


the Food Industry

Phosphoric acid
Pyrophosphates
Polyphosphates
Hexametaphosphate
Dicalcium phosphate
Disodium phosphate
Sodium
Polyphosphate
Phosphate

Hypoallergenic Diet as a Treatment


for Idiopathic Nephrotic Syndrome

Over the past three decades, several clinical stud-


ies and case reports have documented the success-
ful treatment of nephrotic syndrome with a
hypoallergenic diet. Cow’s milk in particular and
more recently gluten have been implicated in
some cases of idiopathic nephrotic syndrome.
However, the use of elimination diets in the man- Fig. 3 Sample food label: low-sodium turkey breast
agement of nephrotic syndrome has received little from Deli
attention from clinicians [75].
To date, there have been five published reports
that document the successful treatment of children ages 10–13 years old with frequently
nephrotic syndrome with a hypoallergenic diet relapsing, steroid-responsive nephrotic syndrome.
[76–80]. The first study to investigate the link During the study period, prednisone was
between nephrotic syndrome and cow’s milk sen- discontinued and the subjects were placed on a
sitivity was conducted by Sandberg et al. in 1977 liquid elemental diet. Remission occurred quickly
[76]. Sensitivity to cow’s milk was studied in six within 10 days in three of the six patients. Protein
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 357

Table 7 Variation in phosphorus absorption from food the diet. Full remission was achieved within
and preservatives 2–3 weeks. More recently, Rasoulpour described
Gastrointestinal four patients with steroid-dependent nephrotic
Source of phosphorus absorption % syndrome ages 4–10 years who responded to a
Animal protein (organic) 40–60 milk protein-free diet [80].
Cheese, milk, yogurt, beef, turkey, The resolution of nephrotic syndrome in
chicken, fish, egg
response to diet therapy supports the concept
Vegetarian protein 10–30
Bread, nuts, beans, vegetables,
that food sensitivities may trigger the disease in
chocolate some cases. Despite the promising results of these
Food additives and 80–100 studies, very little attention has been given to diet
preservatives (inorganic) in the treatment of nephrotic syndrome over the
Cola, packaged food, processed past 20 years. There has been a renewed interest
meat within the nephrology community about the asso-
ciation between food allergy and sensitivity, par-
ticularly milk protein and gluten with the
excretion decreased to less than 500 mg/24 h. development of nephrotic syndrome [75]. This
Significant proteinuria and edema returned when coincides with the large increase in awareness
cow’s milk was reintroduced. Four of the six and prevalence of food allergies. The prevalence
patients were able to maintain remission on a of food allegories has increased 18 % from
milk protein-free diet, while the other two subjects 1997 to 2007 (http://www.cdc.gov/nchs/data/
experienced relapses. When 20 mg of prednisone databriefs/db10.pdf). In addition, children with
was added and the diet was restricted further to food allergies are two to four times more likely
exclude grains, relapses were controlled. This to have other immune-mediated related health
suggests a possible sensitivity to wheat or gluten problems such as asthma and eczema. Children
in these two subjects. now require more time to outgrow the allergies
In 1989, Laurent et al. investigated the connec- [81, 82]. There is increased attention from the
tion between idiopathic nephrotic syndrome and a research community in nonceliac gluten sensitiv-
wide range of food allergies in 26 patients ages ity (NCGS), and the effects of NCGS on the
7–72 years old [77]. The foods investigated were immune system are still elucidated [83]. The
cow’s milk, egg, chicken, beef, pork, and gluten. effect of diet on the immune system is complex
Subjects were given skin testing with various food and may be mediated in part by the amount and
allergens. Patients were instructed to follow spe- type of bacteria in the gut. The influence of diet on
cific diet restrictions based on these results. Six of the gut microbiome may lead to the development
the 26 patients successfully responded to diet of allergies or immune system dysfunction.
treatment with resolution of nephrotic syndrome. More studies are needed to address the poten-
Two patients were successfully treated with the tial of using hypoallergenic diets as treatment for
elimination of gluten and one patient with the idiopathic nephrotic syndrome. Clinical trials are
exclusion of diary. Three patients required the currently underway investigating the effects of
elimination of multiple foods to achieve gluten on nephrotic syndrome. While the
remission. published literature at this time is limited, diet
In the early 1990s, Sieniawska et al. evaluated therapy is a treatment that confers no risk to the
the effects of a dairy-free diet in 17 patients ages patient and may decrease or eliminate their expo-
1–15 years with steroid-resistant nephrotic syn- sure to potentially harmful and toxic medications.
drome [78]. Six of the 17 subjects responded to Therefore, nutritional intervention with a hypoal-
the milk protein-free diet with remission of lergenic is a therapy that should be offered to
nephrotic syndrome. In all six subjects, protein- patients with steroid-resistant and frequently
uria resolved quickly – within 3–8 days of starting
358 L. Graf et al.

Table 8 Diet modification for nephrotic syndrome


Age Responsible foods (n = patients Length of time between initiating
Studies range with sensitivity in various studies) Diet therapy diet therapy and remission of NS
Sandberg >3 Cow’s milk (10) Specific Remission occurred within 3 days
et al. [76] years avoidance to 1 month
of age
Laurent Possible gluten/wheat (4)
et al. [77]
Rasoulpour Egg, chicken, and cow’s milk (1)
et al. [80] Gluten, pork, and cow’s milk (1)
Beef, pork, and egg (1)
Sieniawska <3 Cow’s milk (7) Cow’s milk Remission occurred within 3 days
et al. [78] years protein-free to 1 month of starting the diet
Salazar de of age diet
Sousa
et al. [79]

relapsing forms of idiopathic nephrotic syndrome 3. Foster BJ, McCauley L, Mak RH. Nutrition in infants
(Table 8). and very young children with chronic kidney disease.
Pediatr Nephrol. 2012;27(9):1427–39.
4. Group KW. KDOQI clinical practice guideline for
nutrition in children with CKD: 2008 update. Execu-
Conclusion tive summary. Am J Kidney Dis. 2009;53(3 Suppl 2):
S11–104.
5. Bunchman TE. Nutritional delivery in infants with
This chapter has reviewed the most recent CKD: techniques to avoid hyperkalemia. J Ren Nutr.
advances in nutrition and CKD over the past two 2013;23(5):387–8.
decades. There is an emphasis on maintaining 6. Thompson K, et al. Pretreatment of formula or
appropriate growth and electrolyte homeostasis expressed breast milk with sodium polystyrene sulfo-
nate (Kayexalate((R))) as a treatment for hyperkalemia
during infancy and childhood, as well as in infants with acute or chronic renal insufficiency.
preventing early onset cardiovascular disease. J Ren Nutr. 2013;23(5):333–9.
The goal of the chapter is to expand the awareness 7. Kovacs CS. Bone development and mineral homeosta-
of the importance of nutrition in improving CKD sis in the fetus and neonate: roles of the calciotropic and
phosphotropic hormones. Physiol Rev. 2014;94
outcomes throughout the life course journey. The (4):1143–218.
topics covered in the chapter are in accordance 8. Kovacs CS. Bone metabolism in the fetus and neonate.
with current NIH research goals [84]. When pos- Pediatr Nephrol. 2014;29(5):793–803.
sible, the focus should be not only on slowing the 9. Abrams SA. Calcium and vitamin d requirements of
enterally fed preterm infants. Pediatrics. 2013;131(5):
progression of CKD but also on regression of e1676–83.
disease to the greatest extent possible. 10. Hicks PD, et al. Total calcium absorption is similar
from infant formulas with and without prebiotics and
exceeds that in human milk-fed infants. BMC Pediatr.
2012;12:118.
References 11. Alon U, Chan JCM. Phosphate in pediatric health and
disease. Boca Raton: CRC Press; 1993. p. 327.
1. Ingulli EG, Mak RH. Growth in children with chronic 12. Bailey DA, et al. Calcium accretion in girls and boys
kidney disease: role of nutrition, growth hormone, during puberty: a longitudinal analysis. J Bone Miner
dialysis, and steroids. Curr Opin Pediatr. 2014;26 Res. 2000;15(11):2245–50.
(2):187–92. 13. Karlen J, Aperia A, Zetterstrom R. Renal excretion of
2. Graf L, et al. Nutrition assessment and hormonal influ- calcium and phosphate in preterm and term infants.
ences on body composition in children with chronic J Pediatr. 1985;106(5):814–9.
kidney disease. Adv Chronic Kidney Dis. 2007;14 14. Aladangady N, et al. Urinary excretion of calcium and
(2):215–23. phosphate in preterm infants. Pediatr Nephrol. 2004;19
(11):1225–31.
12 Nutrition Management in Childhood Kidney Disease: An Integrative and Lifecourse Approach 359

15. In: Dietary reference intakes for calcium, phosphorus, 33. Tuso PJ, et al. Nutritional update for physicians: plant-
magnesium, vitamin D, and fluoride. Washington, DC: based diets. Perm J. 2013;17(2):61–6.
National Academy Press; 1997. 34. Krishnamurthy VM, et al. High dietary fiber intake is
16. Gattineni J, Baum M. Developmental changes in renal associated with decreased inflammation and all-cause
tubular transport-an overview. Pediatr Nephrol. 2013. mortality in patients with chronic kidney disease.
17. Takaiwa M, et al. Fibroblast growth factor 23 concen- Kidney Int. 2012;81(3):300–6.
trations in healthy term infants during the early post- 35. Ramezani A, Raj DS. The gut microbiome, kidney
partum period. Bone. 2010;47(2):256–62. disease, and targeted interventions. J Am Soc Nephrol.
18. Raaijmakers R, et al. Pre-treatment of dairy and breast 2014;25(4):657–70.
milk with sevelamer hydrochloride and sevelamer car- 36. Brown K, et al. Diet-induced dysbiosis of the intestinal
bonate to reduce phosphate. Perit Dial Int. 2013;33(5): microbiota and the effects on immunity and disease.
565–72. Nutrients. 2012;4(8):1095–119.
19. Parekh RS, et al. Improved growth in young children 37. De Filippo C, et al. Impact of diet in shaping gut
with severe chronic renal insufficiency who use spec- microbiota revealed by a comparative study in children
ified nutritional therapy. J Am Soc Nephrol. 2001; from Europe and rural Africa. Proc Natl Acad Sci
12(11):2418–26. U S A. 2010;107(33):14691–6.
20. Ivanova E, et al. Cystinosis: clinical presentation, path- 38. Vaziri ND, et al. Chronic kidney disease alters intesti-
ogenesis and treatment. Pediatr Endocrinol Rev. nal microbial flora. Kidney Int. 2013;83(2):308–15.
2014;12 Suppl 1:176–84. 39. Niwa T. Indoxyl sulfate is a nephro-vascular toxin.
21. Nesterova G, et al. Cystinosis: renal glomerular and J Ren Nutr. 2010;20(5 Suppl):S2–6.
renal tubular function in relation to compliance with 40. Evenepoel P, et al. Uremic toxins originating from
cystine-depleting therapy. Pediatr Nephrol. 2014. colonic microbial metabolism. Kidney Int Suppl.
22. Meyer KB, Levey AS. Controlling the epidemic of 2009;114:S12–9.
cardiovascular disease in chronic renal disease: report 41. Vanholder R, et al. The uremic toxicity of indoxyl
from the National Kidney Foundation Task Force on sulfate and p-cresyl sulfate: a systematic review. J Am
Cardiovascular Disease. J Am Soc Nephrol. 1998;9 Soc Nephrol. 2014;25(9):1897–907.
(12 Suppl):S31–42. 42. Moe SM, et al. Vegetarian compared with meat dietary
23. Foster BJ, et al. Change in mortality risk over time in protein source and phosphorus homeostasis in chronic
young kidney transplant recipients. Am J Transplant. kidney disease. Clin J Am Soc Nephrol. 2011;6(2):
2011;11(11):2432–42. 257–64.
24. Mitsnefes MM. Cardiovascular disease in children 43. Noori N, et al. Organic and inorganic dietary phospho-
with chronic kidney disease. J Am Soc Nephrol. rus and its management in chronic kidney disease. Iran
2012;23(4):578–85. J Kidney Dis. 2010;4(2):89–100.
25. Flynn JT, et al. Blood pressure in children with chronic 44. Fukagawa M, Komaba H, Miyamoto K. Source mat-
kidney disease: a report from the Chronic Kidney Dis- ters: from phosphorus load to bioavailability. Clin J
ease in Children study. Hypertension. 2008;52(4): Am Soc Nephrol. 2011;6(2):239–40.
631–7. 45. Kalantar-Zadeh K, et al. Understanding sources of
26. Saland JM, et al. Dyslipidemia in children with dietary phosphorus in the treatment of patients with
chronic kidney disease. Kidney Int. 2010;78(11): chronic kidney disease. Clin J Am Soc Nephrol.
1154–63. 2010;5(3):519–30.
27. Oh J, et al. Advanced coronary and carotid arteriopathy 46. Gutierrez OM, et al. Dietary patterns and risk of
in young adults with childhood-onset chronic renal death and progression to ESRD in individuals with
failure. Circulation. 2002;106(1):100–5. CKD: a cohort study. Am J Kidney Dis. 2014;64(2):
28. Foley RN, Parfrey PS, Sarnak MJ. Clinical epidemiol- 204–13.
ogy of cardiovascular disease in chronic renal disease. 47. Dekker MJ, et al. Fructose: a highly lipogenic nutrient
Am J Kidney Dis. 1998;32(5 Suppl 3):S112–9. implicated in insulin resistance, hepatic steatosis, and
29. Shroff R, et al. HDL in children with CKD promotes the metabolic syndrome. Am J Physiol Endocrinol
endothelial dysfunction and an abnormal vascular phe- Metab. 2010;299(5):E685–94.
notype. J Am Soc Nephrol. 2014;25(11):2658–68. 48. Carvalho CR, et al. Fructose alters adiponectin, hapto-
30. Evenepoel P, Meijers BK. Dietary fiber and protein: globin and angiotensinogen gene expression in 3T3-L1
nutritional therapy in chronic kidney disease and adipocytes. Nutr Res. 2010;30(9):644–9.
beyond. Kidney Int. 2012;81(3):227–9. 49. Johnson RJ, Sanchez-Lozada LG, Nakagawa T. The
31. Uribarri J, Oh MS. The key to halting progression of effect of fructose on renal biology and disease. J Am
CKD might be in the produce market, not in the phar- Soc Nephrol. 2010;21(12):2036–9.
macy. Kidney Int. 2012;81(1):7–9. 50. Khitan Z, Kim DH. Fructose: a key factor in the devel-
32. Cordain L, et al. Origins and evolution of the Western opment of metabolic syndrome and hypertension.
diet: health implications for the 21st century. Am J Clin J Nutr Metab. 2013;2013:682673.
Nutr. 2005;81(2):341–54. 51. Madero M, et al. Dietary fructose and hypertension.
Curr Hypertens Rep. 2011;13(1):29–35.
360 L. Graf et al.

52. DiNicolantonio JJ, O’Keefe JH, Lucan SC. An unsa- 69. Qamar IU, et al. Effects of 3-month amino acid dialysis
vory truth: sugar, more than salt, predisposes to hyper- compared to dextrose dialysis in children on continu-
tension and chronic disease. Am J Cardiol. 2014; ous ambulatory peritoneal dialysis. Perit Dial Int.
114(7):1126–8. 1994;14(1):34–41.
53. Shroff R. Phosphate is a vascular toxin. Pediatr 70. Qamar IU, et al. Effects of amino acid dialysis com-
Nephrol. 2013;28(4):583–93. pared to dextrose dialysis in children on continuous
54. Shuto E, et al. Dietary phosphorus acutely impairs cycling peritoneal dialysis. Perit Dial Int. 1999;19(3):
endothelial function. J Am Soc Nephrol. 2009;20(7): 237–47.
1504–12. 71. Goraya N, et al. A comparison of treating metabolic
55. Tonelli M, et al. Relation between serum phosphate acidosis in CKD stage 4 hypertensive kidney disease
level and cardiovascular event rate in people with cor- with fruits and vegetables or sodium bicarbonate. Clin
onary disease. Circulation. 2005;112(17):2627–33. J Am Soc Nephrol. 2013;8(3):371–81.
56. Chang AR, et al. High dietary phosphorus intake is 72. Goraya N, et al. Dietary acid reduction with fruits and
associated with all-cause mortality: results from vegetables or bicarbonate attenuates kidney injury in
NHANES III. Am J Clin Nutr. 2014;99(2):320–7. patients with a moderately reduced glomerular filtra-
57. Sullivan CM, Leon JB, Sehgal AR. Phosphorus- tion rate due to hypertensive nephropathy. Kidney Int.
containing food additives and the accuracy of nutrient 2012;81(1):86–93.
databases: implications for renal patients. J Ren Nutr. 73. Nevin KG, Rajamohan T. Beneficial effects of virgin
2007;17(5):350–4. coconut oil on lipid parameters and in vitro LDL oxi-
58. Uribarri J. Phosphorus additives in food and their effect dation. Clin Biochem. 2004;37(9):830–5.
in dialysis patients. Clin J Am Soc Nephrol. 2009;4(8): 74. Noori N, et al. Dietary omega-3 fatty acid, ratio of
1290–2. omega-6 to omega-3 intake, inflammation, and sur-
59. Sherman RA, Mehta O. Phosphorus and potassium vival in long-term hemodialysis patients. Am J Kidney
content of enhanced meat and poultry products: impli- Dis. 2011;58(2):248–56.
cations for patients who receive dialysis. Clin J Am Soc 75. Uy N, et al. Effects of gluten-free, dairy-free diet on
Nephrol. 2009;4(8):1370–3. childhood nephrotic syndrome and gut microbiota.
60. Azocar MA, et al. Body composition in children on Pediatr Res. 2014;77:252.
peritoneal dialysis. Adv Perit Dial. 2004;20:231–6. 76. Sandberg DH, et al. Severe steroid-responsive nephro-
61. Quan A, Baum M. Protein losses in children on con- sis associated with hypersensitivity. Lancet. 1977;
tinuous cycler peritoneal dialysis. Pediatr Nephrol. 1(8008):388–91.
1996;10(6):728–31. 77. Laurent J, Lagrue G. Dietary manipulation for idio-
62. Uribarri J. The obsession with high dietary protein pathic nephrotic syndrome. A new approach to therapy.
intake in ESRD patients on dialysis: is it justified? Allergy. 1989;44(8):599–603.
Nephron. 2000;86(2):105–8. 78. Sieniawska M, et al. The role of cow’s milk protein
63. Wolfson M, Jones MR, Kopple JD. Amino acid losses intolerance in steroid-resistant nephrotic syndrome.
during hemodialysis with infusion of amino acids and Acta Paediatr. 1992;81(12):1007–12.
glucose. Kidney Int. 1982;21(3):500–6. 79. de Sousa JS, et al. Cow’s milk protein sensitivity: a
64. Chazot C, et al. Dialytic nutrition: provision of amino possible cause of nephrotic syndrome in early infancy.
acids in dialysate during hemodialysis. Kidney Int. J Pediatr Gastroenterol Nutr. 1995;21(2):235–7.
1997;52(6):1663–70. 80. Rasoulpour M, Dalidowitz C. 170: resolution of
65. Ikizler TA, et al. Amino acid and albumin losses during steroid-dependency by a dairy/hypoallergenic diet in
hemodialysis. Kidney Int. 1994;46(3):830–7. children with nephrotic syndrome. Am J Kidney Dis.
66. Canepa A, et al. Long-term effect of amino-acid dialy- 2007;49(4):B67.
sis solution in children on continuous ambulatory peri- 81. Skripak JM, et al. The natural history of IgE-mediated
toneal dialysis. Pediatr Nephrol. 1991;5(2):215–9. cow’s milk allergy. J Allergy Clin Immunol. 2007;
67. Canepa A, et al. Continuous ambulatory peritoneal 120(5):1172–7.
dialysis (CAPD) of children with amino acid solutions: 82. Wood RA. The natural history of food allergy. Pediat-
technical and metabolic aspects. Perit Dial Int. 1990; rics. 2003;111(6 Pt 3):1631–7.
10(3):215–20. 83. Mansueto P, et al. Non-celiac gluten sensitivity: litera-
68. Hanning RM, Balfe JW, Zlotkin SH. Effectiveness and ture review. J Am Coll Nutr. 2014;33(1):39–54.
nutritional consequences of amino acid-based vs 84. Kaskel F, et al. Improving CKD therapies and care:
glucose-based dialysis solutions in infants and children a national dialogue. Clin J Am Soc Nephrol. 2014;
receiving CAPD. Am J Clin Nutr. 1987;46(1):22–30. 9(4):815–7.
Physiology of the Developing Kidney:
Fluid and Electrolyte Homeostasis and 13
Therapy of Basic Disorders (Na/H2O/K/
Acid Base)

Isa F. Ashoor and Michael J. G. Somers

Contents Oral Rehydration Schemes . . . . . . . . . . . . . . . . . . . . . . . . . . 381


Use and Acceptance of Oral Rehydration
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 362 Solutions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 382
Total Body Water . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363 Choice and Volume of Parenteral Fluid . . . . . . . . . . . . . 383
Extracellular and Intracellular Fluid Colloid Solutions and Volume Resuscitation . . . . . . . . 384
Compartments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 363 Rapid Rehydration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 386
Effective Circulating Volume . . . . . . . . . . . . . . . . . . . . . . . . 365 Symptomatic Hyponatremia . . . . . . . . . . . . . . . . . . . . . . . . . 386
Asymptomatic Hyponatremia . . . . . . . . . . . . . . . . . . . . . . . 387
Water and Electrolyte Requirements . . . . . . . . . . . . . 366 Severe Hypernatremia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 388
Maintenance Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 366
Serum Osmolality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 368 Fluid and Electrolyte Therapy with Renal
Water Homeostasis in Acutely Ill Children Dysfunction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 389
or During the Perioperative Period . . . . . . . . . . . . . . . . . . 368 Impact of Kidney Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . 389
Derangements in Urinary Concentrating Ability Fluid and Electrolyte Therapy in the Pediatric
Affecting Water Homeostasis . . . . . . . . . . . . . . . . . . . . . . . 370 Intensive Care Unit . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 390
Nephrogenic Diabetes Insipidus . . . . . . . . . . . . . . . . . . . . . 370 Abnormalities in Serum Sodium Complicated
Special Considerations in Infants and Children by Kidney Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 391
with Chronic Kidney Disease . . . . . . . . . . . . . . . . . . . . . . . . 371 Potassium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394
Maintenance Electrolyte Therapy . . . . . . . . . . . . . . . . . . . 371
Hyperkalemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 394
Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 372 Causes of Hyperkalemia in Children . . . . . . . . . . . . . . . . 394
Alterations in Water Balance, Serum Sodium, Evaluation of Hyperkalemia in Children . . . . . . . . . . . . 398
and Cell Volume . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 372 Treatment of Hyperkalemia in Children . . . . . . . . . . . . . 399
Hyponatremia: Initial Approach . . . . . . . . . . . . . . . . . . . . . 373
Hypernatremia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 374 Hypokalemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 401
Causes of Hypokalemia in Children . . . . . . . . . . . . . . . . . 401
Fluid Replacement Therapy . . . . . . . . . . . . . . . . . . . . . . . 376 Diagnostic Evaluation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 405
Assessment of Volume Depletion . . . . . . . . . . . . . . . . . . . 376 Treatment of Hypokalemia in Children . . . . . . . . . . . . . 407
Oral Rehydration with Fluids Other Than ORS . . . . . 380
Oral Rehydration and Serum Sodium
Abnormalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 381

I.F. Ashoor
Division of Nephrology, Children’s Hospital, New
Orleans, LA, USA
e-mail: iashoor@chnola.org
M.J.G. Somers (*)
Division of Nephrology, Boston Children’s Hospital,
Harvard Medical School, Boston, MA, USA
e-mail: michael.somers@childrens.harvard.edu

# Springer-Verlag Berlin Heidelberg 2016 361


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_12
362 I.F. Ashoor and M.J.G. Somers

Acid–Base Homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . 407


Introduction
Perturbations in Acid–Base Balance . . . . . . . . . . . . . . . . 408
Simple Acid–Base Disorders . . . . . . . . . . . . . . . . . . . . . . . . 408 The spectrum of clinical conditions requiring the
Respiratory Acid–Base Disorders . . . . . . . . . . . . . . . . . . . 409 prescription of fluid and electrolyte therapy by
Metabolic Acidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 409
pediatric clinicians is vast, ranging from
Respiratory Compensation for Metabolic
Acidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410 rehydrating otherwise healthy children with
Serum Anion Gap . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410 acute gastrointestinal illness to correcting life-
Urine Anion Gap . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 411 threatening abnormalities in children with com-
Urine Osmolal Gap . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 411
plex chronic disease. Recognition of each child’s
Metabolic Alkalosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 411
Mixed Acid–Base Disorders . . . . . . . . . . . . . . . . . . . . . . . . . 412 individual clinical situation, notably any changes
The Δ Anion Gap/Δ HCO3 Ratio . . . . . . . . . . . . . . . . . . . 413 in normal physiology and homeostatic mecha-
Clinical Application and Approach . . . . . . . . . . . . . . . . . . 413 nisms that accompany the acute or chronic illness,
General Considerations for Therapy of
and each situation’s ultimate goal with respect to
Acid–Base Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 414
volume resuscitation or electrolyte correction is
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 415 crucial for the provision of the correct combina-
tion of fluid and electrolytes in the proper amount
of time.
Historically, an understanding of the morbidity
and mortality that accompanies significant pertur-
bations in fluid and electrolyte balance dates to
clinical observations made in epidemics of chol-
era and other diarrheal illness in the eighteenth
century [1]. The recognition that moribund
patients could be saved with provision of salt
solutions spurred interest in defining the fluid
and electrolyte needs in healthy individuals and
in developing clinical parameters for fluid and
electrolyte therapy. Over time, this work helped
to define the threshold for the minimum daily
provision of fluid and electrolytes – so-called
maintenance requirements – as well as a threshold
of maximal tolerance.
In the early twentieth century, clinicians began
to try to restore circulation in children with vol-
ume compromise by intraperitoneal injection of
saline or intravenous infusion of isotonic solu-
tions [2, 3]. Fluid spaces were defined in terms
of intracellular and extracellular compartments,
and the kidney’s role in the regulation of overall
body volume and specific solute gradients
between these fluid spaces became better appreci-
ated [4, 5].
By the middle of the twentieth century, simple
equations to link average daily metabolic rate to
daily fluid requirements were devised, and the
practice of calculating daily fluid and electrolyte
needs for an ill child based on consideration of
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 363

both “maintenance” needs and past and current exacerbates preexisting alterations in body fluid
losses or “deficits” was taught as the best volume, composition, or distribution or creates a
approach to minimize complications and improve new problem.
outcomes [6–8]. Despite caveats by the founders The sections that follow provide information
of this approach to individualize therapy and on fluid and electrolyte homeostasis in
be cognizant of clinical situations where they children, most notably the regulation of water,
may not pertain, emphasis on these empiric sodium, potassium, and acid–base balance.
equations led to formulaic hydration protocols There is additional focus on therapeutic
that became problematic in the setting of reduced approaches to correct basic imbalances as well
urine output or non-osmotically stimulated ADH as discussions on how chronic perturbations
release. in their homeostasis may affect usual approaches
Reassessment of this approach in the late to therapy.
twentieth and early twenty-first centuries has lead
to more widespread understanding that such
elaborate maneuvers are often unnecessary and Total Body Water
that there needs to be ongoing focus on the
child’s response to initial fluid and electrolyte ther- Extracellular and Intracellular Fluid
apy with consideration for adjustments to optimize Compartments
outcome [9]. More widespread recognition that oral
rehydration solutions are simple, safe, and effica- Water makes up a large component of body
cious alternatives for most children has also weight, the exact proportion varying in a child
impacted this tradition grounded on arbitrarily by age, body size, and body composition.
calculated intravenous fluid volumes [10]. In early gestation, up to 90 % of the developing
In more complex disorders of fluid and fetus may be water. By the early third trimester,
electrolyte pathophysiology, such as seen in total body water (TBW) of the developing fetus
critically ill children with sepsis, burns, trauma, approximates 80 % of body weight in kilograms.
or postoperatively or in children and adolescents After birth, this percentage falls further, from
with conditions such as chronic pulmonary 70–75 % in term infants to 65–70 % in toddlers
disease or complex congenital cardiac disease, and young children and, eventually, to 60 % in
an understanding of the distribution of body older children and adolescents. Body habitus also
fluids, usual fluid and electrolyte requirements, affects body water composition, with lean chil-
and the effect of disturbances in normal homeo- dren having higher TBW than obese and more
static balance remains vital to the correct prescrip- muscular children and adolescents manifesting
tion of therapy and the proper assessment of higher TBW than their less well-conditioned
response. To this extent, there continues to be a peers. Although 60 % is the usual benchmark for
place for an understanding of these precise calcu- estimating TBW in older children and adults,
lations and the ability to apply them when appro- there may be the need to adjust the estimated
priate [11]. Similarly, such an understanding percentage based on specific individual somatic
becomes paramount when approaching the care characteristics [12].
of a child with renal disease in whom normal Body water is distributed across intracellular
homeostatic mechanisms may not be able to and extracellular compartments (Fig. 1). The
come into play and with whom a standard intracellular compartment consists of all the
approach to fluid and electrolyte therapy may not water within the cells of the entire body, compris-
be appropriate. In these circumstances, the clini- ing about two-thirds of TBW or 40 % of body
cian needs to combine a systematic approach with weight. The extracellular compartment is smaller,
attention to individual clinical circumstance, comprising one-third of TBW or 20 % of body
thereby reducing the chances that any intervention weight. The extracellular compartment consists of
364 I.F. Ashoor and M.J.G. Somers

Fig. 1 Total body water:


fluid compartments and
solute composition
Plasma
Water

Intracellular Extracellular
Fluid Fluid
Interstitial
Fluid

the interstitial fluid that bathes all cells as well as an increased sodium concentration in the
the intravascular plasma water. The increased interstitium. Thus, potassium serves as the effec-
TBW as a proportion of body weight that is seen tive osmol intracellularly and sodium intersti-
in young children is the result of a relatively tially. Similarly, plasma proteins such as albumin
increased surface area as compared to body exert osmotic force to maintain water intravascu-
weight, resulting in an overall increased extracel- larly. Hydrostatic perfusion pressure counterbal-
lular compartment [13]. ances this osmotic force by pushing water across
The cell membrane separates the intracellular the capillary from the lumen to the interstitium.
and extracellular compartments. Input or output Changes in the distribution of the effective
from the body proceeds via some interface with osmoles or in hydrostatic perfusion can result in
the extracellular compartment, so there needs to redistribution of water between intracellular and
be net transport of fluid or solute across the cell extracellular spaces.
membrane for changes in the composition of the The equilibrium between the intracellular and
intracellular compartment. For instance, intrave- extracellular spaces is dynamic. Diffusional
nous electrolyte solutions are infused into the gradients, osmotic forces, and cellular transporter
extracellular intravascular space, and the subse- activities all combine to establish and maintain
quent delivery of the fluid and electrolyte in the differences in body compartment composition.
solution into the intracellular compartment Because the intracellular space cannot be directly
depends on a host of factors that influence trans- accessed, its composition can be altered only
port across the cell membrane. by affecting the extracellular space and its subse-
Most cell membranes are readily permeable quent communication with the cell. Any intake by
to water, and the distribution of water ingestion or infusion requires a new equilibrium
between the intracellular and extracellular spaces to be established as solute and fluid comes to be
comes to reflect osmotic forces. Each body exchanged from the extracellular space into
space contains a solute that is primarily seques- the cells. The final equilibrium is a result of all
tered within that compartment and that maintains of these complex biochemical, electrical, and
its osmotic gradient [14, 15]. For instance, physical interactions.
sodium–potassium adenosine triphosphatase Communication between the cellular spaces can
activity (sodium–potassium pump) found in be bidirectional so there can also be exchange from
all human cell membranes leads to an increased the intracellular space to the extracellular space,
intracellular concentration of potassium and allowing for transfer or release of cell metabolites.
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 365

Since the extracellular space can communicate with nutrients get delivered to the intracellular space
the external milieu, output from the extracellular and cellular metabolites get cleared from the
space to the external milieu results in effective intracellular space. Effective circulating volume
excretion from the body. Because no direct com- refers to that portion of the extracellular vascular
munication exists between the intracellular space space that actually perfuses the tissues and accom-
and the external milieu, any output from the cells plishes such an exchange.
themselves is mediated via the cell’s direct ability to The body attempts to maintain effective perfu-
interface with the extracellular compartment by sion through the intravascular space since any
way of interstitial fluid or plasma water. compromise proves deleterious to cell homeosta-
Impairment in the patient’s normal fluid and sis. Feedback mechanisms include baroreceptors
electrolyte homeostatic mechanisms will strikingly that respond to the stretch of specialized areas of
impact the patient’s TBW and its extracellular and the carotid arteries and the atrium. Hypoperfusion
intracellular constituents. The development of decreases stimulation of these stretch receptors,
hypertension in chronic kidney disease exemplifies triggering the secretion of vasopressin to increase
this disruption of normal balance. As GFR falls, the distal nephron water reabsorption with subsequent
kidney’s ability to excrete free water (CH2O) also expansion of the vascular volume. Similarly, in
declines, generally in the setting of a decreasing response to glomerular hypoperfusion, there is not
number of functioning nephrons with concomitant only decreased afferent arteriolar stretch but also
impairment of overall tubular solute excretion, most decreased glomerular filtration and presentation
notably salt. Superimposed on this dysregulation of of filtered sodium to the macula densa. These
salt and water balance may be other clinical factors stimuli lead to the secretion of renin from the
such as decreased effective arterial volume from juxtaglomerular cells of the afferent arteriole.
circulatory failure, leading to further renal salt and Renin release initiates a cascade ultimately increas-
water retention. This salt and water overload leads ing aldosterone-mediated sodium and water
to chronic TBW expansion and mediates systemic reabsorption from the kidney as well as increasing
hypertension with expansion of the extracellular angiotensin-mediated vasoconstriction and sodium
volume compartment. Appropriate therapy would and water uptake.
include sodium and fluid restriction and use of Effective circulating volume is impacted by
diuretics to reduce the total body burden of salt multiple factors, not the least of which include
and water and to restore the total body water to a the size of the vascular space and the influence
more physiologic state. Failure to appreciate the of various regulatory hormones. As a component
preexisting expansion of the total body water of the extracellular body water, the size of the
because of chronic salt and water overload could vascular space often parallels the size of the extra-
prove deleterious if management did not include cellular space. The size of the vascular space and
some measure to address this perturbation. This the adequacy of the effective circulating volume
example also highlights that fluid and electrolyte do not, however, always vary coordinately. The
therapy in children may involve the removal of extracellular space may be replete or expanded
solute and water, in contrast to the more common and the actual effective circulating volume
clinical scenario that focuses on correction of elec- decreased. For instance, children with significant
trolyte and volume deficits. liver disease are often edematous, due to sodium
retention and expansion of the interstitial compo-
nent of the extracellular space. The intravascular
Effective Circulating Volume component of their extracellular space may also
be expanded as a result of factors resulting in avid
Effective circulating volume is a more abstract salt and water reabsorption by the kidney. But,
concept than the division of body water into because of portal hypertension, splanchnic vessel
intracellular and extracellular fluid compartments. congestion, and multiple arteriovenous spider
As the vascular volume circulates, oxygen and angiomas that are seen with this condition, much
366 I.F. Ashoor and M.J.G. Somers

of the expanded intravascular volume is ineffec- Table 1 Alterations in effective circulating volume
tive – it does not serve to perfuse the tissues and Cause Mechanism
accomplish effective cellular exchange. Thus, Contracted Water or sodium chloride
these children act as if they are volume depleted: extracellular fluid deficit
they avidly reabsorb any filtered sodium and space
excrete small volumes of urine, and they Massive Loss of vascular tone
vasodilatation sustaining perfusion pressure
vigorously continue to expand their already
Loss of intravascular Osmotic fluid losses into the
overexpanded extracellular space by reabsorbing osmotic pressure interstitium
even more salt and water in response to the effects Overfill of the Hydrostatic fluid losses into
of renin and ADH. Similarly, this paradoxical intravascular space the interstitium
state of sodium avidity and ADH-mediated water Hemorrhage Direct loss of blood and
reabsorption characterizes children with nephrotic plasma water
syndrome or with cardiac failure despite their
preexisting expansion of the extracellular space.
In prescribing fluid and electrolyte therapy, the fluid compartment. In such a circumstance, failure
clinician needs to assess the patient’s current to initiate appropriate fluid and electrolyte therapy
extracellular volume status along with the effec- may result in eventual compromise of the effec-
tive circulating volume and then reconcile these tive circulating volume.
with potential causes of volume loss. At all times, Important initial clinical signs to assess in any
the first maneuver should be to maintain an effec- patient being evaluated for fluid therapy include
tive circulating volume and, to accomplish this pulse rate and capillary refill. Tachycardia and
properly, therapeutic decisions should be based sluggish refill generally precede more obvious
on the patient’s unique clinical circumstances at signs of ineffective circulation such as hypoten-
that point in time. Consequently, effective man- sion and oliguria. Clinical symptoms may also be
agement may require rather disparate therapeutic nonspecific and include fatigue and lethargy that
interventions. For instance, expansion of the are often attributed to an underlying illness rather
extracellular volume with vigorous rehydration than volume depletion. Proper restoration of
may be called for in a child with poor perfusion effective circulating volume or extracellular fluid
secondary to dehydration from diarrhea, whereas compartment depletion requires both an under-
another child with equally poor perfusion due to standing of baseline fluid and electrolyte needs
cardiodynamic compromise may be intravascu- as well as consideration of extenuating clinical
larly replete and require the initiation of pressor circumstances unique to the patient.
therapy, and yet another child with edema from
relapsed nephrotic syndrome may actually
require fluid and salt restriction. These examples Water and Electrolyte Requirements
underscore that loss of effective circulating
volume generally arises as a result of one or Maintenance Therapy
more broad perturbations in the extracellular
fluid compartment that impacts effective Maintenance therapy denotes the amount of water
perfusion (Table 1). In hospitalized children and electrolytes required to replace usual daily
where there may be both aberrant disease-related losses and to maintain an overall balance of
physiology as well as iatrogenic derangements water and electrolytes with no net gain or loss;
of regulatory response, the causes of effective as noted below, “maintenance” fluid therapy
volume perturbations may be even more complex. should not be conflated with simple estimation
The clinical signs and symptoms of effective formulas taught in medical school and residency.
circulating volume loss may be subtle. There may Maintenance needs are a function of homeostatic
even be preservation of effective circulating vol- and environmental factors and vary from day to
ume in the face of an overall depleted extracellular day and from child to child. In the average child
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 367

with adequate access to water and food, these Table 2 Relationship of body weight to metabolic and
maintenance needs are generally readily met maintenance fluid needs
[6, 16]. In the ill or hospitalized child who requires Maintenance
therapeutic intervention and in whom there may Body weight Metabolic needsa fluid needsb
be ongoing aberrant physiology, these needs must (kg) (kcal/day) ml/day ml/h
be considered more carefully. 5 500 500 20
10 1,000 1,000 40
Maintenance fluid and electrolyte requirements
15 1,250 1,250 50
are typically calculated based on weight or surface
20 1,500 1,500 60
area, but individual clinical circumstance must be
30 1,700 1,700 70
considered when making such calculations
40 1,900 1,900 80
[17]. For instance, the 20-kg child who is well 50 2,100 2,100 90
will require a far different “maintenance” quantity 60 2,300 2,300 95
of fluid and electrolytes than the 20 kg child who is 70 2,500 2,500 105
tachypneic and febrile or the 20 kg child who is As described in Holliday and Segar [6]
anuric and on a ventilator in the pediatric intensive a
Based on 100 kcal/kg for the first 10 kg of body weight +
care unit. Careful assessment of the patient’s vol- 50 cal/kg for the next 10 kg of body weight + 20 cal/kg
ume status and attention to the balance of overall for the next 50 kg of body weight
b
Based on need of 1 ml of water to metabolize 1 kcal of
daily input and output will prove more useful at energy
arriving at a correct estimate of daily fluid and
electrolyte needs than using mathematical equa-
tions without clinical correlation. Despite these
caveats, it is widespread clinical practice to make
Table 3 Factors affecting insensible water losses
certain empirical assumptions regarding daily
needs for water and the major electrolytes. Decreased
Increased Losses % change losses % change
Historically, daily maintenance water needs
Prematurity 100–300 Enclosed 25–50
were first derived based on measurements in hos- incubator
pitalized patients suggesting that water needs Radiant warmer 50–100 Humidified 15–30
could be linked to energy expenditure (Table 2) air
[6, 16]: for the first 10-kg of body weight, 100 ml Phototherapy 25–50 Sedation 5–25
of water per kg is provided daily; for the next Hyperventilation 20–30 Decreased 5–25
10 kg of body weight, 50 ml of water per kg is activity
provided daily; and for every kg of body weight in Increased 5–25 Hypothermia 5–15
activity
excess of 20 kg, 20 ml of water per kg is provided
Hyperthermia 12 %/ C
daily. In addition, in the process of oxidation of
carbohydrate and fat, approximately 15 ml of
water is generated for every 100 kcal of energy Clinical factors can have a striking impact on
produced. This water of oxidation contributes sig- insensible water losses (Table 3). Fever increases
nificantly to overall water balance. insensible losses by >10 % for each degree Cel-
Maintenance water losses occur from urine sius. Premature infants with relatively increased
output and from insensible sources that are almost surface areas for size can have insensible losses
exclusively evaporative and respiratory losses. In two- to threefold higher than baseline, especially
the child with average metabolic demands, for if they are on open warmers or under photother-
every 100 kcal of energy expended, 100 ml of apy. On the other hand, children on ventilators
water must be ingested. Of this 100 ml of water, providing humidified oxygen may have half the
40 ml is lost insensibly and 75 ml is lost as urine insensible losses of a non-ventilated child.
output. Since oxidative metabolism generates Similarly, urinary water output can vary tre-
15 ml of water in the course of producing the mendously. A child with a renal concentrating
100 kcal of energy, net water balance is zero. defect or ADH unresponsiveness may have
368 I.F. Ashoor and M.J.G. Somers

obligate urinary water losses of several liters per small variations outside of the normal range by
day, whereas an oligoanuric child will have no adjusting posterior pituitary ADH release. With
appreciable urinary water losses regardless of hypovolemia, changes in osmolality augment
input. In any child with normal renal function, ADH release further. ADH effect on water perme-
even in the setting of maximal ADH stimulation, ability of the collecting tubule is a principal influ-
there is a minimal volume of urinary water neces- ence on the regulation of water balance.
sary to excrete the osmotic load arising from diet Sustained alterations in water intake or excre-
solute and basal metabolism. As a result, even the tion result in hypo- or hyperosmolality developing
child concentrating urine to 1,200–1,400 mOsm/L as the usual ratio of extracellular solute to water is
will lose nearly 25 ml of urinary water per perturbed. Since sodium is the largest component
100 kcal of energy expended [6]. of extracellular osmolality, its concentration can
Recently, there has been recognition that the be influenced profoundly by changes in water
relationships between energy expenditure and metabolism. An understanding of this link
water requirements demonstrated by stable hospi- between water regulation and serum sodium
talized children at bed rest do not apply to anesthe- values is crucial when prescribing fluid and elec-
tized or critically ill children [18]. For instance, in trolyte therapy. Most importantly, the clinician
infants and children studied during general anes- must recognize that hypo- or hypernatremia is
thesia, energy expenditure was half that of awake usually a manifestation of impaired water regula-
inactive children. On the other hand, water needs tion and that therapy must address regulation of
for cell metabolism were increased over baseline by water balance rather than alterations in body
about 60%, leaving the overall relationship sodium stores.
between water needs and caloric expenditure sim-
ilar in both situations. In critically ill children,
however, the overall relationship may be affected Water Homeostasis in Acutely Ill
and volumes may need to be reduced by 40–50% to Children or During the Perioperative
prevent positive water balance [19]. Period

In ill children, there are multiple causes of both


Serum Osmolality physiologic and aberrant vasopressin effect as listed
in Table 4 [23]. As a result, if these children
Serum osmolality is determined by concentrations receive hypotonic intravenous fluids for prolonged
of sodium, glucose, and urea nitrogen in the serum periods of time or in volumes exceeding those
and maintained by water homeostasis [20]. Osmo- generally recommended, there is the risk of acute
lality is estimated by the equation: (2  serum hyponatremia. After volume resuscitation with
Na) + (serum glucose/18) + (BUN/2.8), where isotonic fluids, most hospitalized children were
the serum sodium is measured in mEq/L and the
glucose and BUN in mg/dl. In the majority of Table 4 Common causes of vasopressin effect in hospi-
children with no functional renal impairment and talized children
normal glucose metabolism, the contributions of Category Specific etiology
BUN and glucose to the effective osmolality are Physiologic Hyperosmolar state, hypovolemia
negligible, and the serum osmolality can be Pulmonary Pneumonitis, pneumothorax, asthma,
bronchiolitis, cystic fibrosis
estimated by doubling the serum sodium concen-
Drug effect Narcotics, barbiturates, carbamazepine,
tration [21, 22]. Most children have a serum
vincristine, cyclophosphamide
osmolality between 270 and 290 mOsm/kg, Metabolic Hypothyroidism, hypoadrenalism,
corresponding to normal serum sodium values porphyria
between 135 and 145 mEq/L. CNS Infection (meningitis or encephalitis),
Chemoreceptors in the hypothalamus constantly tumor, trauma, hypoxia, shunt
monitor serum osmolality, responding to even malfunction, nausea, pain, anxiety
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 369

traditionally provided hypotonic fluids for their hyponatremic after hospital admission, most as a
maintenance therapy. Given the tendency for many consequence of aggressive intravenous hydration
of these children to manifest vasopressin effect inde- with hypotonic fluid. These hyponatremic chil-
pendent of the usual osmotic and volume-related dren received on average three times more the
stimuli, isotonic fluids may be a safer alternative as volume of electrolyte free water than their
the source of maintenance fluid even after acute eunatremic peers and were also three times more
volume repletion in this setting [24]. likely to receive fluids at rates exceeding
Similarly, some have called for using isotonic recommended maintenance rates [30].
fluids whenever a maintenance infusion is needed A retrospective analysis of postoperative
in the perioperative period when oral hydration admissions to a pediatric ICU found that 11 % of
has been held and when high ADH levels may be children became hyponatremic (serum sodium
expected because of pain or anxiety. In these <130 mEq/L) [31]. In the hyponatremic children,
instances, children could receive intravascular there was a trend towards an increased likelihood
volume expansion with an initial infusion of of hypotonic fluid provision, although there was
20–40 ml/kg of isotonic fluid over a period of a no statistical difference demonstrated in the
few hours and then, rather than a transition to incidence of either moderate (<130 mEq/L) or
hypotonic fluid, continued on isotonic fluid as severe (<125 mEq/L) hyponatremia. None of
dictated by clinical circumstance. Proponents these children developed any neurologic sequelae
believe it both decreases the number of hospital- or other morbidity, but serum electrolytes were
ized children who develop hyponatremia and assayed four times daily likely resulting in clinical
prevents hyponatremia-related central nervous interventions to prevent hyponatremia from
system damage [25]. exacerbating.
Others have claimed that maintenance infusion A prospective randomized study of 102 chil-
of isotonic saline to all ill or perioperative dren with gastroenteritis demonstrated that the
children results in sodium loading in those with- risk of developing hyponatremia decreased with
out any trigger for water retention [8, 26]. In fact, provision of isotonic saline [32]. Eunatremic chil-
hypernatremia has been described in some dren at presentation were most protected from
children receiving even less sodium than that subsequent falls in serum sodium when provided
provided in maintenance isotonic infusions isotonic (0.9 % NaCl) versus hypotonic (0.45 %
[27]. Such cases of hypernatremia are often a NaCl) solutions. Urinary biochemistries demon-
result of an underlying renal concentrating defect, strated that hyponatremic children were more
significant free water loss from sources other than likely to retain sodium and increase serum sodium
urine, or aggressive fluid restriction [28]. Children levels with isotonic fluids, whereas children with
with certain renal or cardiopulmonary problems normal serum sodiums at presentation were
may be especially sensitive to such sodium loads able to excrete excess sodium appropriately and
and may more readily develop unintended prevent hypernatremia.
sequelae of such sodium provision [29]. A meta-analysis of six studies comparing
Several studies have shown that children with hypotonic and isotonic maintenance intravenous
acute illness requiring emergency department solutions in children suggested that no single fluid
evaluation or hospitalization do seem to be at composition or rate is ideal for all children but that
risk for hyponatremia. In one report of 103 an isotonic or nearly isotonic solution may prove
children, nearly 80 % had elevated serum ADH less likely to cause symptomatic hyponatremia in
levels and increased plasma renin activity, inde- acute illness and the perioperative period [33].
pendent of the underlying illness. As expected Nonetheless, in many clinical situations where
with ADH release, plasma osmolality was non-osmotic ADH effect can be expected,
reduced significantly [23]. In another report, children are still frequently receiving hypotonic
fewer than 5 % of children were hyponatremic fluid therapy, and such iatrogenic hyponatremia
at presentation, but nearly 10 % became may not be unusual [34].
370 I.F. Ashoor and M.J.G. Somers

Derangements in Urinary mechanism is seen in conditions with defective


Concentrating Ability Affecting Water sodium reabsorption from the thick ascending
Homeostasis limb of the loop of Henle (TAL) and distal neph-
ron required to create the medullary osmotic gra-
The maximum ability of the kidneys to concen- dient [37], hence the coexistence of a solute
trate urine determines the obligatory urine volume diuresis component to the polyuria in these set-
required to excrete the daily solute load. On aver- tings. Chronic polydipsia, either as a primary phe-
age, 600 mOsm are generated daily which would nomenon or as a response to polyuria, can lead to
require approximately 2 L of urine (at a concen- “washout” of the renal medullary concentration
tration of 300 mOsm/kg) to be excreted. When the gradient further worsening the polyuria [38].
kidneys are faced with a higher solute load, or
the maximum concentrating ability is reduced
despite a stable solute load, the daily urine output Nephrogenic Diabetes Insipidus
will increase to maintain equilibrium. This clini-
cally manifests as polyuria. Polyuria may be very Nephrogenic diabetes insipidus (NDI) refers to a
difficult to assess in infants and toddlers outside state of water diuresis secondary to resistance to the
the hospital setting. However, certain clues action of ADH in the distal nephron. Under condi-
include a history of polyhydramnios in conditions tions of volume depletion or hyperosmolality,
that are hereditary in nature [35], excessive thirst, ADH acts on arginine vasopressin receptor
recurrent episodes of dehydration, and in the most 2 (AVPR2) on the basolateral membrane of princi-
severe cases failure to thrive [36]. In toilet-trained pal cells leading to insertion of aquaporin 2 (AQP2)
children and adolescents, nocturia and secondary channels in the luminal membrane, thereby increas-
enuresis can be the first manifestation of an ing water reabsorption [39, 40]. An intact medul-
acquired urinary concentrating defect. lary concentration (osmotic) gradient is critical to
Polyuria can result from solute (or osmotic) fully realize the ADH effect on this segment [41].
diuresis, water diuresis, or a combination of In infants and young children, NDI is usually
both. Assessment of a random spot urine sample hereditary, secondary to genetic mutations affecting
osmolality should facilitate the distinction the AVPR2 (X linked) [42, 43] or the AQP2 gene
between the two entities in most cases. The classic (autosomal) [40, 42, 44, 45]. The condition com-
case of osmotic diuresis, where urine osmolality is monly manifests early with failure to thrive, poly-
usually equal to or exceeds the serum osmolality, uria, polydipsia, nocturia, and enuresis [36].
can be seen in the child presenting with new-onset Recurrent episodes of hypernatremic dehydration
diabetes mellitus. The hyperglycemia produces are common in the setting of impaired thirst mech-
a high filtered glucose load that exceeds the anism or lack of free access to water, particularly in
proximal tubule transport maximum for glucose. infants before their condition is recognized.
Other osmotic agents such as urea and mannitol The diagnosis of hereditary NDI is typically
can cause a similar picture. suspected on clinical grounds and supported by
Water diuresis encompasses conditions where high normal serum Na concentration, concomitant
the distal nephron mechanisms for urinary con- hypoosmotic urine, and lack of response to
centration are impaired leading to the loss of large desmopressin administration, without resorting to
amounts of dilute urine where the osmolality a water deprivation test. A water deprivation test
is typically far less than serum osmolality. should be reserved for older children with equivo-
This can be a result of deficient ADH secretion cal clinical findings and only performed under
(central diabetes insipidus), impaired renal ADH close medical supervision. Molecular genetic test-
responsiveness (nephrogenic diabetes insipidus), ing is available to confirm the diagnosis [42].
or reduced renal medullary concentration gradient In older children and adolescents, NDI is usu-
required to reabsorb water from the distal nephron ally acquired secondary to conditions compromis-
under the influence of ADH. The latter ing the renal medullary concentration gradient,
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 371

which reduces water reabsorption under the influ- tubulointerstitial injury from chronic pyelonephri-
ence of ADH. Such conditions include, among tis, interstitial nephritis, calcium deposition in
others, hypercalcemia [46], hypokalemia [47, nephrocalcinosis [46], and sickle-cell disease
48], drug-induced NDI [49–52], and chronic [62] reduces the medullary concentration gradient
tubulointerstitial injury seen in some forms of leading to ADH hyporesponsiveness. Children
chronic kidney disease as discussed in the next with congenital or hereditary disorders primarily
section. In addition to nocturia, polyuria, and affecting the tubulointerstitium and medullary
polydipsia, children with acquired NDI will also regions (e.g., nephronophthisis [63] and medul-
manifest symptoms of the underlying disorder. lary cystic kidney disease [64]) or solute transport
Unlike hereditary NDI, the onset of symptoms is in the distal nephron required to generate the
typically gradual and, oftentimes, at least partially medullary concentration gradient (e.g., Bartter
reversible with correction of the underlying cause. syndrome) [35, 65, 66] will also manifest
In the absence of an identifiable acquired impaired urinary concentrating ability. In children
reversible cause for NDI, treatment aims at mini- with chronic kidney disease secondary to obstruc-
mizing polyuria and providing sufficient fluids to tive uropathy, a superimposed urinary tract infec-
prevent recurrent episodes of hypernatremic tion may be followed by a brief period of polyuria
dehydration. This is particularly challenging if secondary to solute diuresis. The urinary tract
the thirst mechanism is impaired or in infants infection is postulated to induce a state of transient
and small children who rely on caregivers for pseudohypoaldosteronism that promotes salt
access to water. In some cases, continuous tube wasting and resolves with treatment [67, 68].
feedings may be required to facilitate normal Also, worsening azotemia from an acute on
growth. Polyuria can be reduced by minimizing chronic obstruction is usually followed by a brief
the dietary solute load that allows the kidneys to period of post-obstructive diuresis when the
excrete it in a smaller volume given the fixed obstruction is relieved, secondary to the osmotic
concentration. This can be accomplished by salt diuretic effect of the filtered urea load [69].
restriction and in selected cases, where failure to The mainstay of treatment in polyuric children
thrive is not a concern, protein restriction under with chronic kidney disease is provision of suffi-
close dietary supervision [53]. Another strategy to cient free water to prevent dehydration and
reduce the daily urine output involves using hypernatremia. In contrast to children with hered-
low-dose thiazide diuretics to induce a state of itary NDI, they may require salt supplementation
mild volume depletion which increases sodium if the salt wasting is the underlying mechanism for
and water reabsorption in the proximal nephron impaired urinary concentration. If these children
[54]. The use of nonsteroidal anti-inflammatory are admitted for intravenous fluid therapy, assess-
drugs (NSAIDs) to reduce renal prostaglandin ment of urine osmolality can be a valuable tool to
synthesis [55, 56] and ADH analogs (e.g., guide fluid tonicity choice to correct or maintain
desmopressin) may be effective in selected normal serum sodium concentration.
patients [57].

Maintenance Electrolyte Therapy


Special Considerations in Infants
and Children with Chronic Kidney Estimates for the maintenance requirements of the
Disease major electrolytes sodium, potassium, and chloride
can also be made based on metabolic demands and
Several mechanisms contribute to impaired daily water needs [6]. For sodium and chloride,
urinary concentrating ability in chronic kidney approximately 2–3 mEq/100 ml of daily water
disease [58–60]. In vitro studies have suggested requirement is needed with 1–2 mEq of potassium
a role for decreased expression of AVPR2 mRNA for each 100 ml of daily water need. Again, these
in rats with chronic renal failure [61]. Progressive estimates require adjustment based on clinical
372 I.F. Ashoor and M.J.G. Somers

circumstance, but the daily intake of most healthy Since sodium is the major extracellular osmol,
individuals contains more than adequate electro- alterations in serum sodium can result in water
lytes for maintenance needs. Although at times flux between the intracellular and extracellular
there can be significant electrolyte losses through spaces. Because significant water movement into
the skin or the gastrointestinal tract, most electro- or out of cells could prove deleterious to cell
lyte losses are urinary [17, 70]. In the setting of function, cell volume is closely regulated to min-
anuric renal failure and no electrolyte losses from imize such shifts [71]. For instance, with
other sources, lower levels of electrolyte supple- hyponatremia there is decreased effective osmo-
mentation may be needed, and water supplementa- lality in the extracellular space. As a result, water
tion alone to replace insensible fluid losses may can shift from the plasma into the intracellular
maintain adequate electrolyte balance. space and cell swelling will occur. To counterbal-
As with provision of water, electrolyte therapy ance such swelling, the cell acutely regulates its
should be tailored to individual need. For sodium volume by transporting electrolytes, especially
and chloride, this requires careful assessment of potassium, from the intracellular space into the
the extracellular fluid space, especially the effec- extracellular space, thereby decreasing the
tive circulating volume. Providing too little osmotic gradient for water transfer. Over several
sodium chloride results in volume contraction days, when faced with chronic hyponatremia or
and circulatory compromise; providing too much chronic hypoosmolality, the cells will also achieve
causes volume overload and sequelae such as effective volume regulation by losing organic
hypertension and edema. osmolytes such as taurine and inositol, thereby
Similarly, inappropriate potassium supplemen- further diminishing the osmotic gradient for
tation may have significant clinical ramifications. water transfer into the cell [72].
In children with diminished renal function or who With hypernatremia, the osmotic gradient
are at risk of hyperkalemia for other reasons, it favors water movement out of the cells and into
may be appropriate to forego maintenance potas- the extracellular space, and cells will shrink
sium supplementation. When supplementation is without protective mechanisms. Acutely, there is
given, there needs to be timely reassessment of transport of electrolytes intracellularly, whereas
serum levels. Oral administration of supplemental with chronic hypernatremia production of intra-
potassium is safer than bolus injection and intra- cellular organic idiogenic osmoles is enhanced
venous potassium supplements rarely need to [72, 73]. Together, these act to blunt the loss
exceed 0.5 mEq/kg/h [20]. of intracellular volume that would otherwise
occur.
Changes in serum sodium values that evolve
Sodium slowly and reflect chronic processes tend to allow
for maximal counter-regulation and fewer clinical
Alterations in Water Balance, Serum sequelae. On the other hand, sudden profound
Sodium, and Cell Volume alterations – especially with serum sodium values
below 120 mEq/L or above 160 mEq/L – are often
Since the regulation of osmolality is achieved by accompanied by neurologic complications
changes in water intake and excretion, serum directly related to the acute changes in cell volume
sodium levels can vary with these alterations in in the central nervous system.
water balance. Generally, serum sodium is regu- These regulatory mechanisms must also be
lated between 135 and 145 mEq/L. A serum kept in mind when formulating specific therapeu-
sodium concentration below 130 or above tic intervention. Acute perturbations can be
150 mEq/L is out of the range of normal homeo- corrected more rapidly than chronic conditions
stasis and most often indicates a problem with because the full gamut of responses to the imbal-
water balance. ance has yet to come into play.
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 373

Hyponatremia: Initial Approach Table 5 Etiology of hyponatremia


Urinary Na (mEq/L)
Hyponatremia is commonly defined as a serum Circulating
sodium concentration less than 135 mEq/L, though volume 20 20
it is rare for there to be any clinical effect in levels Decreased Burns Adrenal
between 130 and 135 mEq/L. Low serum sodium insufficiency
Cystic fibrosis Diuretics – early
values are usually the result of persistent ADH
Diuretics – late Salt wasting
effect and a relative surfeit of water for solute in
Gastroenteritis
the extracellular space; hyponatremia uncommonly
Normal or Cardiac failure Renal failure
arises secondary to depleted salt stores alone. increased Hepatic SIADH
Infrequently, pseudohyponatremia may be seen. cirrhosis
Pseudohyponatremia is not a depletion of sodium Nephrotic Water
stores but a change in the usual makeup of the syndrome intoxication
extracellular space such that there is a pathologic
elevation of another solute that decreases the rela-
tive concentration of sodium. Common etiologies of aldosterone and angiotensin. As a result, urine
of pseudohyponatremia include hypergylcemia, sodium values are generally low (<20 mEq/L)
hyperlipidemia, and hyperproteinemia. In these and water reabsorption in the distal nephron is
clinical conditions, serum sodium values tend to facilitated by high levels of ADH. In the face of
be only modestly depressed. Since there is no continuing sodium losses exceeding intake, this
underlying true anomaly in sodium or water stores state of vigorous ADH effect leads to a relative
in these conditions, the serum sodium need not be excess of water and concomitant hyponatremia.
addressed with any therapeutic maneuvers. With A similar clinical state of hyponatremia can
the introduction of ion-sensitive electrodes for the also be seen in cystic fibrosis where there are
measurement of plasma sodium concentration, increased skin losses of sodium and chloride,
pseudohyponatremia related to the presence of with bleeding, with burns, and with certain losses
confounding factors in the laboratory assay is of fluid from the intravascular space of the extra-
much less commonly encountered. cellular fluid into the interstitial space as can occur
To clarify the etiology of the hyponatremia, the postoperatively, in conditions of vascular leak
clinician should assess the patient’s extracellular such as sepsis, or with peritonitis. Such
volume status and determine if it is decreased, hyponatremia can also be seen following a period
normal, or increased. Then, by measuring urine of diuretic therapy. In response to chronic
sodium excretion and determining the renal diuretic-mediated volume contraction, the mech-
response to the hyponatremia, it becomes easier anisms outlined above come into play. Thiazide
to determine if the patient should receive sodium diuretics, especially in combination with a loop
and water or if sodium and water restriction is the diuretic such as furosemide, are particularly prone
appropriate therapy (Table 5). to inducing such hyponatremia.
The appropriate therapeutic response to these
Hyponatremia: Decreased Volume Status conditions is the provision of sodium and water
and Urine Na < 20 mEq/L either by use of intravenous or oral electrolyte
Decreased circulating volume is usually seen with solutions. This results in restoration of sodium
states of significant sodium loss. Most commonly, balance and volume expansion.
loss is from the gastrointestinal tract as a result of
vomiting, diarrhea, or tube drainage. The renal Hyponatremia: Decreased Volume Status
response to the decreased effective circulating and Urine Na > 20 mEq/L
volume involves increased activity of the Decreased circulating volume with a random
renin–angiotensin axis and relatively high levels urinary sodium excretion >20 mEq/L is
374 I.F. Ashoor and M.J.G. Somers

indicative of renal salt wasting, either from an Hyponatremia: Normal or Expanded


intrinsic tubulopathy or from early diuretic Volume and Urine Na > 20 mEq/L
effect. Less commonly, adrenal insufficiency Hyponatremia in the setting of normal or
can cause sodium wasting from the cells of the increased effective circulating volume is always
distal nephron. Such a deficiency can arise from related to persistent ADH effect [15]. If the ran-
an intrinsic endocrine defect such as congenital dom urine sodium value is >20 mEq/L, the most
adrenal hyperplasia related to 21-hydroxylase common clinical scenario is the syndrome of inap-
deficiency, from some secondary impairment of propriate antidiuretic hormone secretion or
adrenal function caused by infection, bleeding, SIADH. SIADH can arise from disparate clinical
or malignancy or from pharmacologic adrenal conditions including the postoperative child, the
suppression without adequate replacement child with significant pain, or the child with pul-
therapy. In the setting of adrenal insufficiency, monary disease. Ill children may be at risk for
provision of appropriate adrenal hormone both inappropriate ADH secretion and inappropri-
replacement as well as adequate sodium and ate ADH effect [74]. In SIADH, despite a state of
water proves therapeutic. With renal salt hypoosmolality, the urine is inappropriately con-
wasting, supplementation with sodium and any centrated as a result of ongoing ADH secretion
other electrolytes exhibiting impaired renal and ADH-mediated water reabsorption from the
reabsorption is useful. distal nephron. Appropriate therapy for SIADH
includes restricting water intake and attending to
Hyponatremia: Normal or Expanded any underlying clinical factors predisposing to
Volume and Urine Na < 20 mEq/L this syndrome. Provision of fluids containing
Normal or increased circulating volume and higher concentrations of sodium will not neces-
random urine sodium excretion <20 mEq/L sarily increase serum sodium without attention to
can be seen in conditions where there is an concomitant water restriction.
excess of both total body water and total body Normal or increased extracellular volume and
sodium. The three major disorders that cause high urine sodium concentration can also be seen
this type of hyponatremia are the nephrotic in the setting of renal failure as both glomerular
syndrome, hepatic failure related to cirrhosis, filtration and water clearance falls, while the frac-
and cardiac failure. In all these conditions, tional excretion of sodium rises. A more unusual
there is a state of sodium and water avidity cause of this form of hyponatremia is polydipsia,
related to high levels of ADH and aldosterone. usually psychogenic in nature. Such water intox-
Most commonly, this is in the setting of ication is rare in children but can occasionally be
preexisting total body sodium overload as seen with emotional or psychiatric illness in older
evidenced by edema. In all of these conditions, children or with infants inappropriately provided
despite the increased extracellular or circulating with large volumes of water or very hypotonic
volume, the effective circulating volume is fluid in a repetitive fashion by a caretaker. In
often depressed. As a result of this ineffective both these circumstances, restriction of the vol-
perfusion of the tissues, sodium and water ume of free water ingested on a daily basis may be
avidity is only heightened by stimulation of beneficial.
the renin–aldosterone–angiotensin axis, further
exacerbating the total body excess of salt and
water. Appropriate therapy includes striking a Hypernatremia
balance between interventions promoting the
maintenance of effective circulating volume Hypernatremia is defined as a serum sodium con-
and restricting the provision of excess water centration greater than 150 mEq/L. Generally,
and sodium which will only contribute to further even higher serum sodium values can be tolerated
total body water and sodium overload. with the most significant clinical effects not
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 375

occurring until levels exceed 160 mEq/L. As with have hypertension or symptoms of pulmonary
hyponatremia, hypernatremia is more commonly edema. These children can respond to therapy
a reflection of a problem with water homeostasis aimed at augmenting sodium elimination. The
than sodium balance [75]. In most instances, the use of diuretics and the provision of adequate
patient has a relative deficiency of water for nor- free water decrease the total body sodium burden.
mal extracellular solute content. Rarely, dialysis may be necessary when the
Since sodium is the major determinant of hypernatremia must be corrected rapidly [77, 78].
plasma osmolality, as serum sodium levels rise, Hypernatremia as a result of salt loading is rare
serum osmolality concomitantly increases. and the pediatric clinician is much more likely to see
Increases in serum osmolality are sensed by hypo- hypernatremia stemming from a free water deficit or
thalamic osmoreceptors, triggering ADH release a combined water and sodium deficit where the
from the posterior pituitary as serum osmolality water losses exceed the sodium losses.
increases over 280 mOsm [76]. In the setting of Hypernatremia secondary to a water deficit arises
high ADH, there can then be increased in the setting of inadequate access to water or some
reabsorption of free water from filtrate in the cor- impairment in ADH release or response. It is
tical collecting duct. Increased serum osmolality uncommon to see hypernatremia secondary to
also causes a sensation of thirst and triggers poor water intake except in infants or young chil-
increased fluid intake. All of these mechanisms dren who cannot get water for themselves in
serve to re-equilibrate serum osmolality and response to their sense of thirst [79]. As for
sodium levels before clinically significant ADH-related anomalies, there are many causes of
hyperosmolality or hypernatremia occurs. central or nephrogenic diabetes insipidus [80].
Outside infancy, hypernatremia as a result of Again, given normal access to water, it is rare for
sodium excess or salt poisoning is infrequent. Its the older child to develop hypernatremia even with
major cause is improper preparation of powdered impairment in the ADH axis because of the strong
or liquid concentrated formula resulting in a drive to drink in response to thirst [81]. In very
hypertonic, hypernatremic solution. Since infants young children with diabetes insipidus, however,
do not have free access to water, they cannot the issue of access to water arises and hypernatremia
respond to their increasing sense of thirst as they may be a concern. Such hypernatremia can also be
develop such hypernatremia. As caregivers con- seen in the postoperative state in children with con-
tinue to provide the same incorrectly prepared centrating defects who are not allowed to drink by
formula for further feedings, there is further salt mouth and who are receiving a prescribed volume
loading. In addition, young infants are unable to of fluid based on a presumed ability to concentrate
excrete sodium loads as efficiently as older chil- urine and conserve water.
dren and this limits the intrinsic renal response. The most common etiology of hypernatremia
Iatrogenic sodium loading can also be seen in in children is the loss of hypotonic fluid, which is
children who receive large doses of sodium bicar- fluid with a relative excess of water for its sodium
bonate during resuscitation, because of persistent content. In these situations, the total body water is
acidosis, or in children who have been given decreased more than the total body sodium. The
inappropriate amounts of sodium in peripheral usual clinical scenario leading to such a condition
nutrition. Iatrogenic sodium loading can also be is viral diarrheal illness with poor water intake or
seen in the child receiving repeated large volumes persistent vomiting. In this condition, there is loss
of blood products since these are generally iso- of stool with sodium content typically <60
tonic or sodium-rich solutions. mEq/L. These children tend to excrete small vol-
Children who have hypernatremia from umes of concentrated urine with urine sodium
sodium excess should exhibit the physical signs content <20 mEq/L, underscoring the fact that
and symptoms of an expanded extracellular space. they are conserving both water and sodium.
They frequently have peripheral edema and may Their hypernatremia is not a manifestation of a
376 I.F. Ashoor and M.J.G. Somers

total body excess of sodium but a depletion of with marked clinical improvement. In the setting
sodium that is overshadowed by a larger relative of more significant compromise of effective vol-
depletion of body water. Therapy is aimed at ume such as with loss of vascular tone in sepsis or
restoring water and sodium balance by providing a systemic inflammatory response, more than
back the hypotonic fluid which was initially lost 200 ml/kg may be needed to achieve hemody-
either by the use of intravenous saline solutions or namic stability and effective perfusion. In most
with oral electrolyte therapy. situations other than outright shock, the clinician
can approach fluid resuscitation with either intra-
venous or oral rehydration therapy.
Fluid Replacement Therapy

Most commonly, the primary goal of fluid replace- Assessment of Volume Depletion
ment therapy is restoration of an adequate effec-
tive circulating volume. In its absence, significant In estimating the severity of dehydration, a change
metabolic derangements can occur that then exac- in weight from baseline is the best objective mea-
erbate perturbations in fluid and electrolyte sure [82]. As rehydration proceeds, following
homeostasis. The volume of fluid replacement weights on a serial basis becomes an important
required varies with the extent and etiology of adjunct in assessing the efficacy of fluid repletion.
the compromised circulation. In many children If no baseline weight is known, most clinicians
with acute illness, there may be an element of use various parameters based on history and phys-
decreased effective volume that is mild and diffi- ical examination to judge the severity of dehydra-
cult to appreciate by clinical examination. Expan- tion (Table 6). Children with mild dehydration
sion of extracellular volume with infusion or will have few clinical signs and only a modest
ingestion of 20–40 ml/kg of fluid over a few decline in urine output. As dehydration becomes
hours often results in better perfusion and more significant, classic findings such as dry
improved clinical appearance from presentation. mucous membranes, tenting skin, sunken eyes,
Urine output tends to remain normal in these and lethargy become prominent. With profound
situations speaking against persistent dehydration, there is anuria, marked alterations in
non-osmotic ADH activity that would encourage consciousness, and hemodynamic instability.
volume overload [29]. Without access to prior weight values, most
Children with mild dehydration, up to about clinicians find it difficult to estimate accurately the
5 % weight loss, will usually respond to the pro- degree of dehydration when it is mild or moderate.
vision of 30–50 ml/kg of fluid over several hours A capillary refill time greater than 2 seconds has

Table 6 Clinical assessment of dehydration


Degree of dehydration
Mild Moderate Severe
Vital signs
Pulse Normal Rapid Rapid and weak
Blood pressure Normal Normal to slightly low Shock
Weight loss
Infant <5 % 10 % >15 %
Older child <3 % 6% >9 %
Mucous membranes Tacky Dry Parched
Skin turgor Slightly decreased Decreased Tenting
Eye appearance Normal tearing Decreased tearing  sunken No tears + very sunken
Capillary refill Normal Delayed (>3 s) Very delayed (>5 s)
Urine output Decreased Minimal Anuric
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 377

long been touted as a useful physical finding 168 dehydrated children, elevated serum urea
suggesting effective volume depletion [83]. Unfor- levels and depressed serum bicarbonate
tunately, delayed capillary refill is neither a sensi- levels were found to be useful adjuncts to clinical
tive nor specific marker of dehydration [84, 85]. It evaluation in accurately assessing the degree of
may be most useful if normal, as this does seem to dehydration but were not by themselves
exclude reliably severe dehydration. In a prospec- predictive [90].
tive cohort study of dehydrated children between In fact, with viral gastroenteritis, the most com-
3 and 18 months of age, the best correlation mon cause of dehydration in children, there rarely
between clinical assessment of degree of dehydra- is a significant laboratory anomaly despite clini-
tion and actual volume depletion came in children cally detected volume depletion, as underscored
who had obvious clinical parameters of significant by the report of a cohort of children from the
dehydration such as prolonged skinfold tenting, a United Kingdom admitted for rehydration due to
dry mouth, sunken eyes, and altered sensorium viral gastroenteritis in which only 1 % had an
[86]. Similarly, in a review of preschool children electrolyte derangement [91–93].
with dehydration, the best clinical indicators of In children with volume depletion accompany-
volume depletion – decreased skin turgor, poor ing trauma, sepsis, surgery, or underlying renal
peripheral perfusion, and Kussmaul breathing – dysfunction, it would be more likely to find per-
accompanied more significant dehydration where turbations in electrolyte and acid–base status.
there was little clinical question of volume Thus, in the absence of a straightforward case of
depletion [87]. mild to moderate diarrheal dehydration, it is gen-
A study of 97 American children given intra- eral consensus that blood should be obtained for
venous fluids for rehydration in an emergency assessment of electrolytes, bicarbonate, and renal
department underscored the difficulty in assessing function to help guide specific fluid and electro-
even severe dehydration by standard clinical lyte therapy [94, 95].
approaches [88]. Physicians’ initial estimate of As the child is volume resuscitated, it is impor-
dehydration compared to the actual percent loss tant to reassess the child’s clinical status. Initial
of body weight varied dramatically, with a sensi- estimates of degree of dehydration may need to be
tivity of 70 % for severe dehydration (>10 % loss) adjusted if the child is not showing progressive
but only 33 % for moderate dehydration (6–10 % improvement. Most clinicians follow parameters
loss). This study suggested that adding a serum such as general appearance and sensorium,
bicarbonate level to the assessment may be useful, change in weight from initiation of rehydration,
increasing the sensitivity of the clinical scales to and urine output and urine osmolality. In children
100 % in severe dehydration and 90 % in moder- with some types of renal dysfunction, there may
ate dehydration if both clinical features and a often be an underlying chronic urinary concen-
serum bicarbonate <17 mEq/L were found. trating defect. In these children, relatively dilute
Other studies have found that laboratory stud- urine flow may be maintained even in the face of
ies by themselves are poor indicators of dehydra- clinical dehydration and markers other than urine
tion. In 40 children receiving intravenous fluids output and osmolality should be followed.
for dehydration, serum BUN, creatinine, uric acid,
anion gap, venous pH, venous base deficit, uri- Oral Rehydration Therapy
nary specific gravity, urinary anion gap, and frac- Although oral rehydration with electrolyte solu-
tional excretion of sodium were all assessed from tions is a safe, convenient, and effective way
prehydration blood and urine samples. Only the to treat volume depletion, parenteral fluid and
serum BUN/Cr ratio and serum uric acid signifi- electrolyte therapy has been the mainstay of
cantly correlated with increasing levels of dehy- treatment for most children presenting with fluid
dration, but both lacked sensitivity or specificity and electrolyte imbalances [96–99]. Especially
for detecting more than 5 % dehydration underutilized in North America, oral therapy has
[89]. Similarly, in a retrospective review of proved successful in clinical settings worldwide
378 I.F. Ashoor and M.J.G. Somers

in resuscitating children of all ages with profound If this regimen is tolerated with no vomiting, the
fluid and electrolyte anomalies. Short of signifi- aliquots may be gradually increased in volume
cant circulatory compromise, oral rehydration can and the frequency reduced, aiming to deliver at
be used as first-line therapy in all fluid and elec- least the prescribed total volume over about
trolyte aberrations [100, 101]. An example of a 4 h. After her rehydration, the child should sub-
situation calling for oral rehydration is the follow- sequently continue to be provided free access to
ing clinical scenario: fluid and resume an age-appropriate diet. If, on
A healthy 4-year-old girl presents to her pedi- the other hand, there are any further episodes of
atrician’s office following 3 days of a febrile ill- vomiting, then for each episode of emesis, an
ness. Her appetite has been severely depressed additional 120–240 ml of oral rehydrating solu-
and her parents estimate that she has only had a tion should be given, again with the goal to com-
few cups of fluid in the last 12 h. She has vomited plete rehydration and resume usual fluid intake
once daily. She has continued to urinate, although and nutrition.
less frequently and with smaller volumes. On The initial provision of oral fluid given often in
physical examination, the girl looks unhappy but small volumes is far more likely to be well toler-
nontoxic and alert. Her pulse is 100 beats per ated by the dehydrated child than larger aliquots.
minute and her sitting blood pressure is 80/50 If families are unwilling to provide the fluid in this
mmHg. She will not cooperate with attempts at manner, a nasogastric tube may be placed for
orthostatic vital signs. Her mucous membranes continuous infusion of rehydrating solution.
are somewhat dry and her weight today is 15 kg, Although occasional children may fail this
exactly the same as her weight at a well child approach and require intravenous rehydration,
examination 6 months previously. The parents most children with mild to moderate rehydration
are concerned that she looks dehydrated. can be rehydrated orally. A guide for the volumes
Primary care and emergency department phy- of fluid to provide and the duration of rehydration
sicians face such clinical scenarios daily. Other- can be found in Table 7.
wise healthy children with viral illness causing The first oral rehydration solutions were devel-
mild to moderate dehydration will frequently be oped in the 1940s at academic medical centers.
treated by intravenous fluids with the contention Within 10 years, a commercial preparation formu-
that oral rehydration will be too labor intensive or lated as a powder to be mixed with water was
will take too much time. In fact, these children are available, but its use became associated with an
excellent candidates for oral rehydration, and, in increased incidence of hypernatremia [106]. Sev-
most developed countries where there is little eral factors contributed to the development of this
concern for cholera-like enteritis, oral solutions problem: the preparation was sometimes incor-
with sodium contents from 30 to 90 mEq/L have rectly administered as the powder itself or improp-
been shown for years to be efficacious for rehy- erly diluted with too little water; when correctly
dration [102–105]. reconstituted, the solution had a final carbohy-
With this girl, given the history and physical drate concentration of 8 % predisposing to an
examination, it is unlikely that any clinically sig- osmotic diarrhea; and it was a common practice
nificant electrolyte perturbations will be found, so at that time for parents to use high solute fluids
there is little indication for assaying electrolytes such as boiled skim milk as an adjunctive home
or renal function prior to starting oral rehydra- remedy. Taken together, these early experiences
tion [91–93]. The family is given a commercially contributed to reluctance by many clinicians to
available oral rehydration solution containing use oral rehydration solutions, especially since
75 mEq/L Na, 20 mEq/L K, 30 mEq/L citrate, intravenous rehydration was becoming more stan-
and 2.5 % glucose. They are asked to provide 1 l dard in practice.
of fluid (50 ml/kg) to the child over the next Over time, there came to be a better under-
4 h. The child should be offered small aliquots of standing of the physiology of water and solute
fluid very often – 5 mls every 1–2 min at initiation. absorption from the gut. Of prime importance
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 379

Table 7 Oral rehydration for previously healthy, well-nourished children


Type of Rehydration Replacement of ongoing
dehydration phase Rehydration duration losses Nutrition
Mild (<5 %) 30–50 ml/kg 3–4 h 60–120 ml ORS for each Continue breast
ORT diarrheal stool or episode of milk or resume
emesisa age-appropriate
usual diet
Moderate 50–100 ml/kg 3–4 h As above As above
(5–10 %) ORT
Severe (>10 %) 100–150 3–4 h As above with use of As above
ml/kg ORT nasogastric tube if needed
Evidence of 20 ml/kg 0.9 Repetitive infusions until As above with use of As above
shock % NaCl or perfusion restored then nasogastric tube if needed
lactated transition to ORT 100 ml/kg or consideration of further
Ringer’s IV over 4 h IV therapy
Accompanying Per type of At least 12 h for ORT As above As above
hypernatremia dehydration Monitor fall in serum Na
ORT oral rehydration therapy with fluid containing 45–90 mmol/l Na, 90 mmol/l glucose, 20 mmol/l K, and 10–30 mmol/l
citrate
IV intravenous infusion
a
For children >10 kg, aliquots for replacement of ongoing losses should be doubled to 120–240 ml rehydration solution
for each stool or emesis

was the recognition that many substances actively carbohydrate to sodium ratio than WHO solution
transported across intestinal epithelium had (Table 8). Some preparations are available as
an absolute or partial dependence on sodium powder and other as ready-to-drink formulations.
for absorption and that sodium itself was Some manufacturers have also used rice solids as
actually better reabsorbed in their presence a carbohydrate source instead of glucose.
[107–109]. Moreover, it became clear that the Many of these formulation changes arose from
sodium/glucose cotransporter remained intact concerns that using an oral rehydrating solution
not only in the face of enterotoxic gastroenteritis with a sodium content >60 mmol/L would prove
such as seen with cholera or Escherichia coli but problematic in developed countries where most
also in more common viral and bacterial enteriti- gastroenteritis is viral in nature and has a lower
des [100, 101]. This led to the routine introduction sodium content than the secretory diarrheas seen
of glucose into oral rehydration solutions in a in less developed areas. Some feared that mini-
fixed molar ratio of no more than 2:1 with sodium. mally dehydrated children losing small amounts
The World Health Organization (WHO) and of sodium in their stools would become
the United Nations Children’s Fund champion hypernatremic if exclusively provided WHO
the use of a rehydrating solution that includes Na solution and a few studies did document such
90 mmol/L, Cl 80 mmol/L, K 20 mmol/L, base iatrogenic hypernatremia [110]. Subsequently,
30 mmol/L, and glucose 111 mmol/L (2 %). solutions with sodium content ranging from
This WHO solution has proved useful in many 30 to 90 mmol/L have proved quite effective in
pediatric clinical trials and has also been shown to cases of mild dehydration stemming from causes
reduce the morbidity and mortality associated other than secretory diarrhea [104, 105, 111].
with diarrheal illness regardless of its etiology A meta-analysis of studies focused on the safety
[102, 103], although it may not be readily and efficacy of oral rehydration solution in well-
commercially available in many locales. nourished children living in developed countries
Most commercially available oral rehydration documented little evidence that WHO solution
solutions have somewhat higher carbohydrate was more likely to cause aberrations in serum
content, a lower sodium content, and a higher sodium than lower sodium containing oral
380 I.F. Ashoor and M.J.G. Somers

Table 8 Oral rehydration solutions


Concentration, mmol/L
Product Na Sugar K Cl Base Osmolarity (mOsm/L)
WHO ORSa 90 111 20 80 30 311
CeraLyte 90a 90 220b 20 80 30 275
Low-Na ORSa 75 75 20 65 30 245
Rehydralyte 75 140 20 65 30 300
CeraLyte 70a 70 220b 20 60 30 230
CeraLyte 50a 50 220b 20 40 30 200
CeraLyte 50 lemon 50 170b 20 40 30 200
Enfalyte 50 170 25 45 34 167
Pedialyte 45 140 20 35 30 254
a
Provided as powder. Needs to be reconstituted with water
b
Contains rice syrup solids substituted for glucose

Table 9 Composition of common oral fluidsa


Fluid Na (mEq/L) K (mEq/L) Source of base Carbohydrate (g/100 ml)
Apple juice <1 25 Citrate 12
Orange juice <1 55 Citrate 12
Milk 20 40 Lactate 5
Cola 2 <1 Bicarbonate 10
Ginger ale 4 <1 Bicarbonate 8
Kool-Aid <1 <1 Citrate 10
Gatorade 20 2.5 Citrate 6
Powerade 10 2.5 Citrate 8
Jell-O 25 <1 Citrate 14
Coffee <1 15 Citrate <0.5
Tea 2 5 Citrate 10
a
Adapted in part from data found in Feld et al. [113]

rehydration solutions [112]. Why ingestion of solutions. In children with dehydration and elec-
lower tonicity oral rehydration fluids would be trolyte losses from vomiting or diarrhea, most
less problematic than infusion of similar tonicity common beverages do not contain adequate
intravenous fluid is not clear, but does again sodium or potassium supplementation, and the
underscore the safety of oral rehydration. base composition and carbohydrate source are
also often suboptimal (Table 9). Similarly, most
sports drinks for “rehydration” following exer-
Oral Rehydration with Fluids Other cise are also depleted of sufficient electrolytes
Than ORS for gastrointestinal disease given sweat is many
fold lower in electrolyte composition than gas-
Despite the efficacy and availability of commer- trointestinal fluid. In prescribing oral rehydration
cial oral rehydration solutions and the ease to children in an ambulatory setting, the clinician
with which other electrolyte solutions can be should specify the appropriate fluid and volume
mixed at home with recipes requiring few ingre- for the child to ingest, emphasizing the need to
dients other than water, sugar, and salt, many use a fluid with appropriate electrolyte content if
children are still given common household bev- there is concern about evolving imbalances in
erages for rehydration instead of oral rehydration sodium, potassium, or bicarbonate homeostasis.
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 381

Oral Rehydration and Serum Sodium fluids by mouth have nasogastric tubes placed.
Abnormalities With this approach, successful oral rehydration
is the rule and 95 % of children are fully
Although oral rehydration is often considered for rehydrated without the need for intravenous
children with modest dehydration and no presumed therapy.
electrolyte anomalies, oral rehydration with WHO An alternative approach has been to have the
or WHO-like solution has also been used in cases of child begin by taking 15 ml/kg/h of a 60–90
dehydration accompanied by hyponatremia or mmol/L Na rehydration solution by mouth or
hypernatremia [114, 115]. Although most children nasogastric tube [117]. The solution is given in
with severe hypernatremia (>160 mEq/L) can be small frequent quantities and increased up to
successfully rehydrated orally, there have been 25 ml/kg/h until hydration has improved at
reports of seizures, generally as a result of too which point solid feedings are reintroduced and
rapid correction of serum sodium from the provi- volumes of 5–15 ml/kg of rehydration solution
sion of supplemental water along with the offered after feeds until the volume deficit have
glucose–electrolyte solution [114, 115]. In those been delivered.
cases, the average serum sodium fell by 10–15 Over two decades ago, the American Academy
mEq/L over 6 h rather than over 24 h as advised. of Pediatrics issued guidelines for the treatment of
In follow-up studies, no seizure activity was seen in fluid and electrolyte deficits with oral rehydration
a similar cohort of hypernatremic children who solutions [100]. Children with acute dehydration
received 90 mmol/L Na rehydration solution alone and extracellular volume contraction were to be
at a rate calculated to replace the infant’s deficit over provided 40–50 ml/kg of a glucose–electrolyte
24 h [114]. It is important for the practitioner to solution containing in each liter 75–90 mmol Na,
remember that once peripheral perfusion has been 110–140 mmol glucose (2–2.5 %), 20 mmol
stabilized with initial volume expansion, there is no potassium, and 20–30 mmol base. This volume
benefit to correcting any deficit rapidly and taking was to be administered over 3–4 h and then once
24–48 h may be a more prudent course in the face of there has been amelioration of the extracellular
significant electrolyte anomalies. volume contraction; the child would be changed
to a maintenance solution with 40–60 mmol/L Na
at half the rate. If the child was still thirsty on
Oral Rehydration Schemes this regimen, there should be free access to
supplemental water or low-solute fluid such as
Several oral rehydration schemes have been breast milk.
shown to be quite effective and well tolerated. In Based on much of this published clinical expe-
one approach used extensively in developing rience, an evidence-based guideline for treating
countries, the patient’s volume deficit is calcu- dehydration in children from industrialized
lated on the basis of weight loss and clinical European countries was created in the late
appearance [116]. The volume deficit is doubled 1990s, recommending oral rehydrating solution
and this becomes the target rehydration volume to containing 60 mmol/L of sodium, 90 mmol/L of
be given over 6–12 h. Two-thirds of this volume is glucose, 20 mmol/L of potassium, and 10–30
given as a glucose–electrolyte solution containing mmol/L of citrate with rehydration occurring
90 mmol/L Na over 4–8 h; once this has been over 3–12 h utilizing from 30 to 150 ml/kg of
ingested, the remaining volume is provided as fluid depending on the degree and type of
water alone over 2–4 h. In cases of suspected or dehydration [82].
confirmed hypernatremia with serum sodium In 2004, the American Academy of Pediatrics
exceeding 160 mEq/L, the volume deficit would updated its oral rehydration recommendations and
not be doubled and would be administered as endorsed guidelines promulgated by the Center
90 mmol/L Na glucose–electrolyte solution for Disease Control and Prevention [118,
alone over 12–24 h. Patients who refuse to take 119]. Minimal dehydration in children weighing
382 I.F. Ashoor and M.J.G. Somers

less than 10 kg was to be treated with provision of despite lengthy shelf storage, its ability to be
60–120 ml of oral rehydration fluid for each administered readily by the child’s caretaker in
watery stool or each episode of vomiting. In larger nearly any locale, and avoidance of the discomfort
children, twice this volume would be provided. and potential complications associated with intra-
For children with more moderate dehydration, venous catheter placement [124]. In developed
50–100 ml/kg of oral rehydration solution would countries, there has been the concern that some
be given over 2 to 4 h to account for estimated powdered ORS formulations may not be looked
fluid deficit, and ongoing losses would be treated upon by parents as convenient since they must be
with the 60–240 ml per stool or emesis depending mixed with water prior to provision, but a ran-
on size. Nursing babies would continue to receive domized controlled trial comparing an urban pedi-
breast milk as desired and formula-fed babies atric clinic and a suburban medical practice found
would be provided age-appropriate diet as soon that parents were equally satisfied with the ease and
as they had been rehydrated. For severely effectiveness of a powdered solution as a commer-
dehydrated children, a combination of intrave- cially prepared ready-to-drink solution [125].
nous hydration with isotonic fluids and prompt Oral rehydration is somewhat less successful in
transition to oral rehydration solution by mouth hospitalized children than in children treated in an
or nasogastric tube was recommended. Overly ambulatory setting [10]. This difference may be
restricted diets were to be avoided during gastro- directly related to the degree of dehydration or
intestinal illness and attention to adequate caloric other complicating clinical issues leading to hos-
intake emphasized (see Table 7). pital admission. Moreover, the relatively labor-
intensive slower approach to oral rehydration
may be problematic in medical facilities with
Use and Acceptance of Oral time constraints or space limitations [124, 126].
Rehydration Solutions Frozen-flavored oral rehydration solutions
may be more readily accepted than conventional
Despite the availability of guidelines for oral rehy- unflavored liquid electrolyte solutions. Their use
dration and their endorsement by professional resulted in higher rates of successful rehydration
organizations, oral rehydration solutions continue in children with mild to moderate dehydration,
to be underutilized by clinicians in both the devel- even if these children initially failed conventional
oped and undeveloped world [104, 120]. Even in oral rehydration [124]. Frozen-flavored rehydra-
Bangladesh where oral rehydration has been tion solution is now commercially available in
closely studied and championed by both local many parts of the world, as is a variety of flavored
and international medical agencies for decades, rehydration solutions.
its use is still suboptimal [121, 122]. Some studies Another potential issue with oral rehydration is
do suggest that familiarity with a specific scheme that its use does not alter the natural course of the
for oral rehydration increases its use in the emer- child’s illness. For instance, in gastroenteritis with
gency department and that younger graduates of dehydration, by far and away the most common
training programs are more likely to use oral rehy- illness requiring rehydration in children, oral rehy-
dration for advanced cases of clinical dehydration dration does not lower stool output or change the
than their older colleagues, even when there was duration of diarrheal illness [127]. As a result,
no generational difference in the baseline knowl- caretakers may abandon oral rehydration because
edge of published data in this field or acceptance the child continues to have symptoms, failing to
of its validity [123]. appreciate the benefits of ongoing hydration. Oral
When utilized according to recommendation, rehydrating solutions have been formulated with
oral rehydration has been demonstrated to be lower electrolyte composition and different carbo-
almost universally successful in achieving some hydrate moieties with the goal to reduce the osmo-
degree of volume repletion [10]. Other advantages larity of solutions and potentially augment
to oral therapy include the stability of the product fluid absorption from the small intestine [112].
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 383

The rice-based oral solutions have been studied Choice and Volume of Parenteral Fluid
most extensively. In these solutions, glucose is
substituted with 50–80 g/l of rice powder. In a Children with significant extracellular volume con-
meta-analysis of 22 randomized clinical trials com- traction (greater than 10 % acute weight loss in an
paring rice-based solution to conventional glucose- infant or 6 % weight loss in an older child) should
containing solutions, stool output dramatically receive an isotonic crystalloid solution such as 0.9
decreased in children with cholera given rice- % saline (154 mEq/L NaCl) or lactated Ringer’s
based hydration but did not change in children (130 mEq/L NaCl) at a rate of 20 ml/kg over 30–60
with other bacterial or viral enteritides [128]. min. In some children, even more rapid infusions or
There are some reports that suggest that pro- serial provision of such aliquots may be necessary
viding children with non-cholera enteritis with to restore effective volume. Children with less pro-
reduced osmolarity rehydration solution may be nounced dehydration may not exhibit signs or
beneficial. A study of 447 boys less than 2 years of symptoms of volume contraction. In certain situa-
age admitted for oral rehydration compared WHO tions, however, it may be clinically warranted to
solution (osmolarity 311 mmol/l) and a solution provide them with an initial rapid intravenous bolus
containing less sodium and chloride (osmolarity to initiate rehydration therapy.
224 mmol/l). Children who received the lower Concomitant with the placement of intravenous
osmolarity solution had reduced stool output, access, blood should be obtained for determination
reduced duration of diarrhea, reduced rehydration of serum electrolytes, osmolality, and renal func-
needs, and reduced risk of requiring intravenous tion. Given that dehydrated children often have
fluid infusion after completion of oral hydration high levels of vasoactive hormones and high vaso-
[129]. A meta-analysis of nine trials comparing pressin levels, it is most prudent to establish base-
WHO solution to reduced osmolarity rehydration line electrolyte levels since it is possible to alter
solution concluded that children admitted for electrolyte balance rapidly with intravenous ther-
dehydration had reduced needs for intravenous apy. In the face of inadequate tissue perfusion, a
fluid infusion, lower stool volumes, and less parenteral fluid infusion should begin immediately
vomiting when receiving the reduced osmolarity prior to the return of any pertinent laboratory
solution [130]. results. If hemorrhagic shock is suspected, resusci-
tation with packed red blood cells is optimal. In
Intravenous Therapy cases of severe volume depletion, if the child does
Although absolute indications for parenteral intra- not improve with the initial 20-ml/kg crystalloid
venous therapy are limited, they do include sig- bolus, this should be repeated up to two additional
nificantly impaired circulation or overt shock. In times. In children who have not improved despite
addition, there are occasional children who are administration of 60 ml/kg of total volume over an
truly unable to sustain an adequate rate of oral hour or in children in whom underlying cardiac,
fluid intake despite concerted effort or have such pulmonary, or renal disease may make empiric
persistent losses that parenteral therapy comes to aggressive rehydration more problematic, consid-
be necessary. The mainstays of fluid therapy in eration should be given to placement of a central
children are saline or buffered saline crystalloid monitoring catheter to more accurately assess intra-
solutions. Isotonic versions of these crystalloids vascular volume and cardiac dynamics [131]. In
are used for volume resuscitation and hypotonic some instances of profound ineffective circulating
saline solutions may be used in addition to pro- volume, such as might accompany certain cases of
vide supplemental maintenance hydration. In sepsis, initial volume resuscitation may require
addition to crystalloid solutions, there are several sequential infusions of fluid ultimately exceeding
colloid fluids that are also used by many clini- 100 ml/kg.
cians. Table 10 lists the electrolyte content of Within minutes of infusion of a crystalloid
some of the more common intravenous solutions fluid, it becomes distributed throughout the extra-
used for pediatric fluid therapy. cellular space. Since this involves equilibration of
384 I.F. Ashoor and M.J.G. Somers

Table 10 Composition of common intravenous fluids


Buffer
Osmolarity Na K Cl (source) Mg Ca Dextrose
Fluid (mOsm/l) (mEq/l) (mEq/l) (mEq/l) (mEq/l) (mEq/l) (mEq/l) (g/l)
Crystalloids
0.9 % saline 308 154 0 154 0 0 0 0
Lactated 275 130 4 109 28 (lactate) 0 3 0
Ringer’s
D5 0.45 % 454 77 0 77 0 0 0 50
saline
D5 0.22 % 377 38 0 38 0 0 0 50
saline
5 % dextrose 252 0 0 0 0 0 0 50
water
Normosol 295 140 5 98 27 (acetate) 3 0 0
23
(gluconate)
Plasma-Lyte 294 140 5 98 27 (acetate) 3 0 0
23
(gluconate)
Colloids
5 % albumin 309 130–160 <1 130–160 0 0 0 0
25 % albumin 312 130–160 <1 130–160 0 0 0 0
Fresh frozen 300 140 4 110 25 0 0 0
plasma (bicarbonate)
3.5 % 301 145 5 145 0 0 6 0
Haemaccel
6% 310 154 0 154 0 0 0 0
hetastarch
Dextran 40 or 310 154 0 154 0 0 0 0
70

the fluid between the two components of the prior to the availability of blood and for
extracellular space – the intravascular and inter- nonhemorrhagic shock as an adjunct to crystalloid
stitial spaces – actually only one-third to use [135]. Types of colloid utilized included 5 %
one-quarter of infused crystalloid stays in the albumin, fresh frozen plasma, modified starches,
blood vessels [132]. This accounts for the need dextrans, and gelatins. These guidelines, gener-
to give large volumes of crystalloid in the setting ally aimed towards the fluid resuscitation of
of circulatory collapse and leads some to suggest adults, were composed despite the prior publica-
that colloid solutions such as 5 % or 10 % albumin tion of a systematic review of randomized con-
should play a role in resuscitation [133, 134]. trolled trials that demonstrated no effect on
mortality rates when colloids were used in prefer-
ence to crystalloids [136]. Moreover, there is a
Colloid Solutions and Volume distinct cost disadvantage to using colloid
Resuscitation solutions.
Subsequent systematic reviews have looked at
The use of colloid solutions for volume resuscita- this issue anew. In one meta-analysis of 38 trials
tion is controversial. Colloids were once included comparing colloid to crystalloid for volume
in a number of widely promulgated guidelines for expansion, there was no decrease in the risk of
the care of patients in emergency facilities and death for patients receiving colloid [135]. In the
intensive care units both for hemorrhagic shock other review, albumin administration was actually
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 385

shown to increase mortality by 6 % compared to driven acidosis [144]. This hyperchloremic


crystalloid [137]. Proposed mechanisms contrib- acidosis is seen less frequently when Ringer’s
uting to this worse outcome include anticoagulant lactate solution is used as the resuscitation
properties of albumin [138] and accelerated fluid because of the metabolic conversion of
capillary leak [139]. lactate to bicarbonate. In the setting of significant
A drawback of all these systematic reviews, preexisting acidosis or underlying hepatic
however, has been the limited number of studies dysfunction preventing the metabolism of lactate,
that included children other than ill premature infusion of Ringer’s lactate solution may,
neonates. As a result, generalization of these however, exacerbate an acidosis.
results from ill adults may not be germane to all With the recent suggestion that some children
critically ill volume-depleted children. For with acute illness or following surgery may ben-
instance, a report of 410 children with meningo- efit from a prolonged period of isotonic fluid
coccal disease suggests that albumin infusion in infusion given high levels of ADH and the possi-
this population may not have been harmful, as bility for hyponatremia developing with hypo-
case fatality rates were lower than predicted tonic fluid therapy, some have expressed concern
[140]. Overall, however, there seems to be no that a hyperchloremic acidosis may develop in
substantive data to support the routine use of these children. In a prospective randomized
colloid to complement or replace crystalloid in study of more than 100 children with gastroenter-
fluid resuscitation. Rather, repetitive infusions of itis given isotonic intravenous rehydration and
large volumes of crystalloid seem to be well tol- maintenance therapy, there was no tendency for
erated in volume-depleted children, do not seem the development of hyperchloremic acidosis even
to predispose to excessive rates of acute respira- after a day of isotonic fluid provision. Although
tory distress syndrome or cerebral edema, and in serum chloride levels did tend to increase in these
some conditions, such as sepsis, play an important children, serum bicarbonates also improved,
role in improved survival [141]. A recent survey potentially related to improved effective volume
of pediatric anesthesiologists in Western Europe and subsequent better tissue perfusion [145]. Sim-
reported that colloid solutions are being used less ilarly, in children who underwent cardiac surgery
frequently in infants and older children and who were given isotonic solutions for their main-
suggested that familiarity with some of the issues tenance fluid needs, although there was a ten-
raised in these systematic reviews are affecting dency for a hyperchloremic acidosis to develop,
practice patterns [142]. there seemed to be no significant clinical ramifi-
Repetitive infusions of crystalloid may also cations and long-term outcomes were similar to
prove problematic in some children. Most nota- children who did not develop hyperchloremic
bly, if very large volumes of 0.9 % NaCl are used acidosis [146].
acutely for volume resuscitation, it is not unusual Large volume infusion of blood may also pre-
for children to develop a hyperchloremic meta- dispose to electrolyte anomalies as well as mani-
bolic acidosis. This occurs as acidotic peripheral festations of citrate toxicity. If aged whole blood is
tissues begin to reperfuse and already depleted infused, there is the possibility that a large potas-
extracellular bicarbonate stores are diluted by a sium load will be delivered to the patient as potas-
solution with an isotonic concentration of chloride sium may have moved from less viable
[131, 143]. This acidosis can be ameliorated by erythrocytes into the plasma. Since most patients
supplemental doses of bicarbonate as well as the receive packed red blood cells instead of whole
addition of supplemental potassium as needed. blood, this potential problem is minimized, and
There is sometimes a tendency for clinicians to the potassium in the small volume of plasma in a
react to the hyperchloremic metabolic acidosis packed cell transfusion can generally be accom-
with further saline bolus infusions. In the face of modated by intracellular uptake.
corrected hypoxia or hypovolemia, however, such Citrate is used as the anticoagulant in stored
maneuvers may only exacerbate the chloride- blood. Since citrate complexes with calcium, there
386 I.F. Ashoor and M.J.G. Somers

can be a fall in ionized calcium levels if large very sudden fluxes in electrolytes may become
volumes of citrate-containing blood are infused symptomatic earlier. On the other hand, children
rapidly or if there are concomitant perturbations whose severe sodium abnormalities are thought to
in calcium homeostasis. Similarly, citrate may be more chronic in nature must be treated in a
complex with magnesium and magnesium deple- more controlled fashion since they are at higher
tion may occur. The liver usually metabolizes risk for developing CNS symptoms during
infused citrate into bicarbonate and alkalosis can treatment.
also occur if large volumes of citrate are delivered. The vast majority of children treated in emer-
In the setting of hepatic dysfunction, however, gency facilities for volume repletion do well with
citrate will not be metabolized and serves as an such rapid rehydration. These children are gener-
acid load and will help create an acidosis or exac- ally healthy with normal cardiac and renal func-
erbate any underlying acidosis. tion and have developed extracellular volume
Regardless of the initial infusion with either depletion relatively rapidly. As a result, they suf-
colloid or crystalloid, once sufficient volume to fer no ill effects from rapid rehydration. In fact,
restore circulatory integrity has been infused, less the clinical success of this aggressive restoration
rapid volume expansion is necessary. During this of extracellular volume underlies the calls to
phase, the rapidity of fluid repletion is most prob- reexamine or abandon the traditional deficit ther-
ably not a concern unless there are severe under- apy approach to rehydration with its tedious cal-
lying aberrations in the serum sodium or serum culations of fluid and electrolytes losses and
osmolality. In the absence of these derangements requirements, especially in the setting of other-
or profound volume deficit, if the child has wise healthy children who are acutely ill [9, 148].
improved significantly with the initial parenteral
volume expansion, attempts should be made to
reinstitute oral rehydration. Prolonged intrave- Symptomatic Hyponatremia
nous therapy should be rarely necessary.
In the setting of symptomatic hyponatremia, espe-
cially if the child has seizures, it is important to
Rapid Rehydration raise the serum sodium concentration by approx-
imately 5 mEq/L in an urgent fashion. Generally,
In an attempt to minimize the duration of hospital- this results in stabilization of the clinical situation
based care, a scheme of rapid intravenous resus- and allows for further more considered evaluation
citation and follow-up oral rehydration has been and treatment of the child. This is one of the few
adopted by many pediatric emergency depart- situations in which hypertonic saline (3 % NaCl,
ments to treat children with up to 10 % dehydra- 514 mEq/L) should be utilized.
tion secondary to vomiting and gastroenteritis To calculate the proper volume of 3 % saline to
[147]. After infusion of 20–30 ml/kg of intrave- infuse, the child’s TBW must be multiplied by the
nous crystalloid, the child is allowed to take up to 5 mEq/L desired increase in serum sodium to
several ounces of a standard oral rehydration fluid, determine the amount of sodium (in mEq) to
and if this intake is tolerated without vomiting for infuse. Since every ml of 3 % saline contains
30–60 min, then the child discharged home to 0.5 mEq of sodium, doubling the number of
continue rehydration, initially with a prescribed mEq of sodium needed results in the proper mil-
volume of standard rehydration solution. liliter volume of 3 % saline to infuse. Thus, in the
If the child does not tolerate oral rehydration or 20-kg child, the TBW is approximately 12 L (0.6
if there are such significant electrolyte anomalies L/kg  20 kg) and the desired sodium dose
that there are concerns regarding potential adverse would be 60 mEq (12 L  5 mEq/L). If 120 ml
CNS sequelae of too rapid rehydration, then intra- of 3 % saline were infused, the serum sodium
venous rehydration may be the best route for concentration would be expected to rise by
continued hydration. Children who have had approximately 5 mEq/L. The infusion should be
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 387

given at a rate to increase the serum sodium by no and current serum sodium is multiplied by the
more than 3 mEq/L/h and is often given more child’s estimated TBW. This product represents
slowly over the course of 3–4 h [149]. If the the hyponatremic sodium losses that must be
child continues to be symptomatic from added to the maintenance sodium needs, any
hyponatremia after this infusion, additional 3 % ongoing losses, and the sodium losses that accom-
saline may be given until the symptoms improve panied weight loss. An example of the calcula-
or the serum sodium is in the 120–125 mEq/L tions and therapeutic maneuvers that need to be
range. At that point, further correction of the considered with significant hyponatremia is
hyponatremia should consist of a slower infusion presented in the following case study:
of more dilute saline to cover the sodium deficit, A girl who normally weighs 10 kg suddenly
the sodium maintenance needs, and any volume develops generalized seizures and is brought by
deficit. Consideration of the role of ADH and ambulance to the emergency department. She has
prior excess free water provision should also be had a week of gastroenteritis, has felt warm to
considered in determining the volume and tonicity touch, and has been drinking water and apple
of fluid to be provided. juice only, refusing any other liquids or any
solid food for several days. Intravenous access is
placed and lorazepam is administered and the
Asymptomatic Hyponatremia seizure activity stops. The emergency department
physician orders serum chemistries and the child
If a child has severe hyponatremia but is not is weighed and found to be 8.8 kg. A bolus infu-
symptomatic, there is no need to administer sion of 200 ml of 0.9 % NaCl is administered over
hypertonic saline based solely on a laboratory the next 30–60 min after which the girl appears
anomaly. With or without symptoms, in cases of well perfused but she is still lethargic. The serum
severe hyponatremia, the child should be carefully sodium is then reported to be 112 mEq/L. While
evaluated as to the etiology of the hyponatremia, further evaluation of the child’s overall status is
keeping in mind that hyponatremia tends to result ongoing, it is important to begin correcting the
from an imbalance of water regulation. If this is symptomatic hyponatremia.
the case, free water should be restricted and appro- The child has actually already received
priate supplementation with intravenous saline approximately 30 mEq of sodium in the 0.9 %
solutions begun to provide maintenance sodium NaCl bolus given because of her dehydration
requirements of approximately 2–3 mEq/kg/day and poor perfusion. Given her TBW of roughly
and any ongoing losses of sodium. 5.5 L (wt in kg  0.6 L/kg), this should result in
Besides these maintenance sodium needs, if an increase in her serum sodium by approxi-
the child has an element of dehydration, every mately 5 mEq/L. Since the child has had
kilogram of body weight lost from baseline rep- hyponatremic seizures and is still exhibiting
resents a 1-L deficit of nearly normal saline from some central nervous system effect with her leth-
the total body water as well. These losses are often argy, it is prudent to raise the serum sodium by
referred to as isotonic losses. These account for a 5 mEq/L so that it will be in the 120–125 mEq/L
sodium deficit of 154 mEq/L that also must be range. Since she is hemodynamically stable, it is
included in the calculations for sodium also best not to provide an excess of further vol-
replacement. ume until the child undergoes imaging to assess
In the setting of hyponatremic dehydration, for cerebral edema, especially given the history of
there have been additional sodium losses as well. seizures, lethargy, and hyponatremia. By using a
Generally, these occur as viral diarrheal stool small volume of hypertonic saline, the serum
losses with a sodium content 60 mEq/L are sodium can be raised in a controlled manner
replaced with fluids with lower sodium concen- while further evaluation of the child continues. It
tration. To estimate these sodium losses, the dif- would take about 28 mEq of sodium (TBW 
ference between the child’s desired serum sodium desired increase in serum sodium = 5.5 L  5
388 I.F. Ashoor and M.J.G. Somers

mEq/L) to accomplish the desired elevation. Since ensuing 16 h. Although such a plan can be
each ml of 3 % saline contains about 0.5 mEq of followed, there is little evidence that more rapid
sodium, a total of 56 ml of 3 % saline could be correction of the hyponatremia is harmful except
infused over approximately 3–4 h. if the patient has been symptomatic with
In addition to this acute management to restore hyponatremia or has profound asymptomatic
initial circulation and perfusion and to raise the hyponatremia of chronic duration. In these
serum sodium to a safer level, plans must be cases, it is safest to plan to correct the serum
formulated to attend to the patient’s overall vol- sodium by no more than 12–15 mEq/L over
ume and sodium deficit. To prescribe the proper 24 h. More rapid correction has resulted in
follow-up intravenous fluid, the patient’s water osmotic demyelination injury to the brain with
and electrolyte deficits at presentation must be devastating long-term neurologic outcomes
reconciled with her therapy thus far. [149–151].
The child’s water deficit is 1.2 l, reflecting the
1.2-kg weight loss. She has “maintenance” water
needs of an additional 1 l/day based on her nor- Severe Hypernatremia
mal weight of 10 kg. She is having no other
ongoing water losses and has already received With hypernatremia, therapy is again guided by
nearly 250 ml in intravenous fluid in the form of the clinical situation and provision of intravenous
0.9 % NaCl and 3 % NaCl. Her current water fluid is usually reserved for those children with
needs are thus 1,950 ml. very elevated serum sodium values who are not
The child’s normal “maintenance” sodium considered candidates for oral rehydration ther-
needs are 30 mEq/day (3 mEq/kg/day). She has apy. In cases of hypernatremia due to salt poison-
lost 1.2 kg of isotonic fluid in body weight that ing, there should be signs of overhydration and
represents 185 mEq of sodium. In addition, she volume expansion. Excretion of sodium should be
has hyponatremic sodium losses that have arisen enhanced by using a loop diuretic to augment
as her diarrheal stool that contained sodium was urine sodium losses and by replacing urine output
replaced with water alone. To calculate these with free water. If the patient has significant
needs, her normal total body water needs to be underlying renal or cardiac compromise, dialysis
multiplied by the difference in her serum sodium and ultrafiltration may be necessary to correct the
from a normal value of 135 mEq/L. Her TBW is water and electrolyte imbalance [77, 78]. With
6 L (TBW = 0.6 L/kg  10 kg) and the difference hypernatremia and volume expansion from salt
in serum sodium is 23 mEq/L (135 – 112 mEq/L); excess, it will be detrimental to provide further
her hyponatremic losses are therefore 138 mEq intravenous saline.
(6 L  23 mEq/L). Total sodium needs are thus In hypernatremia accompanied by volume
30 mEq of maintenance, 185 mEq of isotonic loss, any significant alterations in effective circu-
losses, and 138 mEq of hyponatremic losses or a lation should be addressed with 20 ml/kg bolus
total of 353 mEq. She has already received infusions of an isotonic crystalloid solution until
52 mEq of sodium from the 400 ml of 0.9 % effective peripheral perfusion is restored. Then,
NaCl given in the emergency department. Her further provision of water and sodium should be
current sodium needs are thus just about provided based on calculated water and sodium
300 mEq. needs. In the majority of cases, with mild eleva-
To choose the proper solution for this child, the tions in serum sodium and minimal degrees of
deficit of 1,950 ml of water should contain dehydration, the actual calculation of deficits is
300 mEq of sodium. This is best approximated probably unnecessary since the child will be
by 0.9 % NaCl with its NaCl content of hemodynamically stable and a candidate for
154 mEq/L NaCl. In the past, it has been exclusive oral rehydration. In situations where
suggested that half of the fluid and sodium deficit there is profound hypernatremia or circulatory
be replaced over 8 h and the remainder over the compromise, it remains necessary, however, to
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 389

be able to calculate a free water deficit to tailor about 1,750 ml. Of this volume, the baby has
intravenous rehydration therapy. An example of already received 300 ml of fluid in the emergency
such a situation is outlined in the following clin- department so a net deficit of 1,450 ml now exists.
ical scenario: The baby has maintenance sodium needs of
After 2 days of refusing to nurse, a 5-kg infant 15 mEq/day (3 mEq/kg/day). His sodium deficit
boy with a viral syndrome presents to an emer- reflects only the isotonic fluid losses that have
gency department in shock, 15 % dehydrated with been estimated above at 240 ml of normal saline
a weight of 4.25 kg and a serum sodium of or 37 mEq of sodium. Thus, over the next 2 days,
170 mEq/L. He receives 300 ml of 0.9 % NaCl his sodium needs are 67 mEq of which he has
urgently and further therapy is now planned. already received more than 45 mEq in the emer-
The child has lost 750 g of weight. Since this is gency department due to initial volume
hypernatremic dehydration, there has been loss of expansion.
water in excess to salt. Thus, part of the weight Initiating an infusion of 30 ml an hour of free
loss represents isotonic losses but a larger pro- water should result in the slow and steady correc-
portion represents free water loss. The child’s free tion of the hypernatremia over 2 days by provid-
water deficit can be calculated by the equation ing the nearly 1.5 l of free water that the child
requires to replace losses and provide ongoing
needs. The serum sodium should be monitored
½ðserum Na actualÞ=ðserum Na desiredÞ
every 4 h initially, and if it is falling faster than
 total body water  total body water
desired (about 0.5 mEq/h), then the sodium
should be added to the rehydration fluid.
Substituting the appropriate data for this baby,

½ð170=145Þ  ð0:6  4:25Þ  ð0:6  4:25Þ Fluid and Electrolyte Therapy


¼ ½1:2  2:55  2:55 ¼ 0:51 L with Renal Dysfunction

Thus, of this baby’s 750-ml fluid deficit due to Impact of Kidney Disease
dehydration, 510 ml is free water and 240 ml is
normal saline. Children with compromised renal function often
Too rapid correction of the baby’s serum manifest a reduced tolerance for changes in total
sodium with free water could result in cerebral body water as well as changes in the composition
edema as the water infused into the extracellular or distribution of volume between the intracellular
space follows osmotic forces and moves into the and extracellular body spaces. Similarly, alter-
intracellular space. In cases of hypernatremia ations in electrolyte balance are more likely prob-
where the serum sodium exceeds 160 mEq/L, it lematic because normal homeostatic mechanisms
is considered safest to correct the serum sodium are frequently perturbed. Especially in the child
by no more than 15 mEq/day. In this boy’s case, with marked nephrosis or significant impairment
this would mean that correction to a serum in renal clearance, it becomes vital to approach
sodium in the normal range would take about the provision of fluids and electrolytes with
2 days. great care.
If the fluid and electrolyte therapy must be As far as fluid therapy is concerned, of utmost
given intravenously, the appropriate prescription importance is the recognition that the concept of
again depends on calculation of water and maintenance fluids or electrolytes presupposes
sodium requirements and deficits. His original normal renal function. Roughly two-thirds of
fluid deficit was 750 ml and his maintenance any daily maintenance fluid prescription is to
water needs are estimated at 500 ml/day. Thus, replace urinary water losses. Similarly, urinary
over the next 2 days, the fluid needed to replace electrolyte losses figure prominently in daily elec-
the deficit and provide maintenance water is trolyte balance. In the setting of oliguria or anuria,
390 I.F. Ashoor and M.J.G. Somers

provision of maintenance fluids could contribute understanding of the pathophysiology underlying


to and, potentially, exacerbate volume overload, the child’s renal dysfunction will also be useful.
and maintenance electrolyte therapy could result The child who has profound tubular electrolyte
in electrolyte anomalies. losses will require more sodium on a daily basis
Fluid and electrolyte needs of the child with than the child who is edematous and total body
renal dysfunction are better considered in the con- salt overloaded from his nephrotic syndrome. The
text of the child’s current volume status and elec- child with chronic renal insufficiency and hyper-
trolyte needs. For instance, in symptomatic tension mediated by long-standing salt and water
volume depletion with decreased circulatory per- overload may actually benefit from diuretic ther-
fusion, volume expansion would be initiated apy to remove salt and water rather than any
regardless of urine output. Once volume is further volume expansion with saline.
repleted, the child’s needs could be reassessed Certainly the provision of supplemental potas-
along with his current renal function. The child sium to the child with renal dysfunction must be
who is volume overloaded would best be man- done judiciously. The oliguric or anuric child
aged by fluid restriction and provision of only should receive no potassium until it is well
insensible losses of approximately 300 ml/m2. documented that serum potassium levels are low
Insensible fluid losses should be considered or that there are extrarenal potassium losses (for
essentially electrolyte free water. The child who instance losses from diarrheal stool). The child
is volume replete should be kept volume replete. with marginal renal function should receive
This is most readily accomplished by providing a small amounts of potassium (approximately
combination of insensible losses as free water and 1 mEq/kg/day) with at least daily assessment of
any other volume losses (urine output, diarrheal electrolyte balance to determine adequacy and
stool, surgical drain output, emesis) on an addi- appropriateness of continued potassium
tional milliliter-for-milliliter basis. If there are supplementation.
significant ongoing losses from a single source,
the electrolyte composition of this fluid can be
assayed so that the replacement fluid may more Fluid and Electrolyte Therapy
accurately reflect the electrolyte losses. Other- in the Pediatric Intensive Care Unit
wise, a solution of 0.45 % NaCl can be used
initially and altered as the clinical situation con- Critically ill children present a challenge to the
tinues to develop and further electrolyte determi- clinician attempting to prescribe appropriate fluid
nations are made. and electrolyte therapy. Oftentimes, there may be
If the child’s volume status or the adequacy of acute kidney injury or multiorgan failure compli-
renal function is difficult to discern initially, it is cating management decisions. With such children,
best to provide the child with replacement of both rote reliance on standard equations or practice
insensible and ongoing losses. This approach guidelines to prescribe fluid and electrolyte ther-
should maintain the child’s current volume status apy may create significant fluid and electrolyte
and allow for further determination of the appro- anomalies. Rather than prescribing set mainte-
priateness of more vigorous hydration or con- nance requirement of fluid or electrolytes, the
versely fluid restriction as the clinical situation clinician should assess the patient’s individual
clarifies. Monitoring the child’s weight on at fluid and electrolyte needs in the context of the
least a daily basis and documenting the child’s underlying pathophysiology, the current volume
total fluid intake and output will also assist in status, the efficacy of tissue perfusion, the current
arriving at a proper hydration regimen. ventilatory requirements, and the current renal
Assessing the child’s current electrolyte status function. Whenever there is concern about incip-
and monitoring the loss of electrolytes in the urine ient or exacerbating fluid overload, it is important
or in any other source of significant output will to review the volume and type of fluids being
help tailor the daily electrolyte prescription. An provided. Maximizing the concentration of
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 391

continuous medication drips and assessing medi- azotemia or frank renal failure. Special care must
cation compatibility for simultaneous infusions be taken with the continuous modalities to insure
are important steps in limiting total daily fluid that ultrafiltration rates are periodically reassessed
input. Initially, it is crucial in these critically ill and readjusted. Furthermore, because the electro-
children to ascertain that their intravascular space lyte losses that accompany the ultrafiltration of
is replete to help maintain hemodynamic stability. fluid are isotonic, the electrolyte content of
Once a patient is felt to be intravascularly replete, infused fluids must be adjusted to match the com-
maintaining euvolemia by providing insensible position of the ultrafiltrate, especially if there is no
water losses as well as replacing any ongoing component of dialysis ongoing that may blunt the
fluid and electrolyte losses should maintain fluid development of serum electrolyte anomalies. As a
and electrolyte balance. result, serum electrolyte values need to be
Oftentimes, despite a desire to limit fluids in followed in a serial fashion with periodic review
the critically ill child, medication requirements, and readjustment of the composition of supple-
nutritional needs, and hemodynamic insufficiency mental intravenous fluids.
may result in very large daily fluid loads. There
may also be situations in which increased vascular
permeability causes a critically ill child to become Abnormalities in Serum Sodium
massively volume overloaded but with a Complicated by Kidney Injury
decreased effective circulating volume. In other
words, renal and tissue perfusion may be sluggish Because of the important contribution of serum
because fluid has moved from the intravascular sodium to serum osmolality, alterations in serum
space into the interstitial space. In this setting, sodium, especially coupled with alterations in
there may be a need to continue to administer BUN related to renal failure, can complicate the
large volumes of fluid to maintain circulatory usual approach to a child with fluid and electrolyte
integrity with the knowledge that such infusions anomalies. Generally, there are greater concerns
will only exacerbate the total body fluid overload. with hypernatremia and renal failure since the
Aggressive diuretic therapy may prove useful need to correct the sodium in a slow fashion can
especially if renal function is not compromised. be problematic when renal replacement therapy
Combination diuretic therapy utilizing agents that needs to be initiated for clearance of urea.
work at separate sites along the renal tubule may Balancing the correction of sodium and the
be necessary. Ultimately, the use of either periodic hyperosmolar state with the clearance of urea
or continuous ultrafiltration may be beneficial to requires a carefully considered plan that is
these patients by allowing ongoing fluid adminis- grounded in a firm understanding of fluid and
tration but limiting the daily imbalance between electrolyte homeostasis.
fluid intake and output. Ultrafiltration may be In most cases of hypernatremia related to
accomplished via peritoneal dialysis, by intermit- severe dehydration, some degree of acute kidney
tent hemodialysis with ultrafiltration or by utiliz- injury is present. This renal dysfunction is usually
ing one of the slow continuous ultrafiltration “prerenal” in nature, a result of a decreased effec-
techniques now known as continuous renal tive circulating volume rather than an intrinsic
replacement therapy (CRRT)). The recognition glomerular or tubular disorder. Most often, in the
that volume overload has a deleterious effect on course of rapid restoration of perfusion and early
many aspects of patient management and seems to rehydration, urine output increases and azotemia
be a strong prognostic indicator of poor ultimate begins to resolve.
outcome suggests that early ultrafiltration should Alternatively, there are occasional cases in
be considered in critically ill patients [152]. which due to intrinsic renal dysfunction or acute
If ultrafiltration is initiated, extreme vigilance tubular necrosis, the renal insufficiency will not
is necessary to prevent exacerbation of intravas- respond to volume infusion, and, in fact, the pro-
cular depletion and the development of prerenal vision of excess volume may contribute to
392 I.F. Ashoor and M.J.G. Somers

significant volume overload. In these cases, there boy has free water needs of 7.5 L to lower his
may be need to consider some form of renal serum sodium to the 140 mEq/L range
replacement therapy to assist in the controlled ([165/140  42]  42). Since the patient is now
correction of fluid and electrolyte derangements, significantly hypernatremic and has been subject
especially if the renal failure is oliguric or anuric to various fluid and electrolyte shifts as his dia-
in nature. Such an example is detailed in the betic ketoacidosis has been treated, it would be
following case study: prudent to correct his serum sodium by no more
A 15-year-old boy presents with several weeks than 10–12 mEq/day over the course of 3 days.
of polyuria, severe weight loss, fatigue, and poor Thus, if the boy undergoes ultrafiltration with a
oral intake. He is diagnosed as having diabetes goal of 2.5 l removed daily, and the ultrafiltration
mellitus with ketoacidosis by his pediatrician and volume each day is replaced back totally as free
referred to an emergency department for manage- water, the serum sodium should be in the normal
ment. At this point, his serum sodium is range in 3 days time. The ultrafiltration goal
154 mEq/L, his creatinine is 3.0 mg/dl, and his could be achieved over the course of a few hours
BUN is 30 mg/dl. In the emergency department, each day if the patient were hemodynamically
the child receives several bolus infusions of nor- stable or over a more prolonged period of time
mal saline supplemented with sodium bicarbon- each day if there were concerns regarding hypo-
ate and is started on an insulin drip. He is tension. Thus, either a conventional hemodialysis
admitted and continues to receive brisk intrave- setup could be used for relatively rapid ultrafil-
nous hydration with normal saline with bicarbon- tration only or a continuous filtration circuit for
ate supplementation per a practice guideline for either rapid or slow filtration.
treating children with diabetic ketoacidosis. He is Since the fluid removed in ultrafiltration is
noted to be oliguric and this does not improve isonatremic to the serum sodium, the sodium con-
with several more hours of hydration with normal centration of each liter of ultrafiltrate should mir-
saline following the guideline hydration recom- ror the serum sodium concentration at the time of
mendations. The next morning, laboratory values ultrafiltration. Thus, on the initial day of ultrafil-
reveal a serum sodium of 165 mEq/L, a creatinine tration, each liter of ultrafiltrate would contain a
of 4.5 mg/dl, and a BUN of 50 mg/dl. He has made sodium content of 165 mEq/L. By providing back
only 75 ml of urine in the last 8 h and is develop- the volume ultrafiltered each day as free water, the
ing some mild peripheral edema. serum sodium content could be expected to fall, in
In this case, the renal insufficiency and poor this case, by about 8–10 mEq/L/day.
urine output have complicated the usual manage- It is important to recognize that free water must
ment of diabetic ketoacidosis and has exacerbated be provided back to the patient to make up for the
an underlying hypernatremia. Given the patient’s ultrafiltration losses. Otherwise, since the ultra-
evolving renal failure, it is not feasible to provide filtrate is isotonic, there will be no change in the
the necessary volume of free water to correct the serum sodium concentration, and the ultrafiltra-
hypernatremia without contributing to further tion may potentially exacerbate the renal failure
volume overload. Because of the apparent pro- by depleting the intravascular space and the effec-
gressive renal failure, it would also be useful to tive circulating volume.
correct the hypernatremia in case dialysis Moreover, it is also important to recognize that
becomes necessary for urea clearance. By the boy’s overall daily fluid needs will be greater
performing controlled ultrafiltration on the than the daily ultrafiltration volume alone since
patient and replacing back the volume maintenance fluid requirements and any ongoing
ultrafiltered with free water, the serum sodium fluid losses must also be considered. Since the boy
could be corrected without exacerbating the vol- is in renal failure, his maintenance fluid needs can
ume status. be scaled back to insensible losses of 300 ml/m2/
With a serum sodium of 165 mEq/L and an day and, in this case, there are no ongoing losses.
estimated TBW of 42 L (70 kg  0.6 L/kg), this Thus, each day for the next 3 days, this 70-kg
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 393

patient needs to receive approximately 500 ml/day serum sodium concentration, the diffusional gra-
of insensible losses and 2,500 ml/day of ultrafil- dient for sodium clearance could be minimized.
tration replacement or a total of 3,000 ml/day. His Then, by making appropriate adjustments in the
maintenance sodium requirements are 3 mEq/kg/ sodium content of the dialysate as the serum
day. Although it may seem counterintuitive to sodium falls, the serum sodium levels could be
provide a hypernatremic patient with mainte- reduced gradually by 10–12 mEq/L/day, while at
nance sodium, disregarding these requirements the same time adequate urea clearance and ultra-
will result in a more rapid correction of the filtration for most situations would be achieved.
hypernatremia than desired. If the child were to Peritoneal dialysis has also been used in cases
receive a saline infusion of 0.45 % NaCl at a rate of severe hypernatremia [153–155]. Again, the
of 125 ml/h, this will provide just over 3 mEq/kg/ concentration of sodium in the dialysate may
day of sodium in a total volume of 3 L/day. need to be adjusted upwards in severe
If the child with hypernatremia has profound hypernatremia to prevent too rapid clearance of
renal failure and requires dialysis for urea clear- sodium. In addition, since the degree of clearance
ance, the dialysis prescription must take into and ultrafiltration may not be as precisely con-
account the need to correct the serum sodium trolled as with hemodialysis or hemodiafiltration,
slowly. Normally, regardless of the modality of frequent assessment of electrolyte values will be
renal replacement therapy, most dialysate contains necessary. Manipulation of dwell volumes and
sodium isotonic to the normal serum sodium dwell times will also influence overall clearance
range. It may prove detrimental, however, to dia- and the use of smaller dwell volumes for longer
lyze a patient who is very hypernatremic against a periods of time will help to minimize sodium
dialysate with a sodium concentration that is clearance.
30 mEq/L lower than the patient’s serum sodium In contradistinction to hypernatremia, since
concentration. The diffusional gradient during hyperosmolality is less common with
dialysis would lead to more rapid correction of hyponatremia, in some ways it is easier to employ
the serum sodium than the desired drop of approx- renal replacement therapy in the setting of severe
imately 1 mEq every 2 h. hyponatremia and concomitant renal insuffi-
Although most hemodialysis machines can be ciency. Again, the focus needs to be on the rapid-
readjusted so that the dialysate produced will have ity of the correction of the serum sodium. In
a sodium content as high as the low to mid-150s, conditions of severe but asymptomatic
this still may not reduce the gradient sufficiently hyponatremia of some chronicity, the rate of cor-
in cases of severe hypernatremia. In those situa- rection of serum sodium should parallel the rate of
tions, by maximizing the sodium concentration of correction recommended in hypernatremia 
the dialysate and by performing dialysis for lim- approximately 10–12 mEq/L/day. Correction of
ited amounts of time, one could minimize the drop chronic hyponatremia at a more rapid rate has
in serum sodium. Still, there would need to be been associated with the development of central
frequent assessments of the serum sodium con- pontine myelinolysis.
centration, and overall clearance may need to be All of the manipulations described above for
sacrificed to prevent too rapid correction of the hypernatremia and renal failure can be utilized
serum sodium and a rapid concomitant decrease in with hyponatremia and renal failure, with the
the serum urea that may increase the chances for understanding that the dialysate sodium concen-
dialysis disequilibrium. tration should now not exceed the serum sodium
Alternatively, a continuous hemodiafiltration value by 10–12 mEq/L. Conventional hemodial-
technique such as continuous venovenous ysis machines can be adjusted to produce dialy-
hemodiafiltration (CVVHDF) could be sate with a sodium concentration as low as the
performed. By asking the hospital pharmacy to mid-120s. In the very rare situation in which a
increase the sodium content of the dialysate and child with profound hyponatremia (<110 mEq/L)
replacement fluid to within 10–12 mEq/L of the was being hemodialyzed, brief hemodialysis runs
394 I.F. Ashoor and M.J.G. Somers

may be necessary initially to prevent too rapid needs to consider the possibility of transcellular
correction of the serum sodium level and the redistribution when interpreting any aberrant
attendant risk of central pontine myelinolysis. If serum potassium level before making decisions
dialysate is being custom prepared for peritoneal to supplement or restrict its provision.
dialysis or hemodiafiltration, precise alterations in In children, abnormalities in potassium homeo-
the electrolyte content can be made more readily stasis are uncommon but, as seen with other elec-
to reduce the sodium gradient. trolytes like sodium, physiologic perturbations that
The local resources, the training of ancillary may occur with acute illness or certain chronic
staff, the unique circumstances of each patient, medical conditions can interfere with normal regu-
and the comfort of the clinician with different latory processes and lead to clinically significant
modalities of renal replacement therapy will imbalances in potassium. As with other electrolyte
guide the choice of therapy when faced with issues, the ramifications of any abnormality gener-
renal failure and significant serum sodium anom- ally reflects on how quickly it has evolved, how
alies. The actual modality of renal replacement extreme it has become, and whether other medical
therapy utilized is less important than careful conditions are more likely to be adversely affected
attention to the rate of correction of the electrolyte or even perpetuate the abnormality.
anomaly, to the rate of urea clearance being
achieved, and to the clinical response of the
patient to ongoing therapy. Hyperkalemia

Hyperkalemia is usually defined as a serum potas-


Potassium sium concentration exceeding 5.5 mEq/L. In most
children, intact homeostatic mechanisms keep
There are several homeostatic mechanisms in serum potassium levels less than 5 mEq/L. With
place to maintain the usual high concentration of infants, there can be a higher range of normal
potassium in the intracellular space. These rely on potassium values because of baseline reduced
the sodium–potassium ATPase found on the cell GFR and some insensitivity to aldosterone, with
membrane, the effects of insulin and adrenergic levels as high as 6 mEq/L not uncommon. There is
hormones on transcellular potassium movement, generally little clinical consequence to serum
and the effects of nephron mass, GFR, hydration, potassium values up to 6 mEq/L, and most children
urinary flow, and especially aldosterone on renal rarely manifest any significant symptoms related to
potassium excretion and net body potassium hyperkalemia until serum potassium levels exceed
balance. 7 mEq/L. At that level, there may be lethargy and
In normal circumstances, potassium distribu- discernible muscle weakness or even paralysis,
tion is extremely well regulated since the gradient often initially affecting the legs. There can also be
between the intracellular and extracellular spaces sporadic heart palpitations or arrhythmias that can
plays a crucial role in the process of nerve excita- progress to asystole. As with any electrolyte aber-
tion and myocyte contraction. This explains why ration, high serum potassium levels should be con-
nearly 99 % of total body potassium can be found sidered in the clinical context of the child’s current
intracellularly and why aberrations in potassium medical status, and significantly elevated levels
handling that disrupts the usual gradient can be require urgent assessment and management tai-
not only detrimental to growth and development lored to take into account any comorbid conditions.
but also life threatening if cardiac conduction and
contractility or respiratory muscle function is sig-
nificantly affected. The intracellular localization Causes of Hyperkalemia in Children
of most body potassium stores also explains why
serum potassium levels are not a good estimate of The causes of hyperkalemia in children can be
total body potassium stores, and the clinician grouped into several broad categories based on
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 395

Table 11 Causes of hyperkalemia in children towards some impairment in renal potassium


Category Cause excretion.
Pseudohyperkalemia Technical issue with
phlebotomy Pseudohyperkalemia
Hemolysis of specimen in In pseudohyperkalemia, the elevated potassium
handling level does not reflect the child’s true potassium
Release of potassium from
balance. Especially in small children, there can be
leukocytes or platelets in blood
sample hemolysis of a blood sample due to technical
Increased potassium IV fluids, peripheral nutrition, reasons, with release of erythrocyte potassium
intake or potassium-containing stores [156, 157]. Smaller gauge needles must
medications often be used or samples withdrawn through
Potassium-based table salt indwelling small gauge intravenous catheters,
substitutes
with the need to apply significant suction to the
Transcellular Conditions with loss of cell
potassium membrane integrity attached syringe to obtain necessary sample vol-
redistribution Rhabdomyolysis ume. These techniques are up to sixfold more
Tumor lysis likely to result in a hemolyzed sample than with
Hemolytic states the use of vacuum flow devices [156].
Acidosis Children are also often less cooperative with
Periodic paralysis the phlebotomy procedure and may require phys-
Decreased potassium Reduced GFR ical restraint or may repetitively contract muscles
excretion Decreased effective circulating in an extremity while undergoing phlebotomy,
volume leading to local potassium release from myocytes
Impaired tubular secretion
and a hyperkalemic blood sample not reflective of
Reflux nephropathy
Obstructive uropathy
true systemic potassium levels. Additionally,
Type IV renal tubular hyperventilation seen with intense crying can
acidosis mediate a very acute respiratory alkalosis with
Sickle cell nephropathy potassium shifting out of cells and a rapid
Impaired increase in serum potassium by as much as
renin–angiotensin–aldosterone 1 mEq/L [156, 158].
axis
In children with marked leukocytosis or
Congenital adrenal
hyperplasia
thrombocytosis such as seen with leukemias or
Pseudohypoaldosteronism myeloproliferative disorders, there may also be
Drug effect (ACE/ARB, potassium egress from white cells or platelets as
eplerenone, spironolactone, the blood sample clots. A plasma potassium assay
aliskiren) decreases the chances of such a confounding
result.
In most cases of pediatric pseudohy-
underlying pathophysiology (Table 11). In chil- perkalemia, an elevation in serum potassium
dren, hyperkalemia may often be spurious and levels is unexpected on clinical grounds, and
related to technical issues commonly seen with details of the phlebotomy procedure or laboratory
pediatric phlebotomy. Such pseudohyperkalemia confirmation of hemolysis in the sample allow the
needs to be distinguished from true hyperkalemia result to be put into proper perspective. It does
that usually arises related to a disturbance in the become more difficult in children with preexisting
renal excretion of potassium or movement of nor- alterations in homeostatic mechanisms due to
mally sequestered intracellular potassium into the acute illness or chronic disease to disregard high
extracellular space. Hyperkalemia in children is potassium levels, and although there can always
rarely due to excess potassium ingestion alone and be an element of pseudohyperkalemia, it is judi-
sustained high potassium levels usually point cious to repeat another sample to determine that a
396 I.F. Ashoor and M.J.G. Somers

normal level exists before attributing high levels causes extracellular potassium shift to maintain
to pseudohyperkalemia exclusively. electroneutrality. In children, metabolic acidosis
is often mediated by decreased effective circulat-
Increased Potassium Intake ing volume from either severe dehydration or
In normal children, even very large dietary potas- infection. This volume imbalance may exacerbate
sium loads are unlikely to result in sustained the hyperkalemia by reducing GFR and reducing
hyperkalemia. Homeostatic mechanisms that pro- renal potassium excretion. The restoration of ade-
mote potassium influx into cells and aldosterone- quate intravascular volume and tissue perfusion
mediated enhanced kaliuresis tend to prevent generally corrects the acidosis and hyperkalemia.
hyperkalemia from intake of most foods. In A similar transcellular shift can be seen with insu-
small children with smaller volumes of distribu- lin deficiency and diabetic ketoacidosis, though
tion, exposure to some nutritional supplements the hyperkalemia can sometimes be blunted early
such as salt substitutes that can contain up to in this process by urinary potassium losses from
80 mEq potassium chloride/teaspoon can result an osmotic diuresis and excreted keto acids.
in potassium loading that exceeds by many fold In hyperkalemic periodic paralysis,
normal dietary potassium intake and can over- hyperkalemia is also related to transcellular relo-
whelm homeostatic mechanisms [159]. cation of potassium. This is an autosomal domi-
Children with impaired GFR may, however, nant condition in which a voltage-gated sodium
require dietary potassium restriction, though gut channel near the neuromuscular junction is
excretion of potassium increases with chronic affected and allows for ongoing movement of
renal dysfunction as a compensatory mechanism, potassium ions from the muscle into the blood-
even in the setting of children maintained chron- stream in response to stimuli that usually reduce
ically normokalemic on dialysis [160]. potassium flux [161]. Hyperkalemic periodic
In hospitalized children, iatrogenic errors in the paralysis may present as early as infancy and
concentration of potassium in intravenous fluids, need to be considered in any child with
peripheral nutrition, or certain intravenous medi- hyperkalemia and muscle weakness.
cations can result in the inadvertent provision of
large potassium loads, and hospitalized children Abnormalities in Renal Excretion
receiving potassium supplementation either orally
or in some intravenous form need to be monitored Intrinsic or Acquired Glomerular or Tubular
for the development of hyperkalemia. Dysfunction
Intrinsic anomalies in the renal handling and
Transcellular Redistribution of Potassium excretion of potassium or acquired conditions
Breakdown of normal tissue or rapid cell lysis can impairing normal excretion are a common cause
result in the release of large amounts of intracel- of significant pediatric hyperkalemia. Although
lular potassium along with other intracellular elec- aldosterone effect on the distal nephron is a key
trolytes into the extracellular space and plasma to potassium homeostasis, any renal disease that
water. This often occurs in the setting of rhabdo- impairs effective glomerular filtration will limit
myolysis from crush injury sustained in accidents absolute potassium clearance. As GFR falls in a
or natural disasters or after periods of extreme child with chronic kidney disease, the ability to
exercise, in tumor lysis syndrome in leukemia or excrete potassium tends to stay relatively well
lymphoma, or with severe hemolytic processes. compensated, especially when the GFR exceeds
There may often be accompanying acute kidney 30 ml/min/1.73 m2. With more profound func-
injury that exacerbates the hyperkalemia and com- tional impairment, especially in the setting of
plicates its management. lower urine output, hyperkalemia may be more
Transcellular redistribution may also occur in commonly encountered.
the absence of cellular damage. In metabolic aci- With children with obstructive uropathy or
dosis, the movement of hydrogen intracellularly reflux nephropathy, there is often a distal tubular
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 397

resistance to aldosterone that results in a type IV see significant hyperkalemia related to effective
renal tubular acidosis (RTA) and associated volume depletion alone and the effects described
hyperkalemia, even when the GFR is well pre- above [163].
served. In infants, there can also be immature In some children with chronically marginal
tubular responsiveness to aldosterone that is hydration and a diet that is low in sodium content,
developmental during early life, and this helps to this same situation of low distal delivery of
explain why babies have a tendency towards sodium exists and can result in high potassium
higher baseline serum potassium levels than levels. This is most often seen in babies with
older children. In children with sickle-cell disease feeding difficulties since both breast milk and
who have impaired medullary blood flow and formula contain little sodium.
ensuing hypoxic renal tubular injury, there is
also a tendency towards impaired aldosterone sen- Decreased Activity
sitivity over time. of Renin–Angiotensin–Aldosterone System
One study of children with acute febrile urinary Although infrequently encountered, endocrine
tract infection but no concomitant urinary tract anomalies impacting the RAAS can result in
obstruction or chronic kidney disease demonstrated infants or young children presenting with
significantly increased rates of hyperkalemia com- hyperkalemia. Congenital adrenal hyperplasia
pared to children with other febrile illnesses (CAH), and especially its most common form
[162]. This would suggest that, even in structurally 21-hydroxylase deficiency, results in impaired
normal distal renal tubules, conditions with intersti- mineralocorticoid production, salt wasting, and
tial inflammation such as seen with pyelonephritis hyperkalemia. CAH is part of the newborn screen
can impact normal potassium handling. across the entire United States and is included in
Certain drugs, most notably potassium-sparing newborn screening in many foreign countries,
diuretics, can similarly impact normal tubular leading to its identification prior to significant
renal excretion of potassium. These medications electrolyte anomalies in many infants. Primary
are often used for disease states such as heart or adrenal insufficiency is a rarer condition than
liver failure in which effective circulating volume CAH and affected children may present with
is also impaired and GFR may be suboptimal, hyperkalemia, though hyponatremia and hypoten-
further exacerbating potential kaliuresis. sion are more common [164].
Pseudohypoaldosteronism, an abnormality of
Decreased Effective Circulating Volume mineralocorticoid receptor activity, is an even
A much more common reason to see rarer endocrine condition. Children present char-
hyperkalemia in children is a state of low effective acteristically with hyponatremia, hyperkalemia,
circulating volume that results in either ineffective and metabolic acidosis, but serum aldosterone
tissue perfusion and the generation of a metabolic levels are elevated, underscoring that the issue is
acidosis or sodium and water avidity in the prox- in the cell receptor and not in mineralocorticoid
imal portion of the nephron. As a result, there is production. An autosomal dominant form only
decreased distal flow of filtrate in the nephron and affects the kidney and, over time, affected chil-
a decreased concentration of sodium in any filtrate dren may show spontaneous improvement due to
actually presented to the distal nephron. This leads maturational enhancement of tubular sodium
to what is often termed a functional renal tubular transport. In the autosomal recessive form, there
acidosis or impaired aldosterone-mediated is generally pronounced distal tubular epithelial
kaliuresis and acid excretion that is not related to sodium channel dysfunction that presents early in
any inherent anomaly in the channels or receptors childhood and requires significant ongoing
in the distal nephron. Especially in young chil- sodium supplementation [165].
dren, there is an increased tendency to develop Children being treated with spironolactone,
significant dehydration and hemodynamic insta- eplerenone, aliskiren, or one of the angiotensin-
bility with acute illness, and it is not uncommon to converting enzyme inhibitors or angiotensin
398 I.F. Ashoor and M.J.G. Somers

receptor blockers will also have decreased miner- Table 12 Diagnostic studies to assess unexplained
alocorticoid production or activity and may hyperkalemia in children
develop hyperkalemia directly related to their Laboratory test Key findings
use. Many of these drugs are used in children Complete blood count, Hemolysis, thrombotic
with medical conditions that already predispose platelet count microangiopathy, blood
dyscrasias
to hyperkalemia due to adverse affects on effec-
Creatinine Reduced GFR
tive volume, GFR, and baseline renal tubular han-
Electrolytes Metabolic acidosis or other
dling of solute. electrolyte aberration
Urine electrolytes and Evidence of inappropriate
urine creatinine (best urinary K excretion
Evaluation of Hyperkalemia in Children done concomitantly with (UK < 20 mEq/L in setting
serum electrolytes and of hyperkalemia)
creatinine) Evidence of reduced
In many children, the clinical presentation or effective volume or
known preexisting medical conditions will point reduced distal tubule
clearly to the underlying etiology of the sodium delivery
hyperkalemia and its proper treatment, and there (UNa < 20 mEq/L)
Lactic dehydrogenase Elevated levels suggesting
is little need for an extensive diagnostic evalua-
hemolysis
tion. For instance, children with tumor lysis, rhab-
Creatine kinase Elevated levels suggesting
domyolysis, structural or functional kidney muscle injury
disease, or markedly decreased effective circulat- Serum aldosterone and Aberrant levels suggest
ing volume all are likely to have correction in their plasma renin activity endocrine abnormality
potassium imbalance as the underlying condition
is treated. In children with no obvious reason for
hyperkalemia and normal renal function and vol-
ume status, a high index of suspicion for potassium in conjunction with reabsorption of
pseudohyperkalemia must exist and potassium sodium, it is often necessary to also measure uri-
values should be confirmed prior to any further nary sodium and urinary osmolality to more
evaluation. completely assess the appropriateness of random
In some children, however, the cause of a per- urine potassium results. Low random urinary
sistently high potassium level will not be obvious, sodium values (<20 mEq/L) demonstrate that
and it is important to assess the renal handling of there is renal sodium avidity, and this may limit
sodium and potassium as well as the renin–angio- the potential secretion of potassium in the distal
tensin–aldosterone system carefully. The direc- nephron even in the face of a normal hormonal
tion of any further evaluation is then guided both milieu and normal renal tubular cell receptors and
by the medical history and initial results from channels. Measures that improve the effective
typical screening laboratories of blood and urine volume and provide more sodium for distal tubu-
(Table 12). lar delivery augment kaliuresis.
In most children, hyperkalemia should result in For some time there was interest in calculation
a random urinary potassium level that exceeds of the transtubular potassium gradient (TTKG) to
20 mEq/L and often exceeds 40 mEq/L. Very help determine if there was appropriate mineralo-
high random urinary potassium values (>80 to corticoid effect in states of hyperkalemia in pedi-
100 mEq/L) almost always point towards intact atric patients [166]. The TTKG estimated the ratio
mechanisms to achieve kaliuresis, and diagnostic of the potassium concentration at the end of the
focus should shift to an issue with potassium intake cortical collecting tubule, the site responsible for
or cellular release rather than renal excretion. most of potassium secretion, to blood potassium
Since random urinary potassium levels may be levels. The TTKG was predicated on the assump-
affected by the degree of urinary concentration tion that changes in the osmolality in the
and by the ability of the distal tubule to secrete collecting duct were only due to water
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 399

reabsorption, but there is now evidence of urea Table 13 Treatment of hyperkalemia in children
cycling in the collecting duct that invalidates this Potassium level <6 Limit potassium intake
premise [167]. Accordingly, the TTKG is no lon- mEq/L Optimize effective volume
ger considered a reliable assessment of mineralo- Potassium levels 6–7 Stop potassium intake
corticoid effect and, when there is clinical mEq/L Optimize effective volume
No ECG changes or only Consider loop diuretic
suspicion of an endocrine issue interfering with peaked T waves therapy such as
potassium handling, serum aldosterone and If other ECG changes, furosemide 1 mg/kg q 6 h
plasma renin activity should be measured. consider therapy for Consider sodium
K > 7 mEq/L polystyrene sulfonate
1 g/kg up to 40 g q 4 h
given po, pg, or pr
Treatment of Hyperkalemia in Children Potassium levels >7 Stop potassium intake
mEq/L Optimize effective volume
The degree of potassium elevation and its likeli- Nebulized beta agonist
such as albuterol
hood to cause immediate cardiac arrhythmias
Calcium gluconate (10 %):
guide the urgency of treatment and the therapies 10 mg/kg IV to stabilize
employed. Specific interventions range from lim- myocardium
iting further potassium intake, to facilitating intra- Insulin and glucose:
0.1–0.3 units/kg regular
cellular shifts of potassium from the extracellular
insulin and 2–4 ml/kg D25
space, to augmenting potassium excretion in the Consider sodium
urine or stool, to actually removing potassium bicarbonate (only after
from the extracellular space by dialysis. Outside calcium provision)
1 mEq/kg
of the significant clinical evidence that exists to
Sodium polystyrene
demonstrate that dialysis is an effective way to sulfonate 1 g/kg up to 40 g
correct hyperkalemia and prevent life-threatening q 4 h given po, pg, or pr
complications, there have been few pediatric- Consider loop diuretic
therapy such as
specific studies that have addressed the overall
furosemide 1 mg/kg IV q
efficacy of some of these maneuvers, although 6h
all of them are commonly employed in clinical Urgent dialysis if no
practice [168] (Table 13). improvement or
concomitant significant
renal failure
General Approach to Therapy
In children with potassium levels >6.5 mEq/L or
specific immediate clinical concern for cardiac fibrillation, there may come to be a sinusoidal
effects of hyperkalemia prior to the laboratory pattern as the QRS complex merges with the T
value having returned, an electrocardiogram wave. Not all children with hyperkalemia will
should be obtained to look for abnormal atrial manifest ECG changes and absence of ECG
(P wave) or ventricular (QRS complex) depolari- change does not preclude the need for potential
zation or abnormal repolarization (T wave). In therapy for significant potassium elevation.
children, ECG changes are more likely to be In children with no ECG changes or peaked T
related to serum potassium levels than in adults, waves alone and more moderate levels of
who are more prone to preexisting cardiac con- hyperkalemia (<7 mEq/L), initial therapy
duction abnormalities or cardiovascular disease. includes limiting further potassium intake and
As hyperkalemia develops, initial ECG findings promoting potassium loss in the stool with an
include tall, peaked, symmetric T waves and enteral cation exchange resin, with consideration
shortened QT intervals. With more severe of augmenting urinary potassium losses with the
hyperkalemia, P waves become flattened, PR use of a loop diuretic.
intervals prolonged, and QRS complexes wid- In children with ECG changes other than
ened. Eventually, as a harbinger to ventricular peaked T waves or with more pronounced
400 I.F. Ashoor and M.J.G. Somers

hyperkalemia (>7 mEq/L), calcium infusion is intraventricular hemorrhage when two nebuliza-
used to stabilize the cardiac membrane excitabil- tions with albuterol were compared to two saline
ity that comes with hyperkalemia. Although its nebulization treatments [170].
protective effects begin quickly, its duration may With intravenous access, other temporizing
be short-lived and further infusions may be nec- maneuvers such as infusion of insulin and glucose
essary. Concomitantly, therapies that promote the or systemic alkalinization with sodium bicarbonate
shift of potassium into the intracellular space such can be attempted. Rates of these infusions must be
as nebulized beta agonists or intravenous insulin calculated on a case-by-case basis according to the
and glucose are begun and enteral cation child’s weight, given the broad spectrum of body
exchange resin given. sizes in pediatric patients. If insulin is provided,
In children unresponsive to such maneuvers or glucose should be given concomitantly to prevent
with concomitant severe renal dysfunction, dialy- hypoglycemia, and the potassium level should start
sis may be necessary. Generally, hemodialysis is to fall within 15 min of insulin provision with peak
the preferred modality to reduce potassium levels insulin effect by an hour. When sodium bicarbon-
most quickly and in the most controlled fashion. ate is infused repetitively, the risk of developing
In some centers, local resources may make forms hypernatremia exists.
of CRRT a better option than conventional hemo- There has been limited experience with a solution
dialysis. Although peritoneal dialysis can be used, containing fixed concentrations of calcium gluco-
potassium loss will be less efficient and less well nate, insulin, dextrose, and sodium acetate in treating
controlled. hyperkalemic children [171]. An advantage to such a
solution is that it could be readily prepared by hos-
Therapies Redistributing Potassium pital pharmacies to be available in hyperkalemic
These therapies are rapid in onset but generally emergencies and could then be infused continuously
have limited duration since no net potassium is to allow for more sustained effect, although the caus-
removed from the child and, as the therapeutic tic properties of such a solution may prevent its
effect of intervention wanes, potassium can redis- provision through peripheral vasculature.
tribute back to the extracellular space. As a result,
most of these therapies are done along with other Therapies Removing Potassium from
maneuvers to either remove potassium from the the Body
body or to adequately reverse whatever is causing Diuretics – Although loop and thiazide diuretics
the hyperkalemia to develop. By themselves, they can promote kaliuresis, given that a large propor-
are less effective in providing a management tion of children with hyperkalemia will have
solution. decreased effective circulating volume or renal
Since securing intravenous access in young dysfunction, the effectiveness of diuretic therapy
children can be more problematic than with ado- in most hyperkalemic children is limited, espe-
lescents or adults, using inhaled beta agonists can cially in the urgent setting. In children with ade-
be an attractive initial option to move potassium quate effective volume, using these diuretics and
intracellularly. This maneuver should be avoided maintaining a diuresis with ongoing optimization
in children manifesting any preexisting cardiac of effective volume could be considered as an
arrhythmia and children should be on a cardiac adjunct. Such therapy can especially be consid-
monitor during its provision. Tachycardia and ered in children with persistently high but clini-
tremors are common, but are usually short-lived. cally less urgent hyperkalemia (5.5–6.5 mEq/L).
Reductions of serum potassium concentrations by Enteral cation exchange resins – Polystyrene
1.5 mEq/L have been obtained as quickly as sulfonates have been used for decades in children
within an hour [169]. Such therapy has been with hyperkalemia. Dissolved in water and taken
shown to be safe and effective even in premature orally or given as a retention enema, they work by
infants, with no differences in heart rate, CNS exchanging a counterion such as sodium or
symptoms, ECG anomalies, hyperglycemia, or calcium for potassium across the large intestine.
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 401

The potassium polystyrene sulfonate complex that turnaround of results. One study of 60 children
results cannot be digested and gets excreted in demonstrated that samples done by blood gas
stool, thereby effectively removing potassium analyzers averaged nearly 0.5 mEq/L lower than
from the body. Each gram of resin may bind as conventional assays [175].
much as 1 mEq of potassium. During this process, Clinically, hypokalemia is encountered more
the resin releases an equivalent amount of its coun- commonly than hyperkalemia, especially in hos-
terion, and provision of large amounts of these pitalized children. In studies of children in pedi-
resins can lead to sodium or calcium imbalances. atric intensive care unit, up to 20 % had
To prevent constipation or fecal impaction, hypokalemia on admission and up to 40 % ulti-
polystyrene sulfonates were mixed for many mately had hypokalemia identified during the
years with sorbitol that would serve as an osmotic course of their ICU stays [176–178]. Hypokalemia
laxative. Reports of colonic necrosis and perfora- was more likely seen in children with cardiac or
tion in patients receiving such mixtures, espe- renal disease or with hemodynamic collapse, as
cially in postsurgical settings where there was well as in children with infections or who required
impaired gastrointestinal mobility, prompted the calcium supplementation [176].
FDA to warn that sodium polystyrene sulfonate Children are often asymptomatic until serum
should not be administered concomitant with sor- potassium levels approach 2.5 mEq/L. Since low
bitol [172]. To counteract its constipating effects, extracellular potassium levels alter the usual
polystyrene powder or powder mixed with water transcellular potassium gradient, significant clinical
can be given with lactulose or polyethylene glycol manifestations of hypokalemia often relate to
3,350 preparations as laxatives. changes in muscle contraction or cardiac conduction
Although widely used in children with [179]. Muscle weakness develops starting in the legs
hyperkalemia both acutely and chronically, one and then may ascend. With profound hypokalemia,
comprehensive review found no randomized evi- respiratory muscles can become involved and there
dence for the efficacy of these binders in the may even be rhabdomyolysis. Smooth muscle dys-
emergency situation [173], and most of the limited function in the gastrointestinal tract can present as
studies of its efficacy have been in individuals nausea, constipation, or frank ileus. In terms of
with chronic kidney disease or after multiple cardiac effects, children with hypokalemia often
doses [173, 174]. Although the only therapy manifest characteristic ECG changes with PR inter-
short of dialysis that can remove potassium from val prolongation, dampening of the T waves, and ST
the body, in most hyperkalemic children, how- depression. As hypokalemia exacerbates, there can
ever, its provision in the setting of significant be the appearance of U waves as well [179, 180].
hyperkalemia is still regarded as a reasonable With chronic hypokalemia there can also be
therapy by most pediatric clinicians. direct renal effects leading to polyuria. This poly-
uria arises related to several mechanisms, includ-
ing the stimulating effect of hypokalemia on thirst
Hypokalemia and other neuroendocrine factors as well as the
impact of persistent hypokalemia on aquaporin
Hypokalemia is usually defined as a serum potas- protein depletion in the cortical collecting duct
sium level less than 3.5 mEq/L, although, in most limiting the number of effective water channels
children and adolescents, levels between 3 and 3.5 [48, 181, 182].
mEq/L are asymptomatic, and some reference
laboratories even set the lower limit of normal in
this range. There may also be measurement dif- Causes of Hypokalemia in Children
ferences between samples processed by conven-
tional laboratory AutoAnalyzers and samples The causes of hypokalemia in children can be
assayed by point-of-care blood gas analyzers that grouped into four broad categories based on
are often used in critical care areas for rapid underlying pathophysiology: decreased
402 I.F. Ashoor and M.J.G. Somers

Table 14 Causes of hypokalemia in children (ROMK) within the renal tubule and resistance
Decreased potassium intake to the insulin-mediated intracellular flow of potas-
Chronic dietary restriction sium [183]. Mild hypokalemia from simple
Malnutrition, eating disorders decreased intake can, however, be exacerbated
Iatrogenic errors in nutritional supplementation by diuretic use, diarrhea, or eating disorders such
Transcellular redistribution of potassium as anorexia or bulimia. States of chronic malnu-
Alkalosis trition are especially prone to severe hypokalemia
Endocrine effects if potassium loss is then superimposed. For
Insulin, catecholamines, thyroid hormone instance, one report of malnourished children in
Periodic paralysis Kenya demonstrated a baseline rate of hypokale-
Medications mia of 10 % from decreased intake, but this
Beta-adrenergics, chloroquine, antipsychotic agents
increased more than threefold in the setting of
Extrarenal potassium losses (usually GI tract)
concomitant diarrhea [184]. In hospitalized chil-
Diarrhea
dren, inadequate potassium intake may be a reflec-
Vomiting
tion of acute or chronic illness preventing
Nasogastric tube losses
Renal potassium losses
sufficient provision of general nutrition, or there
Increased delivery of sodium and water to distal tubule can be iatrogenic oversights such as the incorrect
Diuretics, mannitol, keto acids formulation of intravenous peripheral nutrition
Renal tubular acidosis solutions. Keeping in mind concomitant clinical
Hyperaldosteronism factors that may be influencing potassium balance
Related to effective volume depletion should facilitate the provision of proper potassium
Glucocorticoid-remediable aldosteronism (GRA) supplementation.
Apparent mineralocorticoid excess (AME)
Congenital adrenal hyperplasia Transcellular Redistribution of Potassium
Tubulopathies
Bartter syndrome Metabolic Alkalosis
Gitelman syndrome Alkalosis promotes hypokalemia through two
Medications mechanisms. As systemic pH increases, hydrogen
Amphotericin B, cisplatin ions in the intracellular space move extracellularly
to help blunt the alkalosis. To maintain
electroneutrality, extracellular potassium then
potassium intake, transcellular redistribution of shifts intracellularly and this will be reflected by
potassium, extrarenal potassium losses, and renal a fall in serum potassium levels.
potassium losses (Table 14). Clinical history With alkalosis, there is also increased potas-
makes the etiology apparent and renders extensive sium excretion in the urine. With the increase in
diagnostic evaluation moot in many situations. serum bicarbonate, there is an increased load of
Children with a history of recurrent hypokalemia, bicarbonate filtered and eventually passed in the
especially in the absence of accompanying acute urine. Since a cation must accompany the filtered
illnesses, need to be carefully considered for bicarbonate, this means that there are also
underlying derangements in renal potassium increased urinary loads of potassium and sodium.
handling. Some of this potassium will get directly excreted
along with the bicarbonate. Additionally, the
Decreased Potassium Intake increased concentration of sodium that is also
By itself, decreased dietary intake of potassium is presented to the distal nephron allows there to be
a rare cause of hypokalemia in children. Ongoing more ready exchange of sodium for potassium in
potassium restriction leads to activation of potas- the distal nephron under the influence of aldoste-
sium conserving mechanisms, such as a reduction rone, with even further urinary potassium losses.
in the number of excretory potassium channels This mechanism is most pronounced when there is
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 403

concomitant effective volume depletion from gas- Drugs


trointestinal losses or diuretic use or if hyperaldos- In addition to beta-adrenergic agents as discussed
teronism exists for other reasons. above, redistributive hypokalemia has also been
described in chloroquine intoxication and also
Endocrine Effects with the provision of risperidone and quetiapine
Transcellular redistribution of potassium can also [188, 189]. In children who have inadvertently
arise from the effects of certain hormones. This is ingested barium salts found in certain fireworks
most commonly seen from the effects of insulin or rodent poisons, hypokalemia has also been
and from catecholamines with beta-adrenergic described as a result of inhibition of normal potas-
effects, both of which promote cell uptake of sium egress from cells [190, 191]. In these cases,
potassium. The ability of these substances to the clinical history and consultation with local
accomplish effective transcellular shift is toxicology resources should help guide appropri-
underscored by the common use of both insulin ate diagnosis and management.
and nebulized albuterol to treat hyperkalemia.
Insulin-mediated hypokalemia can also be seen Extrarenal Potassium Losses:
in children with refeeding syndrome or with eat- Gastrointestinal Tract
ing disorders where insulin release may exacer- Hypokalemia can evolve from upper or lower
bate a preexisting tendency towards hypokalemia gastrointestinal tract losses, and gastrointestinal
from long-term poor nutritional intake. Children illness with unreplaced losses of potassium
receiving intermittent nebulized albuterol are accounts for most cases of hypokalemia in other-
unlikely to present with significant alterations in wise healthy children. At up to 50 mEq/L,
serum potassium levels, but with continuous neb- the potassium content of diarrhea is high
ulization there can be a tendency for lower potas- compared to other body fluids, including
sium levels to develop [185]. In critically ill vomit or gastric secretions [192]. Although
children receiving intravenous catecholamines, both types of gastrointestinal loss can cause
either for bronchodilation or for hemodynamic hypokalemia, the underlying pathophysiology
reasons, there can be an augmented effect; up to differs.
50 % of children with these treatments have been With diarrheal illness, there are not only sig-
reported to develop hypokalemia [186]. nificant potassium losses from the stool, but with
In hyperthyroidism, hypokalemia can develop, the evolution of concomitant volume depletion,
mediated in part by increased expression and an aldosterone-mediated kaliuresis can ensue.
activity of the sodium–potassium ATPase This can be exacerbated in the early phases of
[187]. Thyrotoxic periodic paralysis is a rare con- rehydration since, with improved effective circu-
dition, almost always seen in adults, where lating volume and augmented GFR, there will be
patients with hyperthyrodism develop profound increased delivery of filtered sodium to the distal
hypokalemia and ensuing muscle dysfunction. nephron and more substrate for aldosterone-
This condition is in contradistinction to hypoka- mediated potassium secretion.
lemic periodic paralysis caused by autosomal dom- In contrast, upper gastrointestinal losses
inant mutations in voltage-gated calcium channels from vomiting or nasogastic drainage may
found in the skeletal muscle and in voltage-gated contain less than 10 mEq/L of potassium. The
sodium channels at the neuromuscular junction. metabolic alkalosis that can develop from these
Affected individuals can present throughout child- losses will, however, lead to increased distal tubu-
hood but especially in adolescence, have normal lar delivery of sodium bicarbonate, and with the
thyroid function, and often have their episodes of secondary upregulation of aldosterone from vol-
pronounced hypokalemia and paralysis instigated ume losses, the increased availability of sodium in
by an event causing insulin or adrenergic hormone distal tubular filtrate will allow for increased
release to initiate a fall in serum potassium levels exchange for potassium and an effective
that then gets exacerbated by their channelopathies. kaliuresis.
404 I.F. Ashoor and M.J.G. Somers

Renal Potassium Losses can be seen, generally accompanied by hyperten-


sion. In autosomal dominant glucocorticoid-
Increased Distal Sodium Delivery remediable aldosteronism, there is aldosterone
Principal cells in the collecting duct contain apical synthase hyperactivity due to genetic mutations
epithelial cell sodium channels (ENaC) where that allow aldosterone production to become reg-
sodium is reabsorbed under the influence of ulated by ACTH. The provision of glucocorticoid
mineralocorticoid (primarily aldosterone) and suppresses the mineralocorticoid excess and ensu-
exchanged electroneutrally for potassium via ing high blood pressure and any associated hypo-
ROMK channels. For potassium excretion to kalemia [193]. In apparent mineralocorticoid
occur effectively, sodium delivery to these neph- excess (AME), cortisol degradation is impaired
ron segments must be adequate and mineralocor- by genetic mutations that alter the efficacy of
ticoid activity must be present. Increased delivery 11-beta-hydroxysteroid dehydrogenase, and cor-
of sodium distally augments potassium excretion, tisol can then bind to mineralocorticoid receptor
especially in clinical situations where there are and mediate signs and symptoms mimicking aldo-
also effective circulating volume depletion and sterone excess. This same enzyme can be
increased aldosterone activity. This physiology inhibited by glycyrrhizic acid that can be found
helps to account for the hypokalemia that can be in natural licorice and this explains why excess
seen with diuretic provision, with mannitol provi- ingestion of natural licorice has been associated
sion, with diabetic ketoacidosis, and, as explained with hypokalemia as well. In forms of congenital
above, with metabolic alkalosis from vomiting or adrenal hyperplasia such as 17-alpha-hydroxylase
nasogastric tube drainage. deficiency and 11-beta-hydroxylase deficiency,
there can also be excess mineralocorticoid pro-
Renal Tubular Acidosis duction and hypokalemia and hypertension.
Both type 1 and type 2 RTA are associated with
hypokalemia, although from varying physiologic Renal Tubulopathies
mechanisms. With type 1 or distal RTA, the intrin- Certain genetic tubular disorders are characterized
sic inability of the distal nephron to acidify the by impaired sodium reabsorption, increased distal
urine by secreting hydrogen ions leads to potas- tubular sodium delivery, and ensuing urinary
sium secretion to maintain electroneutrality. There potassium losses with the development of an asso-
may also be associated tubular cellular membrane ciated alkalosis. In the various types of Bartter
permeability that allows for further potassium syndrome, abnormalities in the electrolyte trans-
loss. With type 2 or proximal RTA, there is porters or channels in the thick ascending limb
impaired proximal tubular reabsorption of filtered help mediate a kaliuresis and alkalosis as well as a
bicarbonate, leading to increased urinary losses of tendency towards hypercalciuria. In Gitelman
bicarbonate and associated cations. The increased syndrome, the thiazide-sensitive sodium chloride
distal tubular presentation of sodium facilitates symporter in the distal convoluted tubule is
potassium secretion, exacerbated by the aldoste- impaired, and there is often concomitant hypo-
rone effect from any effective volume depletion magnesemia and normal or lower urinary calcium
caused by the augmented sodium and water excretion. With both syndromes volume depletion
losses. and physiologic aldosterone stimulation will only
serve to aggravate any underlying tendency to
Hyperaldosteronism waste potassium in the urine, but provision of
Normal physiologic stimuli for aldosterone secre- volume will not ameliorate baseline potassium
tion, most notably volume depletion, commonly wasting. In Liddle syndrome, gain-of-function
play a contributing role to many of the causes of mutations in ENaC result in dysregulated sodium
pediatric hypokalemia. In comparison, primary reabsorption with ensuing hypertension, hypoka-
abnormalities in aldosterone secretion are rare in lemia, and alkalosis as seen with more classic
childhood, though certain inherited conditions cases of mineralocorticoid excess.
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 405

Any Fanconi syndrome, whether idiopathic in the low serum potassium such as cardiac arrhyth-
etiology, related to drug exposure, or related to mias or alterations in muscle tone or strength.
renal conditions such as cystinosis, can also result There should also be some clinical assessment of
in hypokalemia as part of the diffuse proximal effective circulating volume and respiratory
tubule solute losses that ensue. Children with effort, since severe abnormalities will influence
hypokalemia from proximal tubular dysfunction immediate management decisions and may
will also lose large amounts of bicarbonate in their impact acid–base balance and, as a result,
urine and will be prone to a metabolic acidosis, in transcellular potassium distribution. Blood pres-
distinction to the metabolic alkalosis that charac- sure should also be carefully measured since the
terizes Bartter and Gitelman syndrome. presence or absence of hypertension helps distin-
guish certain of the renal tubulopathies.
Drugs
Diuretics are by far and away the most commonly Laboratory Testing
prescribed drug that can mediate hypokalemia In the event that the cause of the hypokalemia is
through direct urinary potassium losses, by clear-cut – for instance vomiting with dehydration
increasing distal sodium presentation, and by con- or diuretic therapy in the setting of poor potassium
tributing to effective volume depletion and phys- dietary intake – it is probable that follow-up
iologic aldosterone activity. In addition, assessment of serum potassium after proper ther-
amphotericin and some chemotherapy drugs, apy will show resolution of the problem and no
most notably cisplatin, can also cause increased reason to do further testing. On the other hand, in
renal potassium wasting. With amphotericin, cases of less clear etiology, in cases of recurrent
there is actual disruption of cellular membrane hypokalemia, or when hypokalemia recurs after
integrity and ensuing loss of potassium from the initial supplementation is completed, there needs
intracellular space. Some studies have suggested to be more specific assessment of renal handling
that up to half of the children receiving of potassium to determine if there are inappropri-
amphotericin will become hypokalemic, though ate renal losses ongoing.
the provision of liposomal amphotericin seems to With profound hypokalemia (<2.5 mEq/L)
substantially mitigate this problem [194]. With and especially when there is evidence or suspicion
cisplatin, there can be direct tubular toxicity that of adverse cardiac effect, steps to ameliorate the
interferes with the handling of potassium as well hypokalemia should begin immediately and
as wasting other electrolytes, with effects that can should not be delayed by further diagnostic eval-
be quite long lived [195]. uation. Fortunately, the blood and urine samples
necessary to guide the evaluation can often be
readily collected contemporaneously with initia-
Diagnostic Evaluation tion of therapy (Table 15).

History and Physical Examination Urine Potassium Quantification


The patient’s history is important in guiding the A 24-h urine collection will give the most accurate
diagnostic evaluation. An otherwise healthy child assessment of the degree of urinary potassium
with hypokalemia accompanying an acute gastro- losses since it can be directly compared to potas-
intestinal illness requires less of an evaluation sium intake to assess a net balance. Timed urine
than a child with recurrent bouts of unexplained collections are difficult to perform in many chil-
hypokalemia. Ascertaining any underlying dren, so spot urine chemistries with concomitant
predisposing medical conditions, medications, or serum chemistries are generally substituted and,
dietary restrictions may help clarify the cause of in most cases, will provide enough information to
the child’s hypokalemia. allow for accurate assessment of renal response.
The physical examination should key on find- In the setting of a serum potassium level less
ings suggesting significant adverse effects from than 3 mEq/L, random urinary potassium values
406 I.F. Ashoor and M.J.G. Somers

Table 15 Initial diagnostic studies to assess unexplained Table 16 Hypokalemia and acid–base disorders
hypokalemia in children
Hypokalemia and metabolic acidosis
Laboratory test Key findings Low urinary losses of potassium
Creatinine, electrolytes, Derangement in addition Diarrhea
phosphorus, magnesium to potassium High urinary losses of potassium
Venous pH Systemic acidosis or Renal tubular acidosis
alkalosis
Proximal tubulopathy (Fanconi or Fanconi like)
Aldosterone and plasma Variation from normal
Hypokalemia and metabolic alkalosis
renin activity levels
Low urinary losses of potassium
Urinalysis Appropriateness of pH
and presence of Diuretics (chronically)
glycosuria High urinary losses of potassium
Urine chemistries and urine Urinary K excretion Hyperaldosteronism (often concomitant
creatinine (best done Random UK > 20 hypertension)
concomitantly with serum mEq/L suggests renal Diuretics (acutely)
electrolytes and creatinine) loss Bartter syndrome (often concomitant
U K/Cr > 15 mEq/g hypercalciuria)
suggests renal loss
Gitelman syndrome (often concomitant
Hypercalciuria
hypomagnesemia)
Urinary Cl excretion

Aldosterone and plasma renin activity will help


usually are less than 20 mEq/L and values signif- determine if there is hyperaldosteronism. Urinary
icantly higher suggest renal wasting. Electrolyte calcium excretion should also be measured to look
concentration in a random urine void will be for hypercalciuria. Urinary chloride levels will
affected by water handling at that point in time, help in the differential diagnosis of hypokalemia
so spot samples in particularly dilute or concen- with alkalosis (Table 16).
trated samples may provide values that are lower In the hypertensive child with urinary potas-
or higher than if urine output were normal. sium wasting, both low blood aldosterone and
Accordingly, urinary potassium-to-creatinine renin levels should make apparent mineralocorti-
ratios can be done since urinary creatinine excre- coid excess, Liddle syndrome, or a form of adre-
tion is constant in children with normal renal nal hyperplasia the most suspected pathologies.
function and this will correct for water excretion. With high aldosterone but low renin levels,
In the setting of hypokalemia, a urine potassium- hyperaldosteronism should be suspected.
to-creatinine ratio <15 mEq/g creatinine (<1.5 In the normotensive child with urinary potas-
mEq/mmol Cr) suggests the hypokalemia is not sium wasting, the presence of an acidosis suggests
due to renal potassium wasting at that time and an RTA. With an alkalosis, urinary chloride levels
focus should shift to GI losses, poor intake, cellu- less than 15 mEq/L speak to renal potassium
lar shift, or prior diuretic use. Ratios >15 mEq/g wasting from volume depletion in the setting of
creatinine suggest renal wasting. a metabolic alkalosis as seen with emesis, naso-
gastric tube losses, or diuretics. With an alkalosis
Further Testing with Renal Potassium and higher urinary chloride levels, a renal
Wasting tubulopathy such as Bartter or Gitelman syn-
In the setting of urinary potassium wasting, drome is suspected. Hypercalciuria would point
assessment of the child’s blood pressure along towards Bartter syndrome, and hypomagnesemia
with a few other specific urine and blood tests is more frequent with Gitelman, though genetic
helps to clarify the reason for the kaliuresis. Spe- testing to clarify specific mutations and which
cifically, a venous pH and serum electrolytes and channels or transporters are affected allows for
serum magnesium level will allow the identifica- there to be a better understanding if the potassium
tion of an acid–base disorder, the presence of an wasting arises from the thick ascending limb or
anion gap, and the presence of hypomagnesemia. the distal convoluted tubule.
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 407

Treatment of Hypokalemia in Children individual response to potassium supplementa-


tion. For instance, the provision of amphotericin
Urgency of Potassium Supplementation or furosemide to children receiving potassium
In patients with significant cardiovascular or supplementation results in more moderate
respiratory compromise related to hypokalemia, increases in serum potassium because of urinary
management must proceed emergently and be potassium losses. Conversely, children receiving
focused on acutely increasing the serum potas- ACE inhibitors often exhibit somewhat higher
sium level by 0.3–0.5 mEq/L and then serum potassium levels after supplementation as
maintaining supplementation until a level of a result of the effect of angiotensin blockade on
3 mEq/L is reached. These interventions should GFR [197].
proceed with ongoing cardiac monitoring to The type of potassium preparation given for
detect new or exacerbating arrhythmia. supplementation can be tailored to the need for
Since intravenous potassium can be irritating other specific electrolyte supplementation. Since
to vessels, to prevent phlebitis or ongoing pain, many hypokalemic children have concomitant
concentrations exceeding 40 mEq/L are generally alkalosis with hypochloremia, potassium chloride
given centrally. When giving intravenous potas- is most frequently used for both enteral and intra-
sium, it is important for patient safety to limit the venous supplementation. Potassium chloride sup-
absolute amount of potassium available to be plements have also been shown to accomplish
delivered in one single infusion in case the rate more efficient repletion than potassium phospho-
or volume of the infusion is inadvertently altered rus or potassium citrate or bicarbonate, although
from what is intended. Hourly rates of infusion of with concomitant acidosis or hypophosphatemia,
0.5–1 mEq/kg will generally result in serum their provision does make sense [198].
potassium levels rising by nearly 0.5 mEq/L in Any supplementation will be less effective if
the face of no new increased potassium losses. there are ongoing potassium losses that can con-
tinue to vary independently. In children with
Enteral Potassium Supplementation Bartter or Gitelman syndromes, use of
In children with levels between 2.5 and 3 mEq/L potassium-sparing diuretics may help limit the
and no clinical signs or symptoms of hypokale- extent of renal losses, and in those children with
mia, potassium supplementation is warranted, such tubulopathies and hypomagnesemia, magne-
although enteral supplementation is generally pre- sium provision can help blunt the attenuating
ferred to avoid the complications of intravenous effects of hypomagnesemia on potassium
provision. Although intravenous supplementation reabsorption [199].
is commonly provided to critically ill children due
to restrictions in enteral intake or other
confounding factors, there is no evidence to sug- Acid–Base Homeostasis
gest that enteral supplementation is inferior to its
intravenous provision. In fact, a study of children Acid–base homeostasis involves an intricate inter-
in a pediatric cardiac intensive care unit showed play between multiple regulatory mechanisms to
that the provision of 1 mEq/kg of potassium either maintain the extracellular arterial pH between
by intravenous or enteral routes resulted in no 7.35 and 7.45. The typical diet for infants and
difference in efficacy or side effects, with both children in much of the developed world is rich
groups demonstrating an average improvement in animal proteins and cereal grain resulting in
in serum potassium of 0.65 mEq/L [196]. generation of net acid, as opposed to the genera-
tion of net base with vegetarian diets
Factors Influencing Efficacy [200–202]. When adjusted for body weight,
of Supplementation infants produce about 2–3 mEq/kg/day of nonvol-
Concomitant administration of other medications atile acid, which is significantly higher than adults
that may alter renal potassium handling may affect where the net nonvolatile acid production
408 I.F. Ashoor and M.J.G. Somers

system and 0.03 is the solubility constant of CO2


in the extracellular fluid.

Perturbations in Acid–Base Balance

Acid–base disorders result from processes that


lead to changes in the serum bicarbonate (HCO3)
concentration, partial pressure of carbon dioxide
(PCO2), or both. The disorder is considered met-
abolic when the inciting event primarily alters
HCO3 concentration and respiratory if changes
in PCO2 were primarily responsible for altering
systemic pH. Newborns and infants are more sus-
ceptible to develop acid–base disorders given
their baseline higher acid load and the time
required for full maturation of various transporters
involved in acid–base homeostasis in the proxi-
Fig. 2 Acid–base handling in the distal nephron
mal and distal nephron [205–208].
averages 50–100 mEq/day [203]. The additional
acid load in children is, in part, secondary to rapid
skeletal growth which also produces a net acid Simple Acid–Base Disorders
excess as calcium is incorporated into the skeleton
to form hydroxyapatite [204]. Under normal conditions, a metabolic acid–base
This daily acid load is handled by initial buffer- disorder secondary to a change in serum HCO3
ing with extracellular buffers (primarily the HCO3 concentration will be associated with a respiratory
buffer system) followed by intracellular buffers compensation that changes the PCO2 in the same
(proteins and phosphate). The respiratory system direction as the change in HCO3 concentration to
adjusts the ventilation rate to excrete volatile acids bring the pH towards normal. For example, a child
as CO2. The kidneys handle the excretion of non- with metabolic acidosis secondary to diarrheal
volatile acids by secreting H+ in the proximal HCO3 losses and resulting lowering serum
tubule, which reclaims filtered HCO3, and by HCO3 concentration will hyperventilate to
secreting H+ in the distal nephron, which com- decrease the PCO2 and raise the depressed sys-
plexes with phosphate to form titratable acid temic pH towards normal. On the other hand, a
(H2PO4) or with ammonia (NH3) to form ammo- child with alkalemia secondary to H+ losses from
nium (NH4) salts. The H+ secreted distally is vomiting leading to an increase in serum HCO3
derived from breakdown of water molecules in concentration will hypoventilate to increase the
the cell, which generates a hydroxyl ion (OH) PCO2 and lower the systemic pH towards normal.
that produces new HCO3 in a reaction with CO2 Similarly, a respiratory acid–base disorder sec-
catalyzed by carbonic anhydrase in α -intercalated ondary to a change in PCO2 will be associated
cells (Fig. 2). The relationship between pH, serum with a metabolic compensation that changes the
HCO3, and PCO2 can be expressed logarithmically HCO3 concentration in the same direction as the
as the Henderson–Hasselbalch equation as follows: change in PCO2 to bring the pH towards normal.
For example, an infant with respiratory acidosis
pH ¼ 6:1 þ Log ð½HCO3 =½0:03  PCO2 Þ
secondary to hypercarbia from a process such as
chronic lung disease of prematurity will increase
where 6.1 is the pKa (log of the dissociation net renal H+ secretion over time, thereby raising
constant for the reaction) of the CO2-HCO3 buffer the serum HCO3 concentration and the systemic
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 409

pH towards normal. On the other hand, a child disorder, compensation is accomplished by extra-
with respiratory alkalosis secondary to anxiety- cellular and intracellular buffering mechanisms.
induced hyperventilation lowering PCO2 will As detailed in Table 17, in acute respiratory
increase the fractional excretion of HCO3 in the acidosis, compensatory response is expected to
urine, thereby lowering serum HCO3 concentra- raise the serum HCO3 concentration by about
tion and lowering systemic pH towards normal. 1 mEq/L in the absence of coexisting acid–base
The degree by which the body compensates for disorders. In comparison, as full compensatory
a primary metabolic or respiratory process alter- responses develop in response to a chronic respi-
ing pH can be deduced from various formulas ratory acidosis, the serum HCO3 concentration
[209–212]. A simple acid–base disorder exists will rise by 4–5 mEq/L [212, 213]. Similarly, in
when the change in pH can be explained by a acute and chronic respiratory alkalosis (Table 18),
primary metabolic or respiratory disturbance and the serum HCO3 concentration is expected to fall
an appropriate degree of compensation. by about 2 and 4–5 mEq/L, respectively [213,
214]. The same underlying etiology may cause
respiratory acidosis or alkalosis depending on
Respiratory Acid–Base Disorders the chronicity, degree of ventilation–perfusion
mismatch, and respiratory muscle fatigue.
A respiratory acidosis exists when a low pH is
secondary to a process elevating arterial PCO2,
while respiratory alkalosis refers to an elevated Metabolic Acidosis
pH secondary to a process reducing arterial PCO2.
A compensatory metabolic response to a primary Metabolic acidosis refers to conditions where a
respiratory acid–base disorder will evolve over low systemic pH is secondary to a process
time; the full renal compensatory response decreasing HCO3 concentration. In children, the
requires up to 3–5 days to develop [213]. In the clinical presentation of metabolic acidosis is var-
initial stages of a primary respiratory acid–base ied and can range from a completely

Table 17 Representative etiologies of respiratory acidosis in children


Mechanism Acute Chronic
Airway obstruction Foreign body aspiration Bronchopulmonary dysplasia
Status asthmaticus Persistent asthma
Epiglottitis Obstructive sleep apnea
Bronchospasm/laryngospasm Hilar lymphadenopathy
Pulmonary – intrinsic Pneumonia Persistent asthma
Respiratory distress syndrome Cystic fibrosis
Pneumothorax/hemothorax Interstitial lung disease
Status asthmaticus Bronchopulmonary dysplasia
Pulmonary – extrinsic Erroneous ventilator settings Chest wall deformity
Diaphragmatic paralysis
Neurologic – central Head injury Congenital CNS malformation
Coma CNS tumor
Stroke Chronic sedative use
Sedative overdose Central sleep apnea
Meningitis
Spinal cord injury
Neurologic – peripheral Guillain–Barre syndrome Muscular dystrophies
Myasthenia gravis Chronic myopathies
Organophosphate poisoning Polio
Familial periodic paralysis
Botulism
410 I.F. Ashoor and M.J.G. Somers

Table 18 Representative etiologies of respiratory alkalo- Table 19 Representative etiologies for metabolic acidosis
sis in children in children
Mechanism Acute Chronic Classification Mechanism Examples
Direct central Head injury Congenital High anion Increased Diabetic
stimulation of the Anxiety CNS gap acidosis endogenous ketoacidosis
respiratory Hyperventilation malformation acid Lactic acidosis
center Meningitis/ CNS tumor production Inborn errors of
encephalitis Meningitis/ metabolism (e.g.,
Drug side effect: encephalitis organic acidemias)
aspirin, nicotine Medication Ingestions
CNS bleed side effect (methanol, ethanol,
Fever ethylene glycol)
Pain Accumulation Advanced renal
Indirect Pulmonary Heart failure of anionic failure
stimulation of the edema Interstitial compounds
respiratory Pulmonary lung disease Normal Loss of Diarrhea
center via embolus anion gap bicarbonate Type 2 (proximal)
pulmonary acidosis renal tubular
stretch receptors acidosis (RTA)
Indirect Hypotension High altitude Carbonic anhydrase
stimulation of the Tissue hypoxia (low FiO2%) inhibitors
respiratory (e.g., asthma, Impaired renal Type I (distal) RTA
center via pneumonia) H+ excretion Type 4 (distal
peripheral hyperkalemic) RTA
chemoreceptors Early CKD

asymptomatic child with incidental findings on increasing pH towards normal. In the absence of
laboratories obtained for another indication to a severe acidosis (pH < 7.1), Winters’ formula
critically ill child presenting with circulatory col- [210] can predict the expected PCO2 for a given
lapse. The history and physical examination find- primary change in HCO3 concentration as
ings may point towards the physiologic follows:
perturbation resulting in the metabolic acidosis.
Typically, metabolic acidosis is caused by condi- Expected PCO2 ¼ ð1:5  measured HCO3 Þ
tions that can be grouped into three broad catego- þ 8 ðþ=2Þ mmHg
ries: (1) increased endogenous or exogenous acid
generation which produces a high anion gap met- Alternatively, an appropriate respiratory response
abolic acidosis, (2) increased HCO3 loss which to metabolic acidosis results in a fall in PCO2
produces a normal anion gap metabolic acidosis, concentration by 1.2 mmHg for every 1 mEq/L
and (3) impaired renal H+ excretion which also fall in HCO3 concentration from a normal baseline
produces a normal anion gap metabolic acidosis of 24 mEq/L. This relationship tends to be accu-
(see below and Table 19). rate down to a PCO2 of 10–15 mmHg.

Respiratory Compensation Serum Anion Gap


for Metabolic Acidosis
The serum anion gap is a measurement obtained
Simple metabolic acidosis exists when there is an by subtracting the sum of the major measured
appropriate respiratory compensation for the serum anions (chloride and HCO3) from the
degree of reduction in HCO3 concentration. The major measured serum cation sodium [215] and
respiratory response to a fall in HCO3 concentra- is often expressed in the equation form: Serum
tion is hyperventilation, lowering PCO2, and AG = Na+  (Cl + HCO3). The serum anion
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 411

gap reflects the difference between unmeasured will be low (reflecting low urine NH4+) and the
anions and unmeasured cations in the blood. Most UAG calculation will yield a positive value.
children have an anion gap less than 16 mEq/L
[215]; however, it is prudent to review local lab-
oratory normal values for specific electrolyte Urine Osmolal Gap
ranges and, in children with an anion gap, to
monitor its trend over time to facilitate accurate The urine osmolal gap (UOG) is another surrogate
categorization of the acid–base disorder in ques- measure of urinary NH4 excretion. It is a better
tion (Table 3). measure of NH4 excretion when NH4 may be
Under normal acid–base status, the anion gap coupled with other anions besides Cl in the
is largely a result of anionic proteins, primarily urine. This can be seen in ketoacidosis and toluene
albumin. Therefore, the anion gap measurement inhalation where NH4 is excreted in conjunction
has to be adjusted for abnormal albumin concen- with keto acids and hippurate, respectively [218,
tration, where for every 1 g/dL reduction in serum 219]. The UOG can be calculated by subtracting
albumin the normal anion gap is expected to the urine-calculated osmolality from the directly
decrease by 2.5 mEq/L [216]. This adjustment measured urine osmolality as follows:
becomes particularly important to consider in
children with substantial aberrations in serum UOG ¼ urine measuredosmolality
albumin levels, as seen with nephrotic syndrome  urinecalculated osmolality ð2  ½Naþ K
or with malnutrition. þ ½urine ureanitrogen=2:8
þ ½urine glucose=18Þ

Urine Anion Gap Both urine urea nitrogen and urine glucose are
measured in mg/dL. The glucose contribution to
The urine anion gap (UAG) is a useful tool in the urine osmolality is only relevant in the setting of
evaluation of metabolic acidosis with a normal glucosuria and in its absence can be ignored.
serum anion gap and is often expressed by the Similar to the concept of the UAG, a high UOG
equation: UAG = Urine (Na + K) – Cl. It is a (typically greater than 400 mOsm/kg) indicates
surrogate measure of ammonium (NH4) excretion increased NH4 excretion by the distal nephron as
in the urine, as direct urinary NH4 measurement is would be seen in a child with diarrhea who should
not widely available [217]. NH4 is excreted as have intact distal tubular acid–base regulatory
NH4 chloride salts in the urine. Chloride is also mechanisms, whereas a low UOG (typically less
excreted along with sodium and potassium in the than 150 mOsm/kg) would indicate impaired NH4
urine. Therefore, subtracting the urine chloride excretion in the distal tubule as would be seen in a
from the sum of urine sodium and urine potassium child with distal RTA [220].
concentrations should be a rough estimate of NH4
concentration in the urine assuming that it is
exclusively excreted as NH4 chloride salts. Metabolic Alkalosis
The ability of the distal nephron to secrete the
excess H+ as NH4+ remains intact in normal AG Metabolic alkalosis refers to conditions where a high
metabolic acidosis secondary to gastrointestinal systemic pH is secondary to a process increasing
(e.g., diarrhea) or renal HCO3 losses (e.g., proxi- serum HCO3 concentration. Children with alkalosis
mal RTA). The urine Cl concentration will be may be asymptomatic or have signs and symptoms
high (reflecting high urine NH4+) and the UAG reflecting the underlying etiology of alkalosis such
calculation will yield a negative value. Whereas if as vomiting or reflecting other concomitant electro-
the normal AG metabolic acidosis is secondary to lyte abnormalities such as seizures and muscle
impaired renal H+ excretion (e.g., distal RTA and cramps from hypomagnesemia and hypokalemia as
early stages of CKD), the urine Cl concentration seen in Gitelman and Bartter syndrome, respectively.
412 I.F. Ashoor and M.J.G. Somers

Table 20 Representative etiologies for metabolic alkalo- Similarly, dehydration or any state of deceased
sis in children effective circulating volume maintains alkalosis
Mechanism Examples by enhancing proximal tubular HCO3
Excessive loss of H+: Vomiting reabsorption along with the sodium avidity caused
gastrointestinal Nasogastric suction by the ineffective volume, creating what is termed
Excessive loss of H+: Bartter syndrome a “contraction” alkalosis effect.
renal Gitelman syndrome
Primary mineralocorticoid Hypokalemia causes intracellular shift of H+ in
excess exchange for K+ to maintain electroneutrality,
Liddle syndrome effectively adding HCO3 to the ECF
Loop or thiazide diuretic use [229]. Hypochloremia typically accompanies vol-
Excessive gain of Alkali administration
ume depletion; however, it can independently
HCO3 Conversion of potential
bicarbonate precursors to maintain alkalosis by reducing HCO3 secretion
HCO3 (e.g., lactate, citrate, in type B intercalated cells in the cortical
acetate) collecting tubule mediated by the luminal
Intracellular shift of Hypokalemia Cl/HCO3 exchanger, pendrin [230].
H+ Glucose-induced alkalosis in
fasting
Finally, primary activation of the RAS system
Relative increase in Contraction alkalosis contributes to the maintenance of alkalosis by
HCO3 concentration increasing Na reabsorption in the distal nephron
in exchange for H+ and K+ secretion (Fig. 1).
The development of metabolic alkalosis The respiratory compensatory response for
requires a process that generates or adds new metabolic alkalosis is limited to increasing PCO2
HCO3 to the ECF. Most commonly, this is the by about 0.7 mmHg for every 1 mEq/L increase in
result of gastrointestinal H+ loss or excessive HCO3 concentration up to a maximum PCO2 of
renal H+ secretion, which effectively adds HCO3 about 55 mmHg [231, 232]. A higher PCO2 than
to the ECF. Alternatively, intracellular shifting of 55 mmHg in a child with metabolic alkalosis
H+, exogenous alkali administration, or contrac- implies concurrent respiratory acidosis.
tion of the ECF around a relatively fixed HCO3
amount (i.e., contraction alkalosis) will all gener-
ate a metabolic alkalosis [221] (Table 20). Mixed Acid–Base Disorders
The subsequent persistence or maintenance of
metabolic alkalosis will depend on factors A mixed acid–base disorder is typically suspected
impairing net renal HCO3 excretion [222]. Appro- on clinical grounds when the history or physical
priate excretion of excess HCO3 requires normal examination suggests that several physiologic
renal function [223], adequate intravascular vol- processes are perturbed that would alter systemic
ume status [224], normal serum K+ [225] and Cl pH in the same or opposite direction. For example,
concentrations [226], and normal activity of the a child with aspirin toxicity is likely to have a
renin–angiotensin–aldosterone (RAS) combination of both metabolic acidosis and respi-
system [227]. ratory alkalosis and therefore may have a near-
In clinical settings when one of these parame- normal pH. Whereas, a child with renal tubular
ters is altered, there is heightened risk for alkalo- acidosis who suffers from a cardiopulmonary
sis. For example, when a child with chronic arrest has a baseline normal anion gap metabolic
kidney disease stage 4 (GFR 15–30 ml/min/1.73 acidosis that now has a high anion gap metabolic
m2) receives bicarbonate containing intravenous acidosis and respiratory acidosis superimposed,
fluids in an attempt to prevent contrast-induced leading to severe acidemia out of proportion to
nephropathy [228], the low GFR will maintain the what would be expected from each individual
alkalosis that is generated longer compared to a component of his acidosis. Mixed acid–base dis-
child with normal GFR who would readily excrete orders should also be suspected when the
the excess alkali load. observed compensatory response to the primary
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 413

acid–base disturbance is less than expected or progressive respiratory distress and a chest radio-
exceeds what should be expected for compensa- graph reveals a right upper lobe infiltrate. A
tion alone. repeat ABG now demonstrates a pH of 7.2,
PCO2 of 42 mmHg, and HCO3 of 16 mEq/L.
The infant still has metabolic acidosis. How-
The D Anion Gap/D HCO3 Ratio ever, Winters’ formula predicts a compensatory
PCO2 of 30–34 mmHg. The PCO2 is much higher,
The Δ anion gap (change in AG from patient’s however, suggesting a concomitant respiratory
baseline)/Δ HCO3 (change in HCO3 from acidosis, likely due to hypoventilation in the set-
patient’s baseline) ratio is a useful tool to deter- ting of aspiration pneumonia. In this case, both
mine if a mixed acid–base disorder exists. A nor- acid–base disorders altered the pH in the same
mal ratio typically ranges between 1 and 1.6 direction, causing a change greater than expected
[233]. A ratio of >1.6 indicates that the HCO3 with either condition operating independently.
concentration did not fall as much as would be The infant’s pneumonia worsens and he is
expected from the increase in the AG. This can be placed on a ventilator. He remains intubated for
seen when metabolic acidosis coexists with met- a month and, after extubation, he still has an
abolic alkalosis. On the contrary, a ratio of <1 oxygen requirement. A blood gas is obtained and
indicates a coexisting normal AG metabolic aci- demonstrates a pH of 7.32, PCO2 of 60 mmHg,
dosis that lowered HCO3 concentration to a and HCO3 of 32 mEq/L.
greater degree than would be accounted for by The infant has now developed chronic lung
isolated high AG metabolic acidosis. disease as indicated by persistent O2 requirement
for >30 days. In such a clinical setting of chronic
respiratory acidosis, the expected full metabolic
Clinical Application and Approach compensation would be an increase in normal
serum HCO3 concentration by 4–5 mEq/L for
The following case explores the previously each 10 mmHg increase in PCO2 from the
discussed concepts in interpreting acid–base dis- expected normal baseline of 40 mmHg, and,
orders and underscores how, especially in ill chil- indeed, this child demonstrates such a compensa-
dren with chronic medical conditions, multiple tion. If the infant’s serum HCO3 concentration
physiologic perturbations can arise resulting in differed significantly from expected, this would
numerous potential acid–base abnormalities: raise the specter of a coexisting metabolic alkalo-
A newborn boy is delivered prematurely at sis for higher-than-expected HCO3 values or a
34 weeks of gestation secondary to preterm coexisting metabolic acidosis for lower-than-
labor. He does well other than requiring 0.5 expected HCO3.
L/min of oxygen by nasal cannula. An arterial The infant goes on to develop central line-
blood gas demonstrates a pH of 7.33, PCO2 of associated bloodstream infection and hypoten-
33 mmHg, and HCO3 of 17 mEq/L. sion with poor perfusion. His respiratory status
This infant has a simple metabolic acidosis is unchanged. An arterial blood gas demonstrates
(low pH and low HCO3) with appropriate a pH of 7.15, PCO2 of 60 mmHg, and HCO3 of
respiratory compensation as expected from 20 mEq/L. Serum chemistries demonstrate a Na of
Winters’ formula: expected PCO2 = 1.5  17 + 145 mEq/L, K of 4 mEq/L, Cl of 95 mEq/L, and
8+/2 = 33.5+/2 mmHg. The low HCO3 is HCO3 of 20 mEq/L.
most likely secondary to proximal renal tubular The infant now has his known chronic respira-
increased fractional excretion of HCO3, an tory acidosis from his baseline lung disease as
expected maturational finding at this well as a superimposed high anion gap
gestational age. (145 – [95 + 20] = 30) metabolic acidosis
The infant starts trials with oral feeds and likely secondary to lactic acidosis from
experiences an aspiration event. He develops hypoperfusion. The Δ anion gap/Δ HCO3 ratio
414 I.F. Ashoor and M.J.G. Somers

([30–12]/[30–20] = 1.8) is greater than 1.6, In alkalosis, potassium repletion should be


suggesting a coexisting metabolic process that accomplished with potassium chloride salts since
raised HCO3 concentration above what otherwise citrate or acetate preparations will generate addi-
would be expected, in this case the baby’s baseline tional HCO3. The use of the carbonic anhydrase
renal compensatory response for his chronic respi- inhibitor, acetazolamide, can be a useful transient
ratory acidosis. adjunctive treatment in selected patients with
sustained metabolic alkalosis [234]. Acetazol-
amide inhibits proximal reabsorption of
General Considerations for Therapy of NaHCO3 and leads to renal bicarbonate wasting.
Acid–Base Disorders The increased Na+delivery to the distal tubule that
ensues can stimulate distal K+ secretion, however,
The most effective therapy for any acid–base dis- and potentially induce or exacerbate a preexisting
order is treatment of the underlying cause or hypokalemia which could then maintain the alka-
causes of the disturbance and correction of any losis. The use of exogenous acid to treat metabolic
other factors that are helping to maintain the alkalosis in children is rarely required and can be
abnormality. The decision to provide treatment associated with significant morbidity and mortal-
with exogenous alkali or acid to return the arterial ity if not approached carefully and in the hands of
pH to normal while awaiting response to therapy experienced clinicians [235, 236].
generally depends on individual clinical circum-
stance such as disease severity, the duration of the Special Considerations in Infants
acid–base abnormality, and whether the underly- and Children with Chronic Kidney
ing perturbation can be effectively reversed with Disease
therapy. Children with CKD suffer from metabolic acido-
For example, a child with chronic respiratory sis initially secondary to reduced distal H+ excre-
acidosis from bronchopulmonary dysplasia who tion and later due to accumulation of anions such
has normal kidneys will typically develop suffi- as phosphate, sulfate, and urate from reduced
cient renal metabolic compensation to re-establish GFR. As the HCO3 buffer systems become
a near-normal pH, and the only treatment required overwhelmed, excess H+ is buffered by bone,
is ongoing optimization of the primary respiratory contributing to bone resorption and CKD-related
condition, whereas a child with metabolic acidosis metabolic bone disease [237, 238]. Persistent met-
from advanced chronic kidney disease will abolic acidosis can also lead to impaired growth in
require exogenous alkali therapy as the underly- children outside of the deleterious effects of renal
ing cause is irreversible and respiratory compen- osteodystrophy [239]. Various mechanisms have
sation for metabolic acid–base disturbances is been implicated including blunted growth hor-
limited [209]. mone (GH) release, inhibition of pulsatile GH
In treating metabolic alkalosis, interrupting the secretion, and resistance to insulin-like growth
mechanisms responsible for maintaining the alka- factor-1 (IGF-1) action in the growth plate
losis is just as important as targeting the underly- [239–241]. Correction of metabolic acidosis is a
ing cause that generated the alkalosis. In children prerequisite for effective response to recombinant
with normal renal function, reduced renal excre- GH therapy in children with CKD and growth
tion of HCO3 which maintains an alkalosis is failure.
typically a result of decreased effective circulating The 2013 Kidney Disease Improving Global
volume, hypokalemia, chloride depletion, or Outcomes (KDIGO) guidelines recommend
excess mineralocorticoid activity [222]. Effective treating metabolic acidosis in CKD with some
therapy entails attention to volume expansion, sort of alkali therapy to maintain serum HCO3
chloride and potassium replacement, and possible concentration in the normal range [242]. Treat-
pharmacologic inhibition of the RAS system if ment can be provided as sodium bicarbonate or
indicated. sodium citrate, starting at 1 mEq/kg/day and
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 415

titrated to achieve target serum HCO3 concentra- 10. Gavin N, Merrick N, Davidson B. Efficacy of
tion. Newborns and infants will require higher glucose-based oral rehydration therapy. Pediatrics.
1996;98:45–51.
alkali doses than older children and adolescents 11. Schwartz R. Comments from another student of Gam-
with the same degree of renal dysfunction due to ble and Darrow on fluids. Pediatrics. 1996;98:314.
their relatively higher acid generation and reduced 12. Cumlea WC, Schubert CM, Sun SS, et al. A review of
acid excretion ability. body water status and the effects of age and body
fatness in children and adults. J Nutr Health Aging.
Finally, for patients with end-stage renal dis- 2007;11:111–8.
ease (ESRD) receiving renal replacement therapy, 13. Yoshioka T, Iitaka K, Ichikawa I. Body fluid compart-
metabolic acidosis typically improves with the ments. In: Ichikawa I, editor. Pediatric textbook of
additional base load delivered in the dialysate. fluids and electrolytes. Baltimore: Williams and Wil-
kins; 1990. p. 14–20.
Persistent acidosis pre-dialysis may be an indica- 14. Ritchie RF, Ledue TB, Craig WY. Patient hydration: a
tion of insufficient dialysis dose, higher net acid major source of laboratory uncertainty. Clin Chem
generation secondary to high dietary animal pro- Lab Med. 2007;45:158–66.
tein intake [243], or some other coexisting etiol- 15. Rose BD, Rennke HG. Regulation of salt and water
balance. In: Rennke HG, Denker BM, editors. Renal
ogy for metabolic acidosis. On the contrary, a Pathophysiology – the essentials. Philadelphia:
normal or high serum HCO3 concentration Lippincott Williams and Wilkins; 2006. p. 31–69.
pre-dialysis may indicate adequate dialysis, 16. Talbot NB, Crawford JD, Butler AM. Homeostatic
decreased dietary protein intake, or a coexisting limits to safe parenteral fluid therapy. N Eng J Med.
1953;248:1100–8.
metabolic alkalosis. Correction of abnormal 17. Al F, Ray PE. Maintenance fluid therapy: what it is
pre-dialysis serum HCO3 abnormalities entails and what it is not. Pediatr Nephrol. 2008;23:677–80.
addressing the underlying cause and adjusting 18. Murat I, Dubois MC. Perioperative fluid therapy in
the dialysate bicarbonate bath pediatrics. Pediatr Anesth. 2008;18:363–70.
19. Choong K, Bohn D. Maintenance parenteral fluids in
concentration [244]. the critically ill child. J Pediatr (Rio J). 2007;83
(2 suppl):S3–10.
20. Rasouli M, Kalantari KR. Comparison of methods for
calculating serum osmolality: multivariate linear
References regression analysis. Clin Chem Lab Med.
2005;43:635–40.
1. Cosnett JE. The origins of intravenous therapy. Lan- 21. Trachtman H. Sodium and water homeostasis. Pediatr
cet. 1989;1:768–71. Clin North Am. 1995;42:1343–63.
2. Blackfan KD, Maxcy KF. Intraperitoneal injection of 22. Rose BD, Rennke HG. Disorders of water balance:
saline. Am J Dis Child. 1918;15:19–28. hyponatremia, hypernatremia, and polyuria. In:
3. Karelitz S, Schick B. Treatment of toxicosis with the Rennke HG, Denker BM, editors. Renal pathophysi-
aid of a continuous intravenous drip of dextrose solu- ology – the essentials. Philadelphia: Lippincott Wil-
tion. Am J Dis Child. 1932;42:781–802. liams and Wilkins; 2006. p. 69–99.
4. Gamble JL. Early history of fluid replacement ther- 23. Gerigk M, Gnehm HE, Rascher W. Arginine vaso-
apy. Pediatrics. 1953;11:554–67. pressin and renin in acutely ill children: implication
5. Holliday MA. Gamble and Darrow: pathfinders in for fluid therapy. Acta Paediatr. 1996;85:550–3.
body fluid physiology and fluid therapy for children, 24. Halberthal M, Halperin ML, Bohn D. Lesson of the
1914–1964. Pediatr Nephrol. 2000;15:317–24. week: acute hyponatraemia in children admitted to
6. Holliday MA, Segar WE. The maintenance need for hospital: retrospective analysis of factors contributing
water in parenteral fluid therapy. Pediatrics. to its development and resolution. BMJ.
1957;19:823–32. 2001;322:780–2.
7. Holliday MA. Body fluid physiology during growth, 25. Moritz ML, Ayus JC. Prevention of hospital acquired
chapter 13. In: Maxwell MH, Kleeman CR, editors. hyponatremia: a case for using isotonic saline in
Clinical disorders of fluid and electrolyte metabolism. maintenance fluid therapy. Pediatrics.
2nd ed. New York: McGraw-Hill; 1972. 2003;111:227–30.
8. Holliday MA, Ray PE, Friedman AL. Fluid therapy in 26. Holliday MA, Segar WE, Friedman A. Reducing
children: facts, fashions, and questions. Arch Dis errors in fluid therapy management. Pediatrics.
Child. 2007;92:546–50. 2003;111:424–5.
9. Holliday M. The evolution of therapy for dehydra- 27. Franz MN, Segar WE. The association of various
tion: should deficit therapy still be taught? Pediatrics. factors with hypernatremic diarrheal dehydration.
1996;98:171–7. Am J Dis Child. 1972;97:298–302.
416 I.F. Ashoor and M.J.G. Somers

28. Moritz ML, Ayus JC. Preventing neurological com- nephrogenic diabetes insipidus caused by an
plications from dysnatremias in children. Pediatr aquaporin-2 mutation. J Clin Endocrinol Metab.
Nephrol. 2005;20:1687–700. 1997;82(2):686–9.
29. Holliday MA, Friedman AL, We S, et al. Acute 45. Loonen AJ, Knoers NV, van Os CH, Deen
hospital-induced hyponatremia in children: a physio- PM. Aquaporin 2 mutations in nephrogenic diabetes
logic approach. J Pediatr. 2004;145:584–7. insipidus. Semin Nephrol. 2008;28(3):252–65.
30. Horn EJ, Geary D, Robb M, et al. Acute hyponatremia 46. Rosen S, Greenfeld Z, Bernheim J, Rathaus M,
related to IV fluid administration in hospitalized chil- Podjarny E, Brezis M. Hypercalcemic nephropathy:
dren: an observational study. Pediatrics. chronic disease with predominant medullary inner
2004;113:1279–84. stripe injury. Kidney Int. 1990;37(4):1067–75.
31. Au AK, Ray PE, McBryde KD, et al. Incidence of 47. Marples D, Frokiaer J, Dorup J, Knepper MA, Niel-
postoperative hyponatremia and complications in cru- sen S. Hypokalemia-induced downregulation of
cially ill children treated with hypotonic and aquaporin-2 water channel expression in rat kidney
normotonic solutions. J Pediatr. 2008;152:33–8. medulla and cortex. J Clin Invest. 1996;97
32. Neville KA, Verge CF, Rosenberg AR, et al. Isotonic (8):1960–8.
is better than hypotonic saline for intravenous rehy- 48. Berl T, Linas SL, Aisenbrey GA, Anderson RJ. On the
dration of children with gastroenteritis: a prospective mechanism of polyuria in potassium depletion. The
randomized study. Arch Dis Child. 2006;91:226–32. role of polydipsia. J Clin Invest. 1977;60(3):620–5.
33. Choong K, Kho ME, Menon K, et al. Hypotonic ver- 49. Garofeanu CG, Weir M, Rosas-Arellano MP,
sus isotonic saline in hospitalized children: a system- Henson G, Garg AX, Clark WF. Causes of reversible
atic review. Arch Dis Child. 2006;91:828–35. nephrogenic diabetes insipidus: a systematic review.
34. Way C, Dhamrait R, Wade A, et al. Perioperative fluid Am J Kidney Dis. 2005;45(4):626–37.
therapy in children: a survey of current prescribing 50. Grunfeld JP, Rossier BC. Lithium nephrotoxicity
practices. Br J Anaesth. 2006;97:371–9. revisited. Nat Rev Nephrol. 2009;5(5):270–6.
35. Peters M, Jeck N, Reinalter S, Leonhardt A, 51. Schliefer K, Rockstroh JK, Spengler U, Sauerbruch
Tonshoff B, Klaus GG, et al. Clinical presentation of T. Nephrogenic diabetes insipidus in a patient taking
genetically defined patients with hypokalemic salt- cidofovir. Lancet. 1997;350(9075):413–4.
losing tubulopathies. Am J Med. 2002;112 52. Schrier RW, Gross P, Gheorghiade M, Berl T, Verbalis
(3):183–90. JG, Czerwiec FS, et al. Tolvaptan, a selective oral
36. van Lieburg AF, Knoers NV, Monnens LA. Clinical vasopressin V2-receptor antagonist, for
presentation and follow-up of 30 patients with con- hyponatremia. N Engl J Med. 2006;355
genital nephrogenic diabetes insipidus. J Am Soc (20):2099–112.
Nephrol. 1999;10(9):1958–64. 53. Wesche D, Deen PM, Knoers NV. Congenital
37. Bockenhauer D, Bichet DG. Inherited secondary nephrogenic diabetes insipidus: the current state of
nephrogenic diabetes insipidus: concentrating on affairs. Pediatr Nephrol. 2012;27(12):2183–204.
humans. Am J Physiol Renal Physiol. 2013;304(8): 54. Earley LE, Orloff J. The mechanism of antidiuresis
F1037–42. associated with the administration of hydrochlorothi-
38. Moses AM, Clayton B. Impairment of osmotically azide to patients with vasopressin-resistant diabetes
stimulated AVP release in patients with primary poly- insipidus. J Clin Invest. 1962;41(11):1988–97.
dipsia. Am J Physiol. 1993;265(6 Pt 2):R1247–52. 55. Libber S, Harrison H, Spector D. Treatment of
39. Nielsen S, Kwon TH, Christensen BM, Promeneur D, nephrogenic diabetes insipidus with prostaglandin
Frokiaer J, Marples D. Physiology and pathophysiol- synthesis inhibitors. J Pediatr. 1986;108(2):305–11.
ogy of renal aquaporins. J Am Soc Nephrol. 1999;10 56. Berl T, Raz A, Wald H, Horowitz J, Czaczkes
(3):647–63. W. Prostaglandin synthesis inhibition and the action
40. Deen PM, Verdijk MA, Knoers NV, Wieringa B, of vasopressin: studies in man and rat. Am J Physiol.
Monnens LA, van Os CH, et al. Requirement of 1977;232(6):F529–37.
human renal water channel aquaporin-2 for 57. Stasior DS, Kikeri D, Duel B, Seifter JL. Nephrogenic
vasopressin-dependent concentration of urine. Sci- diabetes insipidus responsive to indomethacin plus
ence. 1994;264(5155):92–5. dDAVP. N Engl J Med. 1991;324(12):850–1.
41. Kokko JP. The role of the collecting duct in urinary 58. Fine LG, Schlondorff D, Trizna W, Gilbert RM,
concentration. Kidney Int. 1987;31(2):606–10. Bricker NS. Functional profile of the isolated uremic
42. Fujiwara TM, Bichet DG. Molecular biology of nephron. Impaired water permeability and adenylate
hereditary diabetes insipidus. J Am Soc Nephrol. cyclase responsiveness of the cortical collecting
2005;16(10):2836–46. tubule to vasopressin. J Clin Invest. 1978;61(6):
43. Bichet DG. Vasopressin receptor mutations in 1519–27.
nephrogenic diabetes insipidus. Semin Nephrol. 59. Tannen RL, Regal EM, Dunn MJ, Schrier
2008;28(3):245–51. RW. Vasopressin-resistant hyposthenuria in advanced
44. Hochberg Z, Van Lieburg A, Even L, Brenner B, chronic renal disease. N Engl J Med. 1969;280(21):
Lanir N, Van Oost BA, et al. Autosomal recessive 1135–41.
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 417

60. Kleeman CR, Adams DA, Maxwell MH. An evalua- 75. Conley SB. Hypernatremia. Pediatr Clin North
tion of maximal water diuresis in chronic renal dis- Am. 1990;37:365–72.
ease. I. Normal solute intake. J Lab Clin Med. 76. Robertson GL. Physiology of ADH secretion. Kidney
1961;58:169–84. Int. 1987;32 suppl 21:S20–6.
61. Teitelbaum I, McGuinness S. Vasopressin resistance 77. Yang CW, Kim YS, Park IS, Chang YS, Yoon YS,
in chronic renal failure. Evidence for the role of Bang BK. Treatment of severe acute hypernatremia
decreased V2 receptor mRNA. J Clin Invest. and renal failure by hemodialysis. Nephron.
1995;96(1):378–85. 1995;70:372–3.
62. da Silva Jr GB, Liborio AB, Daher Ede F. New 78. Pazmino PA, Pazmino BP. Treatment of acute
insights on pathophysiology, clinical manifestations, hypernatremia with hemodialysis. Am J Nephrol.
diagnosis, and treatment of sickle cell nephropathy. 1993;13:260–5.
Ann Hematol. 2011;90(12):1371–9. 79. Molteni KH. Initial management of hypernatremic
63. Hildebrandt F, Attanasio M, Otto E. dehydration in the breastfed infant. Clin Pediatr.
Nephronophthisis: disease mechanisms of a 1994;33:731–40.
ciliopathy. J Am Soc Nephrol. 2009;20(1):23–35. 80. Carr M, Gill D. Polyuria, polydipsia, polyposia:
64. Scolari F, Caridi G, Rampoldi L, Tardanico R, Izzi C, “Mummy I want a drink”. Arch Dis Child Educ Pact
Pirulli D, et al. Uromodulin storage diseases: clinical Ed. 2007;92:ep139–43.
aspects and mechanisms. Am J Kidney Dis. 2004; 81. Robertson GL. Abnormalities of thirst regulation.
44(6):987–99. Kidney Int. 1984;25:460–9.
65. Jeck N, Schlingmann KP, Reinalter SC, Komhoff M, 82. Murphy MS. Guidelines for managing acute gastro-
Peters M, Waldegger S, et al. Salt handling in the enteritis based on a systematic review of published
distal nephron: lessons learned from inherited research. Arch Dis Child. 1998;79:279–84.
human disorders. Am J Physiol Regul Integr Comp 83. Saavedra JM, Harris GD, Li S, Finberg L.
Physiol. 2005;288(4):R782–95. Capillary refilling (skin turgor) in the
66. Bockenhauer D, van’t Hoff W, Dattani M, assessment of dehydration. Am J Dis Child.
Lehnhardt A, Subtirelu M, Hildebrandt F, 1991;145:296–8.
et al. Secondary nephrogenic diabetes insipidus as a 84. Gorelick MS, Shaw KN, Baker MD. Effects of ambi-
complication of inherited renal diseases. Nephron ent temperature on capillary refill in children. Pediat-
Physiol. 2010;116(4):23–9. rics. 1993;92:699–702.
67. Bogdanovic R, Stajic N, Putnik J, Paripovic 85. Baraff LJ. Capillary refill: is it a useful clinical sign?
A. Transient type 1 pseudo-hypoaldosteronism: Pediatrics. 1993;92:723–4.
report on an eight-patient series and literature review. 86. Duggan C, Refat M, Hashem M, Wolff M, Fayad I,
Pediatr Nephrol. 2009;24(11):2167–75. Santosham M. How valid are clinical signs of dehy-
68. Rodriguez-Soriano J, Vallo A, Oliveros R, Castillo dration in infants? J Pediatr Gastroenterol Nutr.
G. Transient pseudohypoaldosteronism secondary to 1996;22:56–61.
obstructive uropathy in infancy. J Pediatr. 1983;103 87. Mackenzie A, Barnes G, Shann F. Clinical signs of
(3):375–80. dehydration in children. Lancet. 1989;2:605–7.
69. Harris RH, Yarger WE. The pathogenesis of post- 88. Vega RM, Avner JR. A prospective study of the
obstructive diuresis. The role of circulating natriuretic usefulness of clinical and laboratory parameters for
and diuretic factors, including urea. J Clin Invest. predicting percentage of dehydration in children.
1975;56(4):880–7. Pediatr Emerg Care. 1997;13:179–82.
70. Choi M, Szerlip HM. Sodium and volume homeosta- 89. Teach SJ, Yates EW, Feld LG. Laboratory predictors
sis. In: Szerlip HM, Goldfarb S, editors. Workshops in of fluid deficit in acutely dehydrated children. Clinical
fluid and electrolyte disorders. New York: Churchill Pediatr (Phila). 1997;36:395–400.
Livingstone; 1993. p. 1–24. 90. Yilmaz K, Karabocuoglu M, Citak A, Uzel
71. Pasantes-Morales H, Lezama RA, Ramos- N. Evaluation of laboratory tests in dehydrated chil-
Mandujano G, et al. Mechanisms of cell volume reg- dren with acute gastroenteritis. J Paediatr Child
ulation in hypo-osmolality. Am J Med. 2006;119: Health. 2002;38:226–8.
S4–11. 91. Conway SP, Phillips RR, Panday S. Admission to
72. Strange K. Cellular volume homeostasis. Adv Physiol hospital with gastroenteritis. Arch Dis Child.
Educ. 2004;28:155–9. 1990;65:579–84.
73. DePetris L, Luchetti A, Emma F. Cell volume regula- 92. Jenkins HR, Ansari BM. Management of gastroenter-
tion and transport mechanisms across the blood–brain itis. Arch Dis Child. 1990;65:939–41.
barrier: implications for the management of 93. Ellis ME, Watson B, Mandal BK, Dunbar EM,
hypernatremic states. Eur J Pediatr. 2001;160:71–7. Mokashi A. Contemporary gastroenteritis of infancy:
74. Rose BD. Hypoosmolal states – hyponatremia. In: clinical features and prehospital management. BMJ.
Rose BD, Post TW, editors. Clinical physiology of 1984;288:521–3.
acid–base and electrolyte disorders. 5th ed. New York: 94. Armon K, Stephenson T, McFaul R, Eccleston P,
McGraw-Hill; 2001. p. 696–745. Werneke U. An evidence and consensus based
418 I.F. Ashoor and M.J.G. Somers

guideline for acute diarrhoea management. Arch Dis Antoine M, Mackenzie K, Johnson B. Comparison
Child. 2001;85:132–41. of low and high sodium and potassium content
95. American Academy of Pediatrics. Provisional Com- in oral rehydration solutions. J Pediatr.
mittee on Quality Improvement Subcommittee on 1980;97:848–53.
Acute Gastroenteritis. Practice parameter: the man- 111. Leung AKC, Taylor PG, Geoffroy L, Darling
agement of acute gastroenteritis in young children. P. Efficacy and safety of two oral solutions as main-
Pediatrics. 1996;97:424–36. tenance therapy for acute diarrhea. Clin Pediatr.
96. US Department of Health and Human Services. The 1988;27:359–64.
management of acute diarrhea in children: oral rehy- 112. Hahn S, Kim S, Garner P. Reduced osmolarity oral
dration, maintenance, and nutritional therapy. rehydration solution for treating dehydration caused
MMWR. 1992;41:1–20. by acute diarrhoea in children. Cochrane Database
97. Pierce NF, Sack RB, Mitra R, Banwell JG, Brigham Syst Rev. 2002;1:CD002847.
KL, Fedson DS, Mondal A. Replacement of electro- 113. Feld LG, Kaskel FJ, Schoeneman MJ. The approach
lyte and water losses in cholera by an oral glucose- to fluid and electrolyte therapy in pediatrics. Adv
electrolyte solution. Ann Intern Med. Pediatr. 1988;35:497–536.
1969;70:1173–81. 114. Pizarro D, Posada G, Levine MM. Hypernatremic
98. Mahalanbais D, Wallace CK, Kallen RJ, Mandel A, diarrheal dehydration treated with “slow” (12-hour)
Pierce NF. Water and electrolyte losses due to cholera oral rehydration therapy: a preliminary report.
in small children and infants: a recovery balance J Pediatr. 1984;104:316–9.
study. Pediatrics. 1970;45:374–85. 115. Pizarro D, Posada G, Villavicencio N, Mohs E,
99. Pizzaro D, Posada G, Mata L, Nalin D, Mohs E. Oral Levine MM. Oral rehydration in hypernatremic and
rehydration of neonates with dehydrating diarrhoea. hyponatremic diarrheal dehydration. Am J Dis Child.
Lancet. 1979;2:1209–10. 1983;137:730–4.
100. American Academy of Pediatrics Committee on 116. Pizarro D, Posada G, Levine MM, Mohs E. Oral
Nutrition. Use of oral fluid therapy and posttreatment rehydration of infants with acute diarrhoeal dehydra-
feeding following enteritis in children in a developed tion: a practical method. J Trop Med Hyg.
country. Pediatrics. 1985;75:358–61. 1980;83:241–5.
101. Santosham M, Greenough WB. Oral rehydration ther- 117. Wittenberg DF, Ramji S. Paediatric diarrhoea – rehy-
apy: a global perspective. J Pediatr. 1991;118: dration therapy revisited. Ann Afr Med
S44–51. J. 1995;85:655–8.
102. World Health Organization. Treatment and preven- 118. American Academy of Pediatrics. Managing acute
tion of dehydration in diarrheal disease. A guide for gastroenteritis among children: oral rehydration,
use at the primary level. Geneva: WHO; 1976. maintenance, and nutritional therapy. Pediatrics.
103. World Health Organization. Cholera: can rehydration 2004;114:507.
therapy be improved? WHO Drug Inf. 2000;14:88. 119. King CK, Glass R, Bressee JS, The Centers for Dis-
104. Snyder JD. Use and misuse of oral therapy for diar- ease Control and Prevention, et al. Managing acute
rhea: comparisons of US practices with American gastroenteritis among children: oral rehydration,
Academy of Pediatrics recommendations. Pediatrics. maintenance, and nutritional therapy. MMWR.
1991;87:28–33. 2003;52(RR-16):1–16.
105. Santosham M, Daum RS, Dillman L, et al. Oral rehy- 120. Avery ME, Snyder JD. Oral therapy for acute diar-
dration therapy of infantile diarrhea: a controlled rhea: the underused simple solution. N Eng J Med.
study of well nourished children hospitalized in the 1990;323:891–4.
United States and Panama. N Eng J Med. 121. Tanzi VL. An econometric study of the adoption of
1986;306:1070–6. oral rehydration solution in Bangladesh. Diss Abstr
106. Finberg L. The role of oral electrolyte-glucose solu- Int. 1999;60:1563.
tions in hydration for children – international and 122. Chowdhury AM, Karim F, Sarkar SK, Cash RA,
domestic aspects. Pediatrics. 1980;96:51–4. Bhuiya A. The status of ORT (oral rehydration ther-
107. Nalin DR, Cash RA, Islam R, Molla M, Phillips apy) in Bangladesh: how widely is it used? Health
RA. Oral maintenance therapy for cholera in adults. Policy Plan. 1997;12:58–66.
Lancet. 1968;2:370–2. 123. Bender BJ, Ozuah PO, Crain EF. Oral rehydration
108. Hirschhorn N, Kinzie JL, Sachar D, Northrup RS, therapy: is anyone drinking? Pediatr Emerg Care.
Taylor JO, Ahmad SZ, Phillips RA. Decrease in net 2007;23:624–6.
stool output in cholera during intestinal perfusion 124. Santucci KA, Anderson AC, Lewander WJ, Linakis
with glucose containing solutions. N Eng J Med. JG. Frozen oral hydration as an alternative to conven-
1968;279:176–81. tional enteral fluids. Arch Pediatr Adolesc Med.
109. Phillips RA. Water and electrolyte losses in cholera. 1998;152:142–6.
Fed Proc. 1964;23:705–12. 125. Ladinsky M, Duggan A, Santosham M, Wilson
110. Nalin DR, Harland E, Ramlal A, Swaby D, M. The World Health Organization oral rehydration
McDonald J, Gangarosa R, Levine M, Akierman A, solution in US pediatric practice: a randomized trial to
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 419

evaluate parent satisfaction. Arch Pediatr Adolesc glucose by APA and ADARPEF members: a postal
Med. 2000;154:700–5. study. Paediatr Anaesth. 2001;11:685–9.
126. Provisional Committee on Quality Improvement, 143. Hirschhorn N. The treatment of acute diarrhea in
Subcommittee on Acute Gastroenteritis. Practice children: an historical and physiological perspective.
parameter: the management of acute gastroenteritis Am J Clin Nutr. 1980;33:637–63.
in young children. Pediatrics. 1996;97:424–36. 144. Skellett S, Mayer A, Durward A, Tibby SM, Murdoch
127. Rabbani GH. The search for a better oral rehydration IA. Chasing the base deficit: hyperchloraemic acido-
solution for cholera. N Eng J Med. 2000;342:345–7. sis following 0.9 % saline fluid resuscitation. Arch
128. Fontaine O, Gore SM, Pierce NF. Rice-based oral Dis Child. 2000;83:514–6.
rehydration solution for treating diarrhoea. Cochrane 145. Neville KA, Verge CF, Rosenberg AR, et al. Is
Database Syst Rev. 2000;2:CD000567. hyperchloraemic acidosis a problem in children with
129. International Study Group on Reduced-Osmolarity gastroenteritis rehydrated with normal saline?
ORS Solutions. Multicentre evaluation of reduced- Authors’ reply. Arch Dis Child. 2007;92:466.
osmolarity oral rehydration salts solution. Lancet. 146. Durward A, Tibby SM, Skellett S, et al. The strong
1995;345:282–5. ion gap predicts mortality in children following car-
130. Hahn S, Kim Y, Garner P. Reduced osmolarity oral diopulmonary bypass surgery. Pediatr Crit Care Med.
rehydration solution for treating dehydration due to 2005;6:281–5.
diarrhoea in children: systematic review. BMJ. 147. Reid SR, Bonadio WA. Outpatient rapid intravenous
2001;323:81–5. rehydration to correct dehydration and resolve
131. Brierley J, Carcillo J, Choong K, et al. Clinical prac- vomiting in children with acute gastroenteritis. Ann
tice parameter for hemodynamic support of pediatric Emerg Med. 1996;28:318–23.
and neonatal shock: 2007 update from the American 148. Holliday MA, Friedman AL, Wassner
College of Critical Care Medicine. Crit Care Med. SJ. Extracellular fluid restoration in dehydration: a
2009;37:666–88. critique of rapid versus slow. Pediatr Nephrol.
132. Chappell D, Jacob M, Hoffman-Kiefer K, et al. A 1999;13:292–7.
rational approach to perioperative fluid management. 149. Oh MS, Kim H-J, Carroll HJ. Recommendations for
Anesthesiology. 2008;109:723–40. treatment of symptomatic hyponatremia. Nephron.
133. Awazu M, Devarajan P, Stewart CL, Kaskel F, Ichi- 1995;70:143–50.
kawa I. “Maintenance” therapy and treatment of 150. Pradhan S, Jha R, Singh MN, Gupta S, Phadke RV,
dehydration and overhydration. In: Ichikawa I, editor. Kher V. Central pontine myelinolysis following
Pediatric textbook of fluids and electrolytes. Balti- “slow” correction of hyponatremia. Clin Neurol
more: Williams and Wilkins; 1990. p. 417–28. Neurosurg. 1995;97:340–3.
134. Tullis JL. Albumin. 2. Guidelines for clinical uses. 151. Faber MD, Kupin WL, Heilig CW, Narins
JAMA. 1977;237:460–3. RG. Common fluid-electrolyte and acid–base prob-
135. Alderson P, Schierhout G, Roberts I, Bunn F. Colloids lems in the intensive care unit: selected issues. Semin
versus crystalloids for fluid resuscitation in critically Nephrol. 1994;14:8–22.
ill patients. Cochrane Database Syst Rev. 2000;2: 152. Mehta RL. Fluid balance and acute kidney injury: The
CD000567. missing link for predicting adverse outcomes. Nat
136. Bisonni RS, Holtgrave DR, Lawler F, Marley Clin Pract Nephrol. 2009;5:10–1.
DS. Colloid versus crystalloids in fluid resuscitation: 153. Miller NI, Finberg L. Peritoneal dialysis for salt poi-
an analysis of randomized controlled trials. J Fam soning. N Eng J Med. 1960;263:1347–50.
Pract. 1991;32:387–90. 154. Finberg L, Kiley J, Luttrell CN. Mass
137. The Cochrane Injuries Group Albumin Reviewers. accidental salt poisoning in infancy. JAMA.
Human albumin administration in critically ill 1963;184:187–90.
patients. BMJ. 1998;317:235–40. 155. el-Dahr S, Goez RA, Campbell FG, et al. Rapid cor-
138. Soni N. Wonderful albumin? BMJ. 1995;310:887–8. rection of acute salt poisoning by peritoneal dialysis.
139. Fleck A, Raines G, Hawker F, Trotter J, Wallace P, Pediatr Nephrol. 1987;1:602–4.
Ledingham I, Calman KC. Increased vascular perme- 156. Baer DM, Ernst DJ, Willeford SI, Gambino
ability: a major cause of hypoalbuminaemia in disease R. Investigating elevated potassium values. MLO
and injury. Lancet. 1985;1:781–4. Med Lab Obs. 2006;38:24.
140. Nichani S. Albumin: saint or sinner? Arch Dis Child. 157. Dickinson H, Webb NJA, Chaloner C, Wynn RF,
1999;81:189. Bonney DK. Pseuohypokalemia associated with leu-
141. Carcillo JA, Fields AI, American College of Critical kemic cell lysis during pneumatic tube transport of
Care Medicine Task Force Committee Members. blood samples. Pediatr Nephrol. 2012;6:1029–31.
Clinical practice parameters for hemodynamic sup- 158. Krapf R, Caduff P, Wagdi P, Staubli M, Hulter
port of pediatric and neonatal patients in septic shock. HN. Plasma potassium response to acute respiratory
Crit Care Med. 2002;30:1365–78. alkalosis. Kidney Int. 1995;47:217.
142. Soderlind M, Salvignol G, Izard P, Lonnqvist PA. Use 159. John SK, Rangan Y, Block CA, Koff MD. Life-
of albumin, blood transfusion and intraoperative threatening hyperkalemia from nutritional
420 I.F. Ashoor and M.J.G. Somers

supplements: uncommon or undiagnosed? Am J sodium and potassium estimation in a pediatric inten-


Emerg Med. 2011;29:1237.e1. sive care unit. Indian J Pediatr. 2013;80(9):731–5.
160. Sandle GI, Hunter M. Apical potassium 176. Cummings BM, Macklin EA, Yager PH, Sharma A,
(BK) channels and enhanced potassium secretion in Noviski N. Potassium abnormalities in a pediatric
human colons. Q J Med. 2010;103:85–9. intensive care unit: frequency and severity. J Intensive
161. Fontaine B. Periodic paralysis. Adv Genet. Care Med. 2014;5:269–74.
2008;63:3–23. 177. Singhi S, Marudkar A. Hypokalemia in a pediatric
162. Gil-Ruiz MA, Alcaraz AJ, Maranon RJ, Navarro N, intensive care unit. Indian Pediatr. 1996;33(1):9–14.
Huidobro B, Luque A. Electrolyte disturbances 178. Subba Rao SD, Thomas B. Electrolyte abnormalities
in acute pyelonephritis. Pediatr Nephrol. in children admitted to pediatric intensive care unit.
2012;27:429. Indian Pediatr. 2000;37(12):1348–53.
163. Kim M, Somers MJG. Fluid and electrolyte physiol- 179. Linshaw MA. Potassium homeostasis and hypokale-
ogy and therapy. In: McMillan JA, DeAngelis CD, mia. Pediatr Clin North Am. 1987;34(3):649–81.
Feigin RD, editors. Oski’s pediatrics. 4th 180. Schaefer TJ, Wolford RW. Disorders of potassium.
ed. Baltimore: Lippincott Williams and Wilkins; Emerg Med Clin North Am. 2005;23(3):723–47. 3.
2006. p. 54. 181. Saikaley A, Bichet D, Kucharczyk J, Peterson
164. Hsieh S, White PC. Presentation of primary adrenal LN. Neuroendocrine factors mediating polydipsia
insufficiency in childhood. J Clin Endocrinol Metab. induced by dietary Na, Cl, and K depletion. Am J
2011;96:E925. Physiol. 1986;251:R1071–7.
165. Schweiger B, Mortiarty MW. Cadnapaphornchai. 182. Wilson JLL, Miranda CA, Knepper MA. Vasopressin
Case report: severe neonatal hyperkalemia due to and the regulation of aquaporin-2. Clin Exp Nephrol.
pseudohypoaldosteronism type 1. Curr Opin Pediatr. 2013; 17(6). doi:10.1007/s10157-013-0789-5,
2009;21:269. 10.1007/s10157-013-0789-5.
166. Rodriguez Soriano J, Vallo A. Transtubular potassium 183. Greenlee M, Wingo CS, McDonough AA, Youn JH,
concentration gradient: a useful test to estimate renal Kone BC. Narrative review: evolving concepts in
aldosterone bio-activity in infants and children. potassium homeostasis and hypokalemia. Ann Intern
Pediatr Nephrol. 1990;4:105. Med. 2009;150(9):619–25.
167. Kamel KS, Halperin ML. Intrarenal urea cycling 184. Talbert A, Thuo N, Karisa J, Chesaro C, Ohuma E,
leads to a higher rate of renal excretion of potassium: Ignas J, et al. Diarrhoea complicating severe acute
an hypothesis with clinical implications. Curr Opin malnutrition in Kenyan children: a prospective
Nephrol Hypertens. 2011;20:547–54. descriptive study of risk factors and outcome. PLoS
168. Masilamani K, van der Voort J. The management of One. 2012;7(6):e38321.
acute hyperkalemia in neonates and children. Arch 185. Krebs SE, Flood RG, Peter JR, Gerard JM. Evaluation
Dis Child. 2012;97:376. of a high-dose continuous albuterol protocol for treat-
169. Helfrich E, deVries TW, van Roon EN. Salbutamol ment of pediatric asthma in the emergency depart-
for hyperkalemia in children. Acta Paediatr. ment. Pediatr Emerg Care. 2013;29(2):191–6.
2001;90:1213. 186. Habashy D, Lam LT, Browne GJ. The administration
170. Singh BS, Sadiq HF, Noguchi A, Keenan of beta2-agonists for paediatric asthma and its adverse
WJ. Efficacy of albuterol inhalation in treatment of reaction in Australian and New Zealand emergency
hyperkalemia in premature neonates. J Pediatr. departments: a cross-sectional survey. Eur J Emerg
2002;141:16. Med. 2003;10(3):219–24.
171. Janjua HS, Mahan JD, Patel HP, Mentser M, 187. Chan A, Shinde R, Chow CC, Cockram CS,
Schwaderer AL. Continuous infusion of a standard Swaminathan R. In vivo and in vitro sodium pump
combination solution in the management of activity in subjects with thyrotoxic periodic paralysis.
hyperkalemia. Nephrol Dial Transplant. BMJ. 1991;303(6810):1096–9.
2011;26:2503. 188. Yanturali S, Aksay E, Demir OF, Atilla R. Massive
172. Bomback AS, Woosley JT, Kshirsagar AV. Colonic hydroxychloroquine overdose. Acta Anaesthesiol
necrosis due to sodium polystyrene sulfate Scand. 2004;48(3):379–81.
(Kayexalate). Am J Emerg Med. 2009;27:753.e1. 189. Malik AR, Wolf PK, Ravasia S. Hypokalemia from
173. Mahoney BA, Smith WAD, Lo D, Tsoi K, Tonelli M, risperidone and quetiapine overdose. Can J Psychia-
Clase C. Emergency interventions for hyperkalaemia. try. 2005;50(1):76.
Cochrane Database Syst Rev. 2005; (2), Art. No.: 190. Deepthiraju B, Varma PR. Barium toxicity a rare
CD003235. doi:10.1002/14651858.CD003235.pub2. presentation of fireworks ingestion. Indian Pediatr.
174. Sterns RH, Rojas M, Bernstein P, Chennupati 2014;49(9):762.
S. Ion-exchange resins for the treatment of 191. Glauser J. Cardiac arrhythmias, respiratory failure,
hyperkalemia: are they safe and effective? J Am Soc and profound hypokalemia in a trauma patient.
Nephrol. 2010;21:733. Cleve Clin J Med. 2001;68(5):401, 5–10, 13.
175. Chhapola V, Kanwal SK, Sharma R, Kumar V. A 192. Molla AM, Rahman M, Sarker SA, Sack DA, Molla
comparative study on reliability of point of care A. Stool electrolyte content and purging rates in
13 Physiology of the Developing Kidney: Fluid and Electrolyte Homeostasis and Therapy of. . . 421

diarrhea caused by rotavirus, enterotoxigenic E. coli, 209. Bushinsky DA, Coe FL, Katzenberg C, Szidon JP,
and V. cholerae in children. J Pediatr. 1981;98(5): Parks JH. Arterial PCO2 in chronic metabolic acido-
835–8. sis. Kidney Int. 1982;22(3):311–4.
193. Dluhy RG, Anderson B, Harlin B, Ingelfinger J, 210. Fulop M. A guide for predicting arterial CO2 tension
Lifton R. Glucocorticoid-remediable aldosteronism in metabolic acidosis. Am J Nephrol. 1997;17(5):
is associated with severe hypertension in early child- 421–4.
hood. J Pediatr. 2001;138(5):715–20. 211. Arbus GS, Herbert LA, Levesque PR, Etsten BE,
194. Dutta A, Palazzi DL. Risk factors of amphotericin B Schwartz WB. Characterization and clinical applica-
toxicity in the nonneonatal pediatric population. tion of the “significance band” for acute respiratory
Pediatr Infect Dis J. 2012;31(9):910–4. alkalosis. N Engl J Med. 1969;280(3):117–23.
195. Bianchetti MG, Kanaka C, Ridolfi-Luthy A, Hirt A, 212. Martinu T, Menzies D, Dial S. Re-evaluation of
Wagner HP, Oetliker OH. Persisting renotubular acid–base prediction rules in patients with chronic
sequelae after cisplatin in children and adolescents. respiratory acidosis. Can Respir J. 2003;10(6):311–5.
Am J Nephrol. 1991;11(2):127–30. 213. Adrogue HJ, Madias NE. Secondary responses to
196. Moffett BS, McDade E, Rossano JW, Dickerson HA, altered acid–base status: the rules of engagement.
Nelson DP. Enteral potassium supplementation in a J Am Soc Nephrol. 2010;21(6):920–3.
pediatric cardiac intensive care unit: evaluation of a 214. Krapf R, Beeler I, Hertner D, Hulter HN. Chronic
practice change. Pediatr Crit Care Med. 2011;12(5): respiratory alkalosis. The effect of sustained hyper-
552–4. ventilation on renal regulation of acid–base equilib-
197. Knudson JD, Lowry AW, Price JF, Moffett rium. N Engl J Med. 1991;324(20):1394–401.
BS. Response to intravenous potassium chloride sup- 215. Guignard JP, Santos F. Laboratory investigations. In:
plementation in pediatric cardiac intensive care Avner ED, Harmon W, Niaudet P, editors. Pediatric
patients. Pediatr Cardiol. 2013;34(4):887–92. nephrology. Philadelphia: Lippincott-Williams and
198. Marquardt K, Albertson TE. Treatment of Wilkins; 2004. p. 404.
hydroxychloroquine overdose. Am J Emerg Med. 216. Feldman M, Soni N, Dickson B. Influence of
2001;19(5):420–4. hypoalbuminemia or hyperalbuminemia on the
199. Lin YC, Chen HZ, Chang TJ, Lane HY. Hypokalemia serum anion gap. J Lab Clin Med. 2005;146
following rapid titration of quetiapine treatment. (6):317–20.
J Clin Psychiatry. 2008;69(1):165–6. 217. Batlle DC, Hizon M, Cohen E, Gutterman C, Gupta
200. Vormann J, Remer T. Dietary, metabolic, physiologic, R. The use of the urinary anion gap in the diagnosis of
and disease-related aspects of acid–base balance: fore- hyperchloremic metabolic acidosis. N Engl J Med.
word to the contributions of the second international 1988;318(10):594–9.
acid–base symposium. J Nutr. 2008;138(2):413S–4. 218. Owen OE, Licht JH, Sapir DG. Renal function and
201. Remer T, Manz F. Estimation of the renal net acid effects of partial rehydration during diabetic
excretion by adults consuming diets containing vari- ketoacidosis. Diabetes. 1981;30(6):510–8.
able amounts of protein. Am J Clin Nutr. 1994;59(6): 219. Carlisle EJ, Donnelly SM, Vasuvattakul S, Kamel KS,
1356–61. Tobe S, Halperin ML. Glue-sniffing and distal renal
202. Cordain L, Eaton SB, Sebastian A, Mann N, tubular acidosis: sticking to the facts. J Am Soc
Lindeberg S, Watkins BA, et al. Origins and evolution Nephrol. 1991;1(8):1019–27.
of the Western diet: health implications for the 21st 220. Kim GH, Han JS, Kim YS, Joo KW, Kim S, Lee
century. Am J Clin Nutr. 2005;81(2):341–54. JS. Evaluation of urine acidification by urine anion
203. Palmer BF, Alpern RJ. Normal acid-base balance. In: gap and urine osmolal gap in chronic metabolic aci-
Feehaly J, Feehally J, Johnson RJ, editors. Compre- dosis. Am J Kidney Dis. 1996;27(1):42–7.
hensive clinical nephrology. 3rd ed. Philadelphia: 221. Galla JH. Metabolic alkalosis. J Am Soc Nephrol.
Elsevier; 2007. p. 141–6. 2000;11(2):369–75.
204. Manz F, Kalhoff H, Remer T. Renal acid excretion in 222. Sabatini S, Kurtzman NA. The maintenance of
early infancy. Pediatr Nephrol. 1997;11(2):231–43. metabolic alkalosis: factors which decrease
205. Baum M, Quigley R. Maturation of proximal tubular bicarbonate excretion. Kidney Int. 1984;25
acidification. Pediatr Nephrol. 1993;7(6):785–91. (2):357–61.
206. Schwartz GJ, Evan AP. Development of solute trans- 223. Berger BE, Cogan MG, Sebastian A. Reduced glo-
port in rabbit proximal tubule. III. Na-K-ATPase merular filtration and enhanced bicarbonate
activity. Am J Physiol. 1984;246(6 Pt 2):F845–52. reabsorption maintain metabolic alkalosis in humans.
207. Karashima S, Hattori S, Ushijima T, Furuse A, Kidney Int. 1984;26(2):205–8.
Nakazato H, Matsuda I. Developmental changes in 224. Galla JH, Bonduris DN, Luke RG. Effects of chloride
carbonic anhydrase II in the rat kidney. Pediatr and extracellular fluid volume on bicarbonate
Nephrol. 1998;12(4):263–8. reabsorption along the nephron in metabolic alkalosis
208. Matsumoto T, Fejes-Toth G, Schwartz GJ. Postnatal in the rat. Reassessment of the classical hypothesis of
differentiation of rabbit collecting duct intercalated the pathogenesis of metabolic alkalosis. J Clin Invest.
cells. Pediatr Res. 1996;39(1):1–12. 1987;80(1):41–50.
422 I.F. Ashoor and M.J.G. Somers

225. Adam WR, Koretsky AP, Weiner MW. 31P-NMR 235. Buchanan IB, Campbell BT, Peck MD, Cairns
in vivo measurement of renal intracellular pH: effects BA. Chest wall necrosis and death secondary to
of acidosis and K+ depletion in rats. Am J Physiol. hydrochloric acid infusion for metabolic alkalosis.
1986;251(5 Pt 2):F904–10. South Med J. 2005;98(8):822–4.
226. Wesson DE. Augmented bicarbonate reabsorption by 236. Bushinsky DA, Gennari FJ. Life-threatening
both the proximal and distal nephron maintains hyperkalemia induced by arginine. Ann Intern Med.
chloride-deplete metabolic alkalosis in rats. J Clin 1978;89(5 Pt 1):632–4.
Invest. 1989;84(5):1460–9. 237. Green J, Kleeman CR. Role of bone in regulation
227. Harrington JT, Hulter HN, Cohen JJ, Madias of systemic acid–base balance. Kidney Int. 1991;
NE. Mineralocorticoid-stimulated renal acidification: 39(1):9–26.
the critical role of dietary sodium. Kidney Int. 238. Krieger NS, Frick KK, Bushinsky DA. Mechanism of
1986;30(1):43–8. acid-induced bone resorption. Curr Opin Nephrol
228. Navaneethan SD, Singh S, Appasamy S, Wing RE, Hypertens. 2004;13(4):423–36.
Sehgal AR. Sodium bicarbonate therapy for preven- 239. McSherry E, Morris Jr RC. Attainment and mainte-
tion of contrast-induced nephropathy: a systematic nance of normal stature with alkali therapy in infants
review and meta-analysis. Am J Kidney Dis. and children with classic renal tubular acidosis. J Clin
2009;53(4):617–27. Invest. 1978;61(2):509–27.
229. Capasso G, Jaeger P, Giebisch G, Guckian V, Malnic 240. Challa A, Krieg Jr RJ, Thabet MA, Veldhuis JD, Chan
G. Renal bicarbonate reabsorption in the rat. II. Distal JC. Metabolic acidosis inhibits growth hormone
tubule load dependence and effect of hypokalemia. secretion in rats: mechanism of growth retardation.
J Clin Invest. 1987;80(2):409–14. Am J Physiol. 1993;265(4 Pt 1):E547–53.
230. Schuster VL. Cortical collecting duct bicarbonate 241. Ordonez FA, Santos F, Martinez V, Garcia E,
secretion. Kidney Int Suppl. 1991;33:S47–50. Fernandez P, Rodriguez J, et al. Resistance to growth
231. Javaheri S, Kazemi H. Metabolic alkalosis and hormone and insulin-like growth factor-I in acidotic
hypoventilation in humans. Am Rev Respir Dis. rats. Pediatr Nephrol. 2000;14(8–9):720–5.
1987;136(4):1011–6. 242. KDIGO. KDIGO 2012 clinical practice guideline for
232. Javaheri S, Shore NS, Rose B, Kazemi H. Compen- the evaluation and management of chronic kidney dis-
satory hypoventilation in metabolic alkalosis. Chest. ease. Kidney International Supplements. 2013;3:5–14.
1982;81(3):296–301. 243. Alexy U, Kersting M, Remer T. Potential renal acid
233. Rastegar A. Use of the DeltaAG/DeltaHCO3- ratio in load in the diet of children and adolescents: impact of
the diagnosis of mixed acid–base disorders. J Am Soc food groups, age and time trends. Public Health Nutr.
Nephrol. 2007;18(9):2429–31. 2008;11(3):300–6.
234. Moffett BS, Moffett TI, Dickerson HA. Acetazol- 244. Oettinger CW, Oliver JC. Normalization of uremic
amide therapy for hypochloremic metabolic alkalosis acidosis in hemodialysis patients with a high bicar-
in pediatric patients with heart disease. Am J Ther. bonate dialysate. J Am Soc Nephrol. 1993;3(11):
2007;14(4):331–5. 1804–7.
Part III
Translational Research Methods
Translational Research Methods:
Basics of Renal Molecular Biology 14
Gian Marco Ghiggeri, Maurizio Bruschi, and
Simone Sanna-Cherchi

Contents Genome-Wide Association in Primary


Glomerulonephritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 437
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 426 Copy-Number Variations in Congenital
Genomic and Proteomic Technologies Anomalies of the Kidney and Urinary Tract . . . . . . . . 439
and Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 426 Next-Generation Sequencing in Renal Disease . . . . . 440
Genetics and Genomics for Identification of The Building of the Podocyte Protein Map . . . . . . . . . 440
Susceptibility Alleles for Disease . . . . . . . . . . . . . . . . . . . 426 Proteomic Analysis of Biological Fluids:
DNA Sequencing and Search for Rare Variants . . . . 428 The Normal Urine Map . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 440
Single Nucleotide Polymorphism (SNP) Peptidome, Degradome, and Metabolomics . . . . . . . . . 441
and Genome-Wide Association Studies (GWAS) . . . 430 Autoantibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 441
Structural Variants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 430 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 442
Techniques for Proteome Analysis and Mass
Spectrometry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 431
Bioinformatics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 434
Bioinformatics Tools Applied to DNA . . . . . . . . . . . . . . 434
Bioinformatics Tools for Proteomics . . . . . . . . . . . . . . . . 436
Protein Interaction, Metabolic Pathways,
and Systems Biology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 437
Application Fields to Renal Diseases . . . . . . . . . . . . . . 437

G.M. Ghiggeri (*) • M. Bruschi


Laboratory of Physiopathology of Uremia, Division of
Nephrology, Dialysis and Transplantation, Istituto
Giannina Gaslini, Genoa, Italy
e-mail: gmarcoghiggeri@ospedale-gaslini.ge.it;
mauriziobruschi@ospedale-gaslini.ge.it
S. Sanna-Cherchi
Division of Nephrology, Columbia University, College of
Physicians and Surgeons, New York, NY, USA
e-mail: ss2517@columbia.edu; ss2517@cumc.columbia.
edu

# Springer-Verlag Berlin Heidelberg 2016 425


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_13
426 G.M. Ghiggeri et al.

Introduction Genomic and Proteomic Technologies


and Applications
Molecular biology has developed between years
1940s and 1960s as a branch of science that Genetics and Genomics
spans from genetics to cell biology and biochem- for Identification of Susceptibility
istry with the goal of understanding the interac- Alleles for Disease
tions by which DNA, RNA, and protein
synthesis operate into cells [1]. Since then, The type of approach to solve the genetic under-
numerous technological innovations pushed the pinning of a trait largely depends on the epidemi-
field forward, allowing recombinant DNA tech- ology of the disease, its heritability, race/ethnicity
nology to be applied to a wide variety of biologic differences, the underlying genetic model, and the
problems and to enter the clinical practice. The predicted effect size of the underlying causal var-
discovery of restriction nucleases and DNA iants (Fig. 2). Traditionally, Mendelian diseases
ligases, development of DNA libraries, DNA occurring in families have been associated to very
cloning procedures, nucleic acid hybridization rare variants with large effect on the phenotype
techniques, and the polymerase chain reaction and have been successfully tackled using linkage/
(PCR) together with transgenic animals have positional cloning strategies, while common traits
all represented successive steps for a successful have been thought to be mostly caused by com-
delineation of the role of genes in cellular phys- mon variants with small-effect size that are ances-
iology and pathophysiology. This molecular rev- trally inherited in populations (common disease –
olution has affected all of the sciences, including common variant hypothesis) and have been the
medical and clinical research, and has culmi- object of hundreds of genome-wide association
nated in the completion of the human genome studies.
project [2, 3]. In the past 15 years, we have The recent discovery of the role of rare copy-
witnessed tremendous technological advances number variations in developmental diseases and
for high-throughput analysis of DNA, RNA, the tremendous advances in sequencing technolo-
and protein in a comprehensive and cost- gies, which lead to the vast use of next-generation
effective manner, with great promises to trans- sequencing, boosted the research on rare variants
late the analysis of the central dogma of biology identification and interpretation in complex dis-
into precision diagnosis and treatment for both eases. Below, we will briefly review the basic
rare and common diseases (Fig. 1). The recent approaches to gene mapping and identification in
advances in RNA biology identified numerous research setting and clinical practice. Examples of
noncoding functional regions of the genome, application of different strategies to renal research
which “escape” the central dogma but that will be given in the next section of this chapter.
can be analyzed and studied using a similar
framework. Mendelian Genetics
This entry briefly illustrates the basic concepts Since the landmark paper of Landers in 1986 [4]
of molecular biology that underlie the various and the first discovery of RFLP maps and
strategies that have been developed for studying then microsatellites, traits with strong genetic
living cells and organisms and summarizes molec- determination, segregating in large pedigrees
ular biology techniques that are used in research following clear Mendelian modes of inheritance
and clinical practice. We will show practical appli- (autosomal dominant, autosomal recessive,
cations of genomic and proteomic tools and strat- X-linked), were approached by using logarithm
egies applied to kidney disease research in a of odds score (LOD score) methods. Using these
concise format. For more information, the reader methods, if a family is large enough to provide the
can refer to the many specialized reference text- statistical evidence that a chromosomal region
books that are available. co-segregates with the disease trait under the
14 Translational Research Methods: Basics of Renal Molecular Biology 427

Molecular Diagnosis
∼20,000
DNA Counselling
GENES
Personalized Genomics

Gene Expression Regulation Personalized


∼80,000
RNA Pathways Treatment
TRANSCRIPTS
Molecular Signatures Strategies

Protein Networks
>1,000,000
PROTEIN Pathophysiology
PROTEINS
Biomarkers

TRAIT

Fig. 1 Schematic representation of the central dogma of biology

Fig. 2 Graphic
representation of disease 1
models and gene Unlikely
Mendelian
identification strategies to occur
based on penetrance 0.75
Linkage / de novo
Penetrance

(X-axis) and allele


frequency (Y-axis)
0.5

0.25
Difficult
Complex
to identify
0
Next-generation genomics Association

< 0.001 0.001 - 0.01 0.01 - 0.1 > 0.1


Allelic Frequency

hypothesis of linkage versus no-linkage (null Complex Traits: Common Variants


distribution) at a genome-wide significance The common variant – common disease theory
level [5], the locus containing the disease- stands on the hypothesis that complex traits that
causing gene is identified. Once the disease manifest later in life have been less subjected to
locus is mapped, direct sequencing of the candi- natural selection because they are less likely to
dates in this region is performed to identify the impact fitness compared to rare, lethal Mendelian
gene containing the disruptive segregating muta- diseases. With less selection operating, these phe-
tion. Finally, the proof of genetic causation notypes are maintained at relatively high fre-
requires the identification of independent delete- quency in the general population. Given the high
rious mutations in additional families or patients heritability of many of these traits (hypertension,
with the same phenotype and functional analyses diabetes mellitus, Alzheimer’s disease, Crohn’s
in cellular and animal models. disease, and many others), it was hypothesized
428 G.M. Ghiggeri et al.

that, similarly to the disease epidemiology, the genetic field for approximately 30 years [6].
underlying disease-causing alleles would be fre- This methodology, which is still considered the
quent in the general population and have small to gold standard for its accuracy and reproducibil-
moderate effect size, since purifying selection ity, relies on enzymatic extension of a DNA
would be unlikely to have a major effect on template (usually a PCR amplification product)
eliminating such alleles. Therefore, these vari- using dideoxy terminators initially labeled with
ants, once introduced in a population, would be radioactive, subsequently with fluorescent dyes.
maintained at relatively high frequency. By the The labeled amplicons are then separated on
use of a complete map of SNPs able to effectively agarose gel or, currently, through an automated
tag the haplotype structure of the human genome capillary sequencer. This methodology was the
from a specific population, it is then feasible to main force behind the efforts of the Human and
identify such variants (or the variants in linkage Venter’s Genome Projects [2, 3], which lasted
disequilibrium with the causal allele). The over a decade and resulted in the first draft of
HapMap Consortium greatly facilitated the the human genome at the cost of almost three
development and generation of SNP arrays for billion dollars [7]. After the completion of the
genome-wide association studies (GWAS). The sequence of the human genome, we assisted in
basic hypothesis underlying the GWAS study the international private and academic efforts
design is that variants at the disease locus aimed to bring the whole-genome sequencing
(i) would have significantly different allele fre- (WGS) to a cost and turnaround that would
quencies between cases and controls compared to allow its implementation into clinical practice.
the vast majority of the remaining variants. Usu- The recent advent of next-generation technology
ally, a GWAS is conducted by comparing allele has completely revolutionized genetic research,
frequencies for 0.5–1 million markers across the by increasing sequencing capabilities by five
genome between cases and controls in order to orders of magnitude and decreasing costs by
fully capture the haplotype structure of the nearly six orders of magnitude. It is in fact now
genome. While this approach represents an unbi- possible to effectively sequence the entire
ased, hypothesis-free way to detect association genome of an individual at approximately
signal, the large number of pairwise comparisons 30X coverage in about 2 weeks at a cost of
mandates rigorous correction for multiple testing 1,500 dollars using the new Illumina HiSeq X
in order to avoid spurious, false-positive results. Ten system. This approaches the goal of the X
In fact, the usual threshold for genome-wide sig- Prize Foundation of achieving 100 human
nificance is set at p < 5  108. While the basic genomes sequenced in less than 30 days at a
concept of GWAS is relatively simple and cost of $1,000 or less per genome (http://geno
straightforward, there are numerous technical mic.xprize.org/). The basic idea underlying com-
and analytical issues to be taken into account, monly used next-generation sequencing (NGS)
such as power calculations, population stratifica- technologies is that clonal DNA fragments are
tion, platform and batch effect, and others, that generated and then they are all subjected at once
will be briefly discussed below in the bioinfor- to massively parallel sequencing using
matics section. microfluidics. Sequencing reactions are carried
on by consecutive cycles of base addition by
polymerization or ligation according to the sys-
DNA Sequencing and Search for Rare tem used. Visualization and base calling are then
Variants conducted in different ways: pyrosequencing for
the Roche FLX System, fluorescence for the
DNA sequencing is the backbone of molecular Illumina and Life Technologies SOLID instru-
genetics. Frederick Sanger published his revolu- ments, and changes on pH for the Ion Torrent
tionary paper in 1977, describing a DNA apparatus. More recent sequencing technologies,
sequencing technology that dominated the sometimes referred to as third- and
14 Translational Research Methods: Basics of Renal Molecular Biology 429

fourth-generation sequencing (nanopore, in situ mutations are identified in the discovery studies
sequencing, respectively), are still in their exper- (see below). Therefore, for studies that are prom-
imental phase and will not be discussed here. inently based on singletons or small families, and
Also, since the vast majority of NGS in both in the setting of high genetic heterogeneity, the
academia and private sector is conducted using discovery of independent mutations and the
the Illumina system, and since the authors of this assessment of the mutational load under the null
chapter have most of their experience using this distribution in large populations rapidly became
technology, we will describe data generation and imperative. On one side, publicly available
analysis using the Illumina NGS system. population-based genomes and exomes can be
Targeted enrichment systems. During these very helpful in rapidly identifying “intolerant”
past years of NGS, several ways to selectively genes (genes harboring no or very few loss-of-
enrich portions of the genome have been devel- function variants). Among such databases, partic-
oped to capture regions of interest at higher depth ularly useful are the 1,000 Genome Project, the
of coverage while attempting to further reduce NHLBI Exome Variant Server (http://evs.gs.wash
costs. Based on the estimation that Mendelian ington.edu/EVS/; featuring whole exome data
diseases and traits with stronger genetic determi- from ~6,500 individuals), and the recently
nation are most likely to be caused by protein- released Exome Aggregation Consortium (http://
altering variants, exome sequencing has been exac.broadinstitute.org/; featuring whole exome
developed [8]. By selectively capturing the data from ~63,000 individuals). Nevertheless, to
exons of the nearly 20,000 human genes, which identify rare independent mutations in patients
constitute about 1–2 % of the euchromatic DNA, with the disease object of study, it is becoming
it is possible to sequence at high coverage apparent that candidate genes detected in whole
(60–90X) the entire protein coding region of the genome or exome studies (usually dozen to a few
genome at a cost of $600–800 per sample. This hundreds) must be effectively resequenced in
approach has been tremendously efficient in solv- hundreds or thousands of individuals in order to
ing the molecular diagnosis of a multitude of uncover excess burden of rare variants and, pos-
Mendelian and complex traits [9–21]. The possi- sibly, establish genetic causation. Obviously,
bility of performing rapid diagnosis for known resequencing studies of this scale cannot be
genetic causes for Mendelian diseases led to the performed in a time- and cost- effective manner
development of array-based targeted gene panels using Sanger sequencing or targeted NGS chips at
for diseases (or group of diseases) to use in the an individual DNA level. Therefore, several
clinical settings. These targeted capture chips, methods have been implemented for multiplex
although of relatively recent introduction, are bar coding DNA capture and massively parallel
already been abandoned due to elevated costs resequencing in large cohorts [22]. Among these
(higher than whole-exome sequencing) and to methods, the molecular inversion probes (MIP)
the not comprehensive nature of platform design; and the Fluidigm 48.48 Access Array™ IFC Sys-
therefore, we will not discuss them in this review tem coupled to NGS have been the most success-
since they are not likely to play a significant part fully used [23–25]. Using these methods, it is
in the armamentarium of genetic tools in human possible to sequence 600–1,000 amplicons
disease research and diagnosis. (approximately 40–80 genes) in 1,000–3,000
The availability of individual-level whole patients in few months and at a cost of 50–75
genome or exome data also uncovered that each cents per gene per sample, corresponding to
individual is a carrier of thousands of single nucle- approximately $15,000–25,000 to resequence
otide variants, many of which are very rare or 40 genes in 1,000 patients. As a comparison, the
unique and predicted to be deleterious, thus enor- same undertaking would require approximately
mously complicating the attribution of genetic 0.8–1.5 million dollars by array-based NGS or
causality for both Mendelian and complex traits, Sanger sequencing and significantly longer
especially when no large families or de novo processing time.
430 G.M. Ghiggeri et al.

Single Nucleotide Polymorphism (SNP) prediction. Moreover, functional interpretation of


and Genome-Wide Association these results is often problematic. Nevertheless, it
Studies (GWAS) is of no doubt that many of such studies have shed
light on disease pathogenesis and treatment and
One of the first and most important discoveries provided important clues into population struc-
deriving from the completion of the human ture, disease epidemiology, and natural selection
genome and the first comprehensive catalog of [28–31, 34–36]. The challenge still remains now-
single nucleotide variants was that the euchro- adays, and consists in translating each of these
matic DNA is organized in haplotype blocks. 7,000 robust associations into functional assays
This led to the release of the International to resolve disease pathogenesis and to improve
HapMap Project [26] which is an effort to catalog therapeutic schemes. Finally, despite the success
millions of variants (mostly single nucleotide in the past 10 years of GWAS, a significant frac-
polymorphisms) and to describe their pattern of tion of heritability for most complex traits still
variations and their correlation (linkage disequi- remains missing, suggesting that rare or
librium) in populations from three main ances- low-frequency variants with large or moderate
tries: Europeans, Africans, and Asians. The effect size might account for it, hence the interna-
HapMap work posed the foundation for the devel- tional efforts for systematic resequencing in large
opment of high-density genome-wide SNP arrays populations.
and their application for genome-wide association
study (GWAS) in complex traits. The overarching
hypothesis was in fact that complex traits with Structural Variants
high prevalence in the general population and
high heritability (e.g., diabetes mellitus, hyperten- Variations other than single nucleotide variants,
sion, Alzheimer’s disease, and many others) such as change in DNA copy number, inversions,
would be caused by common variants with and translocations are known as structural vari-
small-effect size (common disease – common var- ants. Structural variants have traditionally been
iant hypothesis) [27]. With the description of the implicated in the pathogenesis of rare genomic
haplotype structure of the human genome, it was disorders [37]. Landmark studies conducted in
then possible to develop SNP arrays containing healthy population controls in mid-2000s showed
only a reduced fraction of the entire genetic vari- that a significant portion of the euchromatic DNA
ation (usually 500,000 to 1 million SNPs) that is subjected to copy-number variations (CNVs)
would efficiently tag these blocks of disequilib- [38–41]. The amount of genetic variation derived
rium and therefore enable genome-wide associa- from CNVs was estimated to be at least one order
tion studies. Since the first successful GWAS of magnitude larger than the one attributable to
reported in 2005 identifying a common variant SNPs. Moreover, given the fact that CNVs are
in the complement factor H as a strong suscepti- often large and contain multiple genes, it was
bility factor for age-related macular degeneration suggested that the effect of gene copy number
[28–30], approximately 7,000 variants have been could be a major driver in the predisposition to
convincingly identified in complex traits genetics complex traits. While common CNPs (copy-
[31]. While this might seem an enormous success, number polymorphisms) did not seem to explain
these studies and the underlying hypothesis have much of the missing heritability of complex traits
also encountered skepticism, especially in regard- [42], subsequent studies clearly showed that rare
ing the potential for utility and translation in clin- CNVs are a major source of genetic variation
ical practice [32, 33]. The reason for this underlying many developmental traits, from cra-
skepticism stands mainly in the fact that most niofacial defects, cardiovascular and kidney
variations, even in aggregate, account for a small malformations, and neurodevelopmental
fraction of the heritability of these traits, thus diseases including autism, schizophrenia, epi-
hampering a direct prognostication and risk lepsy, intellectual disability, and developmental
14 Translational Research Methods: Basics of Renal Molecular Biology 431

delay [43–51]. Importantly, the work on progressively taking place using whole-exome
neurodevelopmental diseases also sets the exam- and whole-genome sequencing. The observation
ple and framework for analysis and interpretation that most CNVs implicated in developmental dis-
of rare variants in diseases with extremely high ease encompass multiple genes, made exome
genetic heterogeneity. sequencing a desirable tool for identification of
CNVs, usually defined as gain or loss of genic CNVs. Many software packages have been
genetic material that spans from 1,000 bp to sev- developed for CNV analysis using exome-
eral megabases, can be identified using different sequencing data (see below), but the capture
technologies. The most used in current research design of this technology results in uneven depth
and in clinical practice are array comparative of coverage across the exome, posing serious con-
genomic hybridization (aCGH), SNP arrays, and cern in accuracy and replication of results. Whole-
next-generation sequencing. genome sequencing on the other hand represents a
Array Comparative Genomic Hybridization very powerful way to accurately score CNVs,
(aCGH). One of the first methods for analysis of identify boundaries, and also to identify other
CNVs was comparative genomic hybridization, structural variants such as inversions, transloca-
which was performed by differential labeling of tions, gene fusions, and complex CNVs by anal-
control and test metaphase chromosomes at low ysis of atypical reads, thus making it the most
resolution. Subsequently, BAC clones and then comprehensive and accurate tool for analysis of
oligonucleotide probes have been implemented structural variations [54]. With the continuous
on a physical support (array) to analyze genomic drop in sequencing costs, WGS will soon repre-
DNA at much higher resolution and at a lower sent the most cost-effective and comprehensive
cost. The aCGH is still currently used, often in the tool for analysis of rare and common variants,
clinical setting, for high-resolution analysis of either SNV or structural variations.
CNV, especially in prenatal care and in the
diagnosis of severe developmental diseases
[52, 53]. One major advantage of aCGH is the Techniques for Proteome Analysis
very high signal-to-noise ratio, which allows and Mass Spectrometry
robust identification of structural variants and
their breakpoints. Principles and techniques. The principle of pro-
Genome-Wide SNP Arrays. While originally tein analysis has considerably evolved over the
developed for SNP genotyping and GWAS appli- years in parallel with key technological advance-
cation, the observation that raw and allelic inten- ments, especially in the field of mass spectrome-
sity data could be used to assess for CNVs made try. The basic principle behind any proteomic
these platform a valid, and sometimes preferred, approach is that the products of a peptide diges-
alternative to aCGH. One of the advantages of tion constitute a fingerprint of the original protein
aCGH compared to SNP chips for CNV analysis which can be defined based on adequate strategies
is the higher signal-to-noise ratio. On the other that include peptide mass identification and bio-
side, SNP chips are usually cheaper and allow informatics analysis. In general, identification of a
better quality control analyses (e.g., identification protein is preceded by a preparative step.
of duplicates or hidden relatedness) and correction Two-dimensional (2D) electrophoresis has now
for population stratification using genotype infor- emerged as a powerful tool to accomplish this
mation, which are critical in large-scale associa- task. LC-Mass and MALDI-TOF are commonly
tion studies. Higher level CNVs (>4 copies) and used for the analysis of trypsin digest.
other structural variants, such as inversions and Denaturing and nondenaturing 2D electro-
translocations, are very difficult or impossible to phoresis. Classic 2D protein electrophoresis is
identify by either aCGH or SNP chips. the technique of choice for the analysis of plasma
Next-Generation Sequencing. Genome-wide proteins or other complex biological samples
analysis of copy-number variation is (Fig. 3a). High resolution is usually achieved by
432 G.M. Ghiggeri et al.

_
a 3.5 non lianer pH 9 b + Native electrophoresis
250 550

Un-folded and reduced electrophoresis


Albumin 200

IgG heavy chain


Mr Haptoglobin β chain Albumin IgG heavy chain
Mr
kDa
kDa

IgG light chain Haptoglobin β chain

IgG light chain

10
5

414m ∼ 100 fmol

414m ∼ 100 fmol


414m ∼ 200 fmol
414m ∼ 200 fmol
414m ∼ 400 fmol

414m ∼ 400 fmol


c 3.5 non lianer pH 9

250

Ang II _ + + + + + + +

Mr
kDa

5 1 2 3 4 5 6 7 8

Fig. 3 (a) Classical 2D polyacrylamide gel electrophore- is by this mean analyzed and quantified. (d) Gel retardation
sis (2D-PAGE) and (b) 2D electrophoresis in assay. The figure demonstrates binding of regulatory
nondenaturing condition (Nat/SDS-PAGE) of the same nuclear proteins to cis-elements in the COL3A1 promoter
serum sample. In the former approach, proteins are first after angiotensin II (Ang II) stimulation. Nuclear extracts
separated according to their charge in the presence of urea were obtained from Ang II stimulated cells (Ang II +) and
(IEF) and are then run in a polyacrylamide gradient that incubated with a 32P-labeled oligonucleotide (414) that
separates on the basis of size. In Nat/SDS-PAGE, protein contains sequences +3 to +20 of the COL3A1 promoter.
complexes migrate unresolved in the first dimension and Ang II stimulates the synthesis of a peptide that binds to the
are then separated in denaturing conditions, in the second target DNA and retards its migration in the gel (lanes 2).
dimension. (c) DIGE systems that utilize thiol-based This reaction can be competed with increasing amounts of
reagents (maleimide, iodoacetamide) increase specificity nonlabeled wild-type oligonucleotide (lanes 3–5) but not
and reproducibility of protein stainings on a quantitative with a cold-mutated analogue sequence (414 m) (lanes
basis. Protein mixtures are labeled separately with different 6–8). In the absence of Ang II stimulation (Ang II -), no
NHS dyes, combined and resolved on a single 2D gel that DNA-protein complex-generating gel retardation is
is analyzed with different fluorescence excitation wave- observed (lane 1)
lengths. The differential expression of individual proteins
14 Translational Research Methods: Basics of Renal Molecular Biology 433

combining separation by charge and separation by that the MALDI sample preparation is followed
mass in denaturing conditions (IEF/PAGE) by TOF mass analysis, while in the surface-
[55, 56]. This is followed by microscale mass enhanced laser desorption and ionization
spectrometry that allows identification of individ- (SELDI) procedure, proteins are immobilized on
ual spots, with reliable sensitivity and reproduc- solid supports or on customized protein chips.
ibility. The development of softgels is a recent In the so-called top-down strategy, intact pro-
evolution that improves the resolution of high- teins are ionized and resolved in the mass ana-
molecular-weight proteins, but requires denatur- lyzer. Alternatively, proteins are predigested into
ing conditions, which do not allow to analyze smaller peptides before the mass analysis, a pro-
protein-protein interactions (Fig. 6a) [57]. For cedure referred to as “bottom-up.” In this case, the
this reason, new techniques that separate protein protein is identified by its pattern of digestion that
mixtures in partially denaturing conditions have creates a “peptide mass fingerprinting” (PMF), or
recently been developed. These include Blue- by “de novo sequencing,” tracking back the pro-
PAGE, which is performed on membranes, and tein sequence from the mass-sequence data using
Nat/SDS-PAGE, which is performed on a poly- protein databases. Often, both “top-down” and
acrylamide substrate (Fig. 3b) [58–60]. “bottom-up” strategies are used to optimize pro-
Protein staining. Traditionally, protein staining tein identification.
after electrophoresis has been performed with Protein-protein interaction. One important
Coomassie R-250 and, from mid-1980s, with sil- question in protein analysis is to identify interac-
ver ions that allow high sensitivity [61]. Nowa- tions between proteins, which regulate most intra-
days, new dyes allow differential protein cellular signaling and are critical to the assembly
expression analysis on 2D gels (DIGE). This tech- of functional peptides.
nique is based on modification of selected amino Different approaches are possible. In one
acid residues and has become a standard applica- approach, proteins are analyzed directly by two-
tion in quantitative proteomics. Peptides are dimensional electrophoresis in nondenaturing and
labeled with matched sets of fluorescent N- in denaturing conditions (Nat/SDS-PAGE) that
hydroxysuccinimidyl ester cyanines (NHS) that allow protein complexes to co-migrate in the
have different excitation-emission wavelengths first dimension (nondenaturing) and then dissoci-
[62]; the use of thiol-based reagents (maleimide, ate in the second (Fig. 3b) [60]. Alternatively,
iodoacetamide) increases specificity and repro- proteins can be extracted from a crude mixture
ducibility (Fig. 3c) [63]. Protein mixtures can be after binding to target proteins linked to solid
labeled separately with different NHS/thiol dyes, supports or interactions that can be identified
combined and resolved on a single 2D gel that is using a yeast two-hybrid system. This methodol-
then analyzed at different fluorescence excitation ogy is based on the modular structure of gene
wavelengths [64]. This allows analysis and quan- activation using the GAL4 transcriptional activa-
tification of the differential expression of individ- tor as reporter. This transcription factor has a
ual proteins. DNA-binding domain and an activation domain,
Mass spectrometry. Proteins are usually char- both of which must be in close association to
acterized by mass spectrometry. As ionized mol- activate transcription. By DNA recombinant tech-
ecules are accelerated through an electric field, niques, two protein sequences acting as bait and
they are separated, reaching the detector at differ- target are fused with sequences encoding with one
ent times, depending on their mass and charge. of the GAL4 domains. When expressed in yeast
This “time-of-flight” (TOF) is specific of a given cells, the activation domain and the DNA-binding
small solute or for a given peptide, allowing its domain are bridged together when the two pro-
identification. Whole proteins are ionized by teins interact and promote transcription of a
electrospray ionization (ESI) or matrix-assisted reporter gene. Similar prokaryotic systems have
laser desorption/ionization (MALDI). The com- been developed based on the modular structure of
monly used “MALDI-TOF” procedure indicates bacterial RNA polymerases, in which target and
434 G.M. Ghiggeri et al.

bait cDNAs are fused to either the core enzyme or analysis is the logarithm of odds (LOD) score, a
a Φ factor. likelihood ratio test, that tests the probability of a
Protein-DNA interaction. Techniques that ana- disease gene to be localized in a certain region
lyze the regulation of DNA expression by tran- (linked to the nearby marker) versus the null
scriptional factors have allowed to identify hypothesis or random segregation. The approach
consensus DNA-binding regions and their regula- hinges on the availability of single, uniquely large
tory proteins. Most of these studies are based on kindreds or large cohorts of small/medium size.
the principle that protein-DNA complexes have Since the joint probability across multiple families
high molecular weights and are therefore retarded is a product of each individual probability, the
when resolved by polyacrylamide gel electropho- LOD scores, which use a logarithmic scale, can
resis. In addition, proteins interacting with DNA be summed. The first linkage package, Liped, was
molecules tend to protect the nucleic acid regions described in 1974, and used the Elston-Stewart
to which they bind from digestion with DNAse, algorithm to compute two-point parametric LOD
allowing their identification [65]. In Fig. 3d, for scores [66]. Subsequent programs, such as LINK-
example, a nuclear extract of proteins obtained AGE and FASTLINK, have allowed to calculate
from cells stimulated with angiotensin II was two-point and multipoint LOD scores; while com-
co-incubated with strings of DNA that encode putation time did not increase significantly with
for the promoter of type III collagen. As shown, the size of pedigrees, these algorithms, however,
angiotensin II stimulations promote the synthesis are not efficient enough for large markers datasets
of a protein that binds to the 32P-labeled DNA [67, 68]. Efficient multipoint analysis of linkage
target, forming macromolecular complexes that for dense marker maps was then developed with
are retarded in the gel. Genehunter [69] and optimized in Allegro and
Merlin software [70, 71]. These linkage packages
allow two-point and multipoint parametric link-
Bioinformatics age analysis, nonparametric, model-free analysis,
haplotype reconstruction, and other analyses. The
Bioinformatics Tools Applied to DNA main limitation of these algorithms is that while
the computation time increases linearly with the
In this section of the entry, we will briefly review number of markers, it increases exponentially
and describe the current methodologies and pub- with pedigree size. Software that can handle larger
licly available software packages that are most genealogies and perform also quantitative traits
commonly used for gene identification in Mende- linkage analysis include SimWalk [72] and
lian and complex traits. The scope is to provide a Loki [73].
short, simple, and user-friendly guide for data
analysis. Large-scale genetic data sets are usually Genome-Wide Association Studies
managed using ad hoc scripts and pipelines (Perl, One of the most user-friendly, versatile, complete,
Python, R, and others), usually in a Unix or Linux and widely used package for association analysis
environment, that are generated in-house based on is PLINK [74]. This package allows rapid data
individual needs, and they will not be discussed in management and analysis for hundreds of thou-
this chapter. sands of markers generated from thousands of
individuals, in a simple research environment
Traditional Positional Cloning/Linkage using a Unix or Linux desktop computer. The
Packages main implemented functions include data
Linkage analysis refers to the analysis of management, summary statistics, population
co-segregation of chromosomal regions with the stratification, association analysis, and identity-
disease trait under a specific mode of inheritance by-descent estimation. More recent additions to
(X-linked, autosomal dominant, autosomal reces- this package include analysis or rare variants,
sive). The statistical inference underlying linkage including CNVs, and next-generation sequencing.
14 Translational Research Methods: Basics of Renal Molecular Biology 435

Fig. 4 Burden analysis


between 1,441 patients with 100%
congenital anomalies of the 75%
kidney and urinary tract 50% CAKUT
(red) and a control dataset
CONTROL
of 21,610 individuals

Population Frequency
25%
(green), examining the
population frequency of the
largest CNV per genome 10%
using a survival function
(logrank P < 5  108;
Sanna-Cherchi, personal
data)

0%

0 1,000 2,000 3,000 4,000 5,000 6,000 7,000 8,000 9,00010,000


Largest CNV Size (kb)

Correction for population stratification is usually analytic dimensions (genotype and intensity) to
conducted in PLINK using multidimensional estimate copy-number state. Association tests for
scaling (MDS) or by principal component analy- common CNVs can be conducted using the same
sis as implemented in other packages such as framework used for GWAS, with the only caveat
Spectral-GEM [75, 76]. that CNVs have often a higher genotyping error
Often, large studies on complex traits combine rate, especially when analyzing data generated
data from multiple cohorts and multiple from different platforms. Analysis of rare CNVs
genotyping platforms, requiring data harmoniza- includes burden tests and single variant tests. To
tion prior to performing joint analyses. To uniform compare the burden of rare CNVs between cases
markers map, genotype imputation can be and controls, the Fisher’s exact test is often used
conducted using Markov chain haplotyping, as for testing differences in the type and size distri-
implemented in MACH software [77]. Imputation butions of CNV. The average CNV rates per
of a few million SNPs such has the HapMap panel genome, the mean CNV size per individual, the
is a computational intensive task that usually largest CNV size per individual, and the total
requires cluster computing. Joint analyses of genomic span per genome are usually calculated.
data generated from multiple cohorts can be Given the positively skewed distributions of these
performed using an inverse variance-weighted metrics, nonparametric statistics are preferred
method (METAL software) [78], and heterogene- (Mann–Whitney U test) and empirical p-values
ity across cohorts can be tested by performing are calculated using permutation tests. The fre-
Cochran’s Q test and deriving heterogeneity quency of the largest CNV per genome in the
index (I2). population can be analyzed using a survival func-
tion (Fig. 4). In this approach, median sizes are
Copy-Number Variation Analysis compared between cases and controls using the
CNV analysis from SNP chips can be performed logrank test, while the proportions of cases and
using multiple publicly available software. controls with CNVs above a given threshold are
Preprocessing and QCs are usually conducted compared using Fisher’s exact test.
using PLINK [74]. The most robust algorithms
for CNV calling, such as QuantiSNP and Next-Generation Sequencing
PennCNV [79, 80], utilize generalized The analysis of NGS data varies significantly
genotyping methods which incorporate the two based on the application, and, besides standard
436 G.M. Ghiggeri et al.

software for base calling and variant calling, most by univariate statistics, such as ANOVA; multi-
of the analyses are conducted using original variate analysis, such as principal component
scripts and pipelines designed ad hoc for each analysis (PCA) and heat maps, allows to better
study. Usually after completion of a sequencing describe trends and to reduce complexity [89].
run, data are automatically transferred to a cluster A combination of both approaches is often used
for base calling. Sequence graph files (.sgr) can be and ad hoc software have been developed [90].
generated to visualize sequencing data. After Adequate data preprocessing is, however, crucial
quality control assessment, sequence reads are to avoid redundancy, data incompleteness, and
converted to FASTQ format and mapped to the errors. Here, we briefly present a statistical
reference genome producing BAM files workflow for maximizing the information
containing all passed filter reads, alignment infor- obtained by quantitative proteomics. The analyses
mation, sequence, and quality score information. are usually conducted with existing statistical
BAM files are then converted in VCF files for packages; one of the most performing is R [91],
downstream analyses [15]. Recalibration and freely available at http://www.r-project.org/. Ana-
local realignment around indels are done using lyses of functional protein interaction networks
GATK [81]. Standard tools for variants’ annota- can be performed with public software; as an
tion include ANNOVAR [82], SNPeff [83], and example, cytoscape, an open-source package for
SeattleSeq (http://snp.gs.washington.edu/ the analysis of protein and genetic interaction
SeattleSeqAnnotation138/). Usually, allele fre- networks, is illustrated.
quencies are tested against dbSNP, 1,000 Sample size. To determine the samples size the
Genome, Exome Variant Server (http://evs.gs. following equation can be utilized:
washington.edu/EVS/), and the recently released
 2
Exome Aggregation Consortium (http://exac. δ
broadinstitute.org/). Metrics for interpretation of n ¼ 1 þ 2c
fc
possible functional relevance are computed using
algorithms that take in account DNA and amino where the quantity c depends on the power of the
acid conservation across species and chemical test and on the significance level. Power is usually
properties of the amino acid substitutions. Some set at a value between 80 % and 90 %, while the
of the most commonly used include SIFT [84], significance level is set at 0.05 % or 0.01 %; δ is
PolyPhen-2 [85], GERP [86], and CADD the standard deviation (SD) of biological replica-
[87]. Several programs to test for rare variant tions, and fc is the difference to be detected that is
association are currently available or underdevel- usually set at twofold changes. To limit the effect
opment; one of the most commonly used is of systematic variation across different experi-
SKAT [88]. mental blocks, protein data must be normalized
and expressed as fold changes relative to other
proteins. Normalization of n spot/peptide abun-
Bioinformatics Tools for Proteomics dance in each experiment is performed after back-
ground subtraction [92].
General considerations. Comparative proteomics Missing values and data filtering. Proteomic
aims at detecting changes in protein expression studies are limited by the high frequency of miss-
profiles between two or more groups of samples ing values, which can be determined by using
(cell extracts, biological fluids, distinct pharmaco- imputation algorithms such as the k-nearest neigh-
logical treatments, etc.). Since an extremely high bors [93]. Filtering outlier decreases artifacts, pre-
number of information derives from mass spec- vents technical biases, and improves the statistical
trometry, bioinformatics data analysis represents a power of the subsequent analyses. In most cases,
crucial aspect. Optimal design of the experiment this also reduces sample noise.
is critical to reduce cost and maximize informa- Data transformation and testing. Usually, a
tion. Quantitative data are traditionally assessed small number of replicates produce nonnormal
14 Translational Research Methods: Basics of Renal Molecular Biology 437

distributions of proteome profiles. In most cases, variables [89]. Pearson’s or Spearman’s correla-
this requires log2 transformation of data sets prior tion coefficient and the Euclidean distance aggre-
of using classical univariate parametric gation method are the most widely used clustering
approaches, such as the student’s t-test or the strategies for proteomics data analysis.
analysis of variance (ANOVA). Often, nonpara- These analyses can also be represented as “heat
metric tests are mandatory; these are based on the maps,” in which the abundance of each variable is
analysis of median and are less sensitive than converted in a color scale (Fig. 5c). This should be
parametric tests [94]. If multiple hypotheses are best performed after Z-Score standardization.
tested simultaneously, a priori control of the error Frequency tests. These tests are used for nom-
is required, generally using the family wise error inal or ordinal scales and include the McNemar,
rate (FWER), the Bonferroni correction, or the Chi-square, and Fisher’s exact tests. Odds ratio
false discovery rate (FDR) procedures (OR) can be measured. When the OR is close to
[95]. After an FDR analysis, the statistical perfor- 1, the odds of healthy controls and patients are
mance is usually expressed as q-values, which similar. OR greater than 1 indicates increased risk,
represent the corrected p-value after FDR and while OR lower than 1 indicates protection against
gives a more accurate indication of the level of the risk. Interpretation of these results requires the
false positivity for a given cutoff value. determination of the confidence intervals.
Volcano plot. The volcano plot is the most
widely used graphical expression to give a quick
glance on results based on univariate statistical Protein Interaction, Metabolic
tests. In this graph representation, the log2 of the Pathways, and Systems Biology
fold change for each spot/peptide is plotted
against the log10 of its p-value (Fig. 5a). Definition of interactive networks further refines
Multivariate statistic: principal component the analysis and is useful to identify pivotal pro-
analysis and heat maps. Multivariate data analy- teins. In Cytoscape, for example, the analysis can
sis allows to reduce the complexity of samples be performed following a simple work flow that
[96]. In the case of biological fluids, it allows to uses plug-in software. The first step consists in
identify combination of spots/peptides that could importing data to generate an interactive network
be of value in identifying and characterizing com- based on univariate/multivariate results. The sec-
plex mixtures. Principal component analysis ond step consists in matching data with public
(PCA) generates new variables, termed “compo- database. Together, this generates an interactive
nents,” which condense the variability of the sam- network where proteins that are linked by nodes
ples (Fig. 5b). Individual components in PCA do and edges represent potential interactions (e.g.,
not correlate with each other are and analyzed physical, co-localization, co-expression, common
separately to see how much they contribute to pathways, etc). Results can be filtered to reduce
the overall variance of data. Plots of each principal complexity. Overall, this approach can provide
component provide graphical views of the data insights on cellular pathways and can identify
structure and allow defining clusters and outliers. molecular mechanisms of diseases (Fig. 5d).
When specific groups of proteins can be distin-
guished, variables that correlate mostly with the
principal component are analyzed, a process Application Fields to Renal Diseases
referred to as loading matrix. Data clustering anal-
ysis is usually performed to identify proteins with Genome-Wide Association in Primary
similar behavior. This technique is complemen- Glomerulonephritis
tary to multivariate statistics. Graphical represen-
tation of pathways and visualization of integrated In recent years, two main glomerular diseases
data sets usually improve data interpretation have been successfully tackled using GWAS
and can also be helpful to select candidate approaches, namely, membranous nephropathy
438 G.M. Ghiggeri et al.

Fig. 5 Analysis of complex sets of data deriving from of the node indicates the degree of overexpression (black)
proteomics: (a) volcano plot; (b) principal component or underexpression (gray) of the proteins in patients com-
analysis (PCA); (c) heat map; (d) example of systems pared to controls. Physical interaction between proteins is
biology of a complex trait that identifies pathways and represented by solid lines while pathways by broken line.
networks associated patterns of protein expression identi- Different shapes represent proteins with different functions
fied by univariate statistical analysis. The network is (diamond = enzymes; triangle = transporters; circle =
graphically displayed with proteins as nodes and the bio- other)
logical relationships between the nodes as lines. The color

and IgA nephropathy. By studying only 556 phospholipase A2 receptor (PLA2R1) and one
European patients affected by membranous on chromosome 6p21 containing the gene
nephropathy and 1,832 controls, Stanescu and encoding HLA complex class II HLA-DQ alpha
colleagues identified genome-wide significant chain 1 (HLA-DQA1) [36]. Interestingly,
signals in two loci: one on chromosome 2q24 PLA2R1 has been previously shown to be the
containing the gene encoding M-type target of the autoimmune response for initiation
14 Translational Research Methods: Basics of Renal Molecular Biology 439

of primary membranous nephropathy [97], thus Copy-Number Variations in Congenital


providing direct functional support to the genetic Anomalies of the Kidney and Urinary
findings. The effect size for both variants was Tract
unusually large for classic GWAS studies, with
an OR of almost 80 when both alleles are present Rare CNVs have been associated to multiple
in homozygosity, supporting a very strong effect human phenotypes, including neurodeve-
of innate immunity and PLA2R1 in the pathogen- lopmental diseases (intellectual disability, autism,
esis of disease. schizophrenia, epilepsy), cardiac defects, lung
IgA nephropathy (IGAN) is the most com- disease, craniofacial malformations, and others
mon primary glomerulonephritis in the world, [43–50]. The role of CNVs in renal disease was
with striking racial differences in prevalence. unknown until 2012, when a first adequately sized
Compared to subject of European descent, the study was conducted on 522 patients with kidney
disease is much more common in Asians and malformations and almost 14,000 controls
rare in Africans, suggesting a strong genetic [51]. Children for this study were recruited in
contribution. A GWAS was conducted in 1,194 multiple centers in Europe and at Children’s Hos-
IGAN cases and 902 controls of Chinese Han pital, Philadelphia. CNV calls were derived from
ancestry and was replicated in 1,950 cases and genome-wide high-density SNP arrays. Burden
1,920 controls from Asia and Europe. These analysis between cases and controls showed a
studies identified five novel loci: three indepen- striking enrichment of large rare CNVs in cases
dent loci were located in the major histocompat- compared to controls, and these structural variants
ibility complex, one tagged a common deletion were mainly deletions containing one or more
of CFHR1 and CFHR3 at chromosome 1q32, coding element. Per-genome analysis suggested
and the last locus was located on chromosome that kidney malformations could be attributable to
22q12 in an intergenic region [34]. These five a CNV of 250 kb or larger in at least 17 % of the
loci were found to explain 4–7 % of the disease cases, indicating that rare, genic submicroscopic
variance and up to a tenfold variation in genomic unbalances are a major source of genetic
interindividual risk. Interestingly, the IgA variation predisposing to renal agenesis and
nephropathy risk allele frequencies mirrored hypodysplasia. Analysis of individual large rare
the known variation in disease prevalence CNVs surprisingly identified 34 independent
among Asian, European, and African known genomic disorders in 55 individuals, con-
populations and uncovered a previously stituting 10.5 % of the entire CAKUT cohort.
unrecognized north–south gradient in IgAN The most frequently identified pathogenic struc-
prevalence across Europe suggesting complex tural variant was the 1.4 Mb deletion at the
selective pressures [34, 98]. Very recently, a chromosome 17q12 locus associated to the renal
novel GWAS on IGAN studying over 20,000 cysts and diabetes (RCAD) syndrome. The other
individuals across three continents identified 33 genomic disorders were present in single
six new genome-wide significant loci: four in individuals or only in few patients (e.g., 1q21
ITGAM-ITGAX, VAV3, and CARD9 and two deletion, Wolf–Hirschhorn syndrome,
new independent signals at HLA-DQB1 and Potocki–Lupski syndrome, DiGeorge syndrome,
DEFA. This study also replicated previously Phelan–McDermid syndrome). Thus, known
reported signals and showed an increased burden copy-number disorders are a frequent cause of
of risk alleles in younger patients. Interestingly, renal malformations. The analysis of cases with-
the genetic risk score computed with all variants out known copy-number syndromes identified
was found to strongly correlate with helminth 38 additional large, rare, genic structural variants
diversity, suggesting a role for the interaction in 32 individuals (6.1 %) representing novel can-
between intestinal pathogens and humans in didate genomic disorders. Altogether, these data
driving the disease prevalence across population show that one in six children with kidney
of different races and ethnicities [35]. malformations carries a large structural variant
440 G.M. Ghiggeri et al.

diagnostic of known or novel syndromes. Inter- libraries are available and partially overcome the
estingly, nearly all of these variants have been limitation due to cell specificity, but they do not
previously associated to neurodevelopmental phe- completely reflect the cell protein and peptide
notypes that develop later in life, with potential repertoire. Advances in proteomics technology
impact on risk stratification and individualization filled this gap allowing to build cell-specific pro-
of care. tein maps; the podocyte is a relevant example.
Podocytes represent partially differentiated epi-
thelial cells expressing a specialized repertoire of
Next-Generation Sequencing in Renal proteins that play a key role in many renal dis-
Disease eases. The full definition of the podocyte protein
composition is currently in progress and will soon
The establishment of NGS techniques and, in be available to study pathological conditions
particular, of exome sequencing has resulted in a [101]. Currently, most data have been obtained
dramatic acceleration in gene identification in on podocyte cell lines [102]. Future studies may
many Mendelian renal phenotypes [10, 14–17, directly use podocytes expanded from kidney
20, 99, 100]. NGS has been particularly critical biopsies or from urines of patients with specific
in facilitating gene discovery for traits character- glomerular diseases. These studies also allow
ized by very high genetic heterogeneity and small defining antigens that are targeted in autoimmune
pedigree size. For example, exome sequencing diseases [97, 103, 104].
coupled to linkage analysis in a pedigree segre-
gating with autosomal dominant urinary tract
malformations has recently allowed to identify a Proteomic Analysis of Biological Fluids:
new gene related to these diseases. Specifically, The Normal Urine Map
genome-wide linkage analysis had identified in
this family five candidate regions, corresponding Biological fluids are potentially an important
to ~2 % of the total genome. Subsequent exome source of information in medicine. The plasma
sequencing identified 24 potentially pathogenic compartment contains important diagnostic
variants shared between two affected individuals, markers or causative molecules of renal diseases.
two of which localized to the regions identified by The expression of most plasma proteins cannot be
linkage analysis; of these, only one variant segre- assayed by recombinant DNA technologies, as
gated with the disease in the entire family, namely, these are generally not synthesized by circulating
a splice mutation in the DSTYK gene, encoding a cells. Proteomics represents, therefore, a major
dual specificity serine/threonine and tyrosine tool to study plasma composition in health and
kinase. Resequencing of the DSTYK gene in in diseases. In nephrotic syndrome, for example,
311 additional patients with congenital abnormal- proteomics has been used to search for permeabil-
ities of the kidneys and urinary tract (CAKUT) ity factors and to characterize oxidized protein
identified other rare damaging variants of this products. Using Nat/SDS-PAGE analysis, albu-
gene in 7/311 (2.3 %) unrelated CAKUT patients. min and other proteins, such as α1-anti-trypsine,
These findings were further supported by experi- have been shown to undergo posttranslational
mental data in mice and zebrafish [15]. modifications that include fragmentation, poly-
merization, and formation of adducts [105]. Oxi-
dation end products [106] in specific conditions
The Building of the Podocyte may also underscore pathologic processes.
Protein Map Likewise, proteomic approaches can also be
used to characterize urine samples (Fig. 6). As
Out of approximately 20,000 genes that are the urine proteomic map is nearly completed
encoded in the human DNA, only a portion is [107], researchers are now attempting to use pro-
ubiquitous. Tissue- and cell-specific cDNA teomics to identify urinary fingerprints of specific
14 Translational Research Methods: Basics of Renal Molecular Biology 441

Precipitate are not enough to be reproducible. Several, if not


most small urinary peptides, originate ex vivo
85 directly from proteolysis of plasma proteins, and
Untreated it is unclear whether they correlate with significant
22
18 Vesicles biological events. A number of these peptides
4 28
12 42 appear, however, to be biologically active, and
18
12 98 some have been shown to correlate with diseases.
154 744
18 25 Only few studies have attempted to analyze
48 404 small urinary peptide as fingerprints in the diag-
76 36
27 nosis and staging of renal diseases. Most are
41 18
20
derived from digestion of urinary albumin.
16 56 Decramer et al. [111] have utilized capillary elec-
172 51
trophoresis and tandem mass to characterize the
37 29 83
349 346 urine peptide composition in newborns with
hydronephrosis secondary to ureteropelvic junc-
CPLL-beads Unbound
tion: quantitative changes of a type V
preprocollagen α2 chain fragment were found to
Fig. 6 Urine protein composition as a result of a multistep be predictive, with a reasonable sensitivity, of the
procedure [107]. The diagram shows proteins identified by clinical evolution. Similarly, by tandem mass
mass spectrometry during successive fractionation steps
spectrometry, protein chip immunoassay, and
SELDI-TOF, O’Riordan et al. have identified
two peptides of 4.7 and 4.4 kDa derived from
renal diseases. Until now, these studies have been defensin1 and α1antichymotrypsin, respectively,
disappointing and have shown that even distantly and showed that their ratio correlates with epi-
related renal diseases share similar patterns of sodes of acute rejection [112]. Obviously, most
urine protein composition. The level of accuracy of these findings need to be confirmed with large-
of urinary biomarkers allowed to differentiate glo- scale prospective studies. Nonetheless, they rep-
merular from tubular involvement but was insuf- resent one of the forefronts of proteomic research
ficient to characterize glomerular lesions and to in human diseases and may provide in the near
guide treatment [108–110]. Recently, it has been future less invasive diagnosis and monitoring
evaluated that the total number of urinary proteins tools.
is in the range of 3–4.000 [107]; it is reasonable to Recent advances in a new science branch,
expect that in the near future, studies based on a referred to as “metabolomics,” also warrant men-
large number of urinary proteins will expand this tion. Several examples suggest that metabolomics
field and will allow to identify patterns of protein may provide in the future new insights in the
excretion that will have clinical applications. diagnosis and understanding of renal diseases
such as in Fanconi syndrome [113] and/or in
various forms of glomerulonephritis [114].
Peptidome, Degradome,
and Metabolomics
Autoantibodies
Plasma and urine also contain small peptides
(<5 Kda) that are difficult to analyze. Several Analysis of tissue obtained from kidney biopsies
methodological hurdles still need to be overcome currently represents a direct approach to human
before applying the analysis of these small pep- autoimmune diseases. Ultraprecise techniques of
tides in human medicine. Currently, results microdissection, such as laser capture, allow
obtained by different techniques, such as SELDI repetitive analysis of minute amounts of renal
and LC-ESI-MS/MS, are rarely concordant and tissue [115]. In combination with proteomics
442 G.M. Ghiggeri et al.

techniques (i.e., characterization of podocyte pro- 11. Ng SB, Bigham AW, Buckingham KJ, et al. Exome
teins recognized by eluted autoantibodies), this sequencing identifies MLL2 mutations as a cause of
Kabuki syndrome. Nat Genet. 2010;42:790–3.
approach has been particularly successful in mem- 12. Ng SB, Buckingham KJ, Lee C, et al. Exome
branous nephropathy [97, 101, 103] and human sequencing identifies the cause of a Mendelian disor-
lupus nephritis [104]. In 2009, the identification of der. Nat Genet. 2010;42:30–5.
IgG autoantibodies against the M-type-phospho- 13. O’Roak BJ, Vives L, Girirajan S, et al. Sporadic
autism exomes reveal a highly interconnected protein
lipase A2 receptor (PLA2R) in patients with network of de novo mutations. Nature. 2012;485:
membranous nephropathy (MN) has represented 246–50.
a landmark discovery; subsequent studies have 14. Sanna-Cherchi S, Burgess KE, Nees SN, et al. Exome
clearly demonstrated that PLA2R positivity iden- sequencing identified MYO1E and NEIL1 as candi-
date genes for human autosomal recessive steroid-
tifies primary MN patients, which has important resistant nephrotic syndrome. Kidney Int. 2011;80:
clinical and therapeutic implications [97]. Addi- 389–96.
tional studies have implicated other proteins that 15. Sanna-Cherchi S, Sampogna RV, Papeta N,
are normally expressed in podocytes, such as et al. Mutations in DSTYK and dominant urinary
tract malformations. N Engl J Med. 2013;369:621.
aldose reductase and superoxide dismutase 2, as 16. Westland R, Bodria M, Carrea A, et al. Phenotypic
novel targets of autoimmunity [103, 115, 116] and expansion of DGKE-associated diseases. J Am Soc
have also shown that different autoantibodies may Nephrol. 2014;25:1408.
be deposited in the same kidney at the same time 17. Boyden LM, Choi M, Choate KA, et al. Mutations in
kelch-like 3 and cullin 3 cause hypertension and elec-
or sequentially [116]. trolyte abnormalities. Nature. 2012;482:98–102.
18. Romberg N, Al Moussawi K, Nelson-Williams C,
et al. Mutation of NLRC4 causes a syndrome of
References enterocolitis and autoinflammation. Nat Genet.
2014;46:1135–9.
1. Astbury WT. Molecular biology or ultrastructural 19. TG, HDL Working Group of the Exome Sequencing
biology? Nature. 1961;190:1124. Project NHL, Blood Institute, et al. Loss-of-function
2. Lander ES, Linton LM, Birren B, et al. Initial mutations in APOC3, triglycerides, and coronary dis-
sequencing and analysis of the human genome. ease. N Engl J Med. 2014;371:22–31.
Nature. 2001;409:860–921. 20. Chaki M, Airik R, Ghosh AK, et al. Exome capture
3. Venter JC, Adams MD, Myers EW, et al. The reveals ZNF423 and CEP164 mutations, linking renal
sequence of the human genome. Science. ciliopathies to DNA damage response signaling. Cell.
2001;291:1304–51. 2012;150:533–48.
4. Lander ES, Botstein D. Mapping complex genetic 21. Keramati AR, Fathzadeh M, Go GW, et al. A form of
traits in humans: new methods using a complete the metabolic syndrome associated with mutations in
RFLP linkage map. Cold Spring Harb Symp Quant DYRK1B. N Engl J Med. 2014;370:1909–19.
Biol. 1986;51:49–62. 22. Metzker ML. Sequencing technologies – the next
5. Lander ES, Schork NJ. Genetic dissection of complex generation. Nat Rev Genet. 2010;11:31–46.
traits. Science. 1994;265:2037–48. 23. O’Roak BJ, Vives L, Fu W, et al. Multiplex targeted
6. Sanger F, Nicklen S, Coulson AR. DNA sequencing sequencing identifies recurrently mutated genes in
with chain-terminating inhibitors. Proc Natl Acad Sci autism spectrum disorders. Science. 2012;338:
U S A. 1977;74:5463–7. 1619–22.
7. International Human Genome Sequencing Consor- 24. Halbritter J, Diaz K, Chaki M, et al. High-throughput
tium. Finishing the euchromatic sequence of the mutation analysis in patients with a nephronophthisis-
human genome. Nature. 2004;431:931–45. associated ciliopathy applying multiplexed barcoded
8. Ng SB, Turner EH, Robertson PD, et al. Targeted array-based PCR amplification and next-generation
capture and massively parallel sequencing of sequencing. J Med Genet. 2012;49:756–67.
12 human exomes. Nature. 2009;461:272–6. 25. Halbritter J, Porath JD, Diaz KA, et al. Identification
9. Choi M, Scholl UI, Ji W, et al. Genetic diagnosis by of 99 novel mutations in a worldwide cohort of 1,056
whole exome capture and massively parallel DNA patients with a nephronophthisis-related ciliopathy.
sequencing. Proc Natl Acad Sci U S A. 2009; Hum Genet. 2013;132:865–84.
106:19096–101. 26. International HapMap Consortium. The International
10. Lemaire M, Fremeaux-Bacchi V, Schaefer F, et al. HapMap Project. Nature. 2003;426:789–96.
Recessive mutations in DGKE cause atypical 27. Risch N, Merikangas K. The future of genetic studies
hemolytic-uremic syndrome. Nat Genet. 2013;45: of complex human diseases. Science. 1996;273:
531–6. 1516–7.
14 Translational Research Methods: Basics of Renal Molecular Biology 443

28. Haines JL, Hauser MA, Schmidt S, et al. Complement 46. Serra-Juhe C, Rodriguez-Santiago B, Cusco I,
factor H variant increases the risk of age-related mac- et al. Contribution of rare copy number variants to
ular degeneration. Science. 2005;308:419–21. isolated human malformations. PLoS One. 2012;7:
29. Edwards AO, Ritter 3rd R, Abel KJ, Manning A, e45530.
Panhuysen C, Farrer LA. Complement factor H poly- 47. Brunetti-Pierri N, Berg JS, Scaglia F, et al. Recurrent
morphism and age-related macular degeneration. Sci- reciprocal 1q21.1 deletions and duplications associ-
ence. 2005;308:421–4. ated with microcephaly or macrocephaly and devel-
30. Klein RJ, Zeiss C, Chew EY, et al. Complement factor opmental and behavioral abnormalities. Nat Genet.
H polymorphism in age-related macular degenera- 2008;40:1466–71.
tion. Science. 2005;308:385–9. 48. Sanders SJ, Ercan-Sencicek AG, Hus V, et al. Multi-
31. Palotie A, Widen E, Ripatti S. From genetic discovery ple recurrent de novo CNVs, including duplications
to future personalized health research. N Biotechnol. of the 7q11.23 Williams syndrome region, are
2013;30:291–5. strongly associated with autism. Neuron.
32. Weiss KM, Terwilliger JD. How many diseases does 2011;70:863–85.
it take to map a gene with SNPs? Nat Genet. 49. Sebat J, Lakshmi B, Malhotra D, et al. Strong associ-
2000;26:151–7. ation of de novo copy number mutations with autism.
33. Terwilliger JD, Hiekkalinna T. An utter refutation of Science. 2007;316:445–9.
the “fundamental theorem of the HapMap”. Eur J 50. Stefansson H, Rujescu D, Cichon S, et al. Large recur-
Hum Genet. 2006;14:426–37. rent microdeletions associated with schizophrenia.
34. Gharavi AG, Kiryluk K, Choi M, et al. Genome-wide Nature. 2008;455:232–6.
association study identifies susceptibility loci for IgA 51. Sanna-Cherchi S, Kiryluk K, Burgess KE,
nephropathy. Nat Genet. 2011;43:321–7. et al. Copy-number disorders are a common cause of
35. Kiryluk K, Li Y, Scolari F, et al. Discovery of new risk congenital kidney malformations. Am J Hum Genet.
loci for IgA nephropathy implicates genes involved in 2012;91:987–97.
immunity against intestinal pathogens. Nat Genet. 52. Reddy UM, Page GP, Saade GR, et al. Karyotype
2014;46:1187–96. versus microarray testing for genetic abnormalities
36. Stanescu HC, Arcos-Burgos M, Medlar A, et al. Risk after stillbirth. N Engl J Med. 2012;367:2185–93.
HLA-DQA1 and PLA(2)R1 alleles in idiopathic 53. Wapner RJ, Martin CL, Levy B, et al. Chromosomal
membranous nephropathy. N Engl J Med. 2011; microarray versus karyotyping for prenatal diagnosis.
364:616–26. N Engl J Med. 2012;367:2175–84.
37. Lupski JR. Genomic rearrangements and sporadic 54. Abel HJ, Duncavage EJ. Detection of structural DNA
disease. Nat Genet. 2007;39:S43–7. variation from next generation sequencing data: a
38. Conrad DF, Pinto D, Redon R, et al. Origins and review of informatic approaches. Cancer Genet.
functional impact of copy number variation in the 2013;206:432–40.
human genome. Nature. 2010;464:704–12. 55. O’Farrell PH. High resolution two-dimensional elec-
39. Sebat J, Lakshmi B, Troge J, et al. Large-scale copy trophoresis of proteins. J Biol Chem. 1975;
number polymorphism in the human genome. Sci- 250:4007–21.
ence. 2004;305:525–8. 56. Anderson L, Anderson NG. High resolution
40. Redon R, Ishikawa S, Fitch KR, et al. Global variation two-dimensional electrophoresis of human plasma
in copy number in the human genome. Nature. proteins. Proc Natl Acad Sci U S A. 1977;74:5421–5.
2006;444:444–54. 57. Bruschi M, Musante L, Candiano G, et al. Soft
41. Iafrate AJ, Feuk L, Rivera MN, et al. Detection of immobilized pH gradient gels in proteome analysis:
large-scale variation in the human genome. Nat a follow-up. Proteomics. 2003;3:821–5.
Genet. 2004;36:949–51. 58. Manabe T, Jin Y. Analysis of protein/polypeptide
42. Wellcome Trust Case Control Consortium, Craddock N, interactions in human plasma using nondenaturing
Hurles ME, et al. Genome-wide association study of micro-2-DE followed by 3-D SDS-PAGE and
CNVs in 16,000 cases of eight common diseases and MS. Electrophoresis. 2007;28:2065–79.
3,000 shared controls. Nature. 2010;464:713–20. 59. Manabe T, Jin Y, Tani O. Assignment of human
43. Cooper GM, Coe BP, Girirajan S, et al. A copy num- plasma polypeptides on a nondenaturing 2-D gel
ber variation morbidity map of developmental delay. using MALDI-MS and PMF and comparisons with
Nat Genet. 2011;43:838–46. the results of intact protein mapping. Electrophoresis.
44. Greenway SC, Pereira AC, Lin JC, et al. De novo 2007;28:843–63.
copy number variants identify new genes and loci in 60. Candiano G, Santucci L, Petretto A, et al. High-
isolated sporadic tetralogy of Fallot. Nat Genet. 2009; resolution 2-DE for resolving proteins, protein
41:931–5. adducts and complexes in plasma. Electrophoresis.
45. Osoegawa K, Vessere GM, Utami KH, et al. Identifi- 2008;29:682–94.
cation of novel candidate genes associated with cleft 61. Panfoli I, Calzia D, Santucci L, Ravera S, Bruschi M,
lip and palate using array comparative genomic Candiano G. A blue dive: from ‘blue fingers’ to ‘blue
hybridisation. J Med Genet. 2008;45:81–6. silver’. A comparative overview of staining methods
444 G.M. Ghiggeri et al.

for in-gel proteomics. Expert Rev Proteomics. 78. Willer CJ, Li Y, Abecasis GR. METAL: fast and
2012;9:627–34. efficient meta-analysis of genomewide association
62. Alban A, David SO, Bjorkesten L, et al. A novel scans. Bioinformatics. 2010;26:2190–1.
experimental design for comparative 79. Colella S, Yau C, Taylor JM, et al. QuantiSNP: an
two-dimensional gel analysis: two-dimensional dif- Objective Bayes Hidden-Markov Model to detect and
ference gel electrophoresis incorporating a pooled accurately map copy number variation using SNP
internal standard. Proteomics. 2003;3:36–44. genotyping data. Nucleic Acids Res. 2007;35:
63. Bruschi M, Grilli S, Candiano G, et al. New iodo- 2013–25.
acetamido cyanines for labeling cysteine thiol resi- 80. Wang K, Li M, Hadley D, et al. PennCNV: an inte-
dues. A strategy for evaluating plasma proteins and grated hidden Markov model designed for high-
their oxido-redox status. Proteomics. 2009;9:460–9. resolution copy number variation detection in
64. Knowles MR, Cervino S, Skynner HA, et al. Multiplex whole-genome SNP genotyping data. Genome Res.
proteomic analysis by two-dimensional differential 2007;17:1665–74.
in-gel electrophoresis. Proteomics. 2003;3:1162–71. 81. McKenna A, Hanna M, Banks E, et al. The Genome
65. Rosenthal N. Molecular medicine. Recognizing Analysis Toolkit: a MapReduce framework for ana-
DNA. N Engl J Med. 1995;333:925–7. lyzing next-generation DNA sequencing data.
66. Ott J. Estimation of the recombination fraction in Genome Res. 2010;20:1297–303.
human pedigrees: efficient computation of the likeli- 82. Wang K, Li M, Hakonarson H. ANNOVAR: func-
hood for human linkage studies. Am J Hum Genet. tional annotation of genetic variants from high-
1974;26:588–97. throughput sequencing data. Nucleic Acids Res.
67. Lathrop GM, Lalouel JM, Julier C, Ott J. Strategies 2010;38:e164.
for multilocus linkage analysis in humans. Proc Natl 83. Cingolani P, Platts A, le Wang L, et al. A program for
Acad Sci U S A. 1984;81:3443–6. annotating and predicting the effects of single nucle-
68. Cottingham Jr RW, Idury RM, Schaffer AA. Faster otide polymorphisms, SnpEff: SNPs in the genome of
sequential genetic linkage computations. Am J Hum Drosophila melanogaster strain w1118; iso-2; iso-3.
Genet. 1993;53:252–63. Fly. 2012;6:80–92.
69. Kruglyak L, Daly MJ, Reeve-Daly MP, Lander 84. Kumar P, Henikoff S, Ng PC. Predicting the effects of
ES. Parametric and nonparametric linkage analysis: coding non-synonymous variants on protein function
a unified multipoint approach. Am J Hum Genet. using the SIFT algorithm. Nat Protoc. 2009;4:
1996;58:1347–63. 1073–81.
70. Gudbjartsson DF, Jonasson K, Frigge ML, Kong 85. Adzhubei IA, Schmidt S, Peshkin L, et al. A method
A. Allegro, a new computer program for multipoint and server for predicting damaging missense muta-
linkage analysis. Nat Genet. 2000;25:12–3. tions. Nat Methods. 2010;7:248–9.
71. Abecasis GR, Cherny SS, Cookson WO, Cardon 86. Cooper GM, Stone EA, Asimenos G, et al. Distribu-
LR. Merlin – rapid analysis of dense genetic maps tion and intensity of constraint in mammalian geno-
using sparse gene flow trees. Nat Genet. 2002; mic sequence. Genome Res. 2005;15:901–13.
30:97–101. 87. Kircher M, Witten DM, Jain P, O’Roak BJ, Cooper
72. Sobel E, Lange K. Descent graphs in pedigree analy- GM, Shendure J. A general framework for estimating
sis: applications to haplotyping, location scores, and the relative pathogenicity of human genetic variants.
marker-sharing statistics. Am J Hum Genet. Nat Genet. 2014;46:310–5.
1996;58:1323–37. 88. Wu MC, Lee S, Cai T, Li Y, Boehnke M, Lin X. Rare-
73. Hinrichs A, Lin JH, Reich T, Bierut L, Suarez BK. variant association testing for sequencing data with
Markov chain Monte Carlo linkage analysis of a the sequence kernel association test. Am J Hum
complex qualitative phenotype. Genet Epidemiol. Genet. 2011;89:82–93.
1999;17 Suppl 1:S615–20. 89. Koskinen VR, Emery PA, Creasy DM, Cottrell
74. Purcell S, Neale B, Todd-Brown K, et al. PLINK: a JS. Hierarchical clustering of shotgun proteomics
tool set for whole-genome association and data. Mol Cell Proteomics. 2011;10:M110 003822.
population-based linkage analyses. Am J Hum 90. Stessl M, Noe CR, Lachmann B. Influence of image-
Genet. 2007;81:559–75. analysis software on quantitation of two-dimensional gel
75. Lee AB, Luca D, Klei L, Devlin B, Roeder electrophoresis data. Electrophoresis. 2009;30:325–8.
K. Discovering genetic ancestry using spectral graph 91. R Core Team. R: a language and environment for
theory. Genet Epidemiol. 2010;34:51–9. statistical computing. Vienna: R Foundation for Sta-
76. Lee AB, Luca D, Roeder K. A spectral graph tistical Computing; 2012.
approach to discovering genetic ancestry. Ann Appl 92. Ejigu BA, Valkenborg D, Baggerman G, et al.
Stat. 2010;4:179–202. Evaluation of normalization methods to pave the
77. Li Y, Willer CJ, Ding J, Scheet P, Abecasis way towards large-scale LC-MS-based metabolomics
GR. MaCH: using sequence and genotype data to profiling experiments. OMICS. 2013;17:473–85.
estimate haplotypes and unobserved genotypes. 93. Pedreschi R, Hertog ML, Carpentier SC, et al. Treat-
Genet Epidemiol. 2010;34:816–34. ment of missing values for multivariate statistical
14 Translational Research Methods: Basics of Renal Molecular Biology 445

analysis of gel-based proteomics data. Proteomics. antitrypsin in glomerular diseases associated with
2008;8:1371–83. nephrotic syndrome. J Am Soc Nephrol.
94. Fay MP, Proschan MA. Wilcoxon-Mann–Whitney or 2006;17:3139–48.
t-test? On assumptions for hypothesis tests and mul- 106. Musante L, Bruschi M, Candiano G, et al. Character-
tiple interpretations of decision rules. Stat Surv. ization of oxidation end product of plasma albumin
2010;4:1–39. ‘in vivo’. Biochem Biophys Res Commun. 2006;
95. Glickman ME, Rao SR, Schultz MR. False discovery 349:668–73.
rate control is a recommended alternative to 107. Santucci L, Candiano G, Petretto A, et al. From hun-
Bonferroni-type adjustments in health studies. J Clin dreds to thousands: widening the normal human
Epidemiol. 2014;67:850–7. Urinome (1). J Proteomics. 2014;112C:53.
96. Jensen KN, Jessen F, Jorgensen BM. Multivariate 108. Ngai HH, Sit WH, Jiang PP, Thongboonkerd V, Wan
data analysis of two-dimensional gel electrophoresis JM. Markedly increased urinary preprohaptoglobin
protein patterns from few samples. J Proteome Res. and haptoglobin in passive Heymann nephritis: a dif-
2008;7:1288–96. ferential proteomics approach. J Proteome Res.
97. Beck Jr LH, Bonegio RG, Lambeau G, et al. M-type 2007;6:3313–20.
phospholipase A2 receptor as target antigen in idio- 109. Shui HA, Huang TH, Ka SM, Chen PH, Lin YF, Chen
pathic membranous nephropathy. N Engl J Med. A. Urinary proteome and potential biomarkers asso-
2009;361:11–21. ciated with serial pathogenesis steps of focal segmen-
98. Kiryluk K, Li Y, Sanna-Cherchi S, et al. Geographic tal glomerulosclerosis. Nephrol Dial Transplant.
differences in genetic susceptibility to IgA nephropa- 2008;23:176–85.
thy: GWAS replication study and geospatial risk anal- 110. Varghese SA, Powell TB, Budisavljevic MN,
ysis. PLoS Genet. 2012;8:e1002765. et al. Urine biomarkers predict the cause of glomerular
99. Saisawat P, Kohl S, Hilger AC, et al. Whole-exome disease. J Am Soc Nephrol. 2007;18:913–22.
resequencing reveals recessive mutations in TRAP1 111. Decramer S, Wittke S, Mischak H, et al. Predicting the
in individuals with CAKUT and VACTERL associa- clinical outcome of congenital unilateral ureteropelvic
tion. Kidney Int. 2014;85:1310–7. junction obstruction in newborn by urinary proteome
100. Gee HY, Ashraf S, Wan X, et al. Mutations in EMP2 analysis. Nat Med. 2006;12:398–400.
cause childhood-onset nephrotic syndrome. Am J 112. O’Riordan E, Orlova TN, Podust VN, et al. Charac-
Hum Genet. 2014;94:884–90. terization of urinary peptide biomarkers of acute
101. Prunotto M, Farina A, Lane L, et al. Proteomic anal- rejection in renal allografts. Am J Transplant.
ysis of podocyte exosome-enriched fraction from nor- 2007;7:930–40.
mal human urine. J Proteomics. 2013;82:193–229. 113. Vilasi A, Cutillas PR, Maher AD, et al. Combined
102. Hays T, Ma’ayan A, Clark NR, et al. Proteomics anal- proteomic and metabonomic studies in three genetic
ysis of the non-muscle myosin heavy chain forms of the renal Fanconi syndrome. Am J Physiol
IIa-enriched actin-myosin complex reveals multiple Renal Physiol. 2007;293:F456–67.
functions within the podocyte. PLoS One. 2014;9: 114. Psihogios NG, Kalaitzidis RG, Dimou S, Seferiadis
e100660. KI, Siamopoulos KC, Bairaktari ET. Evaluation of
103. Bruschi M, Carnevali ML, Murtas C, et al. Direct tubulointerstitial lesions’ severity in patients with glo-
characterization of target podocyte antigens and merulonephritides: an NMR-based metabonomic
auto-antibodies in human membranous glomerulone- study. J Proteome Res. 2007;6:3760–70.
phritis: alfa-enolase and borderline antigens. J Prote- 115. Murtas C, Bruschi M, Carnevali ML, et al. In vivo
omics. 2011;74:2008–17. characterization of renal auto-antigens involved in
104. Bruschi M, Sinico RA, Moroni G, et al. Glomerular human auto-immune diseases: the case of membra-
autoimmune multicomponents of human lupus nous glomerulonephritis. Proteomics Clin Appl.
nephritis in vivo: alpha-enolase and annexin AI. J 2011;5:90–7.
Am Soc Nephrol. 2014;25:2483. 116. Murtas C, Allegri L, Ghiggeri GM. Circulating
105. Candiano G, Musante L, Bruschi M, et al. Repetitive antipodocyte antibodies in membranous nephropathy:
fragmentation products of albumin and alpha1- new findings. Am J Kidney Dis. 2013;62:12–5.
Translational Research Methods:
The Value of Animal Models in Renal 15
Research

Jordan Kreidberg

Contents Introduction
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
Institutional Oversight . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447 The use of animal models has been an essential
Animal Models of Kidney Disease . . . . . . . . . . . . . . . . . . 448 aspect of nearly all areas of nephrological
Genetic Models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448 research since its earliest days. Research on kid-
ney formation and malformation, physiology
Genetic Approaches with Known Genes:
Genotype to Phenotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449 and pathophysiology, immunological injury,
Gene Targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449 and tolerance or transplant rejection all depend
Conditional Gene Targeting . . . . . . . . . . . . . . . . . . . . . . . . . 449 on the use of animal experimentation. This chap-
Animal Models Using RNAi Approaches . . . . . . . . . . . 453
ter will emphasize genetic approaches that uti-
Transgenic Mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456
Mutagenesis in Zebrafish . . . . . . . . . . . . . . . . . . . . . . . . . . . 458 lize animals, as this area has shown the great
progress in the development of novel technolo-
Gene Identification: Phenotype to Genotype . . . . . 458
Mutational Screens to Obtain New Phenotypes gies that have had great impact in all areas of
and Identify Genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 458 nephrology.
Gene Identification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
Implications of Genome Sequencing . . . . . . . . . . . . . . 461
Institutional Oversight
Other Animals in Nephrology Research . . . . . . . . . . 462
Models of Renal Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463 There is an increasing public awareness of the
Models of Immunological Injury . . . . . . . . . . . . . . . . . . 463 use of animals in research, and with this, comes
Transplant Models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 463 an increasing concern about the appropriateness
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 464 of the use of animals and whether much of the
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 464
research that does involve animal models could
be accomplished using nonanimal models.
Therefore, it is important to note that all animal
research in the United States and presumably in
most other countries must be evaluated and
approved by institutional committees, generally
known as the IACUC (Institutional Animal
Care and Use Committee) before any experi-
mentation may commence. These regulatory
J. Kreidberg (*)
Children’s Hospital Boston, Boston, MA, USA committees are charged with evaluating animal
e-mail: jordan.kreidberg@childrens.harvard.edu protocols to make certain of the following
# Springer-Verlag Berlin Heidelberg 2016 447
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_14
448 J. Kreidberg

criteria: (1) that animals are used in an ethical of animals that can be studied consequently
manner, with proper use of anesthetics or anal- decrease. For this reason, some studies may
gesics to minimize or eliminate any source of begin with a zebrafish, Xenopus, or rodent model
pain during experimentation, (2) that animals and then progress to a larger model once the
are indeed required for the specific research in rodent model establishes the feasibility of the
question, (3) that minimal numbers are used, hypothesis under study. The size of an animal
(4) that large animals are not used when smaller may be important to the extent that it affects the
ones would suffice, (5) and that the investiga- ability to perform surgical manipulations or phys-
tors are trained and knowledgeable about proper iological measurements. However, since it has
use of animals. Furthermore, the US Depart- become increasingly desirable to obtain physio-
ment of Agriculture (USDA) and the National logical measurements on various strains of knock-
Institutes of Health Office of Laboratory out mice, the equipment available to perform
Animal Welfare (NIH-OLAW) provide constant these measurements has improved and become
oversight for large animal research, the former commercially available.
through the use of frequent and usually
unannounced visits to animal facilities with
research institutions and the latter by publishing Genetic Models
detailed regulations concerning animal use that
serve as guidelines for IACUCs and serving as Animal models of disease that have a genetic basis
the NIH agency to whom IACUCs report vio- may either result from spontaneous or induced
lations in NIH-funded use of animals in mutations. Spontaneous mutations or phenotypes
research. Additionally, over 900 academic and are those noticed either by chance or through the
industrial animal research facilities in 39 coun- directed observation of large numbers of mice that
tries worldwide are accredited by AAALAC were not otherwise treated to induce a mutation. In
International (Association for Assessment and contrast, induced mutations are those resulting
Accreditation of Laboratory Animal Care Inter- from the treatment of mice with irradiation or
national), a private organization that sends mutagenic agents known to introduce point muta-
reviewers every 3 years to rigorously inspect tions or deletions into the genome.
and certify animal research facilities. Despite The past 25 years have witnessed an explosion
these several layers of oversight, in the end it in the use of genetic approaches to understand
is up to the principal investigator to be thought- development and physiology, and thus they will
ful about whether their intended experimental receive appropriate emphasis in this chapter.
approach will yield sufficiently important and Several genetic approaches are available for use
worthwhile results to justify the use of labora- with animal model systems. A gene of interest
tory animals. may be mutated using gene targeting, or
expressed in transgenic mice in such a way to
interfere with its normal function. On the other
Animal Models of Kidney Disease hand, it is possible to start with a phenotype of
interest, which could either be obtained as a
The selection of an animal model for some aspect spontaneous mutation or from mice treated with
or type of kidney disease takes several factors into a mutagen, and an effort is made to identify the
consideration. Most importantly, the similarity to mutated gene responsible for the phenotype.
human disease that can be observed in a particular Genetic approaches using gene-targeted or trans-
model is taken into account. Other important fac- genic mice are useful for a wide variety of devel-
tors include the cost of the animals involved: the opmental and physiological studies in which
cost of maintaining animals larger than rodents there is a need to study the function of a
increases dramatically with size, and the numbers known gene.
15 Translational Research Methods: The Value of Animal Models in Renal Research 449

Genetic Approaches with Known which renders the gene unable to be expressed.
Genes: Genotype to Phenotype This ES cell would in essence be heterozygous for
a mutation in the targeted gene, and heterozygous
Gene Targeting ES cells can be isolated and expanded to provide a
population for injection into blastocysts. There-
Gene targeting was originally used to introduce a fore, by combining the ES cell technology and
deletion or interruption into a gene of interest, homologous recombination, it became possible
using the scheme shown in Fig. 1, such that it to target mutations into genes in ES cells and
could be determined whether mice would be able then introduce ES cells carrying these mutations
to develop in the absence of that gene’s function. into blastocysts, finally obtaining a mutant adult
In cases where a gene was shown not to be essen- mouse.
tial for development, the homozygous mutant In a typical experiment, gene-targeted ES cells
mouse might serve as a useful model in which to would contain one mutated allele and one normal
study the role of a specific gene in a physiological or wild-type allele for the gene under study. The
or disease process. For example, targeted dele- targeted ES cells would be injected into preimplan-
tions of the Wt1 [1], Pax2 [2], GDNF [3–5], tation blastocysts, and groups of these blastocysts
Wnt4 [6], and BMP7 [7, 8], among others, would be introduced into female mice that were
showed these genes to be essential for various previously hormonally primed to allow implanta-
aspects of early kidney development. On the tion of the injected blastocysts into their uteri, to
other hand, the absence of many immunology- begin a pregnancy. The resultant mice from these
related genes does not result in any developmental injections are termed “chimeras,” because any spe-
impairment, but these mice have served as useful cific cell is either derived from an ES cells or the
models to study the role of the immune system in original injected embryo, i.e., the chimeric mouse
transplant rejection. essentially has four parents, the male and female
The advent of gene targeting was made possi- that provided the blastocyst and the male and
ble through the use of two technologies developed female that provided the embryo from which the
mainly in the 1980s. The first was the develop- ES cell line in use was originally derived. In the
ment of tissue culture conditions that allowed best cases, a chimera might be nearly entirely
embryonic stem (ES) cell lines to be grown indef- derived from the ES cells. Among the tissues that
initely in culture while retaining their totipotency ES cells contribute to are the germ cells: spermato-
[9]. ES cells grown in culture could then be intro- cytes or oocytes. When ES cells heterozygous for a
duced into mouse preimplantation embryos or mutation are used to make a chimera, germ cells
blastocysts and become fully integrated into derived from the ES cells have a 50 % chance of
those embryos such that their descendant cells carrying the mutant rather than the wild-type allele.
would give rise to all developmental lineages Therefore, mating chimeric and wild-type mice can
that are found in adult mice [10]. The second result in some of the offspring being true heterozy-
technology involved the use of homologous gotes for the mutated gene. After obtaining both
recombination to introduce mutations into mam- male and female heterozygotes, they can be mated
malian genes [11–13]. As shown in Fig. 1, when to obtain homozygous mutant embryos or mice,
long stretches of genomic DNA in recombinant depending on whether or not the gene is essential
DNA constructs are introduced into cells in cul- for development.
ture, this DNA will, at variable and often quite low
frequency, recombine into the locus from which
the genomic DNA was originally derived. There- Conditional Gene Targeting
fore, homologous recombination of the correctly
designed genomic fragment can be used to intro- The process described in the preceding section
duce a deletion or insertion into a genomic locus, results in the inactivation of a target gene from
450 J. Kreidberg

Probe
BamH1 BamH1
a Vector
Neo

BamH1 BamH1
Chromosome

BamH1 BamH1 BamH1


Chromosome
Neo

ICM
b Gene-targeted c
ES cells
+/+ +/− −/−

Blastocysts
Reimplantation

Chimeric mouse

d Chimera Wild Type

x
+ / −mix+ / + +/+

Germ-line transmission

x
+/−
+/−

Phenotype of
homozygous mutants ?
−/−

Fig. 1 Gene targeting in mice. (a) The scheme for Restriction sites for restriction enzyme BamH1A are
targeting a deletion of an exon in embryonic stem cells. shown. The replacement vector is constructed such that
Exons are shown as black boxes along a chromosome. the neomycin resistance gene (neo) is shown as an open
15 Translational Research Methods: The Value of Animal Models in Renal Research 451

the beginning of embryogenesis. In this situation, are experimental approaches for expressing Cre in
an embryo will become nonviable at the first point temporally and spatially specific manners or both.
at which expression of the inactivated gene Spatial- or lineage-specific expression of Cre is
becomes essential for survival. However, it may most often obtained by placing the Cre cDNA
be highly desirable to study the function of a gene downstream of a known tissue-specific promoter.
product in many later events during development Sometimes this is achieved by using homologous
or adult life. Conditional gene targeting allows the recombination to insert the Cre gene into the
inactivation of a gene in particular tissues or at genomic locus of a gene with known tissue-
particular times during development or adult life specific expression, such that Cre replaces the
[14–16]. This technology has been developed first exon of that gene. Temporally specific
more recently and has proved more difficult to expression of Cre has proved more difficult to
employ on a widespread basis thus far, for reasons obtain. One approach is to regulate Cre using the
that will be discussed. tetracycline system for inducible gene expression
The general approach to conditional gene [17]. The other approach makes use of a fusion
targeting is shown in Fig. 2. This is a variation protein consisting of Cre and a portion of the
on traditional gene targeting, in that it also relies estrogen receptor that confers steroid-mediated
on homologous recombination to introduce a seg- nuclear localization [18, 19]. The latter is modi-
ment of recombinant DNA into the locus of a gene fied to bind tamoxifen or tamoxifen derivatives
in ES cells. However, whereas traditional instead of estrogen. The Cre-modified estrogen
gene targeting inactivates the gene, conditional receptor fusion protein will remain in the cyto-
gene targeting must modify the gene such that it plasm and therefore not be able to mediate
can be expressed until such time as its inactivation site-specific recombination of LOX sites, until
is desired. The most commonly used approach tamoxifen is administered to the mouse to induce
involved the insertion of LOX sites, which are nuclear translocation of the Cre fusion protein.
34 base pair sites involved in site-specific recom- This system can be used to induce recombination
bination by Cre recombinase, and enzyme origi- in embryos, when tamoxifen is administered to
nally derived from a bacteriophage [15]. Since pregnant mice. The major obstacle to employing
LOX sites are rather small, it is usually possible conditional gene targeting on a widespread basis
to insert them in introns where they have no effect is the availability of promoter/enhancer elements
on gene expression. By placing two LOX sites in a that are able to confer robust tissue- or cell
gene to flank an exon, Cre can be used to inacti- lineage-specific expression of Cre recombinase.
vate a gene by recombining out the DNA segment However, an increasing number of mouse strains
containing the exon that was situated between the are available that express Cre recombinase in var-
two LOX sites, thus inactivating the gene. There ious cell lineages within the developing and adult

Fig. 1 (continued) box, in place of one of the exons. strains with different coat colors, then the chimeric
An external probe specifically does not overlap with the mouse will have a variegated coat color pattern on its fur,
replacement vector. A double homologous recombination providing an indication of its overall extent of chimerism.
results in the integration of the vector into the chromo- In the best cases, the resultant mouse is nearly entirely
some, thus replacing the exon with the Neo gene. The derived from ES cells. (c) Shows a possible pattern
BamH1 site within the neo gene results in a shorter obtained in a Southern blot, based on the scheme shown
BamH1 restriction fragment detected by probe after in (a), using the external probe. A wild-type mouse shows
homologous recombination. (b) ES cells can be injected only the longer band. A heterozygous mouse shows both
through a micropipette into a blastocyst, where they the wild-type and gene-targeted band, and the homozygous
become part of the inner cell mass. The injected blastocyst mutant shows only the shorter band, due to the presence of
is introduced into the uterus of a hormonally primed mouse the BamH1 site in the neo gene. (d) The mating involved in
and gives rise to a chimeric mouse, partially derived from obtaining germ line transmission of the mutation and sub-
the ES cells and partially from the original inner cell mass sequently obtaining homozygous mutant mice
cells. If the ES cells and blastocysts are derived from
452 J. Kreidberg

Vector LOX LOX FRT FRT


Neo

Chromosome

Homologous recombination in ES cells

Chromosome LOX LOX FRT FRT


Neo

1. Inject Es cells into blastocysts to make chimeric mice


2. Mate heterozygous mice with Flp deleter mice to remove neo

Chromosome LOX LOX FRT

1. Breed heterozygous mice with


Cre-expressing mice, and breed to
homozygosity.

X
tissue specific
promoter
Cre

Fig. 2 Conditional gene targeting. The targeting vector is neo gene. Mice without the neo gene, but still containing
different from the previous figure in that LOX sites flank the exon flanked by LOX sites, are mated with mice
the exon that will eventually be deleted and the Neo gene is expressing Cre in a particular tissue or cell type, or
flanked by Frt sites. The vector is incorporated into the expressing an inducible Cre, to obtain the conditional
chromosome through homologous recombination, and ES knockout. The breeding scheme shown in the figure is
cells with this knock-in are used to make chimeric mice, oversimplified. In the actual experiment, a more compli-
and germ line transmission is obtained. Although the LOX cated breeding scheme is required to obtain a mouse that is
sites should not interfere with expression of the gene, the homozygous for alleles with LOX sites and that also has
Neo gene is likely to interfere with normal gene expres- the Cre-expressing transgene. An alternative is to breed
sion. However, in most cases, mice will tolerate one inac- mice with the conditional allele with mice carrying a tra-
tive gene, as long as the other allele is functional. After ditional knockout. This has the advantage that to obtain the
obtaining heterozygous mice, they are mated with conditional knockout, Cre must only recombine one, and
Flp-deleter mice that express Flp recombinase in germ not two, pair of LOX sites in each cell
cells. Flp will recombine the FRT sites and eliminate the
15 Translational Research Methods: The Value of Animal Models in Renal Research 453

kidney [20]. It is possible to obtain conditional ZFNs and TALENs are based on protein back-
knockouts restricted to nephron progenitor cells bones that can be custom designed to bind specific
[21], podocytes [21–24], proximal tubules nucleotide sequences. In the case of ZFNs (Fig. 4a),
[25–27], thick ascending limb [28], ureteric bud algorithms are used to predict amino acid sequences
[29], juxtaglomerular cells, and collecting ducts that will bind different trinucleotide sequences.
[30]. It is also possible to obtain vascular knock- These nucleotide-binding peptides are incorporated
outs [31–33], though not restricted to the kidney. within a zinc finger backbone peptide. With suffi-
As more tissue-promoter elements become cient length, these sequences become unique in the
available, conditional gene targeting promises to target genome. For TALENs (Fig. 4b), the specific-
have a large impact on genetic approaches to kid- ity is similarly conferred by peptide sequence, but
ney disease. As noted above, there are many genes instead of trinucleotide recognition, “TALE” motifs
expressed both in developing and adult kidney, recognize single nucleotides. In both systems, these
where the knockout of the gene results in embry- “designer” peptides are linked to the Fok1 endonu-
onic lethality. This precludes study of how that clease. As shown in Fig. 4, by designing two ZFNs
product of that gene might function in postnatal or TALENs to flank an intended mutagenesis
kidneys, or why a mutation in that gene leads to site, two Fok1 peptides are brought together, thus
kidney disease in humans. It also raises the question creating an active Fok1 enzyme that produces a
of why humans carrying such mutations are able double-stranded break. Repair of this break by
survive, albeit with a genetic disease, when mice nonhomologous end joining typically introduces a
carrying mutations in the same gene do not survive small deletion into the genome.
embryogenesis. Sometimes this is simply because The Crispr/Cas9 system (Fig. 4c) is distinct in
mice and humans differ in their respective require- using a complex of a “guide” or gRNA (Crispr) and
ments for specific genes, but more often, it is protein endonuclease (Cas9) to introduce a targeted
because humans with genetic disease often have cut into a specific sequence within a genome. Thus
point mutations that lead to partial loss of function, far, it appears to be more cost-efficient than the use
whereas mouse knockouts often involve complete of TALENs or ZFNs because it is much easier to
loss of function mutations. Conditional gene synthesize a custom gRNA to target a specific
targeting can sometimes offer a solution to this genomic sequence than to design and synthesize
problem, by allowing normal gene expression dur- sequence-specific peptides as required to use
ing embryogenesis, and then inactivating a gene in TALENs or ZFNs. These systems offer the possi-
adult mice. Alternatively, there are variations on the bility of not only introducing small deletions but
Cre-LOX approach that allow the introduction of also editing the genome by introducing along with
point mutations into mice. The introduction of the TALEN, ZFN, or Crispr/Cas9 reagent, a small
point mutations into mice has been greatly facili- DNAwith flanking sequences homologous to either
tated by recent advancements that facilitate homol- side of the cut, and an internal “edited” sequence.
ogous recombination into BACs (bacterial artificial By homologous recombination, this small piece of
chromosomes) in E. coli (Fig. 3) [34–36]. BACs DNA can be inserted into the site of the cut, thereby
are used as they contain large amounts of genomic placing a new sequence into the genome. This tech-
DNA and thus are ideal for use as gene-targeting nology offers the ability to repair a mutated
vectors. The longer length of BACs compared with sequence or introduce an entirely new sequence
shorter genomic clones should improve the fre- into a specific site in the genome, giving us unprec-
quency of homologous recombination in ES cells. edented ability to repair or alter genomes [40, 41].

Crispr/Cas9 and Talen Knockouts


The zinc finger nuclease (ZFN), TALEN, and Animal Models Using RNAi Approaches
Crispr/Cas9 systems are presently revolutionizing
the ability to place mutations into animal genomes RNAi technologies have had great impact across
[37–39] (Fig. 4). the breadth of molecular biology and the study of
454 J. Kreidberg

SacB Kan DNA fragment with


a b homologous ends
and SacB, Kanamycin
markers

a 2 b 3
1

Select for Kanamycin resistance

X 2 Knock in with
a b mutation in
exon 2.

SacB Kan

a b 3
1

Select against SacB


X

a 2 b 3
1

Additional round of homologous


recombination to place Neo gene
flanked by FRT sites in between two
exons.

1. Linearize BAC and use to target ES cells.


2. Remove Neo gene using Flp deleter mice.
3. Use ES cells to derive chimeric mice.

Fig. 3 Gene targeting using BAC clones. Homologous with the same homologous ends but containing a mutated
recombination is done in E. coli instead of in ES cells. In exon 2, denoted by the “X,” is introduced into the E. coli
the first step, a DNA fragment is prepared that contains the containing the BAC. Selection against SacB will obtain
kanamycin resistance positive selectable marker and the BACs in which the mutated exon 2 has replaced the select-
SacB-negative selectable marker and which also contains able markers. A third round of homologous recombination
homologous ends (A and B, each about 50–60 bp) is is used to insert the neo gene, flanked by FRT sites, so that
introduced into E. coli. This fragment can usually be pre- the BAC can be used for homologous recombination in ES
pared by PCR, using primers that contain the homologies cells. As shown, this scheme is used to introduce point
to the genomic region, and also to a vector containing the mutations or small deletions into a gene. It can also be used
selectable markers. Usually, a strain of E. coli is used that to construct conditional knockout vectors, similar to those
allows transient activation of the enzymes required for shown in Fig. 2. An additional use is to knock in a green
homologous recombination. Selection for kanamycin fluorescent protein (GFP) or β-galactosidase (LacZ)
resistance will obtain BAC clones where the selectable reporter gene into a locus to obtain information about
markers have recombined into the BAC. In a second patterns of gene expression
round of homologous recombination, A DNA fragment
15 Translational Research Methods: The Value of Animal Models in Renal Research 455

Fig. 4 Mutagenesis with a ZFNs ZFN_R


Talens and Crispr/Cas9
(Figure from Ref. [39]). (a) kl
ZFNs targeting a specific Fo
genomic sequence to
position Fok1
endonucleases in order to

l
k
Fo
introduce a double-stranded
break. (b) TALENs
similarly targeting a ZFN_L
sequence. The NI peptide
sequence binds adenosine,
b TALENs
HD binds cytosine, NK TALEN_L
binds guanosine, and NG FokI
5’ GCTTCTGACACAACTGTGTTCACTAGCAACCTCAAACAGACACCATGGTGCATCTG 3’
binds thymidine. By using 3’ CGAAGACTGTGTTGACACAAGTGATCGTTGGAGTTTGTCTGTGGTACCACGTAGAC 5’
specific arrangements of the FokI
FokI
NI, HD, NK, and NG TALEN_R
peptides, a peptide is
constructed to target a RVD: NI HD NK NG
specific sequence. (c) The
Crisrp/Cas9 system uses a Base: A C G T
gRNA designed to target a
c

G
specific sequence CRISPR/CAS9

UCGGUGC
AGCCACGGUGAAAAAAGUUC
(in green). (d) These
technologies can be used to

UAAAAUU CGAU A A
GUUUUAGAGCUA G A
3’-UUUU
disrupt genes by
introducing deletions

GAA
resulting from a double-
stranded break followed by Cas9 Guide
nonhomologous end joining RNA
(NHEJ), genome correction 5’-CNNNNNNNNNNNNNNNNNNN AAGGCUAGUCCGUUAUCAA
by homologous
N N CNNNNNNNNNNNNNNNNNNNNCC N
NN
recombination (HR) to NN NNNNNNNNNNNNNN- 5’
introduce a wild-type (WT) 3’-NNNNNNNN
DNA to repair the double- 5’-NNNNNNNN N N NNNNNNNNNNNNN- 3’
NN NN Chromosome
stranded break, or the N GNNNNNNNNNNNNNNNNNNNNGG N
introduction of a transgene PAM
by homologous
recombination 23 bp genomic target sequence

d Gene targeting by ZFNs, TALENs or CRISPR/Cas9

ZFNs, TALENs or CRISPR/Cas9


targeted DSB

WT donor Transgene donor


DNA DNA

Gene disruption Gene correction Transgene addition


by NHEJ by HR by HR

disease over the past 10 years [42]. RNAi is based complex, hybridizes to a complementary
on a mechanism common to both plant and ani- sequence in a mRNA, causes degradation or inhi-
mals whereby a short strand of RNA, that is asso- bition of translation of the mRNA [43, 44]. In the
ciated with a set of proteins known as the RISC natural setting, these small RNAs are encoded in
456 J. Kreidberg

the genome as microRNAs. The same molecular effects can also be found in cases where two pro-
machinery used by microRNAs to inhibit expres- teins heterodimerize, and an inactive mutant pro-
sion of mRNAs can be co-opted for use by tein is able to complex with its partner protein, but
siRNA (silencing RNA) or shRNA (short hairpin as before, the complex is inactive. Dominant-
RNA) that are ectopically expressed in cells. negative effects can be studied in animal models
siRNA refers to RNA duplexes that are usually using transgenic mice. Although gene-targeted
obtained commercially and transfected into cells. mice discussed in the previous sections can also
shRNA refers to small RNAs that are expressed be considered to be transgenic, because foreign
from plasmids or viral vectors that are introduced DNA is used to disrupt the endogenous gene, here
into cells. A major addition to the arsenal of the term “transgenic” is reserved for those mice in
approaches involving transgenic mice involves which foreign DNA has been inserted into the
the derivation of transgenic mice that express murine genome through pronuclear injection
shRNA molecules capable of reducing levels of [47–50]. In contrast to gene-targeting schemes in
mRNA for specific genes in specific tissues or which genes are modified in ES cells and ES cells
cell lineages [43–46]. shRNAs can be expressed are then used to derive chimeric mice, transgenic
either constitutively or conditionally using the strategies, DNA is directly microinjected into the
Cre-Lox system (Fig. 5). An advantage of pronucleus of a fertilized egg or zygote, and the
shRNA technology over gene knockouts is that injected zygotes are then reimplanted into the
it is quicker and cheaper, as mice do not need to oviduct of a hormonally primed female mouse.
be bred to homozygosity and also to carry the Cre The injected DNA is able to recombine by
transgene. Instead, a resulting phenotype can be nonhomologous or illegitimate recombination
obtained by mating a Cre-expressing mouse with into random locations within the genome and in
a mouse carrying a conditional shRNA transgene variable amounts from zygote to zygote. Once
and examining the first-generation offspring car- mice are derived from the injected zygotes, they
rying both transgenes. The major disadvantage in are tested to determine if they carry the injected
comparison with gene knockouts is that expres- DNA within their genomes as a transgene, and if
sion of the target gene may be variably reduced, they do, whether the transgene is expressed. By
and it may be necessary to examine several injecting DNA constructs that contain a tissue-
shRNA transgenic lines in order to obtain ones specific promoter and a mutated gene of interest,
that demonstrate efficient knockdown of the it is possible to study whether expression of the
desired gene. mutant gene leads to an observable phenotype. In
other instances, the gene to be expressed is not
mutated, and the experiment is designed to deter-
Transgenic Mice mine if overexpression or de novo expression of
the gene results in an observable phenotype or
As mentioned above, many mutations that result disease model. While the original transgenic stud-
in human disease are point mutations that result in ies used relatively short DNA constructs, more
“hypomorphic,” or partial loss of function alleles recent studies have used BAC (bacterial artificial
of a gene. In this case, a disease state may result chromosome)- or YAC (yeast artificial
from decreased activity of a gene product. In other chromosome)-based vectors whose much longer
cases, a point mutation or deletion mutation may stretches of DNA containing promoters and other
produce a protein that interferes with the function regulatory regions will hopefully confer more
of the normal gene; this is referred to as a faithful patterns of expression of the transgene
dominant-negative effect. This could occur in directed by those regulatory elements
instances where a protein requires [47, 51]. The great majority of transgenic work
homodimerization for activity, and dimerization has been done in murine models, but there have
of a wild-type and a mutant form of a protein leads been pioneering efforts in other species such as
to an inactive complex. Dominant-negative pigs and rats [52, 53].
15 Translational Research Methods: The Value of Animal Models in Renal Research 457

a Tissue specific cre transgene shRNA transgene

Promotor Cre U6-Promoter Sense loxP TTTTT loxP Anti-Sense TTTTT

Cre-mediated recombination of loxP sites

U6-Promoter Sense loxP Anti-Sense TTTTT

Sense
loxP
Anti-Sense

Dicer

b Sense
Anti-Sense

Processing to remove hairpin loop

Formation of RISC complex

RISC

Recognition of target mRNA


RISC
5’ 3’
mRNA

Translational repression
mRNA degradation

Fig. 5 Conditional expression of shRNA transgenes. (a) recombination of the first poly-T sequence. After removal
Mice containing a Cre recombinase-expressing transgene of the first poly-T sequence by Cre-mediated recombina-
and a shRNA transgene are crossed to obtain embryos or tion of the two LoxP sites, the transgene is fully transcribed
offspring mice containing both transgenes. The shRNA and terminates at the second poly-T sequence. The resul-
transgene contains a U6 promoter to direct RNA polymer- tant shRNA forms a hairpin loop by base pairing of the
ase III-mediated transcription that is terminated by a sense and antisense sequences. (b) The shRNA associates
poly-T sequence. It also contains a sense and antisense with DICER, an RNase III class enzyme that removes the
sequence, designed to target a mRNA, that will become hairpin loop. The double-stranded RNA then associates
the double-stranded RNA that associates with the RISC with the RISC complex, the sense strand is removed, and
complex to mediate the inhibitory effect on mRNA stabil- the RISC complex with the antisense RNA finds its target
ity or translation. Therefore, transcription of the shRNA mRNA and suppresses translation or decreases mRNA
transgene is interrupted prior to Cre-mediated stability
458 J. Kreidberg

Mutagenesis in Zebrafish Gene Identification: Phenotype


to Genotype
Possibly the single most important advancement in
the development of novel animal models for kidney Mutational Screens to Obtain New
disease over the past 20 years is the establishment Phenotypes and Identify Genes
of zebrafish (Danio rerio) as a major model for
understanding the genetic and physiological basis ENU mutagenesis: At present, several major
for disease. Zebrafish models of glomerular devel- efforts in several countries involve the use of
opment and disease and polycystic kidney disease N-ethyl-N-nitrosourea (ENU) to introduce small
have been particularly useful models for study of mutations throughout the mouse genome
human disease [54–60]. Most recently, the [88–93]. Similar approaches have been used
zebrafish has been more generalized as a model with zebrafish, and many interesting phenotypes
for exploring genetic variation in human disease have been obtained [94–98]. These large genome-
[61, 62]. Zebrafish are much less expensive to scale approaches, which can involve very large
maintain than mice (once a facility has been mouse or zebrafish colonies, are justified by the
constructed), and genetic tools to map zebrafish following arguments: (1) Most disease-related
mutations nearly equal or in some cases extend human mutations are caused by point mutations;
beyond those available for mice [63–65]. The therefore, a ENU-mutagenic approach may have a
zebrafish excretory system involves a pronephric greater chance of producing a phenotype resem-
duct and glomus that bears important similarity to bling a human disease than will gene-targeted
mammalian nephrons and has already been the mutations that usually completely inactivate a
subject of many research studies [54, 66–72]. gene. (2) An ENU-based approach does not rely
Two great advantages of zebrafish are (1) their on previous identification or cloning of the gene, i.
shorter developmental timing and (2) that develop- e., any gene is a theoretical target and can be
ment occurs in nearly transparent embryos that studied, to the extent that some degree of compro-
develop outside the mother, allowing for far greater mise in the gene product’s activity will result in an
observation and intervention than is possible with observable phenotype. The obvious disadvantage
rodent embryos. The zebrafish genome is presently in comparison with gene targeting is that a large
being sequenced at the Sanger Center [73], and amount of work lies between the observation of a
a full set of markers exists for gene mapping phenotype and the final identification of the
[64, 74, 75]. It is possible to make transgenic mutated gene. (3) Given a large enough effort, it
zebrafish [76–78], and gene targeting is now pos- should be possible to eventually “saturate” the
sible using genome editing approaches that utilize genome with mutations, i.e., examination of sev-
Crispr/Cas9 or Talens [79–82]. There have eral hundred thousand mutagen-treated mice or
also been large-scale efforts to saturate the zebrafish is likely to provide the opportunity to
zebrafish genome through insertional mutagenesis observe the effects of placing a mutation in every
[83–85]. An alternative to gene targeting is the use gene capable of causing an observable phenotype.
of morpholino oligonucleotides that inhibit expres- However, one important point remains to be men-
sion of specific genes against which the morpholino tioned that dramatically increases the labor and
is targeted; these can be injected into cells of early expense of an ENU-based effort. Most observable
embryos, and phenotypes can be observed at vari- phenotypes tend to be genetically recessive
ous developmental stages thereafter [86, 87]. In one instead of dominant, meaning that they are not
sense, morpholinos have an advantage over conven- apparent in the first-generation offspring of
tional gene knockouts, in that they can be designed mutagen-treated animals. Instead, it is necessary
to block expression of specific alternative splice to breed a second generation and then backcross it
forms [86], rather than all forms as is often the to the first-generation mice (or zebrafish),
case with gene knockouts in mice. resulting in a third generation (Fig. 6). Doing
15 Translational Research Methods: The Value of Animal Models in Renal Research 459

Screen for recessive phenotypes


ENU

Founder Wild type

Heterozygote Wild type


(with several mutations)

Backcross
to original
heterozygous parent
Heterozygote
(with subset of mutations)

Homozygote
(observable recessive
phenotypes)

Fig. 6 ENU mutagenesis. A scheme is depicted for find- carry a subset of these mutations. These heterozygotes
ing recessive phenotypes through ENU mutagenesis. are mated with wild-type mice to produce a second gener-
Dominant phenotypes require a less complicated approach, ation, which will carry a smaller subset of the original
as phenotypes will be apparent in the first generation mutations. These are then mated to the original heterozy-
derived from crossing founders with wild-type mice. In gous offspring of the founders, and 25 % of the offspring of
this scheme, a mutagenized male founder that probably this cross will be homozygous for any particular mutation
carries many mutations after mutagenesis is mated with a that was present in the second heterozygous generation
wild-type mouse to produce heterozygote offspring that

this on a large scale will result in many third- potential to identify genes involved in disease
generation animals that are now homozygous for progression, as well as those responsible for mor-
mutations resulting from the original mutagenic phogenetic processes.
treatment, and some will have observable pheno-
types that can be studied for biological interest
and to map the responsible gene. Gene Identification
These large-scale genetic approaches are not
only suited to study developmental anomalies. Microsatellite repeats and SNPs: Mapping sites
Some of the large-scale efforts on mouse muta- of induced or spontaneous mutations in mice has
genesis ongoing around the world will involve been greatly aided by the development of sets of
performing basic blood work and a urinalysis on microsatellite repeat markers and more recently
each mouse from the group being screened for by SNP microarray panels. Microsatellite repeats
new phenotypes. Thus, this approach has the used in mapping are stretches of CA dinucleotide
460 J. Kreidberg

repeats that are found interspersed throughout can also be done quantitatively in humans and
mammalian genomes [99, 100]. Typically, these rodents, such that decreased or increased signal
CA repeats contain 10–20 CA dinucleotides. on some SNPS relative to control yields informa-
These CA repeats are flanked by unique tion about genomic deletions or amplifications.
sequences, and thus it is possible to design pairs SNPs can be used as genetic markers similarly to
of PCR primers that correspond to these flanking the microsatellite repeats described in the previ-
sequences that will amplify the (CA)n sequence ous section. Therefore, a single microarray can
between the two primers. Within a genetically provide the same information as hundreds of
inbred strain of mice each individual mouse will PCR reactions. To be used in a genetic mapping
contain the same number of CA dinucleotides at experiment, the same type of interspecific cross
each repeat. However, similarly to the variation would be performed as described in the previous
observed between human individuals, different section, but instead of using PCR reactions to
inbred strains may differ in the number of CA analyze the segregation of microsatellite repeats,
dinucleotides at any particular repeat. By tracking a microarray is used for each individual to analyze
the segregation of a mutant phenotype with a the segregation of SNPs, to narrow the interval
particular microsatellite repeat, it was possible to that contains the gene being mapped.
determine first which chromosome harbored the Using SNP microarray panels and running a
mutation and then focus on microsatellite repeats microarray for each of the many mice produced
within a chromosome to further map the mutation from a cross between mutant and wild-type mice,
and eventually clone the mutated gene. it is now possible to identify SNP polymorphisms
SNPs, or single nucleotide polymorphisms, are that segregate with the mutant phenotype and
single base pair differences found between indi- much more quickly identify the genomic locus
viduals within a species. Among the human pop- containing the mutation.
ulation, SNPs tend to be found every 100–300 QTL and GWAS: Almost all human disease
base pairs within the genome and can be used as has a genetic component, whether it is the relative
a measure of genetic relatedness among, for susceptibility to an infectious agent, at one end of
example, ethnic groups or people in different geo- a spectrum, or a disease that is primarily due to a
graphic areas [101]. SNPs are also found in com- genetic mutation in a single gene, at the other end.
paring different inbred strains of mice and Between these two extremes lies most human
between the commonly used laboratory-inbred disease, whose etiology derives from a combina-
strains that are all derived from Mus musculus tion of genetic and environmental factors. In many
and other mouse strains that are “non-musculus” instances, the genetic component is due to the
that can be intercrossed with Mus musculus strains effects of more than one gene. In other instances,
to aid in genetic mapping, as described above a single gene might be responsible, but the phe-
[102]. In contrast, within a particular inbred strain notype is not absolute, but rather of variable pen-
of mice, there is by definition genetic homogene- etrance or severity. A genetic element that
ity, and SNPs should not be present between indi- contributes to a disease phenotype in a quantita-
viduals of the same strain. SNPs are also used to tive, as opposed to absolute, manner is referred to
map genes in zebrafish [75]. as a quantitative trait locus, or QTL [106]. It is
It is now possible to detect SNPs using probably not an overstatement to say that most
microarrays [103, 104], such that thousands of non-Mendelian genetic components of disease
oligonucleotides complementary to sequences occur as QTLs, and indeed, there are thousands
containing known SNPs can be arrayed on a sin- of known QTLs that have been reported [107].
gle microarray chip, and hybridized to an individ- A current approach to identifying QTLs in the
ual human or animal’s DNA to determine which human genome that increase (or decrease) risk of
of thousands of particular SNPs that individual the disease is referred to as GWAS (genome-wide
has in their genome [105]. Using comparative association studies), whereby the genomes of
genomic hybridization (CGH) microarrays, this many (up to thousands) of individuals are
15 Translational Research Methods: The Value of Animal Models in Renal Research 461

interrogated for SNP polymorphisms across the incompatibility between individuals with different
genome to determine the association of specific HLA haplotypes (in the case of humans) or
SNPs with the presence, absence, or severity of between mouse strains of different H2 haplotypes.
disease. This approach has found a wide use in the It is now known that haplotype units exist through-
nephrology community, examples being a search out mammalian genomes and that SNPs can be
for risk variants in diabetic nephropathy used as markers to define haplotypes [115–117]. It
[108–110] and search for additional variants that then follows that inbred mouse strains that more
further increase risk of chronic kidney disease in closely genetically related are likely to have the
individuals with APOL1 risk alleles [111–113]. same haplotype at a particular genomic location,
The disease severity in many animal models of whereas more distantly related strains would be
human disease also appears to be influenced by more likely to have different haplotypes
QTLs, and the identification of the responsible [115, 116]. Thus, when SNPs are being analyzed
gene(s) is a pursuit of many research laboratories in the progeny of a genetic cross in an attempt to
(e.g., [114]). In many examples, the presence of a narrow down the location of a candidate gene,
trait or disease may be determined in a Mendelian haplotype analysis provides an alternative to con-
pattern by a mutation in a single known gene, but sidering each SNP individually (see Fig. 7). For
the severity of the trait or disease may be example, if strains A and B are being studied with
influenced by so-called “modifier” genes that the aim of identifying a disease-related QTL that
show up as QTLs in a GWAS-type screen and confers greater disease severity in strain A than in
whose identification may be challenging. Impor- strain B, then genomic areas where strains A and B
tantly, comparisons of syntenic regions between share the same haplotype are unlikely to harbor the
human and rodent genomes have aided the search disease locus, whereas areas of the genome where
for modifier genes at QTLs, as it is often apparent they have differing haplotypes are more likely to
that a QTL identified in humans corresponds to a harbor the disease locus. Therefore, consideration
QTL for a similar phenotype in rodents of haplotypes, each of which may contain several to
[115]. Comparisons of syntenic regions can also hundreds of SNPs, may allow a means of
be complicated, as a region harboring a QTL in a narrowing down the location of candidate genes,
rodent genome, if not already delimited to a nar- by focusing on areas of the genome where the
row interval, may be syntenic with several regions stains differ in haplotype. In practice, such an anal-
of the human genome that are found on different ysis may involve multiple strains of mice and
chromosomes. On the other hand, comparisons of crosses between them in an effort to narrow down
syntenic regions between humans and rodents a genetic interval and physically locate a QTL
may also serve to delimit regions of interest and [120, 121]. Indeed, a recent effort known as the
facilitate the identification of the gene of interest. “collaborative cross” involves the intercrosses of
Haplotype analysis: Haplotype analysis has the several inbred strains of mice and then genotyping
potential to make gene identification by SNP map- and phenotyping resultant recombinant inbred
ping more efficient [115–117]. Haplotypes are strains [122, 123]. This represents an animal
defined as a group of genetic markers that are model version of a GWAS-like approach to identify
physically linked on chromosomes and that tend human disease genes.
to be inherited together more often than would be
predicted if genetic recombination events were ran-
dom and evenly distributed along chromosomes. Implications of Genome Sequencing
Human haplotype maps now contain over three
million SNPs [118, 119]. Perhaps the best known Sequencing of the entire human genome was com-
examples of haplotypes are the major histocompat- pleted in 2001 [124, 125] and of the mouse genome
ibility loci, HLA in humans and H2 in mice, in in 2002 [126]. These continue to be refined, with
which there is considerable polymorphism between more detailed reports and annotations of the
haplotypes that is manifested by transplant sequence of specific human chromosomes
462 J. Kreidberg

Strain published from 2003 to the present. In the 2008


a version of this chapter, it was noted that the geno-
1 2 3 4
mic sequences of approximately 180 organisms,
including bacteria, plants, and animals, have been
A A G G SNP1 reported. This number is now in the tens of thou-
Haplotype A T T T T sands, and the new frontier in human biology lies in
assembling complete genome sequences of thou-
C C C C sands of individuals. This rapidly expanding data-
base made possible by high-throughput sequencing
G G G G (also referred to as “next-generation” or “massively
parallel” sequencing) has transformed modern biol-
Haplotype B C C A A SNP2
ogy and the study of disease [127–129]. For exam-
G G G G ple, in disease gene identification studies described
above, it is no longer the situation that a disease
gene might be mapped to an area within a chromo-
A A A A
some that had never been sequenced, and the hunt
T T T C SNP3 for the gene becomes a large-scale sequencing pro-
Haplotype C
ject. Now, once an area is delimited, the candidate
G G G G
genes in that area are largely known from prior
C C C C genome sequencing, allowing much more directed
sequencing to be done in attempts to find disease-
b causing mutations in affected individuals. The
Strains Strains
Haplotype A
1, 2 3, 4
genome sequences for most, if not all, animals
used as disease models is also known, greatly
Strains Strains accelerating studies such as those involving
Haplotype B
1, 2 3, 4 disease-related gene identification in animal
models. In many of these accelerated efforts, only
Strains Strain coding exons are sequenced (“whole exome
Haplotype C
1, 2, 3 4
sequencing”), rather than the entire genome. The
advantage of whole exome sequencing is that it
Fig. 7 SNPs and haplotypes. (a) A highly simplified
schematic of SNPs and haplotypes is depicted. Four greatly reduces the amount of sequencing per indi-
mouse strains, labeled at top, are compared. A three vidual, and the corresponding disadvantage is that
nucleotide stretch is shown for haplotypes A and B and mutations outside coding regions will be missed.
four nucleotides for C, though in reality a haplotype may Thus, a major frontier in genome sequencing
span megabases and be defined by hundreds or more
SNPs. The sequences in haplotypes A, B, and C are not related to animal disease models is not so much
necessarily contiguous and might even be on different the sequencing of additional species as it is the
chromosomes. In the haplotype at locus A, SNP1 is pre- sequencing of multiple genomes among individ-
sent at the first nucleotide, in the haplotype at locus B, uals that vary in their severity of, or susceptibility
SNP2 is present at the second nucleotide, and in the
haplotype at locus C, SNP3 is present at the second nucle- to, a disease, so that we can increase our under-
otide. (b) Haplotype grouping defined by the SNPs. For standing of inter- and intraspecies genetic variation
haplotypes at loci A and B, strains 1 and 2 are the same and how it relates to disease susceptibility [130].
haplotype, and 3 and 4 define a different haplotype. For
haplotype at locus C, strains 1, 2, and 3 are the same
haplotype, and strain 4 is a different haplotype, suggesting
that strain 3 might be more closely related to strains Other Animals in Nephrology Research
1 and 2 than is strain 4. If strains 1 and 2 differ from
3 and 4 for a disease phenotype, it is more likely that the The emphasis on mice and zebrafish thus far in
gene is within the area covered by haplotypes A and B
than within haplotype C that is shared between strains this chapter is not meant to overlook the enormous
1, 2, and 3 benefit that has derived from the use of other
15 Translational Research Methods: The Value of Animal Models in Renal Research 463

species. Since the previous edition of this text- form of tubular injury involves temporary
book, gene targeting has been extended to rats obstruction of a ureter [189–191]. Chronic kidney
and rabbits, and the advent of TALENs and the disease has also been studied using a variety of
Crispr/CAS9 system will accelerate efforts in mouse and rat models, including Adriamycin-
other species. ENU mutagenesis has also been induced podocyte injury [192–194] and
used to obtain rat mutant models of disease and glomerular injury from nephrotoxic serum
gene mapping of disease phenotypes in rats has [161, 164, 195]. There are also several mouse,
yielded important insights [131–135]. Rats have rat, and other mammalian models for dominant
also been widely used in studies of nutrition as it and recessive forms of polycystic kidney disease
related to kidney development and disease that will be discussed in the respective chapters on
[136]. Furthermore, their larger size makes rats those diseases [196–198]. Combining these injury
more amenable to studies that require precise models with genetic models is an emerging fron-
physiological monitoring or imaging, though the tier in nephrology research.
technology to perform such studies in murine
models, to take advantage of knockout mouse
models, has also improved dramatically Models of Immunological Injury
[137–144].
Large animal research has also had an impor- There are many models of autoimmune injury to
tant legacy in nephrology that continues to this the kidney. A traditional model for a lupus-like
day. Historically, several animal models have autoimmune disease is the NZB mouse that has
been used to study renal physiology, including been studied for many years [199–203]. These
swine, sheep, guinea pigs, rabbits, rats, and mice develop autoantibodies similar to those
mice. Fetal lambs have been a particularly impor- observed in humans with systemic lupus
tant model in which developmental aspects of erythematosus and other related autoimmune dis-
physiology have been studied, particularly relat- orders. More recently, many strains of mice car-
ing to obstructive uropathy [145–157]. Studies in rying mutations in genes involved in the
large animals are also vital in efforts to use tissue regulation of the immune response have been
engineering to develop artificial tissues or in used to increase our understanding of the role
models of tissue regeneration [158–160]. As in the immune system plays in the onset and pro-
other situations, there is a constant need to balance gression of kidney disease [204–214]. These
the advantages of a large animal model with the knockout strains have allowed investigators to
lower costs of smaller animal models. begin a genetic dissection of genes involved in
autoimmune and other disorders. Transgenic tech-
nologies have also had an important impact in the
Models of Renal Failure development of new immunological models. One
particular contribution is the use of live-cell imag-
Approaches to the study of renal failure include ing that exploits fluorescent transgenic reporter
acute and chronic models. Acute renal failure has genes which mimic the expression of genes
been induced using pharmacological agents, expressed in specific immunological cell types
antisera against kidney tissue, or other antigens [215, 216]. These can be used either to trace the
in which immune complex formation leads to location of these cells in animals or to show evi-
glomerular disease [161–167]. Ischemia- dence of gene expression in an in vivo setting.
reperfusion models of acute renal failure are
achieved by temporarily ligating a renal artery
allowing study of the pathological processes Transplant Models
involved in tubular damage, as well as the effect
of various pharmacological treatments on the Animal models have been used extensively to
pathologic process (e.g., [168–188]). An alternate study transplant rejection and in efforts to
464 J. Kreidberg

understand how tolerance to transplanted tissue 3. Moore MW, Klein RD, Farinas I, Sauer H, Armani M,
may be improved. Over the past 20 years, there Philips H, et al. Renal and neuronal abnormalities in
mice lacking GDNF. Nature. 1996;382:76–9.
has been an extraordinary advancement in our 4. Pichel JG, Shen L, Sheng HZ, Granholm A-C,
mechanistic understanding of the immune func- Drago J, Grinberg A, et al. Defects in enteric inner-
tion, and this has been brought to bear on the vation and kidney development in mice lacking
study of transplant rejection and tolerance GDNF. Nature. 1996;382:73–6.
5. Sanchez MP, Silos-Santiago I, Frisen J, He B, Lira
[217–223]. Important models under study include SA, Barbacid M. Renal aegenesis and absence of
skin and heart transplants in mice and kidney enteric ganglions in mice lacking GDNF. Nature.
transplants in rats [224–231]. Additionally, it is 1996;382:70–4.
possible to produce “humanized” mice by 6. Kispert A, Vainio S, McMahon AP. Wnt-4 is a mes-
enchymal signal for epithelial transformation of meta-
transplanting human tissue into immunodeficient nephric mesenchyme in the developing kidney.
or irradiated mice whose immune system has been Development. 1998;125(21):4225–34.
reconstituted with human lymphocytes, thus 7. Dudley AT, Lyons KM, Robertson EJ. A requirement
allowing the study of human immune function in for bone morphogenetic protein-7 during develop-
ment of the mammalian kidney and eye. Genes Dev.
an animal model [232–235]. One area of research 1995;9(22):2795–807.
that remains controversial is that involving xeno- 8. Luo G, Hofmann C, Bronckers AL, Sohocki M,
grafts [236, 237]. Since the supply of human Bradley A, Karsenty G. BMP-7 is an inducer of
kidneys and other organs for transplants continues nephrogenesis, and is also required for eye develop-
ment and skeletal patterning. Genes Dev. 1995;9
to fall far short of the demand, there is a desirabil- (22):2808–20.
ity of determining whether nonhuman animals 9. Robertson EJ. Isolation of embryonic stem cells. In:
provide an alternative source of organs for trans- Robertson EJ, editor. Teratocarcinomas and embry-
plantation. The major concerns here include the onic stem cells: a practical approach. Oxford: IRL
Press; 1987.
strong immunological rejection to a xenograft that 10. Bradley A. Production and analysis of chimeric mice.
must be overcome and the danger that xenografts In: Robertson EJ, editor. Teratocarcinomas and
might serve as vectors for the introduction of embryonic stem cells: a practical approach. Oxford:
novel infectious agents into the human IRL Press; 1987. p. 113–51.
11. Thomas KR, Capecchi MR. Targeting of genes to
population. specific sites in the mammalian genome. Cold Spring
Harb Symp Quant Biol. 1986;51(1):1101–13.
12. Thomas KR, Deng C, Capecchi MR. High-fidelity
Summary gene targeting in embryonic stem cells by using
sequence replacement vectors. Mol Cell Biol.
1992;12(7):2919–23.
Animal models are of increasing importance in 13. Capecchi MR. The new mouse genetics: altering the
the study of kidney disease. An important shift genome by gene targeting. Trends Genet. 1989;5(3):
over the past 20 or more years is the use of rodent 70–6.
14. Orban PC, Chui D, Marth JD. Tissue- and site-
models and the use of genetic models. Large ani- specific DNA recombination in transgenic mice.
mal use remains an important aspect of these Proc Natl Acad Sci U S A. 1992;89(15):6861–5.
studies. The use of large versus small animals 15. Sauer B. Inducible gene targeting in mice using the
must take into account cost and ethical Cre/lox system. Methods. 1998;14(4):381–92.
16. Stricklett PK, Nelson RD, Kohan DE. The Cre/loxP
considerations. system and gene targeting in the kidney. Am J
Physiol. 1999;276(5 Pt 2):F651–7.
17. Furth PA, St. Onge L, Boger H, Gruss P, Gossen M,
References Kistner A, et al. Temporal control of gene expression
in transgenic mice by a tetracycline responsive pro-
1. Kreidberg JA, Sariola H, Loring JM, Maeda M, moter. Proc Natl Acad Sci U S A. 1994;91:9302–6.
Pelletier J, Housman D, et al. WT-1 is required for 18. Sohal DS, Nghiem M, Crackower MA, Witt SA,
early kidney development. Cell. 1993;74(4):679–91. Kimball TR, Tymitz KM, et al. Temporally regulated
2. Torres M, Gomez PE, Dressler GR, Gruss P. Pax-2 and tissue-specific gene manipulations in the adult
controls multiple steps of urogenital development. and embryonic heart using a tamoxifen-inducible
Development. 1995;121(12):4057–65. Cre protein. Circ Res. 2001;89(1):20–5.
15 Translational Research Methods: The Value of Animal Models in Renal Research 465

19. Verrou C, Zhang Y, Zurn C, Schamel WW, Reth recombineering using galK selection. Nucleic Acids
M. Comparison of the tamoxifen regulated chimeric Res. 2005;33(4):e36.
Cre recombinases MerCreMer and CreMer. Biol 35. Zhang XM, Huang JD. Combination of overlapping
Chem. 1999;380(12):1435–8. bacterial artificial chromosomes by a two-step
20. Gawlik A, Quaggin SE. Conditional gene targeting in recombinogenic engineering method. Nucleic Acids
the kidney. Curr Mol Med. 2005;5(5):527–36. Res. 2003;31(15):e81.
21. Park JS, Valerius MT, McMahon AP. Wnt/beta- 36. Testa G, Zhang Y, Vintersten K, Benes V, Pijnappel
catenin signaling regulates nephron induction during WW, Chambers I, et al. Engineering the mouse
mouse kidney development. Development. 2007;134 genome with bacterial artificial chromosomes to cre-
(13):2533–9. ate multipurpose alleles. Nat Biotechnol. 2003;21(4):
22. Moeller MJ, Sanden SK, Soofi A, Wiggins RC, 443–7.
Holzman LB. Two gene fragments that direct 37. Gaj T, Gersbach CA, Barbas 3rd CF. ZFN, TALEN,
podocyte-specific expression in transgenic mice. and CRISPR/Cas-based methods for genome engi-
J Am Soc Nephrol. 2002;13(6):1561–7. neering. Trends Biotechnol. 2013;31(7):397–405.
23. Wong MA, Cui S, Quaggin SE. Identification and [Research Support, N.I.H., Extramural Research Sup-
characterization of a glomerular-specific promoter port, U.S. Gov’t, Non-P.H.S. Review].
from the human nephrin gene. Am J Physiol Renal 38. Peng Y, Clark KJ, Campbell JM, Panetta MR, Guo Y,
Physiol. 2000;279:F1027–32. Ekker SC. Making designer mutants in model organ-
24. Eremina V, Wong MA, Cui S, Schwartz L, Quaggin isms. Development. 2014;141(21):4042–54. [Review].
SE. Glomerular-specific gene excision in vivo. J Am 39. Ramalingam S, Annaluru N, Chandrasegaran S. A
Soc Nephrol. 2002;13:788–93. CRISPR way to engineer the human genome.
25. Li H, Zhou X, Davis DR, Xu D, Sigmund CD. An Genome Biol. 2013;14(2):107. [Research Support,
androgen-inducible proximal tubule-specific Cre N.I.H., Extramural Research Support, Non-U.S.
recombinase transgenic model. Am J Physiol Renal Gov’t Research Support, U.S. Gov’t, Non-P.H.S.].
Physiol. 2008;294(6):F1481–6. 40. Mali P, Esvelt KM, Church GM. Cas9 as a versatile
26. Dworniczak B, Skryabin B, Tchinda J, Heuck S, tool for engineering biology. Nat Methods. 2013;10
Seesing FJ, Metzger D, et al. Inducible Cre/loxP (10):957–63. [Research Support, N.I.H., Extramural
recombination in the mouse proximal tubule. Neph- Research Support, Non-U.S. Gov’t Research Support,
ron Exp Nephrol. 2007;106(1):e11–20. U.S. Gov’t, Non-P.H.S. Review].
27. Rubera I, Poujeol C, Bertin G, Hasseine L, 41. Yang L, Mali P, Kim-Kiselak C, Church G. CRISPR-
Counillon L, Poujeol P, et al. Specific Cre/Lox recom- Cas-mediated targeted genome editing in human
bination in the mouse proximal tubule. J Am Soc cells. Methods Mol Biol. 2014;1114:245–67.
Nephrol. 2004;15(8):2050–6. [Research Support, N.I.H., Extramural].
28. Stricklett PK, Taylor D, Nelson RD, Kohan 42. Cheng JC, Moore TB, Sakamoto KM. RNA interfer-
DE. Thick ascending limb-specific expression of Cre ence and human disease. Mol Genet Metab. 2003;80
recombinase. Am J Physiol Renal Physiol. 2003;285 (1–2):121–8.
(1):F33–9. 43. Campbell TN, Choy FY. RNA interference: past, pre-
29. Marose TD, Merkel CE, McMahon AP, Carroll sent and future. Curr Issues Mol Biol. 2005;7(1):1–6.
TJ. Beta-catenin is necessary to keep cells of ureteric 44. Tijsterman M, Plasterk RH. Dicers at RISC; the mech-
bud/Wolffian duct epithelium in a precursor state. Dev anism of RNAi. Cell. 2004;117(1):1–3.
Biol. 2008;314(1):112–26. 45. Shukla V, Coumoul X, Deng CX. RNAi-based con-
30. Stricklett PK, Nelson RD, Kohan DE. Targeting ditional gene knockdown in mice using a U6 pro-
collecting tubules using the aquaporin-2 promoter. moter driven vector. Int J Biol Sci. 2007;3(2):91–9.
Exp Nephrol. 1999;7(1):67–74. 46. Coumoul X, Deng CX. RNAi in mice: a promising
31. Li WL, Cheng X, Tan XH, Zhang JS, Sun YS, approach to decipher gene functions in vivo.
Chen L, et al. Endothelial cell-specific expression of Biochimie. 2006;88(6):637–43.
Cre recombinase in transgenic mice. Yi Chuan Xue 47. Vintersten K, Testa G, Naumann R, Anastassiadis K,
Bao. 2005;32(9):909–15. Stewart AF. Bacterial artificial chromosome
32. Licht AH, Raab S, Hofmann U, Breier transgenesis through pronuclear injection of fertilized
G. Endothelium-specific Cre recombinase activity in mouse oocytes. Methods Mol Biol. 2008;415:
flk-1-Cre transgenic mice. Dev Dyn. 2004;229 83–100.
(2):312–8. 48. Feng G, Lu J, Gross J. Generation of transgenic mice.
33. Cattelino A, Liebner S, Gallini R, Zanetti A, Methods Mol Med. 2004;99:255–67.
Balconi G, Corsi A, et al. The conditional inactivation 49. Isola LM, Gordon JW. Transgenic animals: a new era
of the beta-catenin gene in endothelial cells causes a in developmental biology and medicine. Biotechnol-
defective vascular pattern and increased vascular fra- ogy. 1991;16:3–20.
gility. J Cell Biol. 2003;162(6):1111–22. 50. Gordon JW, Ruddle FH. Gene transfer into mouse
34. Warming S, Costantino N, Court DL, Jenkins NA, embryos: production of transgenic mice by pronu-
Copeland NG. Simple and highly efficient BAC clear injection. Methods Enzymol. 1983;101:411–33.
466 J. Kreidberg

51. Schedl A, Larin Z, Montoliu L, Thies E, Kelsey G, 67. Liu S, Lu W, Obara T, Kuida S, Lehoczky J, Dewar K,
Lehrach H, et al. A method for the generation of YAC et al. A defect in a novel Nek-family kinase causes
transgenic mice by pronuclear microinjection. cystic kidney disease in the mouse and in zebrafish.
Nucleic Acids Res. 1993;21(20):4783–7. Development. 2002;129(24):5839–46.
52. Nottle MB, Nagashima H, Verma PJ, Du ZT, Grupen 68. Briggs JP. The zebrafish: a new model organism for
CG, Ashman RJ, et al. Developments in transgenic integrative physiology. Am J Physiol Regul Integr
techniques in pigs. J Reprod Fertil Suppl. Comp Physiol. 2002;282(1):R3–9.
1997;52:237–44. 69. Serluca FC, Fishman MC. Pre-pattern in the proneph-
53. Filipiak WE, Saunders TL. Advances in transgenic rat ric kidney field of zebrafish. Development. 2001;128
production. Transgenic Res. 2006;15(6):673–86. (12):2233–41.
54. Majumdar A, Drummond IA. Podocyte differentia- 70. Majumdar A, Drummond IA. The zebrafish floating
tion in the absence of endothelial cells as revealed in head mutant demonstrates podocytes play an impor-
the zebrafish avascular mutant, cloche. Dev Genet. tant role in directing glomerular differentiation. Dev
1999;24(3–4):220–9. Biol. 2000;222(1):147–57.
55. Drummond I. Making a zebrafish kidney: a tale of two 71. Drummond IA. The zebrafish pronephros: a genetic
tubes. Trends Cell Biol. 2003;13(7):357–65. system for studies of kidney development. Pediatr
56. Hostetter CL, Sullivan-Brown JL, Burdine Nephrol. 2000;14(5):428–35.
RD. Zebrafish pronephros: a model for understanding 72. Majumdar A, Lun K, Brand M, Drummond
cystic kidney disease. Dev Dyn. 2003;228(3):514–22. IA. Zebrafish no isthmus reveals a role for pax2.1 in
57. Drummond IA. Zebrafish kidney development. tubule differentiation and patterning events in the
Methods Cell Biol. 2004;76:501–30. pronephric primordia. Development. 2000;127
58. Sun Z, Amsterdam A, Pazour GJ, Cole DG, Miller (10):2089–98.
MS, Hopkins N. A genetic screen in zebrafish iden- 73. Vogel G. GENOMICS: Sanger will sequence
tifies cilia genes as a principal cause of cystic kidney. Zebrafish genome. Science. 2000;290(5497):1671b.
Development. 2004;131(16):4085–93. [Research 74. Stickney HL, Schmutz J, Woods IG, Holtzer CC,
Support, U.S. Gov’t, P.H.S.]. Dickson MC, Kelly PD, et al. Rapid mapping of
59. Hentschel DM, Park KM, Cilenti L, Zervos AS, zebrafish mutations with SNPs and oligonucleotide
Drummond I, Bonventre JV. Acute renal failure in microarrays. Genome Res. 2002;12(12):1929–34.
zebrafish: a novel system to study a complex disease. 75. Bradley KM, Elmore JB, Breyer JP, Yaspan BL,
Am J Physiol Renal Physiol. 2005;288(5):F923–9. Jessen JR, Knapik EW, et al. A major zebrafish poly-
60. Hentschel DM, Mengel M, Boehme L, Liebsch F, morphism resource for genetic mapping. Genome
Albertin C, Bonventre JV, et al. Rapid screening of Biol. 2007;8(4):R55.
glomerular slit diaphragm integrity in larval zebrafish. 76. Damert A, Kusserow H. Generation of transgenic
Am J Physiol Renal Physiol. 2007;293(5):F1746–50. mice by pronuclear injection. Methods Mol Med.
61. Davis EE, Frangakis S, Katsanis N. Interpreting 2003;89:513–28.
human genetic variation with in vivo zebrafish assays. 77. Gaiano N, Allende M, Amsterdam A, Kawakami K,
Biochim Biophys Acta. 2014;1842(10):1960–70. Hopkins N. Highly efficient germ-line transmission of
[Review]. proviral insertions in zebrafish. Proc Natl Acad Sci U
62. Niederriter AR, Davis EE, Golzio C, Oh EC, Tsai IC, S A. 1996;93(15):7777–82.
Katsanis N. In vivo modeling of the morbid human 78. Gaiano N, Hopkins N. Introducing genes into zebrafish.
genome using Danio rerio. J Vis Exp. 2013;(78): Biochim Biophys Acta. 1996;1288(1):O11–4.
e50338. [Research Support, N.I.H., Extramural 79. Bedell VM, Wang Y, Campbell JM, Poshusta TL,
Research Support, Non-U.S. Gov’t Video-Audio Starker CG, Krug 2nd RG, et al. In vivo genome
Media]. editing using a high-efficiency TALEN system.
63. Fishman MC. Zebrafish genetics: the enigma of Nature. 2012;491(7422):114–8. [Research Support,
arrival. Proc Natl Acad Sci U S A. 1999;96 N.I.H., Extramural Research Support, Non-U.S.
(19):10554–6. Gov’t Research Support, U.S. Gov’t, Non-P.H.S.].
64. Shimoda N, Knapik EW, Ziniti J, Sim C, Yamada E, 80. Hwang WY, Fu Y, Reyon D, Maeder ML, Tsai SQ,
Kaplan S, et al. Zebrafish genetic map with 2000 Sander JD, et al. Efficient genome editing in zebrafish
microsatellite markers. Genomics. 1999;58(3):219–32. using a CRISPR-Cas system. Nat Biotechnol.
65. Knapik EW, Goodman A, Ekker M, Chevrette M, 2013;31(3):227–9. [Research Support, N.I.H., Extra-
Delgado J, Neuhauss S, et al. A microsatellite genetic mural Research Support, Non-U.S. Gov’t].
linkage map for zebrafish (Danio rerio). Nat Genet. 81. Cade L, Reyon D, Hwang WY, Tsai SQ, Patel S,
1998;18(4):338–43. Khayter C, et al. Highly efficient generation of heri-
66. Drummond IA, Majumdar A, Hentschel H, Elger M, table zebrafish gene mutations using homo- and
Solnica-Krezel L, Schier AF, et al. Early development heterodimeric TALENs. Nucleic Acids Res. 2012;40
of the zebrafish pronephros and analysis of mutations (16):8001–10. [Research Support, N.I.H., Extramural
affecting pronephric function. Development. Research Support, Non-U.S. Gov’t Research Support,
1998;125(23):4655–67. U.S. Gov’t, Non-P.H.S.].
15 Translational Research Methods: The Value of Animal Models in Renal Research 467

82. Shin J, Chen J, Solnica-Krezel L. Efficient homolo- the development of the zebrafish, Danio rerio. Devel-
gous recombination-mediated genome engineering in opment. 1996;123:1–36.
zebrafish using TALE nucleases. Development. 98. Solnica-Krezel L, Schier AF, Driever W. Efficient
2014;141(19):3807–18. recovery of ENU-induced mutations from the
83. Amsterdam A, Hopkins N. Retrovirus-mediated zebrafish germline. Genetics. 1994;136(4):1401–20.
insertional mutagenesis in zebrafish. Methods Cell 99. Dietrich WF, Copeland NG, Gilbert DJ, Miller JC,
Biol. 1999;60:87–98. Jenkins NA, Lander ES. Mapping the mouse genome:
84. Talbot WS, Hopkins N. Zebrafish mutations and func- current status and future prospects. Proc Natl Acad
tional analysis of the vertebrate genome. Genes Dev. Sci U S A. 1995;92(24):10849–53.
2000;14(7):755–62. 100. Brown DM, Matise TC, Koike G, Simon JS, Winer
85. Amsterdam A, Hopkins N. Mutagenesis strategies ES, Zangen S, et al. An integrated genetic linkage
in zebrafish for identifying genes involved in devel- map of the laboratory rat. Mamm Genome. 1998;9
opment and disease. Trends Genet. 2006;22(9): (7):521–30.
473–8. 101. Wang DG, Fan JB, Siao CJ, Berno A, Young P,
86. Draper BW, Morcos PA, Kimmel CB. Inhibition of Sapolsky R, et al. Large-scale identification, map-
zebrafish fgf8 pre-mRNA splicing with morpholino ping, and genotyping of single-nucleotide polymor-
oligos: a quantifiable method for gene knockdown. phisms in the human genome. Science. 1998;280
Genesis. 2001;30(3):154–6. (5366):1077–82.
87. Scholpp S, Brand M. Morpholino-induced knock- 102. Tsang S, Sun Z, Luke B, Stewart C, Lum N,
down of zebrafish engrailed genes eng2 and eng3 Gregory M, et al. A comprehensive SNP-based
reveals redundant and unique functions in midbrain–- genetic analysis of inbred mouse strains. Mamm
hindbrain boundary development. Genesis. 2001;30 Genome. 2005;16(7):476–80.
(3):129–33. 103. Grant SF, Hakonarson H. Microarray technology and
88. Hrabe de Angelis M, Strivens M. Large-scale produc- applications in the arena of genome-wide association.
tion of mouse phenotypes: the search for animal Clin Chem. 2008;54(7):1116–24.
models for inherited diseases in humans. Brief 104. Patil N, Nouri N, McAllister L, Matsukaki H, Ryder
Bioinform. 2001;2(2):170–80. T. Single-nucleotide polymorphism genotyping using
89. Nolan PM, Peters J, Vizor L, Strivens M, microarrays. Curr Protoc Hum Genet. 2013;Chapter 2:
Washbourne R, Hough T, et al. Implementation of a Unit 2 9.
large-scale ENU mutagenesis program: towards 105. Pletcher MT, McClurg P, Batalov S, Su AI, Barnes
increasing the mouse mutant resource. Mamm SW, Lagler E, et al. Use of a dense single nucleotide
Genome. 2000;11(7):500–6. polymorphism map for in silico mapping in the
90. Chen Y, Yee D, Dains K, Chatterjee A, Cavalcoli J, mouse. PLoS Biol. 2004;2(12):e393.
Schneider E, et al. Genotype-based screen for 106. Abiola O, Angel JM, Avner P, Bachmanov AA,
ENU-induced mutations in mouse embryonic stem Belknap JK, Bennett B, et al. The nature and identi-
cells. Nat Genet. 2000;24(3):314–7. fication of quantitative trait loci: a community’s view.
91. Anderson KV. Finding the genes that direct mamma- Nat Rev Genet. 2003;4(11):911–6.
lian development: ENU mutagenesis in the mouse. 107. Korstanje R, Paigen B. From QTL to gene: the harvest
Trends Genet. 2000;16(3):99–102. begins. Nat Genet. 2002;31(3):235–6.
92. Justice MJ, Noveroske JK, Weber JS, Zheng B, Brad- 108. Pezzolesi MG, Skupien J, Mychaleckyj JC, Warram
ley A. Mouse ENU mutagenesis. Hum Mol Genet. JH, Krolewski AS. Insights to the genetics of diabetic
1999;8(10):1955–63. nephropathy through a genome-wide association
93. Hrabe de Angelis M, Balling R. Large scale ENU study of the GoKinD collection. Semin Nephrol.
screens in the mouse: genetics meets genomics. 2010;30(2):126–40. [Research Support, N.I.H.,
Mutat Res. 1998;400(1–2):25–32. Extramural Research Support, Non-U.S. Gov’t
94. Knapik EW. ENU mutagenesis in zebrafish–from Research Support, U.S. Gov’t, P.H.S. Review].
genes to complex diseases. Mamm Genome. 109. Pezzolesi MG, Poznik GD, Mychaleckyj JC, Paterson
2000;11(7):511–9. AD, Barati MT, Klein JB, et al. Genome-wide asso-
95. Beckwith LG, Moore JL, Tsao-Wu GS, Harshbarger ciation scan for diabetic nephropathy susceptibility
JC, Cheng KC. Ethylnitrosourea induces neoplasia in genes in type 1 diabetes. Diabetes. 2009;58
zebrafish (Danio rerio). Lab Invest. 2000;80 (6):1403–10. [Research Support, N.I.H., Extramural
(3):379–85. Research Support, Non-U.S. Gov’t Research Support,
96. Weinstein BM, Schier AF, Abdelilah S, Malicki J, U.S. Gov’t, P.H.S.].
Solnica-Krezel L, Stemple DL, et al. Hematopoietic 110. Malhotra A, Igo Jr RP, Thameem F, Kao WH, Abboud
mutations in the zebrafish. Development. 1996;123: HE, Adler SG, et al. Genome-wide linkage scans for
303–9. type 2 diabetes mellitus in four ethnically diverse
97. Haffter P, Granato M, Brand M, Mullins MC, populations-significant evidence for linkage on chro-
Hammerschmidt M, Kane DA, et al. The identifica- mosome 4q in African Americans: the Family Inves-
tion of genes with unique and essential functions in tigation of Nephropathy and Diabetes Research
468 J. Kreidberg

Group. Diabetes Metab Res Rev. 2009;25(8):740–7. Non-U.S. Gov’t Research Support, U.S. Gov’t,
[Multicenter Study Research Support, N.I.H., Extra- Non-P.H.S. Review].
mural Research Support, N.I.H., Intramural]. 123. Keane TM, Goodstadt L, Danecek P, White MA,
111. Genovese G, Tonna SJ, Knob AU, Appel GB, Katz A, Wong K, Yalcin B, et al. Mouse genomic variation
Bernhardy AJ, et al. A risk allele for focal segmental and its effect on phenotypes and gene regulation.
glomerulosclerosis in African Americans is located Nature. 2011;477(7364):289–94. [Comparative
within a region containing APOL1 and MYH9. Kidney Study Research Support, N.I.H., Extramural Research
Int. 2010;78(7):698–704. [Research Support, N.I.H., Support, Non-U.S. Gov’t Research Support,
Extramural Research Support, Non-U.S. Gov’t]. U.S. Gov’t, Non-P.H.S.].
112. Wasser WG, Tzur S, Wolday D, Adu D, Baumstein D, 124. Lander ES, Linton LM, Birren B, Nusbaum C, Zody
Rosset S, et al. Population genetics of chronic kidney MC, Baldwin J, et al. Initial sequencing and analysis
disease: the evolving story of APOL1. J Nephrol. of the human genome. Nature. 2001;409
2012;25(5):603–18. [Research Support, Non-U.S. (6822):860–921.
Gov’t Review]. 125. Venter JC, Adams MD, Myers EW, Li PW, Mural RJ,
113. Shlush LI, Bercovici S, Wasser WG, Yudkovsky G, Sutton GG, et al. The sequence of the human genome.
Templeton A, Geiger D, et al. Admixture mapping of Science. 2001;291(5507):1304–51.
end stage kidney disease genetic susceptibility using 126. Waterston RH, Lindblad-Toh K, Birney E, Rogers J,
estimated mutual information ancestry informative Abril JF, Agarwal P, et al. Initial sequencing and
markers. BMC Med Genomics. 2010;3:47. [Research comparative analysis of the mouse genome. Nature.
Support, Non-U.S. Gov’t]. 2002;420(6915):520–62.
114. Tsaih SW, Pezzolesi MG, Yuan R, Warram JH, 127. Wheeler DA, Srinivasan M, Egholm M, Shen Y,
Krolewski AS, Korstanje R. Genetic analysis of albu- Chen L, McGuire A, et al. The complete genome of
minuria in aging mice and concordance with loci for an individual by massively parallel DNA sequencing.
human diabetic nephropathy found in a genome-wide Nature. 2008;452(7189):872–6.
association scan. Kidney Int. 2010;77(3):201–10. 128. Raven P, Fauquet C, Swaminathan MS, Borlaug N,
[Research Support, N.I.H., Extramural Research Sup- Samper C. Where next for genome sequencing? Sci-
port, Non-U.S. Gov’t]. ence. 2006;311(5760):468.
115. DiPetrillo K, Wang X, Stylianou IM, Paigen B. Bio- 129. Bentley DR. Whole-genome re-sequencing. Curr
informatics toolbox for narrowing rodent quantitative Opin Genet Dev. 2006;16(6):545–52.
trait loci. Trends Genet. 2005;21(12):683–92. 130. Schmouth JF, Bonaguro RJ, Corso-Diaz X, Simpson
116. Cuppen E. Haplotype-based genetics in mice and rats. EM. Modelling human regulatory variation in mouse:
Trends Genet. 2005;21(6):318–22. finding the function in genome-wide association stud-
117. Guryev V, Smits BM, van de Belt J, Verheul M, ies and whole-genome sequencing. PLoS Genet.
Hubner N, Cuppen E. Haplotype block structure 2012;8(3):e1002544. [Research Support, N.I.H.,
is conserved across mammals. PLoS Genet. 2006; Extramural Research Support, Non-U.S. Gov’t
2(7):e121. Review].
118. Frazer KA, Ballinger DG, Cox DR, Hinds DA, Stuve 131. Shiozawa M, Provoost AP, van Dokkum RP,
LL, Gibbs RA, et al. A second generation human Majewski RR, Jacob HJ. Evidence of gene-gene inter-
haplotype map of over 3.1 million SNPs. Nature. actions in the genetic susceptibility to renal impair-
2007;449(7164):851–61. [Research Support, N.I.H., ment after unilateral nephrectomy. J Am Soc Nephrol.
Extramural Research Support, Non-U.S. Gov’t]. 2000;11(11):2068–78.
119. Sachidanandam R, Weissman D, Schmidt SC, Kakol 132. Kwitek-Black AE, Jacob HJ. The use of designer rats
JM, Stein LD, Marth G, et al. A map of human in the genetic dissection of hypertension. Curr
genome sequence variation containing 1.42 million Hypertens Rep. 2001;3(1):12–8.
single nucleotide polymorphisms. Nature. 2001;409 133. Stoll M, Jacob HJ. Genetic rat models of hyperten-
(6822):928–33. [Research Support, Non-U.S. Gov’t sion: relationship to human hypertension. Curr
Research Support, U.S. Gov’t, P.H.S.]. Hypertens Rep. 2001;3(2):157–64.
120. Wang X, Korstanje R, Higgins D, Paigen B. Haplo- 134. Stoll M, Cowley Jr AW, Tonellato PJ, Greene AS,
type analysis in multiple crosses to identify a QTL Kaldunski ML, Roman RJ, et al. A genomic-systems
gene. Genome Res. 2004;14(9):1767–72. biology map for cardiovascular function. Science.
121. Wittenburg H, Lyons MA, Li R, Kurtz U, Wang X, 2001;294(5547):1723–6.
Mossner J, et al. QTL mapping for genetic determi- 135. Jacob HJ, Kwitek AE. Rat genetics: attaching physi-
nants of lipoprotein cholesterol levels in combined ology and pharmacology to the genome. Nat Rev
crosses of inbred mouse strains. J Lipid Res. Genet. 2002;3(1):33–42.
2006;47(8):1780–90. 136. Rao GN. Diet and kidney diseases in rats. Toxicol
122. Flint J, Eskin E. Genome-wide association studies in Pathol. 2002;30(6):651–6.
mice. Nat Rev Genet. 2012;13(11):807–17. [Research 137. Ma X, Abboud FM, Chapleau MW. Analysis of affer-
Support, N.I.H., Extramural Research Support, ent, central, and efferent components of the
15 Translational Research Methods: The Value of Animal Models in Renal Research 469

baroreceptor reflex in mice. Am J Physiol Regul 151. Gimonet V, Bussieres L, Medjebeur AA, Gasser B,
Integr Comp Physiol. 2002;283(5):R1033–40. Lelongt B, Laborde K. Nephrogenesis and angioten-
138. Ishii T, Kuwaki T, Masuda Y, Fukuda Y. Postnatal sin II receptor subtypes gene expression in the fetal
development of blood pressure and baroreflex in lamb. Am J Physiol. 1998;274(6 Pt 2):F1062–9.
mice. Auton Neurosci. 2001;94(1–2):34–41. 152. Nguyen HT, Kogan BA. Renal hemodynamic
139. Gross V, Plehm R, Tank J, Jordan J, Diedrich A, changes after complete and partial unilateral ureteral
Obst M, et al. Heart rate variability and baroreflex obstruction in the fetal lamb. J Urol. 1998;160(3 Pt
function in AT2 receptor-disrupted mice. Hyperten- 2):1063–9.
sion. 2002;40(2):207–13. 153. Wang J, Perez FM, Rose JC. Developmental changes
140. Rokosh DG, Simpson PC. Knockout of the alpha 1A/ in renin-containing cells from the ovine fetal kidney. J
C-adrenergic receptor subtype: the alpha 1A/C is Soc Gynecol Investig. 1997;4(4):191–6.
expressed in resistance arteries and is required to 154. Berry LM, Polk DH, Ikegami M, Jobe AH, Padbury
maintain arterial blood pressure. Proc Natl Acad Sci JF, Ervin MG. Preterm newborn lamb renal and car-
U S A. 2002;99(14):9474–9. diovascular responses after fetal or maternal antenatal
141. Besnard S, Bakouche J, Lemaigre-Dubreuil Y, betamethasone. Am J Physiol. 1997;272(6 Pt 2):
Mariani J, Tedgui A, Henrion D. Smooth muscle R1972–9.
dysfunction in resistance arteries of the staggerer 155. Matsell DG, Bennett T, Bocking AD. Characteriza-
mouse, a mutant of the nuclear receptor RORalpha. tion of fetal ovine renal dysplasia after mid-gestation
Circ Res. 2002;90(7):820–5. ureteral obstruction. Clin Invest Med. 1996;19
142. Vecchione C, Fratta L, Rizzoni D, Notte A, Poulet R, (6):444–52.
Porteri E, et al. Cardiovascular influences of alpha1b- 156. Peters CA, Gaertner RC, Carr MC, Mandell J. Fetal
adrenergic receptor defect in mice. Circulation. compensatory renal growth due to unilateral ureteral
2002;105(14):1700–7. obstruction. J Urol. 1993;150(2 Pt 2):597–600.
143. Gross V, Luft FC. Adapting renal and cardiovascular 157. Peters CA, Docimo SG, Luetic T, Reid LM, Retik
physiology to the genetically hypertensive mouse. AB, Mandell J. Effect of in utero vesicostomy on
Semin Nephrol. 2002;22(2):172–9. pulmonary hypoplasia in the fetal lamb with bladder
144. Holschneider DP, Scremin OU, Roos KP, Chialvo outlet obstruction and oligohydramnios: a morpho-
DR, Chen K, Shih JC. Increased baroreceptor metric analysis. J Urol. 1991;146(4):1178–83.
response in mice deficient in monoamine oxidase A 158. Rosines E, Sampogna RV, Johkura K, Vaughn DA,
and B. Am J Physiol Heart Circ Physiol. 2002;282(3): Choi Y, Sakurai H, et al. Staged in vitro reconstitution
H964–72. and implantation of engineered rat kidney tissue. Proc
145. Edouga D, Hugueny B, Gasser B, Bussieres L, Natl Acad Sci U S A. 2007;104(52):20938–43.
Laborde K. Recovery after relief of fetal urinary 159. Fissell WH. Developments towards an artificial kid-
obstruction: morphological, functional and molecular ney. Expert Rev Med Devices. 2006;3(2):155–65.
aspects. Am J Physiol Renal Physiol. 2001;281(1): 160. Hammerman MR. Tissue engineering the kidney.
F26–37. Kidney Int. 2003;63(4):1195–204.
146. Kitagawa H, Pringle KC, Zuccollo J, Koike J, 161. Chugh S, Yuan H, Topham PS, Haydar SA, Mittal V,
Nakada K, Ikoma M, et al. Glomerular size in renal Taylor GA, et al. Aminopeptidase A: a nephritogenic
dysplasia secondary to obstructive uropathy: a further target antigen of nephrotoxic serum. Kidney Int.
exploration of the fetal lamb model. J Pediatr Surg. 2001;59(2):601–13. [Research Support, Non-U.S.
2000;35(11):1651–5. Gov’t Research Support, U.S. Gov’t, P.H.S.].
147. Kitagawa H, Pringle KC, Zucollo J, Koike J, 162. Cook HT, Khan SB, Allen A, Bhangal G, Smith J,
Nakada K, Moriya H, et al. Early fetal obstructive Lobb RR, et al. Treatment with an antibody to VLA-1
uropathy produces Potter’s syndrome in the lamb. J integrin reduces glomerular and tubulointerstitial
Pediatr Surg. 2000;35(11):1549–53. scarring in a rat model of crescentic glomerulonephri-
148. Smith LM, Ervin MG, Wada N, Ikegami M, Polk DH, tis. Am J Pathol. 2002;161(4):1265–72.
Jobe AH. Antenatal glucocorticoids alter postnatal 163. Hiromura K, Haseley LA, Zhang P, Monkawa T,
preterm lamb renal and cardiovascular responses to Durvasula R, Petermann AT, et al. Podocyte expres-
intravascular volume expansion. Pediatr Res. 2000; sion of the CDK-inhibitor p57 during development
47(5):622–7. and disease. Kidney Int. 2001;60(6):2235–46.
149. Kitagawa H, Pringle KC, Zuccolo J, Stone P, Nakada K, 164. Lin F, Emancipator SN, Salant DJ, Medof ME.
Kawaguchi F, et al. The pathogenesis of dysplastic Decay-accelerating factor confers protection against
kidney in a urinary tract obstruction in the female fetal complement-mediated podocyte injury in acute neph-
lamb. J Pediatr Surg. 1999;34(11):1678–83. rotoxic nephritis. Lab Invest. 2002;82(5):563–9.
150. Wang J, Rose JC. Developmental changes in renal [Research Support, U.S. Gov’t, P.H.S.].
renin mRNA half-life and responses to stimulation in 165. Topham PS, Csizmadia V, Soler D, Hines D, Gerard
fetal lambs. Am J Physiol. 1999;277(4 Pt 2): CJ, Salant DJ, et al. Lack of chemokine receptor
R1130–5. CCR1 enhances Th1 responses and glomerular injury
470 J. Kreidberg

during nephrotoxic nephritis. J Clin Invest. 1999;104 179. Lloberas N, Torras J, Herrero-Fresneda I, Cruzado JM,
(11):1549–57. Riera M, Hurtado I, et al. Postischemic renal oxidative
166. Xu Y, Berrou J, Chen X, Fouqueray B, Callard P, stress induces inflammatory response through PAF and
Sraer JD, et al. Induction of urokinase receptor oxidized phospholipids. Prevention by antioxidant
expression in nephrotoxic nephritis. Exp Nephrol. treatment. Faseb J. 2002;16(8):908–10.
2001;9(6):397–404. 180. Megyesi J, Andrade L, Vieira Jr JM, Safirstein RL,
167. Yanagita M, Ishimoto Y, Arai H, Nagai K, Ito T, Price PM. Positive effect of the induction of
Nakano T, et al. Essential role of Gas6 for glomerular p21WAF1/CIP1 on the course of ischemic acute
injury in nephrotoxic nephritis. J Clin Invest. renal failure. Kidney Int. 2001;60(6):2164–72.
2002;110(2):239–46. 181. Meldrum KK, Hile K, Meldrum DR, Crone JA,
168. Carmago S, Shah SV, Walker PD. Meprin, a brush- Gearhart JP, Burnett AL. Simulated ischemia induces
border enzyme, plays an important role in hypoxic/ renal tubular cell apoptosis through a nuclear factor-
ischemic acute renal tubular injury in rats. Kidney Int. kappaB dependent mechanism. J Urol. 2002;168
2002;61(3):959–66. (1):248–52.
169. Chatterjee PK, Brown PA, Cuzzocrea S, 182. Modolo NS, Castiglia YM, Ganem EM, Braz JR,
Zacharowski K, Stewart KN, Mota-Filipe H, Vianna PT, Vane LA. Acute renal ischemia model in
et al. Calpain inhibitor-1 reduces renal ischemia/ dogs: effects of metoprolol. Ren Fail. 2001;23
reperfusion injury in the rat. Kidney Int. 2001;59 (1):1–10.
(6):2073–83. 183. Mister M, Noris M, Szymczuk J, Azzollini N,
170. Fernandez M, Medina A, Santos F, Carbajo E, Aiello S, Abbate M, et al. Propionyl-L-carnitine pre-
Rodriguez J, Alvarez J, et al. Exacerbated inflamma- vents renal function deterioration due to ischemia/
tory response induced by insulin-like growth factor I reperfusion. Kidney Int. 2002;61(3):1064–78.
treatment in rats with ischemic acute renal failure. 184. Okusa MD. The inflammatory cascade in acute ische-
J Am Soc Nephrol. 2001;12(9):1900–7. mic renal failure. Nephron. 2002;90(2):133–8.
171. Gimelreich D, Popovtzer MM, Wald H, Pizov G, 185. Power JM, Tonkin AM. Large animal models of heart
Berlatzky Y, Rubinger D. Regulation of ROMK and failure. Aust N Z J Med. 1999;29(3):395–402.
channel-inducing factor (CHIF) in acute renal failure 186. Textor SC. Pathophysiology of renal failure in reno-
due to ischemic reperfusion injury. Kidney Int. vascular disease. Am J Kidney Dis. 1994;24
2001;59(5):1812–20. (4):642–51.
172. Gretz N. The development of hypertension in the 187. Vaneerdeweg W, Buyssens N, De Winne T,
remnant kidney model after either pole resection or Sebrechts M, Babloyan A, Arakelian S, et al. A stan-
partial infarction of the kidney. J Am Soc Nephrol. dardized surgical technique to obtain a stable and
1995;5(10):1839–40. reproducible chronic renal failure model in dogs.
173. Jia ZQ, Worthington AE, Hill RP, Hunt JW. The Eur Surg Res. 1992;24(5):273–82.
effects of artery occlusion on temperature homogene- 188. Yoshida T, Tang SS, Hsiao LL, Jensen RV, Ingelfinger
ity during hyperthermia in rabbit kidneys in vivo. Int J JR, Gullans SR. Global analysis of gene expression in
Hyperthermia. 1997;13(1):21–37. renal ischemia-reperfusion in the mouse. Biochem
174. Kakoki M, Hirata Y, Hayakawa H, Suzuki E, Biophys Res Commun. 2002;291(4):787–94.
Nagata D, Nishimatsu H, et al. Effects of vasodilatory 189. Chevalier RL. Chronic partial ureteral obstruction and
antihypertensive agents on endothelial dysfunction in the developing kidney. Pediatr Radiol. 2008;38 Suppl
rats with ischemic acute renal failure. Hypertens Res. 1:S35–40.
2000;23(5):527–33. 190. Ma FY, Tesch GH, Flavell RA, Davis RJ, Nikolic-
175. Knoll T, Schult S, Birck R, Braun C, Michel MS, Paterson DJ. MKK3-p38 signaling promotes apopto-
Bross S, et al. Therapeutic administration of an sis and the early inflammatory response in the
endothelin-A receptor antagonist after acute ischemic obstructed mouse kidney. Am J Physiol Renal
renal failure dose-dependently improves recovery of Physiol. 2007;293(5):F1556–63.
renal function. J Cardiovasc Pharmacol. 2001;37 191. Chevalier RL. Pathogenesis of renal injury in obstruc-
(4):483–8. tive uropathy. Curr Opin Pediatr. 2006;18(2):153–60.
176. Kren S, Hostetter TH. The course of the remnant 192. Lee VW, Harris DC. Adriamycin nephropathy: a
kidney model in mice. Kidney Int. 1999;56(1):333–7. model of focal segmental glomerulosclerosis.
177. Kwon O, Phillips CL, Molitoris BA. Ischemia Nephrology (Carlton). 2011;16(1):30–8. [Review].
induces alterations in actin filaments in renal vascular 193. Pippin JW, Brinkkoetter PT, Cormack-Aboud FC,
smooth muscle cells. Am J Physiol Renal Physiol. Durvasula RV, Hauser PV, Kowalewska J,
2002;282(6):F1012–9. et al. Inducible rodent models of acquired podocyte
178. Lieberthal W, Fuhro R, Andry CC, Rennke H, Aber- diseases. Am J Physiol Renal Physiol. 2009;296(2):
nathy VE, Koh JS, et al. Rapamycin impairs recovery F213–29. [Review].
from acute renal failure: role of cell-cycle arrest and 194. Fogo AB. Animal models of FSGS: lessons for path-
apoptosis of tubular cells. Am J Physiol Renal ogenesis and treatment. Semin Nephrol. 2003;23(2):
Physiol. 2001;281(4):F693–706. 161–71. [Review].
15 Translational Research Methods: The Value of Animal Models in Renal Research 471

195. Dai Y, Gu L, Yuan W, Yu Q, Ni Z, Ross MJ, in mice lacking nitric oxide synthase type 2. Kidney
et al. Podocyte-specific deletion of signal transducer Int. 1998;53(4):932–6.
and activator of transcription 3 attenuates nephrotoxic 210. Quigg RJ, Lim A, Haas M, Alexander JJ, He C, Carroll
serum-induced glomerulonephritis. Kidney Int. MC. Immune complex glomerulonephritis in C4- and
2013;84(5):950–61. [Research Support, N.I.H., C3-deficient mice. Kidney Int. 1998;53(2):320–30.
Extramural Research Support, Non-U.S. Gov’t 211. Tang T, Rosenkranz A, Assmann KJ, Goodman MJ,
Research Support, U.S. Gov’t, Non-P.H.S.]. Gutierrez-Ramos JC, Carroll MC, et al. A role for
196. Happe H, Peters DJ. Translational research in Mac-1 (CDIIb/CD18) in immune complex-stimulated
ADPKD: lessons from animal models. Nat Rev neutrophil function in vivo: Mac-1 deficiency abro-
Nephrol. 2014;10(10):587–601. [Review]. gates sustained Fcgamma receptor-dependent neutro-
197. Nagao S, Kugita M, Yoshihara D, Yamaguchi phil adhesion and complement-dependent proteinuria
T. Animal models for human polycystic kidney dis- in acute glomerulonephritis. J Exp Med. 1997;186
ease. Exp Anim. 2012;61(5):477–88. [Research Sup- (11):1853–63.
port, Non-U.S. Gov’t Review]. 212. Ito MR, Terasaki S, Itoh J, Katoh H, Yonehara S, Nose
198. Wilson PD. Mouse models of polycystic kidney dis- M. Rheumatic diseases in an MRL strain of mice with
ease. Curr Top Dev Biol. 2008;84:311–50. [Review]. a deficit in the functional Fas ligand. Arthritis Rheum.
199. Foster MH. Relevance of systemic lupus 1997;40(6):1054–63.
erythematosus nephritis animal models to human dis- 213. Haas C, Ryffel B, Le Hir M. IFN-gamma is essential
ease. Semin Nephrol. 1999;19(1):12–24. for the development of autoimmune glomerulone-
200. Morel L, Wakeland EK. Susceptibility to lupus phritis in MRL/Ipr mice. J Immunol. 1997;158
nephritis in the NZB/W model system. Curr Opin (11):5484–91.
Immunol. 1998;10(6):718–25. 214. Hibbs ML, Tarlinton DM, Armes J, Grail D,
201. Walport MJ, Davies KA, Botto M. C1q and systemic Hodgson G, Maglitto R, et al. Multiple defects in
lupus erythematosus. Immunobiology. 1998;199 the immune system of Lyn-deficient mice, culminat-
(2):265–85. ing in autoimmune disease. Cell. 1995;83(2):301–11.
202. Gavalchin J, Staines NA. T and B cell recognition of 215. Cyster JG. Lymphoid organ development and cell
idiotypes of anti-DNA autoantibodies. Lupus. migration. Immunol Rev. 2003;195:5–14.
1997;6(3):337–43. 216. Reinhardt RL, Khoruts A, Merica R, Zell T, Jenkins
203. Isenberg DA, Ravirajan CT, Rahman A, Kalsi J. MK. Visualizing the generation of memory CD4 T cells
The role of antibodies to DNA in systemic in the whole body. Nature. 2001;410(6824):101–5.
lupus erythematosus–a review and introduction to 217. Gudmundsdottir H, Turka LA. T cell costimulatory
an international workshop on DNA antibodies blockade: new therapies for transplant rejection. J Am
held in London, May 1996. Lupus. 1997;6 Soc Nephrol. 1999;10(6):1356–65.
(3):290–304. 218. Dong VM, Womer KL, Sayegh MH. Transplantation
204. Pickering MC, Cook HT, Warren J, Bygrave AE, tolerance: the concept and its applicability. Pediatr
Moss J, Walport MJ, et al. Uncontrolled C3 activation Transplant. 1999;3(3):181–92.
causes membranoproliferative glomerulonephritis in 219. Bromberg JS, Murphy B. Routes to allograft survival.
mice deficient in complement factor H. Nat Genet. J Clin Invest. 2001;107(7):797–8.
2002;31(4):424–8. 220. Light J, Salomon DR, Diethelm AG, Alexander JW,
205. Salvador JM, Hollander MC, Nguyen AT, Kopp JB, Hunsicker L, Thistlethwaite R, et al. Bone marrow
Barisoni L, Moore JK, et al. Mice lacking the transfusions in cadaver renal allografts: pilot trials
p53-effector gene Gadd45a develop a lupus-like syn- with concurrent controls. Clin Transplant. 2002;16
drome. Immunity. 2002;16(4):499–508. (5):317–24.
206. Tabata N, Miyazawa M, Fujisawa R, Takei YA, 221. Knechtle SJ, Hamawy MM, Hu H, Fechner Jr JH,
Abe H, Hashimoto K. Establishment of monoclonal Cho CS. Tolerance and near-tolerance strategies in
anti-retroviral gp70 autoantibodies from MRL/lpr monkeys and their application to human renal trans-
lupus mice and induction of glomerular gp70 deposi- plantation. Immunol Rev. 2001;183:205–13.
tion and pathology by transfer into non-autoimmune 222. Inverardi L, Ricordi C. Tolerance and pancreatic islet
mice. J Virol. 2000;74(9):4116–26. transplantation. Philos Trans R Soc Lond B Biol Sci.
207. Cruse JM, Lewis RE, Dilioglou S. Fate of immune 2001;356(1409):759–65.
complexes, glomerulonephritis, and cell-mediated 223. Field EH, Strober S. Tolerance, mixed chimerism and
vasculitis in lupus-prone MRL/Mp lpr/lpr mice. Exp protection against graft-versus-host disease after total
Mol Pathol. 2000;69(3):211–22. lymphoid irradiation. Philos Trans R Soc Lond B Biol
208. Ophascharoensuk V, Fero ML, Hughes J, Roberts JM, Sci. 2001;356(1409):739–48.
Shankland SJ. The cyclin-dependent kinase inhibitor 224. Decker CJ, Heiser AD, Chaturvedi PR, Faust TJ,
p27Kip1 safeguards against inflammatory injury. Nat Ku G, Moseley S, et al. The novel IMPDH inhibitor
Med. 1998;4(5):575–80. VX-497 prolongs skin graft survival and improves
209. Cattell V, Cook HT, Ebrahim H, Waddington SN, Wei graft versus host disease in mice. Drugs Exp Clin
XQ, Assmann KJ, et al. Anti-GBM glomerulonephritis Res. 2001;27(3):89–95.
472 J. Kreidberg

225. Yoshimura R, Chargui J, Aitouche A, Veyron P, Tou- tissue-specific immune response to cardiac myosin
raine JL. Induction of hyperacute rejection of skin can prolong survival of allogeneic heart transplants.
allografts by CD8+ lymphocytes. Transplantation. J Immunol. 2002;169(3):1168–74.
2000;69(7):1452–7. 232. Coates PT, Krishnan R, Kireta S, Johnston J, Russ
226. Gardner CR. The pharmacology of immunosuppres- GR. Human myeloid dendritic cells transduced with
sant drugs in skin transplant rejection in mice and an adenoviral interleukin-10 gene construct inhibit
other rodents. Gen Pharmacol. 1995;26(2):245–71. human skin graft rejection in humanized NOD-scid
227. Tepper MA, Linsley PS, Tritschler D, Esselstyn chimeric mice. Gene Ther. 2001;8(16):1224–33.
JM. Tolerance induction by soluble CTLA4 in a 233. Fahy O, Porte H, Senechal S, Vorng H, McEuen AR,
mouse skin transplant model. Transplant Proc. Buckley MG, et al. Chemokine-induced cutaneous
1994;26(6):3151–4. inflammatory cell infiltration in a model of Hu-
228. Sho M, Sandner SE, Najafian N, Salama AD, Dong V, PBMC-SCID mice grafted with human skin. Am J
Yamada A, et al. New insights into the interactions Pathol. 2001;158(3):1053–63.
between T-cell costimulatory blockade and conven- 234. Moulton KS, Melder RJ, Dharnidharka VR, Hardin-
tional immunosuppressive drugs. Ann Surg. Young J, Jain RK, Briscoe DM. Angiogenesis in the
2002;236(5):667–75. huPBL-SCID model of human transplant rejection.
229. Rolls HK, Kishimoto K, Dong VM, Illigens BM, Transplantation. 1999;67(12):1626–31.
Sho M, Sayegh MH, et al. T-cell response to cardiac 235. Briscoe DM, Dharnidharka VR, Isaacs C,
myosin persists in the absence of an alloimmune Downing G, Prosky S, Shaw P, et al. The allogeneic
response in recipients with chronic cardiac allograft response to cultured human skin equivalent in the hu-
rejection. Transplantation. 2002;74(7):1053–7. PBL-SCID mouse model of skin rejection. Transplan-
230. Zhai Y, Meng L, Gao F, Busuttil RW, Kupiec- tation. 1999;67(12):1590–9.
Weglinski JW. Allograft rejection by primed/memory 236. Hammerman MR. Xenotransplantation of renal
CD8+ T cells is CD154 blockade resistant: therapeu- primordia. Curr Opin Nephrol Hypertens. 2002;11
tic implications for sensitized transplant recipients. (1):11–6.
J Immunol. 2002;169(8):4667–73. 237. Palmer DB, Lechler R. Can the thymus be a useful
231. Fedoseyeva EV, Kishimoto K, Rolls HK, Illigens tool to induce specific tolerance to xenoantigens?
BM, Dong VM, Valujskikh A, et al. Modulation of Transplantation. 1999;68(11):1628–30.
Basics of Clinical Investigation
16
Susan L. Furth and Jeffrey J. Fadrowski

Contents Sources of Data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487


Assessing Data Quality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 488
Clinical Research Question . . . . . . . . . . . . . . . . . . . . . . . . 474
Hallmarks of a Good Research Question . . . . . . . . . . . . 474 Analytic Plan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 488
Steps in Refining a Good Research Question . . . . . . . 474 Data Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 488
Statistical Significance and Confidence
Scientific Method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 475 Intervals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 491
Inference . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 475
Validity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 476 Topics Related to Clinical Decision Making . . . . . . 492
Reliability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 477 The Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 492
The Diagnostic Test . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 492
Study Design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 477
Observational Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 477 Screening Programs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
Experimental Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 482 Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
Test . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
Important Issues in Carrying Out a Person/Population . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
Research Plan . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 484
Selection of Subjects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 484 Ethics in Research . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 495
Attrition of Study Subjects . . . . . . . . . . . . . . . . . . . . . . . . . . 486
Evaluating the Literature: Rating the
Data Collection: Measurement . . . . . . . . . . . . . . . . . . . . 487 Strength of Scientific Evidence . . . . . . . . . . . . . . . . . . . . 496
Identifying the Variables to Be Measured . . . . . . . . . . . 487
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 497
Types of Variables . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 487

S.L. Furth (*)


Departments of Pediatrics and Epidemiology, Perelman
School of Medicine at the University of Pennsylvania,
Philadelphia, PA, USA
e-mail: furths@email.chop.edu
J.J. Fadrowski
Department of Pediatrics, John Hopkins University School
of Medicine, Baltimore, MD, USA
e-mail: fadrowsj@jhmi.edu

# Springer-Verlag Berlin Heidelberg 2016 473


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_15
474 S.L. Furth and J.J. Fadrowski

Clinical Research Question Finally, a good research question needs to be


answerable with available resources. These
How can we best evaluate, treat, and assess long- include subjects available for study, technical
term risks for children with kidney disease? Who expertise of the research staff, and the time
is at risk of developing end-stage renal disease and money that can be devoted to the
(ESRD) in childhood or young adulthood? Clini- project. Once a question is framed, the researcher
cians are often faced with questions such as these needs to outline the study protocol or
with uncertain answers in the practice of pediatric methods, which include specifying the recruit-
nephrology. Parents ask, “Why did my child get ment method, number of subjects and how they
this disease?” “What is the most effective method will be recruited, how each variable will be
to treat this condition?” “What’s the prognosis of defined, and the plan for data analysis. A poorly
this condition in my child?” Frequently, these designed study is worse than no study at all
answers are not known, and these questions are because, like imprecise measurements and an
the inspiration for high-quality clinical research. improper analytic plan, it can also lead to false
The first step in developing a valuable clinical conclusions.
study is determining whether the initial query
can be translated into a good research question.
Steps in Refining a Good Research
Question
Hallmarks of a Good Research Question
A good research question usually begins with a
A good research question gives useful informa- broadly stated concept. The initial question is then
tion, is interesting to the researcher, builds on made more specific by identifying independent
what is known, and can be answered with avail- and dependent variables. Often, research
able resources. Research is a labor of love, questions are concerned with causal relationships.
demanding attention to detail, perseverance, hon- Independent variables are those conceptualized to
esty, and imagination. Developing a good be causes; dependent variables are those
research question is an iterative process. One conceptualized to be effects. The research ques-
needs input from knowledgeable colleagues and tion can be modified to ask about the role of
collaborators. The researcher must become thor- multiple potential causes in leading to the specific
oughly familiar with what is already known about outcome. A simple research question asks
the topic by reviewing the literature and consult- whether x (independent variable) causes
ing with experts in the area. Investigating what is y (dependent variable). More complex research
already known has several benefits. First, it can questions could assess the relative importance of
reveal that the candidate research question has x and other variables (e.g., a and b) as causes of y.
already been answered adequately. Second, learn- A different research question might ask how
ing what is already known provides insight into strongly x predicts y in one population versus
potentially useful methods for addressing a another.
research question. For example, previous studies The next step is to translate a research
may demonstrate good ways to measure a variable question into a hypothesis. In our simple
of interest or provide background information for example, the researcher may hypothesize that
determining sample size. Third, a literature review x causes y. In the actual research project, the
may suggest ways to frame the research question information collected is examined to
at hand. For example, a literature review may determine whether it is reasonable to conclude
reveal that particular risk factors are consistently that x does cause y. In examining the data, the
associated with a disease process, and an interven- investigator tests the null hypothesis that x does
tion to modify these risk factors may form a sound not cause y versus the alternative hypothesis that
basis for a clinical trial. it does.
16 Basics of Clinical Investigation 475

Scientific Method systematically from the truth. Three broad catego-


ries of bias include selection bias, measurement
A study’s potential value is determined by the bias, and confounding bias. Selection bias arises if
relevance of the research question. Its ultimate the manner in which subjects with the outcome of
worth is determined by the study methods. interest and the comparison group were selected
Methodologic issues concern the study design, yields an apparent association, when, in reality,
subject selection, data collection techniques, and exposure and disease are not associated. Measure-
the analytic plan. Subsequent sections in this ment bias occurs when the methods of measure-
chapter discuss each of these aspects. As a foun- ment systematically differ between groups.
dation, this section describes the concepts of infer- Confounding occurs when a factor is associated
ence, generalizability, and validity. both with the exposure and the outcome and the
effect of the main exposure under study is con-
fused or distorted by the confounder.
Inference Statistical inference depends on the methods
used to define and sample the population. The
As Fig. 1 shows, scientific research begins with researcher uses inferential statistics to extrapolate
the research question. It then moves (clockwise in the sample findings to the larger population of
the figure) to the controlled arena of the study children from which the sample was drawn. Infer-
design and then through the implementation of ential statistics assume that the studied sample is
the actual study and findings. Inferences from drawn by probability methods and can be used to
the findings in the study approximate the “truth make inferences about the larger population. The
in the study.” From these “truths,” we attempt to size of a probability sample determines the cer-
infer the applicability of the findings to general tainty of inferences from it. All other things being
clinical practice. Researchers describe and explain equal, the larger the sample size, the greater the
reality by sampling a portion of it, measuring certainty of inferences to the population.
characteristics of the sample, analyzing the mea- The researcher’s ability to make a causal infer-
surements, and interpreting the results. Errors in ence from study results depends largely on issues
the design or implementation of the study can lead of study design. Study designs are observational
to false conclusions. The strength of inference when the investigator does not manipulate the risk
depends largely on the research methods used in factor but merely selects children with and with-
the recruitment of study subjects (sampling) and out disease and compares them in terms of the risk
in the choice and integrity of the study design. factor(s). Study designs are experimental when
At any stage in study design and implementation, the investigator not only observes but actually
bias can occur. Bias is the result of any process manipulates the relationship between two vari-
that tends to produce results that depart ables. Observational designs provide somewhat

Fig. 1 The role of


inference in drawing
conclusions from clinical
research studies
(generalizability)
476 S.L. Furth and J.J. Fadrowski

weaker evidence of causation, because they fail to can also be thought of as accuracy. Systematic
rule out explanations other than association bias undermines validity. As a research question’s
between the variables studied. Experimental relevance increases, so does the need for validity.
designs can provide much stronger evidence for The clarity and rigor of study design as well as the
causation. In an experimental study, the investi- careful implementation of the research plan
gator controls the independent variable, which is increase the likelihood that inferences from the
the factor hypothesized to produce change in a study are valid.
dependent variable. In an experimental study, sub- Cook and Campbell [1] define several aspects
jects are randomized to receive or not receive the of validity. Statistical validity is the correctness of
independent variable. The goal of the process of study conclusions regarding the existence of a rela-
randomization is to produce study groups that are tionship between two variables. A study lacks sta-
“balanced” in terms of other factors that could tistical validity when it concludes there is no
influence the dependent variable. Unfortunately, relationship between variables when in fact there
experimental designs often are not feasible, ethi- is one or when it concludes there is a relationship
cal, or desirable. Epidemiologic studies of disease when in fact there is none. Statistical validity is
preclude manipulation of risk factors in humans. jeopardized most often by inadequate sample size
Health services researchers studying the public and by improper use of statistical tests.
health impact of changes in health policy rarely If there is a relationship between two variables,
can control these changes. internal validity is the correctness of conclusions
Within the broad categories of observational about whether the relationship between the inde-
and experimental designs, there are many varia- pendent and dependent variables is causal. Inter-
tions. These variations, distinguishing character- nal validity is jeopardized when a study design
istics, primary uses, strengths, and weaknesses, fails to control for factors that could confound the
are discussed, in the section Study Design. Study hypothesized causal relationship.
design also influences the validity of study results. Finally, given a causal relationship between the
independent and dependent variables, external
validity (Fig. 2) is the correctness of generalizing
Validity to other persons, settings, and times. Poor choice
of a study population and inadequate research
Validity is the extent to which study findings cor- procedures are common challenges to external
rectly reflect and explain reality (Fig. 2). Validity validity.

Fig. 2 External and


internal validity in
experimental designs (From
Fletcher RH, Fletcher SW,
Wagner EH. Clinical
epidemiology: the
essentials, 3rd
ed. Baltimore: Williams &
Wilkins, 1996:12, with
permission)
16 Basics of Clinical Investigation 477

Reliability study is easy to conduct and is useful as a prelim-


inary study to reinforce anecdotal evidence, to
While validity is the degree to which data measure generate hypotheses, or to establish variable dis-
what they were intended to measure, reliability is tributions in planning future research. In a field
the extent to which repeated measurements at like pediatric nephrology which deals with many
different times and places, or by different people, rare diseases, this study design can be quite useful.
are reproducible. An instrument measuring an However, because there is no comparison group,
assay may be accurate if, on average, the measures the case series design cannot provide an estimate
vary around the true value, but may not be reli- of risk. An example of a case series is the report by
able, because the measures are widely scattered Porras et al. examining 53 children presenting to a
around the true value. Reliability and precision are single hospital with midaortic syndrome over the
related concepts. A precise measure is one that has past 53 years [2]. The aim of this study was to
nearly the same value each time it is measured. It better understand the outcomes associated with a
is very reliable. Precision can be described statis- multidisciplinary management approach to
tically using the standard deviation of repeated patients with this diagnosis. The authors described
measures. The standard deviation divided by the clinical characteristics at presentation, cardiac and
mean is the coefficient of variation. Small coeffi- kidney function parameters at follow-up, and
cients of variation imply precise measurements. interventions such a number of blood pressure
In summary, the scientific method involves medications used in treatment, and number of
extrapolating inferences from a study situation to percutaneous and surgical procedures.
the larger world. The value of research depends on
the validity of such inferences, which in turn is Cross-Sectional Design
determined by the researcher’s choice of methods. A cross-sectional study is one in which the disease
The following sections explain the strengths and and risk factors are measured at the same time in a
weaknesses of the methodologic choices sample of subjects. Subjects can be categorized as
available. either having or not having the risk factor. Within
each group, the presence of the disease can be
determined. Analytically, the association between
Study Design a particular risk factor and the disease is measured
as the relative prevalence of the disease among
Observational Studies those with, versus those without, the risk factor.
The cross-sectional study design is superior to
There are four major types of observational stud- the case series in that it provides a means for
ies: case series, cross-sectional, case–control, and comparison. Cross-sectional studies are relatively
cohort. Observational designs are weaker than economic, are easy to conduct, and allow simul-
experimental designs in establishing causation, taneous examination of multiple risk factors.
but they are useful when it is not feasible to Cross-sectional studies have a number of lim-
manipulate the independent variable. Studies of itations. Whether the results of such studies gen-
disease etiology usually are observational. In eralize to the population of interest is significantly
these types of observational studies, risk factors impacted by the population from which a cross-
or exposures are the independent variables, and sectional slice is taken. The simultaneous exami-
disease is the dependent variable. nation of exposure and disease in the study popu-
lation can lead to the introduction of significant
Case Series biases that may directly impact results of the
In a case series study, a sample of patients with study. Perhaps the most important limitation of a
common characteristics are selected to describe a cross-sectional study that must be recognized is
clinical or pathophysiological aspect of a disease, the inability to infer causality. If an exposure and
treatment, or diagnostic procedure. A case series disease are assessed simultaneously, then one
478 S.L. Furth and J.J. Fadrowski

Fig. 3 Cystatin
C-estimated glomerular 130
filtration rate (GFR) by
blood lead levels. The thick
line represents estimates
based on restricted
quadratic spline

Cystatin C–Based GFR, mL/min/1.73 m2


transformation for
log-transformed blood lead
levels with knots at the 110
10th, 50th, and 90th
percentiles. Thin lines
represent corresponding
95% confidence intervals.
Data points for the
scatterplot represent
adjusted blood lead and 90
estimated GFR values. The
horizontal axis is in the log
scale (From Fadrowski and
Navas-Acien [3])

70

0.7 1.7 4.1 10.0


Blood Lead Level, μg/dL

cannot be sure the exposure preceded the disease, Case–Control Studies


which is critical to the inference of causality. Case–control studies are also known as
In the United States, the National Health and case–referent, compeer, retrospective, case his-
Nutrition Examination Survey (NHANES) con- tory, and cohort studies. A case–control study
tinually collects data on various exposures and starts with the identification of persons with the
outcomes simultaneously in a representative sam- disease or other outcome variables of interest in
ple of the general population. This data lends itself the population at risk (Fig. 4). A suitable control
well to cross-sectional studies. The association group of persons without the disease or outcome
between lead levels in children and estimated is also selected from the population at risk. This is
glomerular filtration rate (GFR) was examined pictured on the right side of Fig. 4. To examine the
among 769 children who participated in the sur- possible relation of one or more exposures to the
vey from 1988 to 1994 [3]. As can be observed in given disease or outcome, the researcher then
Fig. 3, higher blood lead levels were associated looks back in time to compare the proportions of
with lower estimated GFR. The authors hypothe- the cases and controls exposed and not exposed to
sized that the toxicity of higher lead levels may be the risk factor in question.
causing the lower GFR, but acknowledged that The case–control design has several advan-
the lower GFR, possibly caused by some other tages. It provides stronger evidence of causation
factor, could be causing the decreased excretion of than the cross-sectional design. In a cross-
lead thus leading to higher lead levels. Given the sectional study, outcomes and exposures are
simultaneous examination of lead and GFR in this assessed simultaneously, and the investigator
study, it is impossible to know if lead was causal must infer cause and effect relationships because
of the lower GFR observed with higher lead the temporal sequence cannot be established. In a
levels. case–control design, an attempt to establish a
16 Basics of Clinical Investigation 479

Fig. 4 Design of a
case–control study (From
Fletcher RH, Fletcher SW,
Wagner EH. Clinical
epidemiology: the
essentials, 3rd
ed. Baltimore: Williams &
Wilkins, 1996:213, with
permission)

temporal relationship between the outcome and elevated circulating PTH levels. The authors
exposure is made by starting with a population suggested these findings could help motivate a
of persons with and without the outcome and then larger multicenter study in which the mechanisms
working backward to examine suspected expo- underlying these abnormalities could be more
sures. Thus, compared to the cross-sectional fully elucidated.
design, the investigator is more confident that Case–control studies yield an odds ratio as an
the exposure of interest came before the outcome, estimate of relative risk. This measure is calcu-
not as a result of the outcome. As compared to lated by dividing the odds that a patient was
other study designs, case–control studies are effi- exposed to a given risk factor by the odds that a
cient in the study of rare diseases or those with control was exposed to the risk factor. It can also
long latent periods between exposure and out- be obtained from logistic regression analysis.
come. Whereas cross-sectional or cohort designs Logistic regression allows the investigator to
would require a large number of subjects and time obtain the odds ratio for a given risk factor inde-
to identify risk factors for a rare disease, a well- pendent of other potential risk factors or con-
designed case–control study can identify similar founders using the technique of adjustment.
risk factors with comparatively fewer subjects and Odds ratios are generally a good approximation
much less time and expense. Also adding to the of relative risk if the outcome is rare.
efficiency of the case–control design, several As with any study design, the case–control
potential risk factors for a disease or outcome method has limitations. Case–control studies
can be examined simultaneously. allow for the study of only one disease at a time,
A recent case–control study by Bettinelli as opposed to cross-sectional or cohort studies.
et al. [4] examined phosphate homeostasis in This design does not allow for the measurement
Bartter syndrome. The authors noted that of incidence, prevalence, or excess risk.
hypercalciuria and nephrocalcinosis were well Case–control studies are also subject to error, or
described in patients with this rare disease, but bias, which can threaten the validity of the study.
characterization of phosphate and calciotropic Selection and information biases, the two major
hormones in this disorder was quite limited. To categories of bias, are possible in the case–control
expand knowledge in this area, the authors com- design. Selection bias arises if the manner in
pared plasma phosphate, calcium, PTH, which cases and controls were selected yields an
25-hydroxyvitamin D, alkaline phosphatase, and apparent association when, in reality, exposure
osteocalcin levels between 15 patients with and disease are not associated. For example,
Bartter syndrome and 15 healthy subjects. It was cases, by definition, include only individuals
found that patients with Bartter syndrome had a who have been identified as having the disease
tendency toward renal phosphate wasting and and who are available for study. Those who have
480 S.L. Furth and J.J. Fadrowski

not been diagnosed, have been misdiagnosed, or Nested case–control studies and nested
have died are excluded. If diagnosis or availability case–cohort studies are alternative case-based
is related to the exposure being studied, the sam- hybrid designs that have many advantages. A
ple of cases will be biased. nested case–control study involves selecting all
Avoiding selection bias can be even more chal- cases and control subjects from a known cohort.
lenging in the selection of controls. The control In this design, the controls are free of the outcome
group must be comparable to the cases. They or disease. Nested case–control studies eliminate
should not be chosen in such a way that important the problem of recall bias, because the exposure
differences between cases and controls exist that information is obtained before the outcome has
might influence exposure history and thus limit developed (cohort design). Also, the temporal
the inferences derived from the study. A number sequence between exposure and outcome is
of strategies exist to select a control group that is defined. This design is also much more econom-
at risk for the disease and otherwise representative ical and efficient; the entire cohort need not be
of the same population as the cases. These include analyzed for a given exposure (e.g., via a labora-
sampling cases and controls in the same way (e.g., tory specimen). Nested case–cohort studies also
from the same clinical setting), matching controls use the selection of cases and controls from a
to cases on key variables related to the disease known cohort. However, in this design, controls
(e.g., age), using multiple control groups, and are randomly selected from the initial cohort
using population-based samples of both cases irrespective of outcome. This design permits the
and controls (e.g., using disease registries). delineation of relative risk for an exposure.
Information bias occurs when the case and
control groups differ in terms of the quality of Cohort Design
the data collected to measure risk factors. The Various names, including prospective, follow-up,
retrospective approach to measuring an exposure and longitudinal, have been used to label cohort
in the case–control design introduces the possibil- studies in the past, reflecting the temporal
ity of differential recall between the cases and sequence of exposure and disease in this category
controls. Cases may have been asked more often of observational studies (Fig. 5). The word cohort
about the presence of a given exposure and/or originated from the Latin word cohors, describing
may be more circumspect in their recall of such a group of warriors that marched together. Clinical
exposures. This introduces recall bias, a form of investigators have adapted this term to a specific
information bias because of better, and sometimes type of research study: a group of individuals free
exaggerated, recollection of exposures by cases as of the disease(s) of interest is assembled, their risk
compared to controls. status is determined, and the group is followed
It can be difficult for an investigator to remain over time to measure the incidence of disease.
objective in collecting exposure information. In Comparison of the incidence of disease (or rate
interviewing subjects and in reviewing records, of death from disease) between those with and
there may be a tendency to look more carefully without the exposure of interest permits measure-
or evaluate evidence differently for cases than for ment of the association between the risk factor and
controls. Strategies for dealing with this problem the disease.
include the use of objective measures and to The significance of the cohort design has been
ensure that the individuals collecting data are emphasized by the wealth of scientific data
unaware of the subject’s group status (blinding/ obtained from famous cohorts such as the Fra-
masking). The more subjective the method for mingham Heart Study or the Physicians Health
measuring the exposure, the more important it is Study. Pediatric nephrology has also benefited
to mask the observer. Blinding as to the specific from studies using the cohort design, including
exposure being studied or study hypothesis is the ongoing Chronic Kidney Disease in Children
useful and also can be used to attempt to control study (CKiD), which is a multicenter, prospective
recall bias. cohort study of greater than 500 children with
16 Basics of Clinical Investigation 481

Fig. 5 Design of a cohort


study (From Fletcher RH,
Fletcher SW, Wagner
EH. Clinical epidemiology:
the essentials, 3rd
ed. Baltimore: Williams
&Wilkins, 1996:102, with
permission)

mild to moderate CKD from 48 pediatric nephrol- lead to a misleading association between the
ogy centers in North America. exposure and the disease. Such a characteristic
The cohort design has an obvious niche in would be a confounder.
clinical research. Ethical and practical consider- To avoid misinterpreting such an association,
ations often do not allow for randomization of the investigator must measure potential con-
individuals to an exposure of interest. Cohort founders and adjust for them in the analysis. Mul-
designs allow for the examination of exposure tivariable analyses are examples of statistical tools
and disease associations under such used to adjust for confounders. However,
circumstances. unsuspected confounders might still jeopardize
Cohort studies can be classified as concurrent the validity of conclusions.
or nonconcurrent. In a concurrent cohort study The relationship between blood pressure con-
(also referred to as a prospective or longitudinal trol and left ventricular hypertrophy was recently
study), the clinical investigator identifies the pop- examined in children in the CKiD cohort
ulation and collects extant exposure information [5]. Using a linear mixed statistical model, the
and then follows the cohort to a designated point study looked at the exposure of casual blood pres-
in the future. Nonconcurrent cohort studies (i.e., sure measurement z scores on the outcome of left
retrospective, historical, and nonconcurrent pro- ventricular mass index (LVMI). In acknowledg-
spective) require the investigator to identify a ment of their potential impact on the exposure
cohort that has been delineated in the past, along and/or outcome, additional variables were
with information regarding the exposure(s) of included in the statistical model including sex,
interest. This population can then be followed race, type of CKD diagnosis, duration of CKD
for the development of a given disease in the diagnosis, age, height at each visit, GFR, use of
more recent past, the present, or into the future. angiotensin-converting enzyme inhibitor (ACEi)
Traditionally, the outcome of interest in cohort or angiotensin receptor blocker (ARB) medica-
studies is the ratio of the incidence of disease in tion, use of other antihypertensive medications,
those with the exposure divided by the incidence and anemia status. In addition to systolic blood
of disease in those without the exposure. This can pressure, predictors of LVMI in this study
be interpreted as the relative risk for disease in included anemia and use of antihypertensive med-
many cases. When calculating and interpreting ications other than ACEi’s or ARBs.
risks in the cohort design, the absence of random- Because risk factors are measured before out-
ization must be taken into account. Because the come or disease, the temporal sequence of risk
investigator is merely observing the exposure and and disease is established, and the potential for
not controlling for it via randomization, subjects biased risk measurement is avoided. Several dis-
with and without the risk factor might differ in eases or outcomes can be measured, and disease
terms of other characteristics that are related to the occurrence can be measured in terms of incidence,
disease. If the characteristic is related to both the not just prevalence. Cohort studies are an ineffi-
exposure being evaluated and the disease, it can cient way to study rare outcomes because in this
482 S.L. Furth and J.J. Fadrowski

Fig. 6 Design of a
randomized trial (From
Fletcher RH, Fletcher SW,
Wagner EH. Clinical
epidemiology: the
essentials 3rd
ed. Baltimore: Williams
&Wilkins, 1996:139, with
permission)

scenario, large sample size and long follow-up are to a decline of 50 % in the GFR or progression to
needed, which significantly increases the cost and end-stage renal disease. In this and other con-
effort associated with the study. A nonconcurrent trolled clinical trials, randomization is the key
cohort design can reduce cost, but it decreases the feature of its experimental nature. Participants
investigator’s control over subject selection and are randomly assigned to the group that will
risk factor measurement. receive the intervention (the intervention, treat-
ment, study or experimental group) or the group
that will not receive the intervention (the control
Experimental Studies or placebo group) (Fig. 6). Through randomiza-
tion, all potential confounders, both those recog-
In an experimental study, the investigator controls nized by the investigator and those that are not
the independent variable, also known as the inter- suspected, are likely to be balanced between the
vention or treatment, and then observes the effect study groups. In other words, the groups in this
on an outcome or series of outcomes. The classic study are considered to be the same except for the
form of experimental design is the clinical trial. treatment they receive. Differences in rates of
progression of kidney disease in the groups are
Randomized Controlled Clinical Trials attributable to the intervention, because the effect
Most investigators, clinicians, and patients are of confounders has been ruled out by the balance
familiar with the “gold standard” of experimental achieved by randomization. Therefore, experi-
designs, the randomized blinded controlled clini- mental designs offer stronger evidence of causal-
cal trial. Such trials are considered “gold stan- ity than do observational designs.
dard” because the rigid design helps minimize In an experimental study, it is important to
the influence of confounding variables and bias, ensure that subject assignment is truly random.
allowing the true effect of the intervention to be This can be achieved by using random numbers,
elucidated. either through computer-assisted assignment or
The Effect of Strict Blood Control and ACE manually, with a table of random numbers. Some-
Inhibition on the Progression of Chronic Renal times blocking is used in conjunction with random
Failure in Pediatric Patients (ESCAPE) trial is an assignment. A block of subjects is simply a set
example of a randomized controlled clinical trial number of consecutive study enrollees. Within
in children with kidney disease [6]. In this study, each block, a predetermined number of subjects
children with CKD received an ACEi at a stan- are randomly assigned to each study group. For
dardized dose for a 6-month period. They were example, if the block size is set at six and two
then randomly assigned to intensified blood pres- study groups of equal size are desired, then three
sure control (target 24-h mean ambulatory blood subjects in each block of six are randomly
pressure below the 50th percentile) or “conven- assigned to one group and three to the other.
tional” (at that time) blood pressure control (mean Blocking is useful when study enrollment is
arterial blood pressure in the 50th to 95th percen- expected to be prolonged. Over extended periods,
tile). The primary end point in this study was time both study procedures and outside conditions can
16 Basics of Clinical Investigation 483

change. Blocking ensures that the study groups blinded. Once completed, the pharmaceutical
will be balanced with regard to such changes. manufacturer may apply to market the drug for
A block randomization scheme, with a block the indication(s) studied in the phase III trial.
size of four, was used in the ESCAPE trial. Phase IV clinical trials are post-marketing studies
Experimental studies, like observational stud- (post-licensure) designed to delineate more infor-
ies, are subject to measurement bias. Research mation about the drug’s risks, benefits, and opti-
staff should be masked or blinded to the subject’s mal use in circumstances approximating “real-
group assignment during data collection, espe- world” conditions.
cially if any outcome measures are not strictly
objective. If outcomes are measured, without sub- Alternative Designs
ject or staff knowledge of the subject’s group It is not always feasible to conduct a traditional
status, they are less likely to be influenced by randomized controlled clinical trial for every
expectations about potential differences between treatment. In such scenarios, multiple alternative
treatment and control group outcomes. Studies in experimental study designs are available and may
which both the participant and the staff are provide useful information regarding the treat-
unaware of the treatment group status are known ment under consideration. A few examples of
as “double blinded.” There may be situations in such alternative designs follow. Crossover trials
which blinding the clinical staff and patients is randomly assign half of the study population to
difficult or impossible. Clinical trials that are not start with the control period and then subsequently
blinded are known as open-label trials. Such switch to active treatment; the other half are on the
designs are common in cancer clinical trials, as opposite schedule. Such trials allow each partici-
treatments being compared are often complex, pant to serve as their own control, allowing for
with different side effect profiles and delivery increased statistical power and thus fewer partic-
protocols, and thus masking is not feasible. The ipants. However, these studies generally take a
medical staff in the ESCAPE trial had to be aware longer period of time. The analysis and
of the assigned blood pressure target as they were interpretation of the results may also be
integral in titrating medications to achieve this complicated if the treatment effects are thought
target. Thus, the ESCAPE trial was not a double- to persist for a period of time even after the
blinded study. intervention has been ceased. A before–after trial
Clinical trials typically provide the necessary compares the outcomes of different types of treat-
medical evidence to bring a new treatment into ments in a group or groups of interest by taking
use, and for drugs, such trials are referred to as advantage of calendar time. In such a study, out-
“phases.” A Phase I trial is most often the first comes in individuals receiving one type of treat-
stage in testing a new drug in humans and may ment during a given period are compared with
include healthy participants and/or patients. In individuals at a subsequent time, who have
these trials, information on the distribution, received a different treatment. Although econom-
metabolism, excretion, and side effects of the ical to perform, the results of such designs may be
drug is investigated. The optimal dose of the more prone to error as it may be difficult to know
drug to deliver is also investigated. Such studies and/or control factors independent of the treat-
are typically not randomized or blinded. Phase II ment that may have also influenced the outcome
clinical trials are designed to evaluate the effec- of interest.
tiveness and short-term safety and side effects
associated with the drug. These trials are generally Issues with Analysis in Experimental
carried out in persons having the disease of inter- Designs
est. Phase III trials are expanded trials to prove the It is not uncommon in clinical trials to have
effectiveness of a drug and provide the informa- patients who were assigned to one group switch
tion necessary for physician labeling of the drug. to a different group. For example, a patient
These studies are most often randomized and assigned to receive the active treatment under
484 S.L. Furth and J.J. Fadrowski

study may discontinue the treatment. Alterna- Important Issues in Carrying Out a
tively, a patient assigned to the control group Research Plan
may end up receiving active treatment. To avoid
introducing bias in the results, it is common prac- Selection of Subjects
tice in clinical trials to analyze the results
by intention-to-treat. In such conservative In any research study, one would like to extrapo-
analyses, every patient is grouped according to late the findings to all patients with the condition
his/her original randomization assignment when of interest (Fig. 2). The study population is the
analyzing the results, regardless of whether the group that is meant to represent the target popula-
patient actually received the assigned treatment tion from which a sample is drawn. Sampling
or not. Intention-to-treat analyses may dilute the decisions involve defining the study population
effect of the treatment of interest, but more impor- and sample.
tantly, likely minimize the introduction of bias
into the study which may lead to erroneous Defining the Target Population
conclusions. Although there is no one single ideal target popu-
To maximize the yield of arduous and costly lation, the investigator needs to consider the ram-
clinical trials, investigators may wish to perform ifications of one definition versus another. If the
subgroup analyses, defined as an evaluation for investigator were interested in studying risk fac-
treatment effects within a subset of patients. tors for a specific disease, the target population
Related to subgroup analyses, post hoc analysis could be defined as all children with this disease
refers to examining the data after the study has or a subset of them (e.g., children of a certain age).
concluded for associations that were not specified The broader the target population, the greater the
a priori. Although subgroup analyses and post hoc generalizability of the study findings. On the other
analyses may provide additional useful informa- hand, the increased heterogeneity of a broadly
tion, due to analytic challenges, particularly in the defined target population could introduce variabil-
area of sample size, results may be misleading. ity among subgroups in terms of the importance of
Given these statistical limitations, it is risk factors. For example, a particular characteris-
recommended that all hypotheses and intended tic could be a major risk factor in some population
analyses be stated prior to the initiation of the subgroups but not in others. Assessing the impor-
study, which may help in planning the design of tance of risk factors within subgroups requires a
the study, including the sample size. In the larger study sample and perhaps a more complex
event that post hoc analyses are performed, sampling design.
they should be clearly labeled as such so that the
reader is able to identify the potential limitations Defining the Study Population
to any conclusions derived from such analyses. A practical consideration in defining the target
A report in the New England Journal of Medicine population is availability of the population for
reviews the challenges of subgroup analyses and study. The investigator could have all children
provides guidelines for their use within the available seen in a particular clinical setting. Inso-
journal [7]. far as children seen in this setting are representa-
In summary, many research designs are avail- tive of the target population, the clinical site
able to the investigator. No single design is best would be a good choice for study; the experience
for all research questions. Although experimental of its enrollees could be considered generalizable
designs are superior to observational designs in to the target population. If children enrolled in the
addressing threats to internal validity, they are not clinical setting differ systematically from the tar-
always feasible or ethical. The most appropriate get population, sampling bias is introduced. For
design for a given research question is the design example, tertiary care pediatric nephrology cen-
that maximizes internal validity within the con- ters might be more likely to serve children with
straints of the research environment. advanced stages of the kidney disease or more
16 Basics of Clinical Investigation 485

severe or complicated cases. Studying only these There are several types of probability sam-
cases may introduce bias toward only studying the pling. In simple random sampling, each element
most complex forms of a particular disease. Sam- has an equal chance of being selected. In system-
pling bias impairs the generalizability of study atic sampling, each element in the population is
findings. Representativeness, therefore, is a assigned a consecutive number, and every nth
prime consideration in defining a study popula- element is sampled. Systematic sampling is easy
tion. Investigators should evaluate the representa- to use, but it will generate a biased sample if the
tiveness of candidate settings and the likely sampling fraction (e.g., every tenth case) is the
implications of potential biases. One possible same as some periodicity in the ordering of cases
approach to this in studies of patients with kidney in the population. For example, if every tenth
disease is to compare the characteristics of partic- patient is sampled in a clinic where ten patients
ipants in a study to known characteristics of the are seen each session and the most complex cases
larger population to whom one would like to are scheduled first, then the sample will contain
generalize the results. either all complex cases or no complex cases,
depending on the first element drawn. Stratified
Defining the Sampling Scheme random sampling is useful when one believes that
Just as we generalize from the study population to population subgroups differ in important ways.
the target population, we generalize from the The population is divided into the subgroups, or
study sample to the study population. Sample strata, of interest. Simple or systematic random
statistics are measures that pertain to the samples samples are then drawn from each stratum. Clus-
that are studied. A sample mean, for example, is ter sampling is useful when it is difficult or costly
the sample’s average score on a particular mea- to sample elements in a population individually.
sure, and a sample standard deviation expresses Instead of elements, groups of elements are sam-
the variability of the sample scores. The sample pled. For example, in a study of school children,
statistics are the investigator’s best estimates of the investigator could take a probability sample of
the population parameters. The sample mean is classrooms and then study all the students within
the best estimate of the population mean; the each selected classroom. The selected classrooms,
sample standard deviation is the best estimate of in combination, must be representative of the
the population standard deviation. overall population. As with stratified sampling,
Extending beyond inference to hypothesis test- formulas for calculating variance must be modi-
ing, sample statistics of the association between fied, and consultation with a statistician is
variables are the best estimates of these associa- recommended.
tions in the target population. The association Nonprobability sampling techniques include
between a hypothesized risk factor and the occur- convenience sampling, quota sampling, and pur-
rence of disease in the study sample (perhaps posive sampling. A convenience sample is one
measured by odds ratios or relative risks) is the that is most readily obtained without die use of
investigator’s best estimate of the association random sampling. A quota sample is a conve-
between the risk factor and disease in the target nience sample drawn to assure specified numbers
population. of subjects in specified strata, without the use of
Probability theory is the rationale for extrapo- random sampling. A purposive sample is one in
lating inferences from a study sample to the refer- which subjects are selected because they are
ence population. A probability sample is one in judged to be representative of the population of
which every subject, or element, in the study pop- interest.
ulation has a known probability of being selected. Probability sampling is preferred but not
A nonprobability sample is one in which the prob- always possible. In clinical research, the investi-
abilities of selection are unknown. It is legitimate to gator is often limited to a particular clinic popula-
extrapolate from a sample to its population only if tion. If a clinic population is believed to be
probability sampling has been used. representative and if it is larger than the number
486 S.L. Furth and J.J. Fadrowski

of subjects needed for study, the investigator Table 1 Example illustrating how a, b, and effect size
should use a probability sampling technique to affect sample size
draw the study sample. Confidence Power
An example of probability sampling using Total
stratified sampling techniques can be seen in a 1  α (%) 1  β (%) Effect size (%) sample no.
survey study of adult and pediatric nephrologists 95 80 10 ! 40 76
95 90 10 ! 40 96
[8]. The authors created a survey containing ten
95 90 10 ! 20 572
case vignettes to assess whether increased experi-
95 99 10 ! 40 156
ence with pediatric patients influenced nephrolo-
Adapted from sample size calculator (Statcalc) in Epi-info
gists’ recommendations for peritoneal or Stat Calc. Available at http://www.cdc.gov/epiinfo
hemodialysis in otherwise identical patients with
ESRD described in the vignettes. Because the
authors wanted the survey respondents to repre-
sent the population of US adult and pediatric or the rate of occurrence of disease (in a cohort
nephrologists, they randomly selected a represen- study). The effect size is an estimate of how much
tative sample of nephrologists in five geographic better than the comparison group you expect a
regions of the United States. Each randomly treatment group to be in a clinical trial or how
selected nephrologist was mailed a survey increased the risk of a particular disease is in the
containing ten case vignettes to assess what fac- setting of a particular risk factor (in a cohort
tors affected the nephrologists’ dialysis study). An illustration of estimated sample sizes
recommendations. for given α, β, and effect sizes is shown in Table 1
for a study comparing differences in proportions
Determining Sample Size in two groups.
In any study, several factors determine the Table 1 illustrates how varying the acceptable
required sample size. This section describes levels of α, β, and effect size influences sample
those that come into play in several common size. For example, if we designed a study to deter-
types of investigations. Detailed sample size for- mine whether a new drug could “cure” 40 % of
mulas and tables are beyond the scope of this patients compared to an old drug that “cured”
chapter, but several excellent references are listed 10 % of patients, we would have 90 % power to
in Suggested Reading. Briefly, to estimate sample see such an effect with 95 % confidence in a total
size, the researcher needs to set the acceptable sample of 96 patients (see row 2 in Table 1). In
level of α (probability of type I error) and β (prob- contrast, to obtain a significant result
ability of type II error) and determine the effect documenting a smaller effect size from the old
size that one is likely to see. In determining sam- drug cure rate of 10 % to a new drug cure rate of
ple size, the probability of making a type I error, α, 20 % with the same α = 0.05 and 90 % power, we
is usually set at 0.05. This is the probability of would need to study 572 patients.
concluding that an association between two vari-
ables exists when it does not. β error is the prob-
ability of concluding that no association exists, Attrition of Study Subjects
when in fact it does. The reader will be more
familiar with β error in terms of its relationship Sample size calculations determine the number of
to “power.” The power of a study is equal to 1  β. subjects needed at the study’s conclusion. In
In many studies, β is customarily set at 0.2 for a determining the number of subjects to enroll, the
power of 80 %. If β is set to 0.1, the power of the investigator must estimate attrition rates and
study is 90 %. enroll a sufficiently large sample to compensate
In addition to specifying α and β, the researcher for study dropout.
must also determine an estimate of the response to Even if probability sampling is used to define
treatment in one of the groups (in a clinical trial) the study, subject attrition could produce a biased
16 Basics of Clinical Investigation 487

sample at the study’s conclusion. An attrition rate male or female gender; nominal, i.e., non-ordered
of more than 25 % is cause for concern. In data categories such as race: White, African American,
analysis, subjects completing the study should be Asian, Native American, or Pacific Islander; or
compared with those who drop out to determine ordinal, i.e., ordinal categories such as a pain
whether the two groups differ in a clinically sig- rating scale: none, mild, moderate, and severe.
nificant way. Such differences must be considered
in interpreting the study findings.
Sources of Data

Data Collection: Measurement Study data can be collected from existing sources
or can be generated specifically for the specific
Decisions on what data to collect and how to do so research hypothesis being tested, using surveys,
begin with specifying the variables that need to be interviews, or observations. Most studies combine
measured and operationally defining each. The both strategies.
investigator will need to evaluate the suitability An enormous variety of existing data sources is
of existing measures and determine whether to use available, including medical records, vital
an existing measure or develop a new one. The records, national and local health surveys, and
data sources for each variable must also be iden- census data. Health programs often keep records
tified. Finally, the investigator should specify the of services provided, and billing records can be
level of measurement of each variable. An effi- especially helpful. Examples of existing data
cient way to document the data collection plan is sources in pediatric kidney disease include the
to make a table with columns listing the variables US Renal Data System (USRDS) (www.usrds.
to be measured, their operational definitions, the org) and data collected by the North American
data source(s) for each variable, and level of mea- Pediatric Renal Trials and Collaborative Studies
surement for each. This section describes issues (NAPRTCS) (web.emmes.com/study/ped/).
pertaining to each of these tasks. Existing sources can provide data for time periods
and individuals otherwise unavailable to the
investigator. The number of studies published by
Identifying the Variables to Be the NAPRTCS and their tremendous contribution
Measured to our understanding of clinical outcomes in pedi-
atric kidney transplantation and dialysis illustrate
Researchers are often tempted to collect as much this point. The chief disadvantage of existing
information as possible. This can be costly, in sources gleaned from registry data is that the
terms of time, money, and data quality. The inves- data are often not collected as systematically as
tigator should be able to justify each variable to be in a prospective research study. Important data
measured. Most important are the hypothesized elements are sometimes missing. Incomplete
independent and dependent variables. In addition, data and inaccuracies are also possible with
identified potential confounders should be mea- registry data.
sured. Finally, data characterizing the study pop- Primary data collection is expensive and is
ulation and sample will be needed to describe the limited to subjects available to the investigator.
study’s generalizability. On the other hand, the investigator’s control over
data collection makes data quality more certain.
Many primary data collection strategies are avail-
Types of Variables able, including mail surveys, mass-administered
questionnaires, telephone and in-person struc-
Variables can be continuous, such as age, height, tured and unstructured interviews, direct observa-
and weight, or categorical. Categorical variables tion, and videotaping and audiotaping. Choosing
can be binary, with two possible outcomes such as a strategy should be based on the research
488 S.L. Furth and J.J. Fadrowski

question, the sensitivity of the data to be collected, Analytic Plan


the literacy of the population to be studied, and the
resources available for the study. The key factor Data Analysis
should be data quality – that is, which method will
provide the most complete and accurate informa- Investigators often defer considering data analysis
tion within budgetary constraints. until the data have been collected. This is a serious
mistake. Study planning should include an ana-
lytic plan of the steps needed to answer the study
Assessing Data Quality questions once the fieldwork is completed.

One strategy to enhance data quality is to train Use of Statistical Tests


research staff thoroughly. Often, data collection As noted earlier, statistical validity is the correct-
staff works independently. To ensure that they ness of study conclusions regarding group differ-
follow study procedures, the protocol for data ences and variable relationships. A key threat to
collection should be detailed in a study manual. statistical validity is the use of inappropriate sta-
Training sessions should be held to explain the tistical tests. Variable types and variable distribu-
study’s aim to staff, as well as how each one’s role tion, as well as the hypothesis being tested,
fits into the big picture. Staff should be given determine the correct type of statistical test. The
ample time to practice their data collection skills. important properties of a variable’s distribution
Once the fieldwork of the main study begins, staff are its location, spread, and shape. Measures of
should be encouraged to bring problems to the location include the median (middle observation),
attention of supervisory staff. Such problems the mean (arithmetic mean or average), and the
should be resolved in a timely fashion, and the mode (most frequent value). Measures of spread
resolution should be documented and added to the of a distribution include the range, which equals
study manual. In this way, research staff are kept the maximum minus the minimum value; the
apprised of changes in the study protocol and are interquartile range, which equals the 75th minus
impressed with the importance of adhering to it. the 25th percentile; and the variance, which
After the instrument pretesting and staff train- equals the average squared deviation from the
ing, the investigator should pilot test the data mean and the standard deviation. The shape of a
collection activities. A pilot test is a dress distribution can be described by its skewness, i.e.,
rehearsal of the activities for selecting study sub- symmetry, and its kurtosis, i.e., “peakedness.”
jects, contacting them, securing informed consent, Parametric statistical tests are based on assump-
and collecting and processing data. Activities that tions about parameters of the population and are
do not work as planned should be modified and the most powerful tests available in situations in
the pilot testing continued until the fieldwork pro- which these assumptions are met. Nonparametric
cedures run smoothly. statistical tests are based on fewer assumptions
Data quality should be monitored during the about the population, so they are appropriate in
main study. Interviews and questionnaires should situations in which the assumptions underlying
be reviewed as they are completed to allow parametric statistics are not met.
recontacting subjects to correct errors. The reliabil- Assumptions vary by statistical test. If a test is
ity of subjective measures and those requiring spe- used in a situation that violates its assumptions, it
cial technical skill should be assessed. For studies will be inaccurate, leading to a misleading mea-
with unsupervised interviewing of subjects, it is sure of statistical significance. This, in turn, will
wise to validate a portion of the completed inter- lead to an incorrect estimate of the likelihood of a
views handed in by fieldworkers. This can be done type I error.
by recontacting a random sample of subjects and In developing the analytic plan, the investiga-
then asking them to verify their responses to a tor should consider the assumptions of
subsample of the interview questions. candidate tests in determining which ones to use.
16 Basics of Clinical Investigation 489

A discussion of specific statistical tests is beyond is the use of a kappa statistic which expresses the
the scope of this chapter, but a framework for degree of concordance beyond that due to chance.
deciding which tests to use can be given. In this Cronbach’s alpha expresses internal consistency
framework, three factors determine the type of test among three or more variables that measure the
to use: the major analytic question to be answered, same general characteristic.
the levels of measurement used, and the number When the study’s purpose is to describe a pop-
and independence of comparison groups. ulation, the investigator makes inferences from
In preparing the analytic plan, the investigator sample statistics to population parameters. Sam-
needs to translate the research question into ana- ple proportions and measures of central tendency
lytic terms. Three major analytic approaches are (mean, median, and mode) and dispersion (stan-
to describe characteristics of the sampled popula- dard deviation, range) are used to estimate these
tion, to compare groups of subjects, and to mea- parameters in the population. Confidence inter-
sure associations among variables. vals can be constructed around proportions and
Where group comparisons are to be made, the means to express the certainty of the sample-
appropriate statistical test is also determined by based population estimates. When the study’s
the number of groups to be compared (two objective is to compare two or more groups, sam-
vs. three or more) and by whether comparison ple group differences in proportions and means
groups are independent or matched. Thus, in a are used to estimate such differences in the popu-
study of cases matched with sibling controls, it lation. Statistical tests of significance can be used
would be inappropriate to use a statistical test for to assess the certainty of sample-based inferences
independent groups. about group differences in the population. The
As decisions about level of measurement and appropriate statistical test depends on the number
the selection of study groups are part of study of groups compared, whether subjects in the
planning, it is easy to see how these decisions groups are matched, and the level of measurement
are better informed if their ramifications for data of the variable on which the groups are being
analysts are considered. Level of measurement, compared. Table 2 displays bivariate statistical
study group formation, and data analysis are all tests commonly used in assessing the significance
interrelated, and all should be considered part of of group differences. For a variable with a normal
study planning. distribution, a t-test compares two means, while
When the study’s purpose is to assess the rela- ANOVA can compare means in three or more
tionship between two continuous variables, the groups. Chi-square compares proportions.
degree of concordance can be expressed as a When normality assumptions cannot be made,
simple correlation coefficient. A related approach the sign test can be used for a single sample or

Table 2 Bivariates Statistical Tests


Two groups Three or more groups
Level of Independent Independent (unmatched
measurement (unpaired groups) Paired groups groups) Matched groups
Nominal Chi-square of McNemar’s test Chi-square Cochran’s test
dichotomy Fisher’s exact test
More than Chi-square McNemar’s test Chi-square Cochran’s test
two categories
Ordinal Mann-Whitney Sign test Kruskal-Wallis one-way Friedman two-
test analysis of variance ANOVA way ANOVA
Wilcoxon matched-
pairs signed-ranks test
Interval t Test for groups t Test for pairs One-way ANOVA ANOVA for
repeated
measures
490 S.L. Furth and J.J. Fadrowski

paired sample to assess whether the medians of population who develop disease, in case–control
the sample and a reference, or two samples being studies, we have the proportion of the cases who
compared differ. The Wilcoxon signed rank test were exposed and the proportion of the controls
can be used, when the data are on an interval scale, who were exposed. In case–control studies we
and makes use of the magnitudes of the differ- utilize the concept of odds to define the odds
ences between measurements and a hypothesized ratio, which approximates the relative risk if the
location parameter. If the variable of interest is incidence of disease is low. We compare the odds
measured on an ordinal scale, the Mann–Whitney of a case having been exposed to a particular
test can be used to assess whether the two factor to the odds of a control being exposed to
populations have different median values. that factor. As in the case of relative risk, if the
When the research aim is to measure the asso- exposure is not related to the disease, the odds
ciation between variables, sample statistics again ratio will equal 1.0. If the exposure is positively
are used to estimate population parameters. The related to the disease, the odds ratio will be greater
variables’ levels of measurement determine the than one.
appropriate statistical measure of the strength of Studies of the combined and relative impacts
their association, the appropriate test of the statis- of multiple independent variables, or the effect of
tical significance of the association, and the cer- an independent variable after controlling for other
tainty of estimates of its strength. For continuous factors, will require multivariable analytic tests.
data, Pearson’s correlation coefficient is used to The appropriateness of a multivariable technique
measure association. For dichotomous variables, is determined by the levels of measurement of the
the odds ratio and relative risk (RR) are measures independent and dependent variables. Multiple
of the degree of association between two factors. linear regression analysis can be used to assess
Analytic studies in the medical literature often are association between a putative exposure and a
designed to determine whether there is an associ- continuous outcome while adjusting for other pos-
ation between exposure to a factor and develop- sibly confounding factors. Multiple logistic
ment of disease. If there is an association, the regression analysis can be used to assess the asso-
question is how strong the association is. To ciation between a putative risk factor and a binary
assess the strength of the association, we measure outcome measure while adjusting for other poten-
the ratio of the incidence of disease in exposed tial confounding factors. These measures can be
individuals to the incidence of disease in used to calculate an adjusted relative risk, or an
nonexposed individuals. This ratio is called the adjusted odds ratio. The references cited at the end
relative risk. If the risk in exposed individuals is of this chapter describe the applicability and inter-
equal to the risk in unexposed individuals, the pretation of the most commonly used multivari-
relative risk is 1.0, and there is no association. If able statistical tests.
the risk in exposed individuals is greater than in In addition to describing association between
unexposed (RR > 1.0), then there is an associa- two variables, and assessing the risk associated
tion that may suggest that the exposure confers with an exposure and an outcome, another com-
risk. If the risk in exposed is less than in mon research question in the medical literature
unexposed (RR < 1.0), the exposure may be includes assessing the time from a particular expo-
protective against risk. The relative risk can only sure until an outcome such as death, or hospitali-
be calculated in a prospective study, as it requires zation or transplantation. A commonly used
incidence of disease. In a case–control study, statistical tool to assess the time to an event is
since we do not know incidence, we cannot cal- survival analysis, or Kaplan–Meier analysis.
culate the RR directly. Instead of the proportion of Kaplan–Meier analyses can be used to compare
the exposed population who develop disease com- survival between two treatment modalities. When
pared to the proportion of the unexposed adjusting for other potential confounding
16 Basics of Clinical Investigation 491

variables in a survival or time to event analysis, seen in the “universe” of pediatric patients with
Cox proportional hazards methods can be used, hyperlipidemia and chronic renal failure. To esti-
yielding a hazard ratio which can be thought of as mate the “true” reduction in lipid levels (which
comparable to a relative risk. can never be directly measured), we can generate
a confidence interval around our estimate.
In any study, construction of a confidence
Statistical Significance and Confidence interval around the point estimate gives us a
Intervals range of values in which we can be confident
that the true value resides. A confidence interval
Most readers of the medical literature will be gives a sense of the estimates precision; it extends
familiar with the term statistical significance, evenly on either side of the estimate by a multiple
which is most often referred to in clinical reports of the standard error (SE) of the estimate. In our
as a “p value <.05.” This highly sought after example, our study might yield an estimate of the
result of a statistical test refers to the probability drop in serum low-density lipoprotein cholesterol
of α, or a type I error. A p value of .05 in a study levels of the treatment group of 30 % with a
means that there is a 5 % chance that the results standard deviation of that estimate of 20 %. One
seen in the study could have occurred by chance. could then use this estimate to generate a confi-
However, the authors have concluded that this dence interval around this estimate. In the medical
probability is low enough for them to accept the literature, one will most often see references to
alternative hypothesis (that there is a real differ- 95 % confidence intervals. The general equation
ence between groups) and to reject the null for a 95 % confidence interval is equal to the
hypothesis (there is no difference between estimate 1.96 times the SE of the estimate. The
groups). It is important to note that the p value in factor 1.96 comes from the standard normal dis-
a study result depends on the size of the observed tribution, in which 95 % of estimates would fall
difference between the groups in question and the within 1.96 SEs of the mean. If one wanted to
size of the sample of patients studied. Standing increase the probability of including the true esti-
alone, the p value does not convey any sense of mate in the confidence interval, one could gener-
the magnitude of the treatment effect seen in the ate a 99 % confidence interval, which would equal
study or the precision of the estimate of the size of the estimate 2.56 times the SE of the estimate.
the treatment effect. Confidence intervals, in con- Because the SE of an estimate is equal to the
trast to p values, can convey this information in a standard deviation of the population divided by
more meaningful way. the square root of the sample size, N, one can see
For any estimated value, it is useful to have an that a larger sample size is needed in a study to
idea of the uncertainty of the estimate in relation generate a precise estimate of a treatment’s effect.
to the true value it is trying to approximate. For Given the same standard deviation, the SE in our
example, if we designed a study to estimate the study would be smaller if we studied 100 children
beneficial effect of a new lipid-lowering medica- compared to 20 children. The larger study would
tion in chronic renal failure in adolescents, we generate a narrower 95 % confidence interval. The
would try to recruit a large representative sample strict interpretation of a 95 % confidence interval
of hyperlipidemic adolescents and randomize is that this is the range of values for the true
them to treatment with a new lipid-lowering med- population estimate that is consistent with the
ication. From our study, we might want to esti- data observed in the study. In our hypothetical
mate the magnitude of lipid level reduction example, the smaller study might give us the
associated with the new medicine. We might also opportunity to conclude that the new lipid lower-
want to use this estimate as an approximation for ing is associated with a 30 % reduction in
the “true” reduction in lipid levels that would be low-density lipoprotein cholesterol with a
492 S.L. Furth and J.J. Fadrowski

relatively broad 95 % confidence interval of million population. This compared to 217 in 1991.
21–39 %, whereas the larger study yields a more The prevalence of ESRD in 2011 (also referred to
precise estimate. The 95 % confidence interval as point prevalence) was 1,924 cases per million
around the same point estimate of a 30 % reduc- population. Based on these definitions, we know
tion is 26–34 % in the study with the larger that 362 (19 %) of the 1,924 prevalent cases of
sample size. ESRD per million population in 2005 were new
cases.

Topics Related to Clinical Decision


Making The Diagnostic Test

Clinicians are routinely faced with patients with When utilizing a diagnostic or screening test to aid
unknown diagnoses, and patients expect that the in the care of a patient, a clinician must know
clinician will know how to efficiently and accu- “how good” or valid is the test. Terms used to
rately diagnose the problem with which they are describe “how good” a test is include sensitivity
presenting. Diagnostic acumen relies in large part and specificity. The sensitivity of a test describes
in understanding the “epidemiology” of the pos- the ability of the test to correctly identify those
sible diagnoses, as well as the strengths and lim- that have the disease. A sensitive test has a low
itations of the various tests that might be false-negative rate, meaning the test result will
performed to diagnose such diseases. The follow- not frequently be negative in those who have the
ing section provides an introduction to concepts disease. The specificity of a test describes the
and terms that are related to the science of clinical ability of the test to correctly identify those who
decision making. do not have the disease. A specific test has a low
false-positive rate, meaning the test result will not
frequently be positive in those who do not have
The Disease the disease.
Table 3 from the American Academy of Pedi-
When describing the burden of a disease in a atrics’ Practice Parameter for the diagnosis of
population, the terms incidence and prevalence initial urinary tract infections in children exam-
are often used. Incidence is defined as the number ines the sensitivity and specificity of various com-
of new cases of a disease that occur in a popula- ponents of the urinalysis [10]. For example, the
tion at risk for developing the disease during a sensitivity of nitrite is reported as 53 %. Thus,
specified period of time. Prevalence is defined as among children with a urinary tract infection,
the proportion of persons present in the population
currently affected by the disease at a specified
point in time. Distinguishing between these
Table 3 Sensitivity and specificity of urinalysis compo-
terms is important when considering the burden nents (13)
of given disease. When prevalence is reported,
Sensitivity % Specificity
people affected by the disease for varying Test (range) % (range)
amounts of time are included; these are not nec- Microscopy: WBCs 73 (32–100) 81 (45–98)
essarily all new cases. Those with severe forms of Microscopy: bacteria 81 (16–99) 83 (11–100)
the disease may have died and thus, depending on Leukocyte esterase 83 (67–94) 78 (64–92)
the time specified in the definition, may not be Nitrite 53 (15–82) 98 (90–100)
included in the prevalence. The incidence and Leukocyte esterase or 93 (90–100) 72 (58–91)
prevalence of end-stage renal disease (ESRD) nitrite positive
are routinely reported by the US Renal Data Sys- Leukocyte esterase or 99.9 (99–100) 70 (60–92)
tem (USRDS) ([9] Annual Data Report). In 2011, nitrite or microscopy
positive
the incidence was 362 (new) cases of ESRD per
16 Basics of Clinical Investigation 493

this test will be positive approximately 53 % of the Likelihood ratios (LR) are another tool to help
time. Knowing the relatively low sensitivity of the the clinician utilize the results of a given test
nitrite test, a clinician would not be reassured that diagnostically. When presented with a patient
a urinary tract infection does not exist if the result with particular signs and symptoms, the clinician
is negative. The specificity of the nitrite test is has an initial assessment of the likelihood (pretest
98 %. Thus, among children without a urinary probability) of a particular disease. A diagnostic
tract infection, this test is negative approximately test ordered to help determine the presence of a
98 % of the time. The high specificity of the nitrite disease may be positive or negative. LRs help
test informs the clinician that a low false-positive inform the clinician as to how much the test result
rate exists. As can be observed in the figure, and as should shift his/her initial assessment of the prob-
often happens in clinical practice, using a combi- ability of the disease being present or absent (post-
nation of diagnostic tests can significantly test probability). Strong, conclusive tests yield
increase the overall sensitivity and specificity, very big or very small LRs. Weak, inconclusive
and thus the accuracy of the diagnosis. tests yield modest LRs, close to 1.0. For example,
Often in clinical medicine, clinicians are faced following the results of a positive test result, if the
with a positive test result, and the next question positive LR is found to equal 1, then there is no
asked is, “among patients with a positive test change in the likelihood of the disease. If the LR
result, what proportion will actually have the dis- was found to be 10 after the positive test result,
ease?” This is known as the positive predictive then the posttest odds of the disease is equal to the
value of the test. The negative predictive value likelihood ratio multiplied by the pretest odds.
of a test relays the probability that if the test is The odds of the disease can be calculated from
negative, the patient does not have the disease. It the probability as probability/1-probability. The
is important to remember that the predictive value higher the LR, the better the test is for ruling in a
of a test is affected by both the prevalence of the diagnosis. These ratios depend upon the validity
disease in the population being considered and, if of the test being ordered in distinguishing who has
the disease in uncommon, the specificity of the the disease in question from who does not; hence,
test being used. Higher disease prevalence gener- these ratios may be derived from sensitivity and
ally leads to an increase in the positive predictive specificity of the test (see below). LRs may be
value of a test. However, as most diseases are rare, more useful than sensitivity and specificity in
tests with higher specificity likely have a greater certain situations. LRs can be calculated for tests
impact on increasing the positive predictive value without dichotomous results such as those that are
for a given test. These values can be easily calcu- resulted as “positive, intermediate, or negative.”
lated as demonstrated in Fig. 7. The results of several diagnostic tests may be

Fig. 7 Positive and


negative predictive values
494 S.L. Furth and J.J. Fadrowski

combined to provide a single LR. Finally, with


some relatively simple calculations, the posttest
probability of a disease can be calculated using the
LR and the pretest odds of the disease. Formulas
for calculating LRs follow.

The Positive LR is the probability of an individual


with the disease having a positive test/proba-
bility of an individual without the disease hav-
ing a positive test.
The Negative LR is the Probability of an individ-
ual with the disease having a negative test/the
probability of an individual without the disease
having a negative test

For dichotomous tests:


Fig. 8 ROC curve for the detection of non-MCNS in
Positive LR: sensitivity/(1  specificity) relation to remission time (14). The graph plots the true-
Negative LR: (1  sensitivity)/specificity positive rate expressed as sensitivity (%) as a function of
the falsepositive rate (100-specificity [%]) at different cut-
off points. Area under the ROC CURVE = 0.859; SE =
Receiver operating characteristic (ROC) 0.057; 95% confidence interval, 0.793 to 0.911. Data in the
curves, originally developed in the field of elec- table present selected cutoff points derived from the graph
tronics, allow for a graphical display of the trade- as mean and 95% CI
off between sensitivity and specificity for diag-
nostic tests with ordinal or continuous results, in Study of Kidney Disease in Children (ISKDC),
which several values of sensitivity and specificity the approach to a child with new-onset nephrotic
are possible. Several cutoff points are determined, syndrome has been to perform a kidney biopsy if
and the sensitivity and specificity are determined the disease was unresponsive to a standard dose of
at each point. The sensitivity (or true-positive corticosteroid therapy of at least 28 days in dura-
rate) is graphed on the Y-axis as a function of tion [12]. Given the increased incidence of FSGS
1-specificity (the false-positive rate) on the observed in their study, Filler et al. considered that
X-axis. Tests with values falling in the upper left kidney biopsies to distinguish FSGS from mini-
corner of the graph are considered ideal (100 % mal change disease may need to be performed
true positives and no false positives). If a test sooner after presentation with the nephrotic syn-
followed the diagonal line from the lower left drome, and based on their data, investigated the
corner to the upper right corner, it would be con- ideal time to perform a kidney biopsy for the
sidered useless – on this diagonal line the true- detection of non-minimal change nephrotic syn-
positive rate equals the false-positive rate. The drome (i.e., FSGS). The clinical feature
area under the ROC curve can range from 0.5 for (as opposed to a “diagnostic” test) represented in
a worthless test to 1.0 for a perfect test. the ROC curve is “time to remission after starting
Filler et al. made use of both an ROC curve and corticosteroid therapy.” By plotting and compar-
LRs in a study examining non-minimal change ing the various sensitivities and specificities of
nephrotic syndrome in children referred to a ter- values for “time to remission,” the authors con-
tiary care medical center in Canada, as can be seen cluded that the cutoff of 28 days was statistically
in Fig. 8 included from this study [11]. The the best point when a biopsy should be consid-
authors noted an increase in the incidence in ered. At this cutoff point, the detection of “true
focal segmental glomerulosclerosis (FSGS) over positives” (non-minimal change nephrotic syn-
a 17-year study period. Based on the International drome) was maximized, and the detection of
16 Basics of Clinical Investigation 495

“false positives” (minimal change nephrotic syn- Do those with earlier diagnosis of the disease
drome) was minimized. The LRs for “time to comply with treatment recommendations and reg-
remission” are also listed. The longer a patient imens – how many of those who screen positive
took to enter remission, the higher the positive receive a final diagnosis and treatment? Is there an
LR for the diagnosis of a non-minimal change improvement in the quality of life in those
pathology underlying the nephrotic syndrome. screened?

Screening Programs Ethics in Research

Screening programs are designed to detect or diag- The foundation of medical research is to help
nose a disease as early as possible, in hopes of improve the lives of patients. In keeping with
improving the prognosis. A common screening this, it is important to be familiar with the various
program is the use of mammography for the earlier terms and concepts related to the responsible and
detection of breast cancer. Detecting breast cancer ethical conduct of medical research.
early, before it is “symptomatic” or advanced, has Conflicts of interest may occur between a
been proven to improve outcomes and survival in researcher’s interest in advancing medical knowl-
older women. A sampling of the many factors that edge and his/her self-interest in fame, prestige,
must be considered when evaluating the feasibility academic, or financial advancement. Transpar-
and effectiveness of a screening program follows. ency of contractual obligations and relationships
is mandatory, and in some circumstances, these
must be dissolved, or the researcher should not
Disease participate in a related project. For example, a
conflict of interest would exist if a physician was
Is there a preclinical phase of the disease – a time paid by a pharmaceutical company as a medical
when the disease is present but clinical symptoms consultant, and this physician also served as a
have not yet manifested? Does intervening earlier primary investigator in a clinical trial of a medi-
in the natural history of the disease make treat- cation produced by the same drug company. Con-
ment easier and/or improve morbidity or mortal- flicts of interest may also occur in the
ity? Is the disease prevalence high enough to make clinician–investigator role when a researcher is
a screening program cost-effective? also the clinician for the research subject. In
such situations, what may be best for the research
project may not be best for the individual patient.
Test In such conflicts, the physician is expected to do
what is best for the patient.
Does a screening test exist with acceptable sensi- Several types of scientific misconduct have
tivity and specificity for a screening program – are been described. Scientists have a responsibility
the false-positive and false-negative rates accept- to report misconduct, and institutions have the
able? Is the test acceptable to the population – will responsibility to investigate the misconduct and
they consent to the test? Do the benefits gained protect the person alleging the misconduct. Fab-
from early diagnosis of the disease outweigh the rication/forgery is the invention and reporting of
cost of the test? data that does not exist from an experiment that
was not performed. Falsification/fraud is the
manipulation of research data such that what is
Person/Population reported misrepresents the actual findings. Pla-
giarism is the presentation of another person’s
Does screening/early diagnosis improve out- words or ideas as one’s own or without giving
comes for an individual, and the population? appropriate credit to the original author(s).
496 S.L. Furth and J.J. Fadrowski

Research on human subjects requires special critically review original articles on etiology,
ethical considerations and protections. Respect diagnosis, prognosis, and therapy [13]. In the
for research participants requires investigators to following decade, the series was widely read and
obtain informed consent or assent, maintain pri- cited, was modified for use by the general public,
vacy and confidentiality, and protect the vulnera- and was published in clinical epidemiology texts
ble. Informed consent involves relaying an [14]. At the same time, clinicians at McMaster
unambiguous description of the research project University and across North America continued
and allowing the subject to make an informed to expand and improve the guidelines. Their focus
decision regarding participation. It must be clearly has expanded to include clinicians’ ability to
stated that the patient will be involved in a access, summarize, and apply information from
research project and participation is entirely vol- the literature to everyday clinical problems,
untary. A clear description of the potential risks transforming the Readers’ Guides to Users’
and benefits, and any compensation, should be Guides [15–38].
provided. For pediatric patients participating in Such systematic approaches have also been
research, assent is also required. Children cannot adapted to assess entire bodies of research on
legally give permission to participate in a research particular subjects. In 1999, the US Congress
study, nor can they give “consent” as consent directed the Agency for Health Care Research
implies full understanding. However, ethicists and Quality to identify methods to assess health-
and medical and legal professionals have agreed care research results. The results of that effort
that children should be routinely asked if they were published in a report entitled “Systems to
agree (assent) to participate in a research project, rate the strength of scientific evidence.” The goals
and their wishes should be respected. Other vul- of this project were to describe systems to rate the
nerable populations that require special attention strength of scientific evidence, including evaluat-
to ensure their safety include prisoners, pregnant ing the quality of individual articles that make up a
women and their fetuses and embryos, and people body of medical evidence related to a particular
with impaired capacity to make decisions, such as disease, allowing for the most informed medical
the mentally ill. During the consent process, con- assessments and decision making. The report pro-
fidentiality and privacy procedures utilized by the vides the framework for the clinician regarding
study should be outlined. The extent of confiden- the evaluation of various types of study design as
tiality should be disclosed – the subject should described in this chapter [39].
understand who will and who will not have access In summary, the busy clinician can afford to be
to the data. selective in reviewing the literature. In rating the
strength of scientific evidence in evaluating a spe-
cific clinical problem or treatment, one needs to
Evaluating the Literature: Rating the be selective. The simplest criteria for choosing
Strength of Scientific Evidence which studies to read in detail or which to weigh
heavily in evidence are clinical relevance and
Health-care decisions should be based on methodologic soundness. This chapter has intro-
research-based evidence. Whether the individual duced a simple framework for evaluating such
nephrologist is making a clinical decision or a features in the context of sound clinical research
national organization is developing clinical prac- methodology and has outlined the most recent
tice guidelines, efforts should be made to system- guidelines for assessing the strength of scientific
atically assess the strengths of scientific evidence evidence for making decisions in clinical care.
related to a particular clinical diagnostic or treat- These tools for systematic assessment of existing
ment plan. Guidelines first developed more than research can also guide the clinical investigator
20 years ago at the Department of Clinical Epide- toward areas that require further study in which
miology and Biostatistics at McMaster University current evidence for treatment or outcomes is
first introduced tools to allow clinicians to scant.
16 Basics of Clinical Investigation 497

References 16. Oxman AD, Sackett DL, Guyatt GH. Users’ guide to
the medical literature: I. How to get started. JAMA.
1. Cook TD, Campbell DT. Quasi’-experimentation: 1993;270:2093–5.
design and analysis issues for field settings. Boston: 17. Guyatt GH, Sackett DL, Cook DJ. Users’ guides to the
Houghton Mifflin; 1979. medical literature: II. How to use an article about
2. Porras D, Stein DR, et al. Midaortic syndrome: therapy or prevention A. Are the results of the study
30 years of experience with medical, endovascular valid? JAMA. 1993;270:2598–601.
and surgical management. Pediatr Nephrol. 2013;28: 18. Guyatt GH, Sackett DL, Cook DJ. Users’ guides to the
2023–33. medical literature: II. How to use an article about therapy
3. Fadrowski JJ, Navas-Acien A, et al. Blood lead level or prevention B. What were the results and will they help
and kidney function in US adolescents. Arch Intern me in caring for my patients? JAMA. 1994;271:59–63.
Med. 2010;170:75–82. 19. Jaeschke R, Guyatt G, Sackett DL. Users’ guide to the
4. Bettinelli A, Vigano C, et al. Phosphate homeostasis in medical literature: III. How to use an article about a
Bartter syndrome: a case-control study. Pediatr diagnostic test A. Are the results of the study valid?
Nephrol. 2014. doi:10.1007/s00467-014-2846-z JAMA. 1994;271:389–91.
(published online). 20. Jaeschke R, Guyatt G, Sackett DL. Users’ guide to the
5. Kupferman JC, Friedman LA, et al. BP control and left medical literature: III. How to use an article about a
ventricular hypertrophy regression in children with diagnostic test B. What are the results and will they help
CKD. J Am Soc Nephrol. 2014;25:167–74. me in caring for my patients? JAMA. 1994;271:703–7.
6. ESCAPE Trial Group, Wuhl E, et al. Strict blood- 21. Levine M, Walter S, Lee H, et al. Users’ guide to the
pressure control and progression of renal failure in medical literature: IV. How to use an article about
children. N Engl J Med. 2009;361:1639–50. harm. JAMA. 1994;271:1615–9.
7. Wang R, Lagakos SW, Ware JH, et al. Statistics in 22. Laupacis A, Wells G, Richardson WS, et al. Users’
medicine – reporting of subgroup analyses in clinical guide to the medical literature: V. How to use an article
trials. N Engl J Med. 2007;357:2189–94. about prognosis. JAMA. 1994;272:234–7.
8. Furth SL, Hwang W, et al. Relation between pediatric 23. Oxman AD, Cook DJ, Guyatt GH. Users’ guide to the
experience and treatment recommendations for chil- medical literature: VI. How to use an overview. JAMA.
dren and adolescents with kidney failure. JAMA. 1994;272:1367–71.
2001;258:1027–33. 24. Richardson WS, Detsky AS. Users’ guide to the medical
9. U.S. Renal Data System. USRDS 2013 annual data literature: VII. How to use a clinical decision analysis.
report: atlas of chronic kidney disease and end-stage A. Are the results valid? JAMA. 1995;273:1292–5.
renal disease in the United States. Bethesda: National 25. Richardson WS, Detsky AS. Users’ guide to the medical
Institutes of Health, National Institute of Diabetes and literature: VII. How to use a clinical decision analysis.
Digestive and Kidney Diseases; 2013. B. What are the results and how will they help me in
10. American Academy of Pediatrics. Subcommittee on caring for my patients? JAMA. 1995;273:1610–3.
Urinary Tract Infection and Steering Committee on 26. Hayward RSA, Wilson MC, Tunis SR, et al. Users’
Quality Improvement and Management. Urinary tract guide to the medical literature: VIII. How to use clin-
infection: clinical practice guidelines for the diagnosis ical practice guidelines. B. What are the recommenda-
and management of the initial UTI in febrile infants and tions and how will they help you in caring for your
children 2 to 24 months. Pediatrics. 2011;128: patients? JAMA. 1995;274:1630–2.
595–610. 27. Hayward RSA, Wilson MC, Tunis SR, et al. Users’
11. Filler G, Young E, et al. Is there really an increase in guide to the medical literature: VIII. How to use clin-
non-minimal change nephrotic syndrome in children? ical practice guidelines. A. Are the guidelines valid?
Am J Kidney Dis. 2003;42:1107–13. JAMA. 1995;274:570–4.
12. International Study of Kidney Disease in Children. The 28. Guyatt GH, Sackett DL, Sinclair JC, et al. Users’ guide to
primary nephrotic syndrome in children Identification the medical literature: IX. A method for grading health
of patients with minimal change nephrotic syndrome care recommendations. JAMA. 1995;274:1800–4.
from initial response to prednisone. A report of the 29. Naylor CD, Guyatt GH. Users’ guide to the medical
International Study of Kidney Disease in Children. literature: X. How to use an article reporting variations in
J Pediatr. 1981;98:561–4. the outcomes of health services. JAMA. 1996;275:554–8.
13. Department of Clinical Epidemiology and Biostatis- 30. Dans AL, Dans LF, Guyatt GH, et al. Users’ guides to the
tics, McMaster University. How to read clinical medical literature: XIV. How to decide on the applicabil-
journals. I. Why to read them and how to start reading ity of clinical trial results to your patient. Evidence-Based
them critically. Can Med Assoc J. 1981;124:555–8. Medicine Working Group. JAMA. 1998;279(7):545–9.
14. Sackett DL, Haynes RB, et al. Clinical epidemiology: a 31. Richardson WS, WiJson MC, Guyatt GH, et al. Users’
basic science for clinical medicine. 2nd ed. Philadel- guides to the medical literature: XV. How to use an
phia: Lippincott Williams & Wilkins; 1991. article about disease probability for differential diag-
15. Guyatt GH, Fvennie D. Users’ guides to the medical nosis. Evidence-Based Medicine Working Group.
literature (editorial). JAMA. 1993;270:2096–7. JAMA. 1999;281(13):1214–9.
498 S.L. Furth and J.J. Fadrowski

32. Berlin JA, Rennie D. Measuring the quality of trials: the Dawson-Saunders B, Trapp RG. Basic and clinical biosta-
quality of quality scales. JAMA. 1999;282(11):1083–5. tistics. 3rd ed. New York: Lange Medical Books-
33. McGinn TG, Guyatt GH, Wyer PC, et al. Users’ guides McGraw-Hill; 2001.
to die medical literature: XXII. How to use articles DeAngelis C. Introduction to clinical research. New York:
about clinical decision rides. Evidence-Based Medi- Oxford University Press; 1990.
cine Working Group. JAMA. 2000;284(1):79–84. Fleiss JL. Statistical methods for rates and proportions.
34. Giacomini MK, Cook DJ. Users’ guides to the medical New York: Wiley; 1981.
literature: XXIII. Qualitative research in health care Fletcher RH, Fletcher SW, Wagner EH. Clinical epidemi-
B. What are the results and how do they help me care ology: the essentials. 3rd ed. Baltimore: Lippincott
for my patients? Evidence-Based Medicine Working Williams & Wilkins; 1996.
Group. JAMA. 2000;284(4):478–82. Glaser AN. High yield biostatistics. Pennsylvania:
35. Richardson WS, Wilson MC, Williams Jr JW, Lippincott Williams & Wilkins; 2001.
et al. Users’ guides to the medical literature: XXIV. Gordis L. Epidemiology. 2nd ed. Philadelphia: Saunders;
How to use an article on the clinical manifestations of 2000.
disease. Evidence-Based Medicine Working Group. Hollander M, Wolfe DA. Nonparametric statistical
JAMA. 2000;284(7):869–75. methods. 2nd ed. New York: Wiley; 1999.
36. Shah NR. What is the best evidence for making clinical Hulley SB, Cummings SR. Designing clinical research.
decisions? JAMA. 2000;284(24):3127–8. 2nd ed. Philadelphia: Lippincott Williams & Wilkins;
37. Airman DG. Poor-quality medical research: what can 2001.
journals do? JAMA. 2002;287(21):2765–7. Kleinbaum DG, Kupper LL. Applied regression analysis
38. Guyatt GH. Users’ guides to the medical literature: and other multivariate methods. 3rd ed. Pacific Grove:
essentials of evidence-hosed clinical practice. Chicago: Duxbury Press; 1998.
AMA Press; 2002. Lilienfeld AM, Lilienfeld DE. Foundations of epidemiol-
39. Agency for Healthcare Research and Quality. Systems ogy. 2nd ed. New York: Oxford University Press; 1980.
to rate the strength of scientific evidence. AHRQ Pub- Meinert CL. Clinical trials: design, conduct and analysis.
lication No. 02-E016. Rockville: Agency for New York: Oxford University Press; 1986.
Healthcare Research and Quality; 2002. Sacketr DL, Haynes RB, Guyatr GH, et al. Clinical epide-
miology: a basic science for clinical medicine. 2nd
ed. Boston: Little Brown; 1991.
Schiesselman JJ. Case control studies: design,
conduct, analysis. New York: Oxford University
Suggested Reading Press; 1982.
Tabachnick BG, Fidel LS. Using multivariate statistics. 4th
Cook TD, Campbell DT. Quad-experimentation: design ed. Boston: Allyn and Bacon; 2001.
and analysis issues for field settings. Boston: Zar JH. Biostatistical analysis. 4th ed. Upper Saddle River:
Houghron-Mifflin; 1979. Prentice-Hall; 1999.
Genomic Methods in the Diagnosis
and Treatment of Pediatric Kidney 17
Disease

Karen Maresso and Ulrich Broeckel

Contents The completion of the Human Genome Project


The Genomics Revolution . . . . . . . . . . . . . . . . . . . . . . . . . . 500 (HGP) in 2003 has laid the foundation and driven
the technological advancements necessary for
The Application of Genomic Methods
to Pediatric Kidney Diseases . . . . . . . . . . . . . . . . . . . . . . . 504
the study of the genetics of complex, multifacto-
Acute Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 504 rial diseases, such as those affecting the kidney.
Hemolytic Uremic Syndrome (HUS) . . . . . . . . . . . . . . . . 506 The International HapMap Project has built upon
Chronic Kidney Diseases and End-Stage Renal the HGP through the systematic identification
Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 508
The Impact of Novel Sequencing Technologies
and cataloging of genetic variation across
on Gene Identification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 512 human populations. Translating the mass of
Next-Generation Sequencing . . . . . . . . . . . . . . . . . . . . . . . . 512 data generated by these studies into useful clini-
Data-Analytic Techniques in NGS . . . . . . . . . . . . . . . . . . 513 cal knowledge is now a major undertaking in
Data Analysis in NGS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 514
NGS in Pediatric Kidney Disease . . . . . . . . . . . . . . . . . . . 514
nearly all areas of medicine, including the field
The Past, Present, and Future Application of pediatric nephrology. Much of this work will
of Genomic Methods in Pediatric Nephrology . . . . . . 516 revolve around linking particular patient pheno-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 517 types to genomic and proteomic data, such as
genotype, expression profile, and protein bio-
markers. As evidenced by the etiological
advances made in various kidney disorders
resulting from the application of genome-wide
linkage analyses in the 1990s, there are a number
of unique aspects of pediatric nephrology that
make it an area particularly suitable to genomic
exploration with such novel technologies such as
genome-wide association and next-generation
sequencing. These aspects relate both to the clin-
ical characteristics and to public health and epi-
K. Maresso demiological concerns of pediatric kidney
Section of Genomic Pediatrics, Medical College of diseases.
Wisconsin, Milwaukee, WI, USA
A number of pediatric kidney diseases are
U. Broeckel (*) considered to have a multifactorial pathogenesis,
Department of Pediatrics, Medical College of Wisconsin
often reflected in their variable clinical presenta-
and Children’s Hospital of Wisconsin, Milwaukee,
WI, USA tions. Such diseases are often difficult to diag-
e-mail: ubroeckel@mcw.edu nosis; and invasive biopsies are often required.
# Springer-Verlag Berlin Heidelberg 2016 499
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_77
500 K. Maresso and U. Broeckel

Genotype sequence and/or expression informa- The Genomics Revolution


tion obtained from DNA analysis and profiling
studies may refine the diagnosis of a number of In 2003, the completion of the Human Genome
kidney diseases and even allow for individual- Project brought the scientific community one step
ized disease management and therapy. Addition- closer to identifying the genes underlying com-
ally, there remains an urgent need for clinically mon, polygenic diseases. Prior to this achieve-
useful biomarkers, which can assist in the ment, the goal of identifying the genetic factors
prevention and early diagnosis of both acute responsible for diseases presenting substantial
and chronic kidney diseases in children. Novel public health burdens was elusive. Research
serum and/or urine biomarkers identified efforts focused on monogenic disorders following
through the application of high-throughput pro- Mendelian patterns of inheritance. Linkage anal-
teomics offer another possibility for enhanced ysis was the main tool for the identification of
diagnosis and treatment. Moreover, from a pub- genes behind these monogenic diseases. Although
lic health standpoint, proper management of linkage analysis proved to be an effective method
renal disease is expensive, particularly renal for the mapping of numerous genes in monogenic
replacement and dialysis. The treatment of kid- disorders during the 1980s and 1990s, including
ney patients based on genomic and proteomic for familiar forms of various kidney diseases
data may help lessen this burden by enhancing [1–5], it’s subsequent application to complex dis-
disease risk prediction and creating disease sub- eases did not meet with the same success. As
sets with associated prognostic and therapeutic linkage analysis is more suitable for the identifi-
implications. While these issues are relevant to cation of genes with relatively large effect sizes,
all of nephrology in general, as well as to many its application to polygenic diseases, where the
other areas of medicine, they are particularly contribution of each locus or allele is believed to
germane to the study of childhood kidney dis- be relatively small, resulted in less than significant
eases where there can be high mortality, the need findings and results that could not be replicated.
for expensive lifetime treatment, as well as seri- Only a handful of genes contributing to common
ous growth and nutrition implications. The diseases have been identified to date through link-
proper application of genomics in pediatric age and subsequent positional cloning methods
nephrology is well suited to address the unique [6–9].
challenges posed by this field. In 1996, Risch and Merikangas demonstrated
This chapter aims to provide an overview of the power of genome-wide association analysis
the application of genomic technologies to the (GWA) over linkage methods for the identification
study of pediatric kidney diseases. We will begin of the genes behind complex diseases [10]. Associ-
with a discussion of the polygenic nature of com- ation analysis relied upon the availability of a stan-
plex diseases and the recent genomic advances dardized dense set of markers covering the genome
enabling their study. This will include outcomes that could be easily and inexpensively typed, as
of the HGP and International HapMap Project. We well as sample sizes of thousands of cases and
will then discuss the application of various geno- controls. Until the technology existed to identify,
mic methods to the more common pediatric kid- accurately map, and then genotype such large num-
ney diseases, covering the range of both chronic bers of markers in such great numbers of individ-
and acute, with a focus on the DNA level. With uals, linkage analysis followed by fine mapping
the recent developments in novel sequencing remained the best option at that time for uncovering
technologies, the field is poised to capitalize on complex disease genes. However, the work of the
these recent advances as demonstrated by most HGP began to drive the technological break-
recent publications. We will end with a discussion throughs needed to map polygenic diseases.
of the future prospects and recommendations for Analysis of the human genome sequence has
applying genomics to pediatric nephrology. demonstrated the abundance and uniformity of
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 501

single nucleotide polymorphisms (SNPs), single Possible Allelic Combinations of Two SNPs in the Absence of LD
base changes in DNA that occur in at least 1 % of SNP1(T/C) SNP2(G/A)

the population. (This is in contrast to a mutation, AC T GGTACGTACCC A ATGTTGCATACGTT


which occurs in <1 % of the population.) It is AC T GGTACGTACCC G ATGTTGCATACGTT
some of these common DNA variants that are AC C GGTACGTACCC A ATGTTGCATACGTT
believed to be responsible for at least some of
AC C GGTACGTACCC G ATGTTGCATACGTT
the phenotypic variation observed within and
between populations, including various diseases. Possible Allelic Combinations of Two SNPs in Presence of LD
The HGP contributed to developing technologies SNP1(T/C) SNP2(G/A)
for the rapid, large-scale identification and scoring AC T GGTACGTACCC A ATGTTGCATACGTT
AC C GGTACGTACCC G ATGTTGCATACGTT
of SNPs and creating the intellectual resources
needed to study sequence variation. The Interna- Fig. 1 The principle of linkage disequilibrium. A set of
tional HapMap Project built upon these discovery DNA sequences illustrating two SNPs (indicated in red and
efforts by characterizing the patterns of linkage blue). In the top half, the red and blue SNP are not in LD
with each other and segregate independently. With two
disequilibrium (LD) and haplotype structure SNPs in linkage equilibrium, four possible allelic combi-
across the human genome. LD occurs when two nations are possible. In the bottom half, two SNPs in the
or more markers segregate together with signifi- presence of LD are illustrated. Notice that LD reduces the
cantly different frequencies than would be number of possible allelic combinations, as the SNPs no
longer segregate independently. Here, the “T” allele of
expected if they segregated independently from SNP1 segregates only with the “A” allele of SNP2, while
each other. LD is generally greater for SNPs in the “C” allele of SNP1 segregates only with the “G” allele
close physical proximity. As LD tends to reduce of SNP2. In such a case, only one of these two SNPs would
the number of possible haplotypes, or set of need to be genotyped, as the genotype at one SNP will
provide the genotype information for the other
alleles, present in a population, it is useful for
association mapping, in that knowing the geno-
type of one marker can predict the genotype of difficult to achieve for various reasons, including
another marker in LD with it (Fig. 1). The work of the small, underpowered sample sizes character-
the HapMap Project, along with others, has istic of many of these types of studies [11].
resulted in the identification, mapping, character- In the second type of association study, the
ization, and the public availability of over four genome-wide strategy is hypothesis-free and
million validated SNPs to date (www.hapmap. does not make any assumptions regarding the
org). The identification, mapping, and cataloging mechanisms behind the phenotype being studied.
of these SNPs in various human populations have Genome-wide association studies are ideally
helped make association studies a reality. In con- suited for common complex diseases, where the
junction with the advent of mass-throughput physiological mechanisms and genetic factors that
genotyping platforms, such data has potential to contribute to them are often not well understood.
contribute extensively to our basic understanding GWA studies are based on hundreds of thousands
of human biology and disease. to millions of SNPs spread across the genome,
The two types of association studies that are each one tested for association with a particular
commonly used are the candidate-gene approach outcome. There are currently two main
and the genome-wide approach (Table 1, Fig. 2). approaches to this type of study. One type
Candidate-gene association studies rely upon a employs an LD-based selection strategy, as exem-
specific hypothesis and are often limited by our plified by the Illumina HumanHap550 and
current knowledge of biological and/or disease Human1M sets of variants. The LD-based strat-
mechanisms. The strategy has been a frequently egy exploits patterns of genomic LD to select a
employed method in the study of numerous traits minimum number of SNPs (tagSNPs) to be
and diseases, including many related to nephrol- assayed, which captures the maximum amount
ogy. Nevertheless, replicable results have been of information across the genome. SNPs on
502 K. Maresso and U. Broeckel

Table 1 Comparison of genome-wide (GWA) and candidate-gene association studies


Advantages Disadvantages
GWA Hypothesis-free Genotyping more technically
May be more economical and efficient when large numbers demanding
of loci need to be worked up May be costly and time-consuming
Ideal in cases where little is known regarding disease Requires sophisticated bioinformatic
mechanisms approaches to data analysis and storage
Often allow for collection of data relating to copy number
polymorphisms (CNPs) in addition to SNPs – two genetic
variants for the price of one
Candidate Less costly and time-consuming if large numbers of loci do Candidate-gene selection constrained
gene not need to be analyzed by known disease biology
Ideal in situations where evidence implicates particular Less economical and efficient when
disease mechanisms and related genes or when following-up testing great numbers of loci
a QTL containing likely functional candidates
Straightforward data-handling and analytical approaches
Various methods of genotyping available; less technically
demanding

Select Study Design: Extract DNA from


Collect Clinical Data:
1) GWA v. Candidate-gene all study participants
1) Determine phenotypes of interest
2) Case-control v.Families
2) Standardized phenotyping
3) Collect data on confounders
Data Analysis:
1) Perform Association Analysis Determine method of Genotyping:
2) Control for Multiple Testing 1) GWA:
3) Determine best p-values a) TagSNPs (Illumina)
4) Replication / Finemapping b) Random selection of SNPs
5) Functional Analysis (Affymetrix)

6) Combine with Transcriptomic/Proteomic Data 2) Candidate-gene: numerous methods

Fig. 2 Workflow of a genetic association study

these arrays serve as proxies for those not assayed. populations, including many outcomes relevant
The second approach ignores patterns of LD and to kidney disease, such as obesity and type II
relies on the random selection of markers. This diabetes [12–14], as well as quantitative traits
approach is the basis for the Affymetrix 500 K and like urinary albumin excretion [15].
1 million SNP array products. A number of GWA Within the field of pediatric nephrology, many
reports have recently been published identifying of the known disease genes were initially identi-
novel genes for various diseases in adult fied through the study of familiar forms of a
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 503

disease using linkage analysis followed by posi- two important points. First, it is unlikely that the
tional cloning methods [16–19]. These genes have often thousands of subjects needed for adequately
become obvious candidates to examine in the powered studies can be collected at a single site;
context of seemingly sporadic occurrence, where and consequently, there is a need for multisite
a patient has no prior family history of the disease collaborative studies. This reflects the fact that
and which constitutes the majority of many large-scale gene association studies of novel
childhood-onset kidney diseases. This is often genes in adult or pediatric populations are criti-
done through mutation scanning in case series. cally required. Second, by adopting strict publish-
While linkage analysis proved to be a useful tool ing guidelines, journals can help assure the proper
for identifying the first disease genes in pediatric design and analysis of association studies. To
kidney disease, allowing for some of the first address the first point, European investigators
insights into the molecular pathogenesis of many interested in nephrology and genetics established
of these diseases, these genes generally only the Renal Genome Network (ReGeNet; www.
account for a minority of cases. New methods regenet.eu) in 2003 to encourage large, collabora-
which result in the identification of novel genes tive clinical studies focused on the genetic epide-
can assist in making further advances. While miology of renal disease [24].
GWA studies are still relatively new, candidate- When many genes are to be tested, or when one
gene studies have been an actively pursued desires a hypothesis-free approach, it may be
approach in nephrology to novel gene identifica- more efficient and economical to perform a
tion. The popularity of this approach is illustrated GWA study. While these studies may assist in
by a review of the 2004 and 2005 abstracts of the the identification of novel genes in acute kidney
American Society of Nephrology (ASN) and the disorders, where currently half or less of all cases
European Renal Association/European Dialysis are explained by known mutations, GWA
and Transplantation Association (ERA/EDTA), approaches in pediatric nephrology are likely to
where over 180 studies encompassing 205 poly- be most beneficial when applied to chronic kidney
morphisms in 92 different genes were submitted disease (CKD) and end-stage renal disease
[20]. The genes most frequently studied were (ESRD) resulting from the increasing rate of
those of the renin–angiotensin system (RAS) and hypertension, diabetes, and obesity in children.
those related to inflammation [20]. The best evi- The mechanisms leading to CKD and ESRD in
dence to date indicates minor statistical associa- these settings are likely to be polygenic in nature
tions and a negligible clinical relevance of the and the current lack of understanding regarding all
most frequently studied RAS polymorphism, the disease mechanisms leading to these outcomes
insertion/deletion (I/D) variant of the angiotensin precludes a selection of appropriate candidate
I-converting enzyme (ACE) gene [21–23]. genes. In addition, the mechanisms of these dis-
Despite their frequent study, a genetic association orders and their precipitating causes may be dif-
has yet to be convincingly demonstrated for any ferent from those occurring in adults. While some
of the RAS genes. (These genes will not be further of the GWA findings from adult populations may
discussed in the chapter.) Of particular note, the be relevant to pediatric populations as well, more
authors highlight the extremely small sample sizes work will be required to determine this. A more
of the vast majority of these studies. In 2004, the in-depth discussion on the application of
average study size was 185 (cases and controls candidate-gene association and GWA approaches
combined), with a slight increase in 2005 to 276. to the more common pediatric kidney diseases
In addition, the authors found that while only will be presented in the following section.
16 % of these studies met the newly adopted While much of this discussion has focused
criteria for publishing candidate-gene association exclusively on the role of DNA sequence varia-
studies in the Journal of the American Society of tion in disease outcomes, there are also extensive
Nephrology (JASN) in 2004, this number jumped research efforts dedicated to the role of mRNA
to nearly half in 2005 [20]. These data highlight and proteins in the prevention, diagnosis, and
504 K. Maresso and U. Broeckel

treatment of kidney disease. Expression profiling, example of where the application of genetic and
as with high-throughput SNP genotyping, has genomic methods can assist in a greater understand-
benefited substantially from recent advances in ing of disease pathogenesis and treatment. See also
microarray technology, which are allowing for ▶ Chap. 27, “Idiopathic Nephrotic Syndrome in
the measurement of genome-wide expression Children: Genetic Aspects” in this text.
levels, also called “transcriptomics,” and their In children, minimal change nephropathy
correlation with various disease states. As with (MCN) is the overwhelming cause of NS, such
cancer, the identification of certain common that renal biopsies are often not performed before
expression “signatures” through the simultaneous beginning treatment with potentially harmful
profiling of thousands of genes from kidney immunosuppressive therapies [27]. Corticoste-
biopsy samples may help determine treatment roids are often prescribed first. The majority of
course and patient prognosis in various renal dis- children respond well to this treatment and they
eases. The genome-wide study of proteins, or are said to have “steroid-sensitive nephrotic syn-
proteomics, has also benefited from recent tech- drome” (SSNS). However, there is a subset of
nological breakthroughs in mass spectrometry, children who do not respond to this therapy and
allowing for a greater number of proteins to be they are said to have “steroid-resistant nephrotic
simultaneously analyzed. There is currently an syndrome” (SRNS). In such cases, a renal biopsy
urgent need in pediatric nephrology for clinically may be indicated and the underlying cause may be
relevant biomarkers which can be obtained non- focal segmental glomerularsclerosis (FSGS).
invasively and which will eventually allow for Current treatment strategies for MCN and FSGS
earlier disease detection [25, 26]. Because these are similar; however, they are generally less
two fields are closely aligned with the study of effective for FSGS; and children with the latter
DNA sequence variation, in that changes affecting may go on to develop ESRD. Despite the exis-
gene sequence can ultimately affect gene expres- tence of these therapeutic strategies, the underly-
sion and protein production, we focus in the fol- ing pathological mechanisms of these two
lowing section on the application of the study of histological classifications are poorly understood.
DNA sequence variation to the various forms of It is a subject of debate in the literature as to
pediatric renal disease. The application of all three whether or not these classifications represent dis-
fields to the study of pediatric nephrology can tinct diseases or subtypes of a single condition
ultimately lead to enhanced prevention efforts, [27]. In addition to a need for a greater under-
diagnoses, and treatment options through the standing of the disease processes involved in NS,
identification of novel genes and genetic markers, less toxic therapies are also needed for treatment
gene expression signatures, and biomarkers asso- in children. In the last decade, much progress has
ciated with various renal disease states and out- been made in the area of NS genetics and
comes in pediatric populations. more can be expected in the coming years with
novel genomic technologies. These advances
should help to clarify NS-related pathologies and
The Application of Genomic Methods offer a pharmacogenetic approach to the treatment
to Pediatric Kidney Diseases of NS.
In the late 1990s, genome-wide linkage analy-
Acute Diseases sis followed by positional cloning was a particu-
larly successful method for the identification of
Table 2 provides a summary of the genes and loci the genes behind the familial form of NS,
implicated in the following conditions, as well as although sporadic cases, those without any previ-
information on known genotype–phenotype ous family history, account for the majority of
correlations. pediatric NS cases. In 1998, Kestila
Nephrotic syndrome (NS) NS as a heteroge- et al. identified the first NS-related gene, NPHS1
neous collection of disorders represents an excellent (nephrin), to be the cause of an autosomal
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 505

Table 2 Genes and genotype–phenotype correlations in selected pediatric renal diseases


Genotype–phenotype
Disease Gene(s)/loci correlations Comments
Nephrotic NPHS1, NPHS2, LAMB2, NPHS1, NPHS2, LAMB2, Additional susceptibility genes
syndrome WT1, PLCE1 WT1 commonly mutated in and modifier genes likely.
(steroid- congenital onset and resistant GWA studies may assist with
resistant) to steroid treatment; type and their discovery
number of NPHS2 mutations
correlate with age of onset
Atypical CFH, CFI, CFB, MCP CFH-severe clinical course, Known complement genes
hemolytic (complement regulatory genes) rapid progression to ESRD, account for ~50 % of cases.
uremic earlier age at onset, disease Additional susceptibility genes
syndrome recurrence after transplant; likely. Modifier genes likely
MCP, most favorable course, due to high incomplete
no recurrence post transplant; penetrance. SNPs may also
CFI, intermediate course play a role along with known
mutations
Autosomal PKD1, PKD2 PKD1 – more severe course PKD1 accounts for ~85 % of
dominant and earlier onset; 50 mutations families. Most mutations in
polycystic may progress to ESRD earlier these genes are unique to single
kidney and be more susceptible to family. Genetic background
disease ICAs may account for variability
within a family. Medical
resequencing can assist in
increasing the numbers of
particular mutational profiles.
GWA studies may help to
uncover modifier genes
Autosomal PKHD1 Presence of two truncating ~33 % of mutations are unique
recessive mutations results in neonatal to a single family. Medical
polycystic death, regardless of position resequencing can assist in
kidney Missense mutations associated increasing the numbers of
disease with milder phenotype particular mutational profiles
IgA 6q22-23; 4q26-31; 17q12-22; Gene verification and No gene(s) yet identified in
nephropathy 2q36; megsin gene, discovery and families. Many results from
uteroglobin, C1GALT1, E- and genotype–phenotype case–control studies need
L-selectins, PIGR, IGHMBP2, correlations still needed verification. GWA studies
TNFα, TGFβ, IL-4, IFγ, and needed
HLA-DRA
End-stage 10p; 18q; see Iyengar et.al. for Gene verification and Genes identified in adult
renal disease additional loci; D10S558, discovery and populations and many in
D10S1435, D6S281, genotype–phenotype setting of DN. Verification in
D4S2937, D2S291, D17S515, correlations in pediatric cohorts adults and children required.
CNDP1, ELMO1, PVT1 still needed GWA studies needed

recessive form of congenital NS in the Finnish descent [18, 19]. This form presented between
population [16, 17]. This was a particularly severe 3 months and 5 years of age and was characterized
form of NS, characterized by heavy proteinuria in by minimal glomerular changes on early biopsy
utero, lesions of the glomerular filtration barrier, samples, FSGS present at later stages, and rapid
and often death within the first 2 years of life progression to ESRD. Both nephrin and podocin
[17]. These findings were quickly followed by localize to the slit diaphragm and are key glomer-
the identification of the NPHS2 (podocin) gene ular filtration barrier proteins.
and its mutations as the cause of autosomal reces- Since the identification of these genes, numer-
sive steroid-resistant NS in older children from ous studies have investigated their role in all
families of Northern European and North African forms of NS. It is now well established that
506 K. Maresso and U. Broeckel

10–30 % of sporadic NS cases with FSGS are due any mutation within the podocin gene, highlight-
to mutations within NPHS2 [28]. Mutations ing the likelihood of as yet undiscovered genes.
within the laminin-β-2 (LAMB2), Wilms’ tumor This is the largest report to date and the first to
1 (WT1), and phospholipase C-ε (PLCE1) genes establish statistical significance of an important
have also been reported in pediatric NS, although genotype–phenotype correlation. It serves as an
they are far less frequent [29–31]. Hinkes example of the advantages of multinational col-
et al. reported that in 89 patients from 80 families laborations and demonstrates that further such
manifesting NS within the first year of life, muta- studies are required in order to identify additional
tions in one of four genes (NPHS1, NPHS2, WT1, correlations with therapeutic and prognostic
and LAMB2) were identified in 66 % of the cases, implications in the treatment of NS.
with nearly 38 % and 23 % due to NPHS2 and Currently, genetic screening is recommended
NPHS1 mutations, respectively [32]. When for those children presenting with NS during their
examined by age of onset, the proportion of con- first year of life, as this could allow carriers of
genital cases explained by mutations in one of the mutations in the NPHS1, NPHS2, LAMB2, and
four genes was nearly twice that of infantile-onset WT1 genes to be spared the potentially harmful
cases. Also of note, the study reported that chil- treatments to which they have been shown not to
dren with mutations in any one of the four genes respond [32, 36]. Genetic testing may eventually
did not respond to steroid treatment. The authors become more widespread in children presenting
concluded that children with onset of NS during with NS, regardless of age, as further studies estab-
the first year of life should be screened for muta- lish links between currently known mutations and
tions within these four genes in order to guide clinically relevant phenotypic outcomes, as well as
therapeutic course and that there are likely addi- with the discovery of novel genes and variants. It
tional unknown genes playing a role in early- should be noted that a different set of genetic var-
onset NS. iants may predispose to the steroid-sensitive form
Previous studies have suggested both allelic and of NS [37]; however, it has not been as intensively
locus heterogeneity of NS. While nephrin muta- studied as SRNS due to its relatively benign clinical
tions have been found only in patients with con- course. While there is currently much research
genital onset, podocin mutations have been found focused on the genes and proteins of the slit dia-
in cases presenting at varying ages [32, 33]. The phragm complex, further inroads may be made in
reasons for this phenotypic variability are not yet both SRNS and SSNS with the application of
completely understood; however, there is data genome-wide association and expression studies
supporting that the specific type or number of to uncover novel susceptibility and modifying
NPHS2 mutations may play a role [34, 35]. genetic variants. Such studies will require large-
Perhaps the largest study to date on podocin scale cohorts resulting from multi-institution
mutations in sporadic cases best demonstrates the and/or multinational collaborations in order to fur-
significant locus and allelic heterogeneity of ther elucidate the pathogenetic mechanisms behind
NS. Using direct sequencing in 430 patients this rare and heterogeneous syndrome. Most
from 404 families from around the world, Hinkes recently, next-generation sequencing (NGS) was
et al. was the first to document that patients with a applied to study NS. As will be discussed below
specific type and number of mutations presented in the section on the application of NGS, this
with symptoms at a significantly earlier age than approach can offer additional novel insights into
patients without such mutations [33]. Neverthe- the genetic basis of this disease.
less, the authors did observe that identical muta-
tions did result in different ages of onset within
their study. Consequently, Hinkes et al. state that Hemolytic Uremic Syndrome (HUS)
there are likely to be additional modifying factors
affecting the phenotype. It must also be noted that Hemolytic uremic syndrome (HUS) is similar to
nearly 300 individuals in this study did not carry NS in that there are different manifestations of the
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 507

disease and it appears to be genetically heteroge- www.fh-hus.org, which catalogs the various
neous. See also ▶ Chap. 47, “Renal Involvement complement activation gene mutations associ-
in Children with HUS” in this text. HUS occurs in ated with aHUS and provides a host of informa-
its typical form following an infection associated tion regarding this disease and its genetic risk
with diarrhea, often from the E. coli serotype factors.
0157:H7. This form is relatively benign, and In order for complement gene mutation status
once resolved, complete renal function is restored. to be translated into clinically useful information,
However, approximately 5–10 % of HUS patients a few reports have examined genotype–phenotype
present with no previous infection. This atypical correlations in aHUS. Although each report is
form, aHUS, can occur at any age, can be based on a small number of children, the results
familiar or sporadic in nature, and is associated are consistent across studies. Carriers of CFH
with a poor clinical outcome, including a 25–30 % mutations have the most severe clinical course,
mortality rate [38]. While a number of environ- demonstrating earlier age of onset, rapid progres-
mental factors triggering aHUS have been sion to ESRD, and disease recurrence after
reported [39–45], a genetic basis for this form kidney transplantation [63, 66]. Those with MCP
has been suggested by familial cases [46–48]. In mutations show the most favorable prognosis,
1998, the first aHUS-associated gene, comple- with no disease recurrence post-transplantation
ment factor H (CFH), was localized [49]. Since [39, 63, 66]. CFI mutation carriers have an
then, aHUS has become largely recognized as a intermediate course, with 50 % quickly
disease of complement alternative pathway progressing to ESRD, 50 % recovering, and a
dysregulation. majority with disease recurrence post-
As with NS, some of the initial advances in transplantation [63, 66]. These data demonstrate
understanding the etiology of aHUS were the that aHUS presentation, response to treatment,
result of genetic linkage and subsequent associa- and long-term outcome are closely linked to geno-
tion studies. Using a candidate-gene linkage type status. This should allow for more tailored
approach in three families, Warwicker et al. first management of children with aHUS and under-
demonstrated linkage of aHUS to a region on chr. scores the urgent need for new and improved
1 containing a number of genes involved in com- therapies for those with CFH and IFH mutations.
plement activation regulation [49]. Subsequent An interesting feature of aHUS is its high rate
mutational screening of the CFH gene identified of incomplete penetrance, approaching 50 %,
two different mutations in one of the three fami- where disease-causing mutations are seen in
lies and in a sporadic aHUS patient. Since this those who do not present with the disease. It has
first identification, numerous studies have been speculated that additional genetic factors
reported various mutations within CFH associ- may be required in addition to the known causa-
ated with aHUS [49–54]. Most mutations are tive mutations in order for the disease to manifest
missense mutations occurring in the C-terminal itself. In addition, approximately half of aHUS
region of the protein, and functional analysis of patients do not carry a known causative mutation,
CFH mutations shows that they result in a suggesting as yet unidentified genetic variants. In
reduced ability of CFH to protect cells from com- this regard, a handful of polymorphisms, or SNPs,
plement lysis [55]. Additional mutations within have also been associated with aHUS [50, 56, 65,
the complement regulatory genes of membrane 67, 68]. Caprioli et al. documented association of
cofactor protein (MCP) [56–58] and factor I three SNPs in CFH with aHUS, concluding that
(CFI) [59, 60], as well as within complement- they may predispose to the disease in those with-
activating components factor B (CFB) [61] and out CFH mutations, as well as contributing to the
C3 genes [62], have also been associated with full manifestation of aHUS in CFH mutation car-
aHUS. Together, these genes are estimated to riers [50]. Findings from Fremeaux-Bacchi
account for approximately 50 % of aHUS cases et al. support these data [56]. A report by
[39, 63–65]. There is now an online database, Esparza-Gordillo identified a SNP haplotype
508 K. Maresso and U. Broeckel

block spanning the complement activation gene known treatments for either form. While the
cluster or chr. 1q32 which was more frequent in genetics of both ADPKD and ARPKD are well
aHUS patients, particularly those with CFH, established, additional advancements await
MCP, or F1 mutations, and which associated through medical resequencing and the identifica-
strongly with the severity of the disease tion of genetic disease modifiers.
[68]. This same group later reported on a pedigree As with the aforementioned acute conditions,
where three independent aHUS genetic risk fac- the genes for ADPKD, polycystin-1 (PKD1) and
tors were segregating, but only those members polycystin-2 (PKD2)/transient receptor potential
who inherited all three manifested aHUS channel (TRPP2), and the gene for ARPKD,
[69]. The authors hypothesize that multiple fibrocystin (PKHD1), were identified through
“hits” are required in complement activation linkage analyses and subsequent positional clon-
genes in order to induce the aHUS phenotype. ing efforts [1–5]. In ADPKD, the PKD1 gene
These data demonstrate the allelic heterogeneity accounts for approximately 85 % of all families
and polygenic nature of aHUS. [72–74] and is associated with both a more
Clearly, more work remains to be done, as severe clinical course and earlier onset
only 50 % of cases are explained by currently [74–77]. There is substantial allelic heterogene-
known mutations. It is likely that more genes and ity of both PKD1 and PKD2, with most muta-
mutations await discovery. Moreover, the role of tions unique to a family. The increased pace of
SNPs and their functional impact alongside the genomic diagnostics is evidenced by evaluation
currently known mutations need to be better elu- of the ADPKD database (http://pkdb.mayo.edu)
cidated. As with NS, large-scale genomic associ- at two points in time. The ADPKD database
ation and expression studies may be helpful in identified an increase from 314 to 929 and
this regard. 91 to 167 likely pathogenic germline mutations
within PKD1 and PKD2, respectively, between
mid-2008 and 2014.
Chronic Kidney Diseases and End-Stage As most mutations are private,
Renal Failure genotype–phenotype studies must generally clas-
sify mutations in some way, usually based on type
As with the acute disorders, Table 2 provides a (missense v. truncating) and position within the
summary of the genes, loci, and genotype–phenotype protein. However, no strong correlations have yet
correlations relevant to the below conditions. been established between either type or position
and phenotypic outcome for either gene. There is
Polycystic Kidney Disease (PKD) some evidence to suggest that patients with muta-
PKD represents a diverse collection of disorders tions in the 50 region of PKD1 progress to ESRD
of the tubules of the kidney, where healthy renal earlier and may be more susceptible to intracranial
tissue is progressively replaced by fluid-filled aneurysms [78, 79]. While these two genes
cysts. See also ▶ Chap. 36, “Childhood Polycys- account for nearly all cases, the disease often pre-
tic Kidney Disease” in this text. Extrarenal man- sents with significant variability within families.
ifestations are also seen in PKD, including hepatic While some of this variability is likely due to
cysts, abnormal heart valves, and intracranial environmental factors, additional genetic factors
aneurysms. Autosomal dominant PKD are also suspected in modifying disease outcomes.
(ADPKD) is the most common form, with symp- Studies have estimated that 18–78 % of the phe-
tom onset usually during the third and fourth notypic variance in PKD1 and PKD2 populations
decades of life, although childhood onset has may be attributable to genetic background
been documented [70, 71]. Autosomal recessive [80–82]. Many candidate-gene studies have been
PKD (ARPKD) is rarer, with symptoms often performed in an attempt to identify some of these
beginning in utero or during the neonatal period. modifying factors; however, the results to date
Aside from palliative care, there are currently no have been inconsistent [23, 83, 84]. Because of
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 509

the diversity of disease mutations and a lack of Primary IgA Nephropathy (IgAN)
phenotypic correlations, the usefulness of Primary IgAN is the most common form of glo-
genetic testing in ADPKD is limited, although it merulonephritis in children worldwide, although
may be helpful in childhood cases with no previ- its prevalence varies according to country
ous family history. This may change as [89–91]. See also ▶ Chap. 32, “Immunoglobulin
genotype–phenotype correlations and new thera- A Nephropathies in Children (Includes HSP)” in
pies are eventually established. this text. Some of this variance is attributable to
In ARPKD, allelic effects play a greater role geographic differences in the policies affecting
than in ADPKD. The ARPKD database provides renal biopsy in the young [92], while some may
an up-to-date infrastructure for the annotation of also be due to geographic differences in genetic
disease variants (http://www.humgen.rwth- and environmental factors [93, 94]. Its incidence
aachen.de/) and noted 748 distinct pathogenic has been estimated as high as 25–30 % in some
germline mutations (accessed 15 April 2015). Asian countries where children routinely undergo
Although genotype–phenotype studies are limited annual urinary screening; however, lower rates
by the substantial amount of allelic heterogeneity have been reported in Europe and the United
present within PKHD1, some conclusions can be States [95–98]. IgAN is characterized by glomer-
drawn from these reports. For example, the pres- ular mesangial deposits of IgA and a widely vary-
ence of two truncating mutations has been shown ing presentation [99]. IgAN often presents with
to result in neonatal death, regardless of their gross hematuria attendant to an upper respiratory
position [85–87]. All ARPKD patients which sur- tract infection [95, 98, 100, 101]. However, it is
vive the neonatal period have been shown to carry estimated that in as many as half of all cases, there
at least one amino-acid substitution, as opposed to is only microscopic hematuria, which can con-
all chain-terminating mutations [85]. While trun- tinue for years before the development of protein-
cating mutations in general are associated with uria [95, 98, 101, 102]. Because symptoms can be
more severe disease, missense mutations have intermittent, and because in most countries there
been shown to result in a milder phenotype are no routine urinary screening programs, a diag-
[85]. Larger cohorts of patients will be required nosis can be missed.
to establish consistent and more detailed IgAN was previously considered a benign dis-
genotype–phenotype correlations that can be ease; however, this outlook is changing based on
used to improve management of the disease and, the recognition of increased morbidity and mortal-
in particular, to assist in the development of novel ity after long-term follow-up [95, 100, 103, 104],
therapeutic approaches. As ARPKD is quite rare, including the finding that approximately 10 % and
and thus the numbers small, no candidate-gene 20 % of children progress to ESRD after 10 and
studies have been carried out to identify genetic 20 years, respectively [95, 100, 105–108]. The dis-
modifiers, although some have been mapped in ease progresses slowly over decades, and with clin-
mouse models [74, 88]. ical symptoms often not presenting until years after
In both ADPKD and ARPKD, the availability onset, it is possible that many adult IgAN cases
of fast, high-throughput medical resequencing, actually had a pediatric onset. Consequently, child-
where patients are resequenced in depth over hood IgAN may be considered an early stage of
long genomic regions in a matter of hours or adult IgAN with lifelong follow-up and evaluation
days, offers a new opportunity for mutation detec- required to detect the signs of disease progression
tion, with a consequent increase in numbers of [95]. Enhanced detection of the disease in child-
patients with specific mutational profiles. This hood may allow for its earlier treatment and more
should eventually aid in establishing clinically aggressive management, offering a reduced risk of
useful genotype–phenotype correlations. More- chronic kidney disease and ESRD at its later stages.
over, the use of GWA studies in ADPKD, where A complete understanding of the pathogenesis
the numbers are larger, may allow for the identifi- of IgAN is still lacking, although abnormally low
cation of novel genetic modifiers. glycosylation of IgAI molecules is a consistent
510 K. Maresso and U. Broeckel

finding in patients [109–111]. Treatment of the association studies have been undertaken for
disease is based upon symptoms, but ACE inhib- IgAN. Some of the genes studied to date, and for
itors are commonly prescribed due to their reno- which significant associations have been reported,
protective effects. Some new drugs are currently include the megsin gene, found in glomerular
under investigation, but more specific novel treat- mesangium and upregulated in IgAN [117, 118];
ments are needed which can be used safely over the uteroglobin gene, a multifunctional anti-
the long term in children. Obtaining a better inflammatory protein [119–121]; the C1GALT1
understanding of this disease from a genetic gene, an enzyme involved in the O-glycosylation
standpoint should allow for insights into IgAN process [122]; and the E- and L-selectin genes,
pathogenesis, as well as offering options for its involved in endothelial and leukocyte cell–cell
improved diagnosis and treatment. interactions, respectively [123]. Associations
As with the diseases previously discussed, have also been reported for a number of inflam-
IgAN is genetically heterogeneous, with familial matory candidates as well, including TNFα,
aggregation identified in a subset of cases, while TGFα, IL-4, IFγ, and HLA-DRA [124–127]. In a
the bulk of cases present with seemingly sporadic partial genome-wide screen, a Japanese group has
onset. Currently, it is estimated that 15 % of cases reported significant associations of the polymeric
are “familial”; however, it may actually be higher immunoglobulin receptor (PIGR) gene [128] and
due to the difficulty in diagnosing the disease. the immunoglobulin mu-binding protein 2 gene
Within families, IgAN appears to follow an [129] using over 80,000 gene-based SNPs in a
autosomal dominant inheritance pattern with stepwise association design. Finally, while not
incomplete penetrance. The most likely genetic associated with IgAN onset, a 32 bp deletion in
model for IgAN resembles that of NS or aHUS, the chemokine receptor 5 (CCR5) gene has been
where there are a handful of primary susceptibility associated with increased renal survival in IgAN
genes with additional modifying genetic and envi- patients [130, 131]. Results for many of the above
ronmental factors also required. Yet the identifi- genes require confirmation in larger, independent
cation of genes for IgAN lags behind that of the cohorts.
acute kidney disorders, with no gene yet identified High-throughput genotyping of dense sets of
in familial cases. However, three genome-wide SNPs within the currently known linkage peaks,
linkage studies have established a number of chro- now made possible through the HapMap Project
mosomal regions linked to the disease. Gharavi and the commercial availability of mass-
et al. identified a locus, termed IGAN1, on chro- throughput SNP genotyping platforms, may assist
mosome 6q22-23 as being linked to IgAN in 60 % in the identification of the genes responsible for
of the families from a Caucasian cohort the signals. GWA studies may also be useful and
[112]. Using 22 informative Italian families from more economical in this regard. Once susceptibil-
the IgAN Biobank [113], Bisceglia et al. reported ity genes are firmly established, the IgAN field
linkage to IGAN1 and also identified two sugges- can progress to identifying genotype–phenotype
tive loci on chromosomes 4q26-31 and 17q12-22 correlations to aid in diagnosis and treatment.
[114]. A genome-wide linkage scan identified a
locus on chromosome 2q36 in a Canadian family End-Stage Renal Disease (ESRD)
of Austrian-German descent with 14 affected sub- While many chronic kidney diseases can lead to
jects [115]. These studies support the likely ESRD, the causes of it differ between children and
genetic heterogeneity of the disease. Review of adults. See also ▶ Chap. 66, “Management of
the genes within these loci does not reveal likely Chronic Kidney Disease in Children” in this text.
candidates; however, this is not unexpected given Although hypertension and diabetes are the pri-
the lack of understanding surrounding IgAN path- mary causes of ESRD in adult populations, FSGS
ological mechanisms [116]. and congenital structural kidney abnormalities are
Not unlike the previous diseases, numerous the primary causes in children. However, the well-
smaller-scale case–control and family-based documented worldwide increase in rates of
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 511

childhood obesity, and its related complications of Although numerous association studies examin-
diabetes and hypertension, has the potential to sig- ing single genes have been published for ESRD, as
nificantly impact the rate of pediatric renal failure in other cases, the small sample sizes and limited
[132–137]. The prevalence of ESRD in pediatric coverage of the candidate genes make results diffi-
cohorts is generally less than that in adults; never- cult to interpret [154]. However, some larger-scale
theless, these children experience significant mor- and more in-depth association scans, including a
bidity and mortality [138–140]. In addition, ESRD GWA study, have been published in this area and
disproportionately affects minorities, with African- warrant discussion [15, 157–160]. In the first com-
American children having nearly double to triple prehensive, family-based screen of candidate genes
the rates of Caucasian children within the same age for ESRD, Ewens identified twenty nominally sig-
categories [140]. nificant genes, including twelve novel ones, in
Due to well-established statistics indicating a 72 type I diabetic trios [160]. McKnight
potentially catastrophic increasing public health bur- et al. used 6,000 microsatellites in 400 Irish patients
den of CKD and ESRD overall, the identification of with and without type I DN to identify two signif-
genes related to these two outcomes represents a icant makers on chromosome 10 and four addi-
substantial opportunity for improvement in under- tional markers worthy of investigation on
standing the complex pathological mechanisms chromosomes 2, 4, 6, and 17 [159]. A Japanese
behind CKD progression. A vast majority of the scan using over 80,000 SNPs in 87 type II diabetics
work in this area has been conducted in adult with nephropathy and 92 diabetic controls without
populations due to the higher prevalence of these nephropathy identified the engulfment and cell
outcomes, as well as to the availability of large motility 1 (ELMO1) gene as a DN susceptibility
cohorts of adults well phenotyped for the more com- gene [158]. In a recent GWA SNP association
mon complex diseases which result in CKD and study, Hanson et al. found a significant association
ESRD within this population. We will briefly review with the plasmacytoma variant translocation
findings from the adult studies here; however, as the (PVT1) gene with ESRD in 207 type II diabetics
precipitating causes differ between adults and chil- with and without DN [157]. Unfortunately, many
dren, disease mechanisms leading to the end point of of the above results still await verification [154].
ESRD may also differ. Therefore, genes identified in It is noteworthy that the above studies all
adults need to be tested for relevance in pediatric examined a qualitative outcome and were
patients. Large, multi-institutional/multinational conducted in the setting of DN. However, there
cohorts of CKD and ESRD pediatric patients have been a number of both large-scale linkage
phenotyped in a standardized manner will be required and association studies published examining
to dissect the genetics of these complex and critical quantitative kidney function traits, such as glo-
outcomes of various pediatric kidney diseases. merular filtration rate and urinary albumin excre-
A number of groups have performed linkage tion, in various populations [15, 141,
analyses for ESRD in the context of one of its 161, 162]. Results from most of these studies
major precipitating causes in adults, diabetic require verification. A number of advantages
nephropathy (DN) [141–153]. Results from have been suggested for the use of quantitative
these linkage studies have identified numerous traits in the genetic dissection of complex diseases
distinct loci; however, the best evidence supports [163–165]. The continued use of large and com-
DN susceptibility loci on chromosomes 10p and prehensive linkage and association scans should
18q [154]. A susceptibility gene, carnosinase-1 assist in the further identification and replication
(CNDP1), has been identified from the 18q of ESRD susceptibility loci. However, their rele-
locus. Two independent groups have demon- vance to children will have to be tested. The
strated diabetic patients harboring a particular leu- establishment of cohorts of children with ESRD
cine repeat polymorphism of CNDP1 to be at a is challenging given the small numbers; neverthe-
significantly reduced risk of developing nephrop- less, such cohorts can be collected through long-
athy and ESRD [155, 156]. term collaborative efforts. Such cohorts are
512 K. Maresso and U. Broeckel

required to map genetic variation, which may portion of the genome is sequenced, offers a less
ultimately allow for more aggressive and tailored expensive alternative to WGS and has been suc-
prevention and treatment efforts in this life- cessfully applied to the identification of unknown
threatening disorder. genetic defects in various Mendelian disorders.
However, the application of either whole-genome
or whole-exome sequencing to common, complex
The Impact of Novel Sequencing diseases, as opposed to Mendelian, is far from
Technologies on Gene Identification straightforward and many challenges remain. Cur-
rently, significant efforts are being devoted toward
Significant resources have been invested in iden- developing the overall infrastructure to process the
tifying the genetic variants underlying common, wealth of DNA information generated by these
complex human diseases and traits. If we look at technologies [169]. Ultimately the challenge will
the genetics field in general, large-scale GWASs be to interpret each individual’s cumulative collec-
have been performed in populations around the tion of polymorphisms and more specifically iden-
world for almost every common disorder and tify disease-causing or disease risk variants.
numerous complex traits. To date, these studies A comprehensive analysis needs to differentiate
have yielded thousands of genomic loci associ- the functional or causative variants from the large
ated with human diseases and traits, and the list amount of variations that are identified using these
will continue to grow (http://gwas.nih.gov/index. NGS techniques. As a first step, bioinformatics
html). However, despite this tremendous success, analyses are a first step to describe the frequency
population genetics analysis clearly demonstrates of variants as well as modeling-based approaches
that only a portion of the overall genetic risk has to predict the functional effects and implications.
been identified using the GWAS approach [166] As these methods will provide a first statistical
of the most widely discussed reasons for this attempt of categorizing each individual’s genome
might lie on the fact that GWASs focus toward information, bioinformatics is limited in the ability
common DNA variants and one apparent source to describe functional effects in a complex biolog-
of this missing heritability can be hidden in less ical system. Consequently, the challenge will be to
frequent variants. This notion is now leading to more comprehensively assess the impact of vari-
the next wave of comprehensive genome analysis ants in a biological system, which captures each
allowing for novel ways to sequence genomes. individual’s unique genome and at the same time
Recent advances in the area of next-generation allows for a comprehensive functional analysis on a
sequencing (NGS) offer the opportunity to build biological level.
and expand upon those findings by creating again While it is apparent that the field is very much
a paradigm shift in the way we approach studying dominated by the technological advancements,
disease at a genetic and genomic level. understanding the current state of sequencing tech-
The area of NGS is rapidly evolving, with con- nology, the necessary steps involved in genome
stant improvements both in instrumentation and in analysis, and the associated limitations will be crit-
downstream data-analytic techniques [167]. While ical for applying these methods for disease identi-
the cost of whole-genome sequencing (WGS) con- fication. Consequently we will discuss examples of
tinues to decrease, it can still be associated with a the application of both whole-genome and whole-
substantial cost. Nevertheless, it is now well in exome sequencing and their challenges and limita-
reach that a human genome can be analyzed for tions for complex disease in general.
around one thousand dollars [168]. One should
note however that this cost estimate often refers
only to the actual sequencing component and the Next-Generation Sequencing
downstream bioinformatics analysis and interpre-
tation will require resources beyond this estimate. Using any of the available next-generation
Whole-exome sequencing, where only the coding sequencing (NGS) platforms which yield millions
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 513

of DNA sequence reads in a single run is now a sequencing technology, the increase in through-
reality [170]. While in the beginning these tech- put, and, at the same time, the reduction in cost
nologies have been limited to larger genome cen- have been truly breathtaking. While sequencing
ters or sequencing groups due to the substantial technologies from companies such as
cost of the equipment, the technology has 454, Illumina, SOLiD, and other more recent plat-
advanced and the cost for the equipment has forms such as PacBio and Ion Torrent are cur-
become in reach for more laboratories. Conse- rently marketed and have captured significant
quently NGS has been spreading rapidly over the market share in both research and diagnostic
past years and many laboratories around the world areas, all methods still present significant chal-
are now using the technology to sequence cohorts lenges to sequence and data analysis. Despite
and individuals in the hopes of identifying novel different approaches to obtaining the sequence
variants responsible for common diseases and data, all current methods have a few characteris-
traits. Data from such studies is revealing a vast tics in common which influence the downstream
amount of rare DNA variation [171, 172], which analysis as well as data interpretation [173]. Cur-
presents specific analytical challenges to our cur- rent methods utilize a shotgun sequencing
rent methodologies but also poses a significantly approach as the target DNA is prepared as a
greater challenge to the overall field of genetics sequencing library which will be more or less
and personalized medicine: exactly how “per- randomly analyzed and numerous reads for each
sonal” does personalized medicine need to be? In base will be obtained. The sequence analysis will
other words, the extent to which risk prediction, assemble the obtained reads and various analyti-
diagnosis, and treatment needs to be tailored is cal approaches are utilized to determine the actual
reflective of the extent to which rare variation base call in addition to the determination of the
plays a role in common disease. If, for example, genotype. The difference between the
each individual and their risks of the different abovementioned methods is determined by the
common diseases are a compilation of rare, very length of a single read which can be obtained, as
rare, and even unique/private mutations, then risk well as the method to obtain or read the sequence
prediction, diagnosis, and treatment will need to itself. Improvement in sequencing technologies
be much more “individualized,” approaching that now allows for increased read lengths, which rep-
seen in cases of Mendelian disease, than if more resent substantial improvements over the initial
common variation were the culprit and risk pre- very short reads obtained just a short time ago.
diction, diagnosis, and treatment could be based The methods also differ in their detection
much more upon overall population inferences. approach. More established light-based detection
With NGS, the field of genomics is now poised is the underlying approach for SOLiD or Illumina,
to answer important questions related to the while newer technologies such as Ion Torrent rely
genetic architecture of disease since in the near on the detection of electrical signals as this method
future it will be possible to comprehensively is based on the use of computer chip technology.
describe the overall polymorphic structure of Numerous other yet less publicized methods are
each individual’s genome. currently under development, and, therefore, at
this time it is hard to predict which method or
platform will be the most widely used in the future.
Data-Analytic Techniques in NGS In fact, with the anticipated broad applications
which can be perceived for sequencing, it appears
With the development of every new technology, likely that a number of robust and cost-effective
significant challenges must be addressed if we approaches will be utilized, rather than just one
want to capitalize upon it. A number of key tech- single method. Different methods will address dif-
nologies, which enable the generation of large ferent needs and applications driven by require-
amounts of sequence data, have emerged over a ments related to throughput, accuracy, ease of
short period of time. The improvement in sample processing, turnaround time, and cost.
514 K. Maresso and U. Broeckel

Data Analysis in NGS assessed the frequency of loss of function variants


in the human genome. The authors estimate that a
A critical next step in next-generation sequencing human genome may contain up to approximately
represents the actual data analysis, sequence 100 loss of function variants, in addition to about
assembly, and the identification and annotation 20 genes which are completely inactivated
of DNA variants. While this field is rapidly devel- [175]. These findings in healthy individuals sug-
oping and novel analysis algorithms are con- gest a surprising tolerance to a reduced function in
stantly being explored and tailored for the genes. While the authors perform a sophisticated
specific NGS methods, a few general issues have and complex analysis to determine which variants
emerged. A number of manufacturers of sequenc- are most likely of greater functional importance,
ing equipment provide software as part of the this clearly demonstrates some of the major chal-
analysis related to their platform; other software lenges: With a large number of rare variants, and an
packages have been developed and can integrate unexpectedly high rate of variants with potentially
the output from the various sequence platforms significant functional effects, it will be challenging
[174]. In general, the programs conduct the to evaluate these variants and their downstream
assembly of the reads in relation to a effect solely based on sequence data. This also
predetermined and assembled human reference demonstrates the difficulties related to diagnostic
sequence. Most commonly, the assembly for the sequencing. Comprehensive whole-genome
genome sequence will be conducted against the sequencing will identify a number of significant
whole human reference genome sequence, even gene variants with significant effects when ana-
for an exome sequencing approach. Variant call- lyzed solely based on sequence prediction. While
ing will be conducted in reference to the baseline the functional effects for some of these variants
sequence and as a starting point for more compre- might be well compensated for due to functional
hensive annotation of downstream functional redundancy, it will be challenging to differentiate
effects. An initial analysis might focus on coding potential disease-causing variants from this loss of
sequence, the identification of synonymous and function background.
non-synonymous variants, as well as the estima-
tion of their likely impact on gene function. More
comprehensive approaches focus also on annotat- NGS in Pediatric Kidney Disease
ing noncoding sequence and the prediction of the
impact of noncoding and copy number variation. As the NGS methodology matures, a number of
In addition, an assessment of the observed popu- recent publications showcase the power of NGS in
lation frequency is often used as a means to cate- the field of pediatric nephrology. Since NGS can
gorize an individual’s variants as common, in lend itself both to targeted candidate-gene
comparison to rare, disease-causing variants. sequencing and whole-genome approaches, the
This approach has proved particularly helpful in published literature reflects all these approaches.
studies focusing on rare diseases. Thus far, many Using NGS, Otto et al. performed targeted
important and unexpected insights into the poly- sequencing in a cohort of 120 patients
morphic structure of the genome have been with nephronophthisis-associated ciliopathies
obtained. First and foremost, initial results of the [176]. For this group of diseases with a recessive
1,000 Genomes Project identified a vast number mode of inheritance, causative mutations have
of rare DNA variants in addition to the previously been previously identified in 18 different genes.
known common variants. This substantially Consequently, clinical mutation analysis for test-
expands the overall catalog of known human ing all genes in patients with a suspected disease
genome variation. Beyond the basic annotation will be expensive and time-consuming. The
with regard to frequency, the functional categori- authors developed a targeted sequencing
zation has provided some interesting insights. approach using PCR amplification covering all
A publication by McArthur et al. comprehensively exomes in the described genes. Subsequent
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 515

pooling of amplified PCR products and NGS targeted sequencing can represent a rapid and
allowed for a rapid, cost-effective, and highly cost-effective approach to test known genes, in
sensitive analysis of the samples. Interestingly in case where this methodology does not yield suf-
75 % of the analyzed cases, this approach did not ficient clarification, expanding to whole exome or
identify a causal variant. Expanding the sample genome will likely provide important additional
size in a follow-up publication, the sequence anal- information.
ysis in 1,056 patients established the molecular Expanding the targeted sequencing results for
diagnosis in 127/1,056 independent individuals CAKUT in including families with VACTERL
(12.0 %) and identified a single heterozygous association, which represents a rare disorder that
truncating mutation in an additional 31 individuals involves congenital abnormalities in multiple
(2.9 %) [177]. Taken together these reports dem- organs including the kidney and urinary tract,
onstrate the power of targeted sequencing for a Saisawat et al. demonstrated that combined
comprehensive analysis of the mutational spec- homozygosity mapping and whole-exome
trum in selected candidate genes and demonstrate resequencing was able to identify recessive muta-
its power for diagnostics. Nevertheless, these tions in the TNF receptor-associated protein
results also demonstrate the substantial heteroge- 1 gene (TRAP1) in two families with isolated
neity of the disease and further sequence analysis CAKUT and three families with VACTERL asso-
in order to identify causal variants in patients ciation. TRAP1 is a heat-shock protein 90-related
without an established genetic diagnosis is mitochondrial chaperone, expressed in renal epi-
warranted. thelia of mouse kidney and in the kidney of adult
Again using a targeted candidate-gene rats [179]. Using a similar approach for nephrotic
approach, Saisawat et al. analyzed a subset of syndrome (NS), Gee at al. aimed to identify
genes for congenital abnormalities of the kidney genetic factors, which might influence the differ-
and urinary tract (CAKUT) [178]. Five heterozy- ence between steroid-sensitive (SSNS) and
gous missense mutations were detected in steroid-resistant forms (SRNS). The authors
two candidate genes (FRAS1 and FREM2), detected biallelic mutations in epithelial mem-
which have not been previously linked to brane protein 2 (EMP2) in four individuals from
non-syndromic CAKUT. three unrelated families affected by SRNS or
In order to further understand the genetic basis SSNS. EMP2 is exclusively localized to glomeruli
of steroid-resistant nephrotic syndrome, in the kidney and subsequent knockdown of emp2
36 patients identified through the UK Renal Reg- in the zebra fish resulted in pericardial effusion,
istry were chosen and next-generation-sequenced supporting the pathogenic role of mutated EMP2
to screen 446 genes, including 24 genes known to in human NS. At the cellular level the authors also
be associated with hereditary steroid-resistant showed that knockdown of EMP2 in podocytes
nephrotic syndrome. The subsequent mutation and endothelial cells resulted in an increased
analysis revealed known and novel disease- amount of CAVEOLIN-1 and decreased cell pro-
associated variations in expected genes such as liferation [180]. To further expand on NGS in
NPHS1, NPHS2, and PLCe1 in 19 % of patients. kidney disease, Malone et al. report using WES
Interestingly, mutations were also detected in and targeted sequencing in families with focal
COQ2 and COL4A4 in two patients with isolated segmental glomerulosclerosis [181]. In seven
nephropathy and associated sensorineural deaf- families they identified rare or novel variants in
ness. Taken together, these studies demonstrate COL4A3 or COL4A4. The predominant clinical
the power of targeted NGS sequencing for the finding at diagnosis was proteinuria associated
analysis of previously identified disease and can- with hematuria. In all seven families, there were
didate genes. However, it also showcases that a individuals with nephrotic-range proteinuria with
candidate-gene approach might be limited in that histologic features of FSGS by light microscopy.
only a small portion of the disease variants might In one family, electron microscopy showed thin
be located in previously identified genes. While GBM, but four other families had variable
516 K. Maresso and U. Broeckel

findings inconsistent with classical Alport nephri- associations. The popularity of this method was
tis. Families with COL4A3 and COL4A4 variants due in large part to its ease of application; how-
that segregated with disease represent 10 % of the ever, the current and widespread use of candidate-
cohort. Consequently COL4A3 and COL4A4 var- gene studies has limited ability to result in the
iants can be considered in the interpretation of novel breakthroughs required to enable paradigm
next-generation sequencing data from such changes in the prevention, diagnosis, and treat-
patients. ment of these often life-threatening conditions.
Taken together, these NGS studies demonstrate Another major step in the field was accomplished
the beginning of what one might anticipate as an through the use of high-throughput genotyping
avalanche of results from large-scale sequencing methods including GWA studies with hundreds
studies. The studies demonstrate a number of of thousands to millions of SNPs. Expanding on
notable findings. First and foremost, NGS and the tremendous pace of technological develop-
targeted NGS can identify and confirm the results ments in DNA analysis, NGS represents another
from previous studies using standard sequencing technical revolution, which is likely to change the
technologies. NGS is likely to emerge as a fast and landscape of genetic research in fundamental
cost-effective methodology not only for research ways again. Beyond all technological advances,
but also for comprehensive, cost-effective, and a few major requirements and concepts remain for
rapid screening of candidate genes. Depending the field to move forward. In order for these
on the genetic architecture, phenotypic disease, genetic studies to become a reality, large, well-
and genetic heterogeneity, a targeted approach phenotyped cohorts of children are necessary. The
might identify genetic disease variants in a limited sample sizes needed for GWA studies to be suc-
subset of patients. Consequently, expanding to an cessful, those samples approaching 1,000 or
unbiased, whole-exome, or whole-genome more, cannot be collected at a single center due
approach is likely to identify novel, potentially to the often modest number of cases of any par-
rare, or fully private mutations, which contribute ticular kidney disorder. For whole-genome
to the disease phenotype. The interpretation of sequencing, sample size requirements might be
rare variants in the context of the overall genome different. While theoretically for rare diseases,
variability will emerge as the main challenge in one single sample might be sufficient to identify
the years to come. This will be of particular impor- potentially underlying disease variants, this needs
tance as we utilize NGS for clinical applications. to be analyzed on the background of many rare
variants. For this, family and pedigree information
will be helpful in addition to larger numbers of
The Past, Present, and Future normal samples in order to estimate whether a rare
Application of Genomic Methods variant is potentially rare and disease relevant or
in Pediatric Nephrology just simply rare.
In any case, it is apparent that investigators
With the application of genome-wide linkage ana- need to form interinstitutional and international
lyses as one of the first genomic technologies, collaborations with standardized methods of
pediatric nephrology witnessed some of the first phenotyping. This is now being pursued for
fundamental advances in the understanding of many diseases including adult renal patients as
many chronic and acute kidney diseases. With well as pediatric diseases. More collaborations
the completion of the HGP and the availability can be expected as the importance of genomic
of genome-wide SNP markers that were easily technologies in pediatric nephrology is increas-
and inexpensively typed, the field witnessed the ingly recognized, as such technologies become
candidate-gene era, where one or a few genes more readily available to investigators and as the
were genotyped for a handful of SNPs in usually infrastructure required for such studies becomes
small- to moderate-size samples with inadequate more firmly established. GWA and NGS studies
statistical power for detecting true disease offer a hypothesis-free design to gene discovery
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 517

and are therefore ideal for many complex kidney 4. Peters DJ, et al. Chromosome 4 localization of a
diseases where there is limited knowledge of dis- second gene for autosomal dominant polycystic kid-
ney disease. Nat Genet. 1993;5(4):359–62.
ease mechanisms. Being able to analyze a genome 5. Ward CJ, et al. The gene mutated in autosomal reces-
in a short time is likely to create a vast amount of sive polycystic kidney disease encodes a large,
genetics information. This poses the obvious receptor-like protein [see comment]. Nat Genet.
questions of data sharing and dissemination. The 2002;30(3):259–69.
6. Hugot JP, et al. Association of NOD2 leucine-rich
amount of data generated through NGS exceeds repeat variants with susceptibility to Crohn’s disease.
by far the amount of data, which was generated as Nature. 2001;411(6837):599–603.
part of the GWAS approach. Novel standards for 7. Horikawa Y, et al. Genetic variation in the gene
data sharing and guidelines for depositing large encoding calpain-10 is associated with type 2 diabetes
mellitus. Nat Genet. 2000;26(2):163–75.
amounts of sequence information are developing. 8. Helgadottir A, et al. The gene encoding
Finally, information on potential disease-causing 5-lipoxygenase activating protein confers risk of
variants needs to be put in context with the poten- myocardial infarction and stroke. Nat Genet.
tial impact and the functional relevance. Just a 2004;3(3):233–9.
9. Ogura Y, et al. A frameshift mutation in NOD2 asso-
short time ago it was hard to imagine that identi- ciated with susceptibility to Crohn’s disease. Nature.
fying a comprehensive set of potential disease 2001;411(6837):603–6.
mutations becomes simply a technical question, 10. Risch N, Merikangas K. The future of genetic studies of
which is solvable in a short time. Consequently complex human diseases. Science. 1996;273:1516–7.
11. Hirschhorn JN, et al. A comprehensive review
the field of genetics will need to expand from of genetic association studies. Genet Med.
DNA analysis and mapping to the functional anal- 2002;4(2):45–61.
ysis of genome variation. At this time, genetics is 12. Salonen JT, et al. Type 2 diabetes whole-genome
likely to incorporate functional genome annota- association study in four populations: the DiaGen
consortium. Am J Hum Genet. 2007;81(2):338–45.
tion, high-throughput phenotyping of targeted cell 13. Wellcome Trust Case Control, C. Genome-wide asso-
systems, and complex tissue models. Animal ciation study of 14,000 cases of seven common dis-
studies and model organisms will gain impor- eases and 3,000 shared controls [see comment].
tance, and model systems, which comprehen- Nature. 2007;447(7145):661–78.
14. Liu YJ, Liu XG, Wang L, Dina C, Yan H, Liu JF,
sively assess the complete human genome, will Levy S, Papasian CJ, Drees BM, Hamilton JJ,
be important. Historically, animal models have Meyre D, Delplanque J, Pei YF, Zhang L, Recker
served as a main platform. Novel technologies RR, Froguel P, Deng HW. Genome-wide association
such as induced pluripotent stem cells hold a scans identified CTNNBL1 as a novel gene for obe-
sity. Hum Mol Genet. 2008;17(12):1803–13.
tremendous potential for disease modeling. This 15. Hwang SJ, et al. A genome-wide association for kid-
field is rapidly developing and the combination of ney function and endocrine-related traits in the
genome analysis, together with the other genome- NHLBI’s Framingham Heart Study. BMC Med
wide technologies of transcriptomics and proteo- Genet. 2007;8 Suppl 1:S10.
16. Kestila M, et al. Congenital nephrotic syndrome of the
mics, offers the best hope for future fundamental Finnish type maps to the long arm of chromosome 19.
advances in the field of pediatric nephrology. Am J Hum Genet. 1994;54(5):757–64.
17. Kestila M, et al. Positionally cloned gene for a novel
glomerular protein–nephrin–is mutated in congenital
nephrotic syndrome. Mol Cell. 1998;1(4):575–82.
References 18. Boute N, et al. NPHS2, encoding the glomerular
protein podocin, is mutated in autosomal recessive
1. Mochizuki T, et al. PKD2, a gene for polycystic steroid-resistant nephrotic syndrome [erratum appears
kidney disease that encodes an integral membrane in Nat Genet 2000 May;25 (1):125]. Nat Genet.
protein. Science. 1996;272(5266):1339–42. 2000;24(4):349–54.
2. Schneider MC, et al. A gene similar to PKD1 maps to 19. Fuchshuber A, et al. Mapping a gene (SRN1) to
chromosome 4q22: a candidate gene for PKD2. chromosome 1q25-q31 in idiopathic nephrotic syn-
Genomics. 1996;38(1):1–4. drome confirms a distinct entity of autosomal reces-
3. Hughes J, et al. The polycystic kidney disease 1 (PKD1) sive nephrosis. Hum Mol Genet. 1995;4(11):2155–8.
gene encodes a novel protein with multiple cell recog- 20. Mondry A, et al. DNA polymorphisms and renal
nition domains. Nat Genet. 1995;10(2):151–60. disease: a critical appraisal of studies presented at
518 K. Maresso and U. Broeckel

the annual ERA/EDTA and ASN conferences in 2004 36. Niaudet P. Utility of genetic screening in children
and 2005 [see comment]. Nephrol Dial Transplant. with nephrotic syndrome presenting during the first
2006;21(10):2775–9. year of life. Nat Clin Pract Nephrol. 2007;3(9):472–3.
21. Frimat L, et al. Polymorphism of angiotensin 37. Ruf RG, et al. Identification of the first gene locus
converting enzyme, angiotensinogen, and angiotensin (SSNS1) for steroid-sensitive nephrotic syndrome
II type 1 receptor genes and end-stage renal failure in on chromosome 2p. J Am Soc Nephrol.
IgA nephropathy: IGARAS–a study of 274 Men. 2003;14(7):1897–900.
J Am Soc Nephrol. 2000;11(11):2062–7. 38. Taylor CM, et al. Clinico-pathological findings in
22. Schena FP, et al. ACE gene polymorphism and IgA diarrhoea-negative haemolytic uraemic syndrome.
nephropathy: an ethnically homogeneous study and a Pediatr Nephrol. 2004;19(4):419–25.
meta-analysis. Kidney Int. 2001;60(2):732–40. 39. Richards A, et al. Implications of the initial mutations
23. Pereira TV, et al. Influence of ACE I/D gene poly- in membrane cofactor protein (MCP; CD46) leading
morphism in the progression of renal failure in auto- to atypical hemolytic uremic syndrome. Mol
somal dominant polycystic kidney disease: a meta- Immunol. 2007;44(1-3):111–22.
analysis.[see comment]. Nephrol Dial Transplant. 40. Kind T, et al. Cobalamin C disease presenting as
2006;21(11):3155–63. hemolytic-uremic syndrome in the neonatal period.
24. Brenchley PE, et al. Translating knowledge of the J Pediatr Hematol Oncol. 2002;24(4):327–9.
human genome into clinical practice in nephrology 41. Dlott JS, et al. Drug-induced thrombotic thrombocy-
dialysis and transplantation: the renal genome net- topenic purpura/hemolytic uremic syndrome: a con-
work (ReGeNet) [comment]. Nephrol Dial Trans- cise review. Ther Apher Dial. 2004;8(2):102–11.
plant. 2006;21(10):2681–3. 42. Becker S, et al. HIV-associated thrombotic
25. Fliser D, et al. Advances in urinary proteome analysis microangiopathy in the era of highly active antiretro-
and biomarker discovery. J Am Soc Nephrol. viral therapy: an observational study. Clin Infect Dis.
2007;18(4):1057–71. 2004;39 Suppl 5:S267–75.
26. Nguyen MT, Devarajan P. Biomarkers for the early 43. Constantinescu AR, et al. Non-enteropathic hemo-
detection of acute kidney injury. Pediatr Nephrol. lytic uremic syndrome: causes and short-term course.
2008;23(12):2151–7. Am J Kidney Dis. 2004;43(6):976–82.
27. Mathieson PW. Minimal change nephropathy and 44. George JN. The association of pregnancy with throm-
focal segmental glomerulosclerosis. Semin botic thrombocytopenic purpura-hemolytic uremic
Immunopathol. 2007;29(4):415–26. syndrome. Curr Opin Hematol. 2003;10(5):339–44.
28. Niaudet P. Genetic forms of nephrotic syndrome. 45. Mungall S, Mathieson P. Hemolytic uremic syndrome
Pediatr Nephrol. 2004;19(12):1313–8. in metastatic adenocarcinoma of the prostate. Am J
29. Hinkes B, et al. Positional cloning uncovers muta- Kidney Dis. 2002;40(6):1334–6.
tions in PLCE1 responsible for a nephrotic syndrome 46. Pichette V, et al. Familial hemolytic-uremic syndrome
variant that may be reversible [see comment]. and homozygous factor H deficiency. Am J Kidney
Nat Genet. 2006;38(12):1397–405. Dis. 1994;24(6):936–41.
30. Hasselbacher K, et al. Recessive missense mutations 47. Pirson Y, et al. Hemolytic uremic syndrome in three
in LAMB2 expand the clinical spectrum of LAMB2- adult siblings: a familial study and evolution. Clin
associated disorders [see comment]. Kidney Int. Nephrol. 1987;28(5):250–5.
2006;70(6):1008–12. 48. Kaplan BS, et al. Renal transplantation in adults with
31. Zenker M, et al. Human laminin beta2 deficiency autosomal recessive inheritance of hemolytic uremic
causes congenital nephrosis with mesangial sclerosis syndrome. Am J Kidney Dis. 1997;30(6):760–5.
and distinct eye abnormalities. Hum Mol Genet. 49. Warwicker P, et al. Genetic studies into inherited and
2004;13(21):2625–32. sporadic hemolytic uremic syndrome [see comment].
32. Hinkes BG, et al. Nephrotic syndrome in the first year Kidney Int. 1998;53(4):836–44.
of life: two thirds of cases are caused by mutations in 50. Caprioli J, et al. Complement factor H mutations
4 genes (NPHS1, NPHS2, WT1, and LAMB2) [see and gene polymorphisms in haemolytic uraemic
comment]. Pediatrics. 2007;119(4):e907–19. syndrome: the C-257T, the A2089G and the
33. Hinkes B, et al. Specific podocin mutations correlate G2881T polymorphisms are strongly associated
with age of onset in steroid-resistant nephrotic syndrome with the disease. Hum Mol Genet. 2003;12(24):
[see comment]. J Am Soc Nephrol. 2008;19(2):365–71. 3385–95.
34. Weber S, et al. NPHS2 mutation analysis shows 51. Caprioli J, et al. The molecular basis of familial
genetic heterogeneity of steroid-resistant nephrotic hemolytic uremic syndrome: mutation analysis of
syndrome and low post-transplant recurrence. Kidney factor H gene reveals a hot spot in short consensus
Int. 2004;66(2):571–9. repeat 20. J Am Soc Nephrol. 2001;12(2):297–307.
35. Ruf RG, et al. Patients with mutations in NPHS2 52. Martinez-Barricarte R, et al. The complement factor
(podocin) do not respond to standard steroid treatment H R1210C mutation is associated with atypical hemo-
of nephrotic syndrome [see comment]. J Am Soc lytic uremic syndrome. J Am Soc Nephrol. 2008;
Nephrol. 2004;15(3):722–32. 19(3):639–46.
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 519

53. Perez-Caballero D, et al. Clustering of missense muta- 67. Pickering MC, et al. Spontaneous hemolytic uremic
tions in the C-terminal region of factor H in atypical syndrome triggered by complement factor H
hemolytic uremic syndrome. Am J Hum Genet. lacking surface recognition domains. J Exp Med.
2001;68(2):478–84. 2007;204(6):1249–56.
54. Richards A, et al. Factor H mutations in hemolytic 68. Esparza-Gordillo J, et al. Predisposition to atypical
uremic syndrome cluster in exons 18-20, a domain hemolytic uremic syndrome involves the concurrence
important for host cell recognition. Am J Hum Genet. of different susceptibility alleles in the regulators of
2001;68(2):485–90. complement activation gene cluster in 1q32 [erratum
55. Sanchez-Corral P, et al. Structural and functional appears in Hum Mol Genet. 2005 Apr 15;14(8):1107].
characterization of factor H mutations associated Hum Mol Genet. 2005;14(5):703–12.
with atypical hemolytic uremic syndrome. Am J 69. Esparza-Gordillo J, et al. Insights into hemolytic ure-
Hum Genet. 2002;71(6):1285–95. mic syndrome: segregation of three independent pre-
56. Fremeaux-Bacchi V, et al. The development of atyp- disposition factors in a large, multiple affected
ical haemolytic-uraemic syndrome is influenced by pedigree. Mol Immunol. 2006;43(11):1769–75.
susceptibility factors in factor H and membrane cofac- 70. Zerres K, Rudnik-Schoneborn S, Deget F. Childhood
tor protein: evidence from two independent cohorts. onset autosomal dominant polycystic kidney disease
J Med Genet. 2005;42(11):852–6. in sibs: clinical picture and recurrence risk. German
57. Noris M, et al. Familial haemolytic uraemic syndrome Working Group on Paediatric Nephrology
and an MCP mutation [see comment]. Lancet. (Arbeitsgemeinschaft fur Padiatrische Nephrologie).
2003;362(9395):1542–7. J Med Genet. 1993;30(7):583–8.
58. Richards A, et al. Mutations in human complement 71. Fick GM, et al. Characteristics of very early onset
regulator, membrane cofactor protein (CD46), predis- autosomal dominant polycystic kidney disease.
pose to development of familial hemolytic uremic J Am Soc Nephrol. 1993;3(12):1863–70.
syndrome. Proc Natl Acad Sci U S A. 2003;100 72. Peters DJ, Sandkuijl LA. Genetic heterogeneity of
(22):12966–71. polycystic kidney disease in Europe. Contrib
59. Fremeaux-Bacchi V, et al. Complement factor I: a Nephrol. 1992;97:128–39.
susceptibility gene for atypical haemolytic uraemic 73. Harris PC, et al. Cyst number but not the rate of cystic
syndrome. J Med Genet. 2004;41(6):e84. growth is associated with the mutated gene in autoso-
60. Kavanagh D, et al. Mutations in complement factor I mal dominant polycystic kidney disease. J Am Soc
predispose to development of atypical hemolytic Nephrol. 2006;17(11):3013–9.
uremic syndrome. J Am Soc Nephrol. 74. Rossetti S, Harris PC. Genotype-phenotype correla-
2005;16(7):2150–5. tions in autosomal dominant and autosomal recessive
61. Goicoechea de Jorge E, et al. Gain-of-function muta- polycystic kidney disease [see comment]. J Am Soc
tions in complement factor B are associated with Nephrol. 2007;18(5):1374–80.
atypical hemolytic uremic syndrome [erratum 75. Hateboer N, et al. Comparison of phenotypes of poly-
appears in Proc Natl Acad Sci U S A. 2007 Jun cystic kidney disease types 1 and 2. European PKD1-
19;104(25):10749]. Proc Natl Acad Sci PKD2 Study Group. Lancet. 1999;353(9147):103–7.
U S A. 2007;104(1):240–5. 76. Michaud J, et al. Autosomal dominant polycystic
62. Fremeaux-Bacchi V, Regnier C, Blouin J, Dragon- kidney disease in the fetus. Am J Med Genet.
Durey MA, Fridman WH, Janssen B, Loirat C. Pro- 1994;51(3):240–6.
tective or aggressive: paradoxical role of C3 in atyp- 77. Torra R, et al. Linkage, clinical features, and progno-
ical hemolytic uremic syndrome. Mol Immunol. sis of autosomal dominant polycystic kidney
2007;44(1):172. disease types 1 and 2. J Am Soc Nephrol.
63. Caprioli J, et al. Genetics of HUS: the impact of MCP, 1996;7(10):2142–51.
CFH, and IF mutations on clinical presentation, 78. Rossetti S, et al. Association of mutation position in
response to treatment, and outcome. Blood. 2006; polycystic kidney disease 1 (PKD1) gene and
108(4):1267–79. development of a vascular phenotype. Lancet.
64. de Cordoba SR, de Jorge EG. Translational mini- 2003;361(9376):2196–201.
review series on complement factor H: genetics and 79. Rossetti S, et al. The position of the polycystic kidney
disease associations of human complement factor disease 1 (PKD1) gene mutation correlates with
H. Clin Exp Immunol. 2008;151(1):1–13. the severity of renal disease. J Am Soc Nephrol.
65. Neumann HP, et al. Haemolytic uraemic syndrome 2002;13(5):1230–7.
and mutations of the factor H gene: a registry-based 80. Paterson AD, et al. Progressive loss of renal function
study of German speaking countries. J Med Genet. is an age-dependent heritable trait in type 1 autosomal
2003;40(9):676–81. dominant polycystic kidney disease. J Am Soc
66. Sellier-Leclerc AL, et al. Differential impact of com- Nephrol. 2005;16(3):755–62.
plement mutations on clinical characteristics in atyp- 81. Fain PR, et al. Modifier genes play a significant role in
ical hemolytic uremic syndrome. J Am Soc Nephrol. the phenotypic expression of PKD1 [see comment].
2007;18(8):2392–400. Kidney Int. 2005;67(4):1256–67.
520 K. Maresso and U. Broeckel

82. Pei Y. Nature and nurture on phenotypic variability of 99. Hogg RJ. IgA nephropathy: what’s new? Pediatr
autosomal dominant polycystic kidney disease [com- Nephrol. 2007;22(11):1809–14.
ment]. Kidney Int. 2005;67(4):1630–1. 100. Ronkainen J, et al. Long-term outcome 19 years after
83. Persu A, et al. Modifier effect of ENOS in autosomal childhood IgA nephritis: a retrospective cohort study
dominant polycystic kidney disease. Hum Mol Genet. [see comment]. Pediatr Nephrol. 2006;21(9):
2002;11(3):229–41. 1266–73.
84. Walker D, et al. The ENOS polymorphism is not 101. Coppo R, et al. Idiopathic nephropathy with IgA
associated with severity of renal disease in polycystic deposits. Pediatr Nephrol. 2000;15(1-2):139–50.
kidney disease 1 [see comment]. Am J Kidney Dis. 102. Coppo R, D’Amico G. Factors predicting progression
2003;41(1):90–4. of IgA nephropathies. J Nephrol. 2005;18(5):503–12.
85. Furu L, et al. Milder presentation of recessive poly- 103. Yoshikawa N, Ito H, Nakamura H. Prognostic indica-
cystic kidney disease requires presence of amino tors in childhood IgA nephropathy. Nephron. 1992;
acid substitution mutations. J Am Soc Nephrol. 60(1):60–7.
2003;14(8):2004–14. 104. Linne T, et al. Course and long-term outcome of
86. Rossetti S, et al. A complete mutation screen of idiopathic IgA nephropathy in children. Pediatr
PKHD1 in autosomal-recessive polycystic kidney Nephrol. 1991;5(4):383–6.
disease (ARPKD) pedigrees. Kidney Int. 105. Levy M, et al. Berger’s disease in children. Natural
2003;64(2):391–403. history and outcome. Medicine. 1985;64(3):157–80.
87. Sharp AM, et al. Comprehensive genomic analysis of 106. Hastings MC, Delos Santos NM, Wyatt RJ. Renal
PKHD1 mutations in ARPKD cohorts. J Med Genet. survival in pediatric patients with IgA nephropathy
2005;42(4):336–49. [comment]. Pediatr Nephrol. 2007;22(2):317–8.
88. Mrug M, et al. Kinesin family member 12 is a candi- 107. Nozawa R, et al. Clinicopathological features and the
date polycystic kidney disease modifier in the cpk prognosis of IgA nephropathy in Japanese children
mouse. J Am Soc Nephrol. 2005;16(4):905–16. on long-term observation. Clin Nephrol.
89. Glassock RJ, et al. IgA nephropathy in Japan. Am J 2005;64(3):171–9.
Nephrol. 1985;5(2):127–37. 108. Wyatt RJ, et al. IgA nephropathy: long-term progno-
90. D’Amico G. The commonest glomerulonephritis in sis for pediatric patients. J Pediatr. 1995;127(6):
the world: IgA nephropathy. Q J Med. 1987;64 913–9.
(245):709–27. 109. Tomana M, et al. Galactose-deficient IgA1 in sera of
91. Levy M, Berger J. Worldwide perspective of IgA IgA nephropathy patients is present in complexes
nephropathy. Am J Kidney Dis. 1988;12(5):340–7. with IgG. Kidney Int. 1997;52(2):509–16.
92. Schena FP. A retrospective analysis of the natural 110. Tomana M, et al. Circulating immune complexes in
history of primary IgA nephropathy worldwide. IgA nephropathy consist of IgA1 with galactose-
Am J Med. 1990;89(2):209–15. deficient hinge region and antiglycan antibodies.
93. Hsu SI, et al. Evidence for genetic factors in the J Clin Invest. 1999;104(1):73–81.
development and progression of IgA nephropathy. 111. Novak J, et al. IgA glycosylation and IgA immune
Kidney Int. 2000;57(5):1818–35. complexes in the pathogenesis of IgA nephropathy.
94. Johnson RJ, et al. Hypothesis: dysregulation of Semin Nephrol. 2008;28(1):78–87.
immunologic balance resulting from hygiene and 112. Gharavi AG, et al. IgA nephropathy, the most com-
socioeconomic factors may influence the epidemiol- mon cause of glomerulonephritis, is linked to 6q22-
ogy and cause of glomerulonephritis worldwide. Am 23. Nat Genet. 2000;26(3):354–7.
J Kidney Dis. 2003;42(3):575–81. 113. Schena FP, et al. The IgA nephropathy Biobank. An
95. Coppo R. Pediatric IgA nephropathy: clinical important starting point for the genetic dissection of a
and therapeutic perspectives. Semin Nephrol. 2008; complex trait. BMC Nephrol. 2005;6:14.
28(1):18–26. 114. Bisceglia L, et al. Genetic heterogeneity in Italian
96. Coppo R, et al. Frequency of renal diseases and clinical families with IgA nephropathy: suggestive linkage
indications for renal biopsy in children (report of the for two novel IgA nephropathy loci. Am J Hum
Italian National Registry of Renal Biopsies in Chil- Genet. 2006;79(6):1130–4.
dren). Group of renal immunopathology of the Italian 115. Paterson AD, et al. Genome-wide linkage scan of a
society of pediatric nephrology and group of renal large family with IgA nephropathy localizes a novel
immunopathology of the Italian society of nephrology. susceptibility locus to chromosome 2q36. J Am Soc
Nephrol Dial Transplant. 1998;13(2):293–7. Nephrol. 2007;18(8):2408–15.
97. Lee YM, et al. Analysis of renal biopsies performed in 116. Beerman I, et al. The genetics of IgA nephropathy.
children with abnormal findings in urinary mass Nat Clin Pract Nephrol. 2007;3(6):325–38.
screening [see comment]. Acta Paediatr. 2006;95(7): 117. Li YJ, et al. Family-based association study showing
849–53. that immunoglobulin A nephropathy is associated
98. Yoshikawa N, Tanaka R, Iijima K. Pathophysiology with the polymorphisms 2093C and 2180T in the 30
and treatment of IgA nephropathy in children [see untranslated region of the Megsin gene. J Am Soc
comment]. Pediatr Nephrol. 2001;16(5):446–57. Nephrol. 2004;15(7):1739–43.
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 521

118. Xia YF, et al. A family-based association study of 133. Ogden CL, et al. Prevalence of overweight and obe-
megsin A23167G polymorphism with susceptibility sity in the United States, 1999–2004. JAMA.
and progression of IgA nephropathy in a Chinese 2006;295(13):1549–55.
population. Clin Nephrol. 2006;65(3):153–9. 134. Chinn S, Rona RJ. Prevalence and trends in over-
119. Kim YS, et al. Uteroglobin gene polymorphisms weight and obesity in three cross sectional studies of
affect the progression of immunoglobulin A nephrop- British Children, 1974–94. BMJ. 2001;322
athy by modulating the level of uteroglobin expres- (7277):24–6.
sion. Pharmacogenetics. 2001;11(4):299–305. 135. de Onis M, Blossner M. Prevalence and trends of
120. Narita I, et al. Role of uteroglobin G38A polymor- overweight among preschool children in developing
phism in the progression of IgA nephropathy in Jap- countries. Am J Clin Nutr. 2000;72(4):1032–9.
anese patients. Kidney Int. 2002;61(5):1853–8. 136. Filozof C, et al. Obesity prevalence and trends in
121. Matsunaga A, et al. Association of the uteroglobin Latin-American countries. Obes Rev. 2001;2(2):
gene polymorphism with IgA nephropathy. Am J 99–106.
Kidney Dis. 2002;39(1):36–41. 137. Wang Y, et al. Epidemic of childhood obesity: impli-
122. Li GS, et al. Variants of C1GALT1 gene are associ- cations for kidney disease. Adv Chronic Kidney Dis.
ated with the genetic susceptibility to IgA nephropa- 2006;13(4):336–51.
thy [see comment]. Kidney Int. 2007;71(5):448–53. 138. McDonald SP, Craig JC, A. Australian and
123. Takei T, et al. Association between single-nucleotide New Zealand Paediatric Nephrology. Long-term sur-
polymorphisms in selectin genes and immunoglobulin vival of children with end-stage renal disease [see
A nephropathy. Am J Hum Genet. 2002;70(3):781–6. comment]. N Engl J Med. 2004;350(26):2654–62.
124. Tuglular S, Berthoux P, Berthoux F. Polymorphisms 139. Groothoff JW. Long-term outcomes of children with
of the tumour necrosis factor alpha gene at position end-stage renal disease. Pediatr Nephrol. 2005;20(7):
-308 and TNFd microsatellite in primary IgA 849–53.
nephropathy. Nephrol Dial Transplant. 2003; 140. Balinsky W. Pediatric end-stage renal disease: inci-
18(4):724–31. dence, management, and prevention. J Pediatr Health
125. Carturan S, et al. Association between transforming Care. 2000;14(6):304–8.
growth factor beta1 gene polymorphisms and IgA 141. Iyengar SK, et al. Genome-wide scans for diabetic
nephropathy. J Nephrol. 2004;17(6):786–93. nephropathy and albuminuria in multiethnic
126. Masutani K, et al. Impact of interferon-gamma and populations: the family investigation of nephropathy
interleukin-4 gene polymorphisms on development and diabetes (FIND). Diabetes. 2007;56(6):1577–85.
and progression of IgA nephropathy in Japanese 142. Imperatore G, et al. Sib-pair linkage analysis for sus-
patients. Am J Kidney Dis. 2003;41(2):371–9. ceptibility genes for microvascular complications
127. Akiyama F, et al. Single-nucleotide polymorphisms in among Pima Indians with type 2 diabetes. Pima Dia-
the class II region of the major histocompatibility betes Genes Group. Diabetes. 1998;47(5):821–30.
complex in Japanese patients with immunoglobulin 143. Moczulski DK, et al. Major susceptibility locus for
A nephropathy. J Hum Genet. 2002;47(10):532–8. nephropathy in type 1 diabetes on chromosome 3q:
128. Obara W, et al. Association of single-nucleotide poly- results of novel discordant sib-pair analysis. Diabetes.
morphisms in the polymeric immunoglobulin recep- 1998;47(7):1164–9.
tor gene with immunoglobulin A nephropathy (IgAN) 144. Yu H, et al. Identification of human plasma kallikrein
in Japanese patients. J Hum Genet. 2003;48 gene polymorphisms and evaluation of their role in
(6):293–9. end-stage renal disease. Hypertension. 1998;31(4):
129. Ohtsubo S, et al. Association of a single-nucleotide 906–11.
polymorphism in the immunoglobulin mu-binding 145. Bowden DW, et al. A genome scan for diabetic
protein 2 gene with immunoglobulin A nephropathy. nephropathy in African Americans. Kidney Int.
J Hum Genet. 2005;50(1):30–5. 2004;66(4):1517–26.
130. Berthoux FC, et al. CC-chemokine receptor five gene 146. Yu H, et al. Linkage analysis between loci in the
polymorphism in primary IgA nephropathy: the 32 bp renin-angiotensin axis and end-stage renal disease
deletion allele is associated with late progression to in African Americans. J Am Soc Nephrol. 1996;
end-stage renal failure with dialysis. Kidney Int. 7(12):2559–64.
2006;69(3):565–72. 147. Freedman BI, et al. Genetic linkage analysis of
131. Panzer U, et al. The chemokine receptor 5 Delta32 growth factor loci and end-stage renal disease in Afri-
mutation is associated with increased renal survival in can Americans. Kidney Int. 1997;51(3):819–25.
patients with IgA nephropathy [erratum appears in 148. Freedman BI, et al. Linkage heterogeneity of
Kidney Int. 2005 Mar;67(3):1213 Note: Kramer, end-stage renal disease on human chromosome 10.
Bernhard H [corrected to Kramer, Bernhard K]]. Kid- Kidney Int. 2002;62(3):770–4.
ney Int. 2005;67(1):75–81. 149. Yu H, et al. Evaluation of markers on human chromo-
132. Ogden CL, et al. Prevalence and trends in overweight some 10, including the homologue of the rodent Rf-1
among US children and adolescents, 1999–2000 [see gene, for linkage to ESRD in black patients. Am J
comment]. JAMA. 2002;288(14):1728–32. Kidney Dis. 1999;33(2):294–300.
522 K. Maresso and U. Broeckel

150. Vardarli I, et al. Gene for susceptibility to diabetic 166. Manolio TA, Collins FS, Cox NJ, Goldstein DB,
nephropathy in type 2 diabetes maps to 18q22.3-23. Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM,
Kidney Int. 2002;62(6):2176–83. Cardon LR, Chakravarti A, Cho JH, Guttmacher AE,
151. Iyengar SK, et al. Linkage analysis of candidate loci Kong A, Kruglyak L, Mardis E, Rotimi CN, Slatkin M,
for end-stage renal disease due to diabetic nephropa- Valle D, Whittemore AS, Boehnke M, Clark AG,
thy. J Am Soc Nephrol. 2003;14(7 Suppl 2): Eichler EE, Gibson G, Haines JL, Mackay TF,
S195–201. McCarroll SA, Visscher PM. Finding the missing heri-
152. Freedman BI, et al. A genome scan for all-cause tability of complex diseases. Nature. 2009;461:747–53.
end-stage renal disease in African Americans. 167. Ng PC, Kirkness EF. Whole genome sequencing.
Nephrol Dial Transplant. 2005;20(4):712–8. Methods Mol Biol. 2010;628:215–26.
153. Osterholm AM, et al. Genome-wide scan for type 168. Mardis ER. Anticipating the 1,000 dollar genome.
1 diabetic nephropathy in the Finnish population Genome Biol. 2006;7:112.
reveals suggestive linkage to a single locus on chro- 169. DePristo MA, Banks E, Poplin R, Garimella KV,
mosome 3q [see comment]. Kidney Int. 2007; Maguire JR, Hartl C, Philippakis AA, del Angel G,
71(2):140–5. Rivas MA, Hanna M, McKenna A, Fennell TJ,
154. Iyengar SK, Freedman BI, Sedor JR. Mining the Kernytsky AM, Sivachenko AY, Cibulskis K, Gabriel
genome for susceptibility to diabetic nephropathy: SB, Altshuler D, Daly MJ. A framework for variation
the role of large-scale studies and consortia. Semin discovery and genotyping using next-generation
Nephrol. 2007;27(2):208–22. DNA sequencing data. Nat Genet. 2011;43:491–8.
155. Janssen B, et al. Carnosine as a protective factor in 170. Pareek CS, Smoczynski R, Tretyn A. Sequencing
diabetic nephropathy: association with a leucine technologies and genome sequencing. J Appl Genet.
repeat of the carnosinase gene CNDP1. Diabetes. 2011;52:413–35.
2005;54(8):2320–7. 171. Mills RE, Walter K, Stewart C, Handsaker RE,
156. Freedman BI, et al. A leucine repeat in the carnosinase Chen K, Alkan C, Abyzov A, Yoon SC, Ye K,
gene CNDP1 is associated with diabetic end-stage Cheetham RK, Chinwalla A, Conrad DF, Fu Y,
renal disease in European Americans. Nephrol Dial Grubert F, Hajirasouliha I, Hormozdiari F,
Transplant. 2007;22(4):1131–5. Iakoucheva LM, Iqbal Z, Kang S, Kidd JM, Konkel
157. Hanson RL, et al. Identification of PVT1 as a candi- MK, Korn J, Khurana E, Kural D, Lam HY, Leng J,
date gene for end-stage renal disease in type 2 Li R, Li Y, Lin CY, Luo R, Mu XJ, Nemesh J,
diabetes using a pooling-based genome-wide single Peckham HE, Rausch T, Scally A, Shi X, Stromberg
nucleotide polymorphism association study. Diabe- MP, Stutz AM, Urban AE, Walker JA, Wu J, Zhang Y,
tes. 2007;56(4):975–83. Zhang ZD, Batzer MA, Ding L, Marth GT,
158. Shimazaki A, et al. Genetic variations in the gene McVean G, Sebat J, Snyder M, Wang J, Eichler EE,
encoding ELMO1 are associated with susceptibility Gerstein MB, Hurles ME, Lee C, McCarroll SA,
to diabetic nephropathy. Diabetes. 2005;54(4): Korbel JO. Mapping copy number variation by
1171–8. population-scale genome sequencing. Nature.
159. McKnight AJ, et al. A genome-wide DNA microsat- 2011;470:59–65.
ellite association screen to identify chromosomal 172. Project TG. A map of human genome variation from
regions harboring candidate genes in diabetic population-scale sequencing. Nature.
nephropathy. J Am Soc Nephrol. 2006;17(3):831–6. 2010;467:1061–73.
160. Ewens KG, et al. Assessment of 115 candidate genes 173. Lee H, Tang H. Next-generation sequencing technol-
for diabetic nephropathy by transmission/disequilib- ogies and fragment assembly algorithms. Methods
rium test. Diabetes. 2005;54(11):3305–18. Mol Biol. 2012;855:155–74.
161. Schelling JR, et al. Genome-wide scan for estimated 174. Stein LD. An introduction to the informatics of “next-
glomerular filtration rate in multi-ethnic diabetic generation” sequencing. Curr Protoc Bioinformatics /
populations: the Family Investigation of Nephropathy editorial board, Andreas D Baxevanis [et al.]. 2011;-
and Diabetes (FIND). Diabetes. 2008;57(1):235–43. Chapter 11:Unit 11 1.
162. Freedman BI, et al. A genome-wide scan for urinary 175. MacArthur DG, Balasubramanian S, Frankish A,
albumin excretion in hypertensive families. Hyper- Huang N, Morris J, Walter K, Jostins L,
tension. 2003;42(3):291–6. Habegger L, Pickrell JK, Montgomery SB, Albers
163. Lander ES, Schork NJ. Genetic dissection of complex CA, Zhang ZD, Conrad DF, Lunter G, Zheng H,
traits [erratum appears in Science 1994 Oct 21;266 Ayub Q, DePristo MA, Banks E, Hu M, Handsaker
(5184):353]. Science. 1994;265(5181):2037–48. RE, Rosenfeld JA, Fromer M, Jin M, Mu XJ,
164. Carlson CS, et al. Mapping complex disease loci in Khurana E, Ye K, Kay M, Saunders GI, Suner MM,
whole-genome association studies. Nature. 2004;429 Hunt T, Barnes IH, Amid C, Carvalho-Silva DR,
(6990):446–52. Bignell AH, Snow C, Yngvadottir B, Bumpstead S,
165. Ghosh S, Schork NJ. Genetic analysis of NIDDM. Cooper DN, Xue Y, Romero IG, Wang J, Li Y, Gibbs
The study of quantitative traits. Diabetes. 1996;45(1): RA, McCarroll SA, Dermitzakis ET, Pritchard JK,
1–14. Barrett JC, Harrow J, Hurles ME, Gerstein MB,
17 Genomic Methods in the Diagnosis and Treatment of Pediatric Kidney Disease 523

Tyler-Smith C. A systematic survey of loss-of-func- Natarajan S, Sperry E, Matassa DS, Stajic N,


tion variants in human protein-coding genes. Science. Bogdanovic R, de Blaauw I, Marcelis CL, Wijers
2012;335:823–8. CH, Bartels E, Schmiedeke E, Schmidt D,
176. Otto EA, Ramaswami G, Janssen S, Chaki M, Allen Marzheuser S, Grasshoff-Derr S, Holland-Cunz S,
SJ, Zhou W, Airik R, Hurd TW, Ghosh AK, Wolf MT, Ludwig M, Nothen MM, Draaken M, Brosens E,
Hoppe B, Neuhaus TJ, Bockenhauer D, Milford DV, Heij H, Tibboel D, Herrmann BG, Solomon BD, de
Soliman NA, Antignac C, Saunier S, Johnson CA, Klein A, van Rooij IA, Esposito F, Reutter HM,
Hildebrandt F. Mutation analysis of Hildebrandt F. Whole-exome resequencing reveals
18 nephronophthisis associated ciliopathy disease recessive mutations in TRAP1 in individuals with
genes using a DNA pooling and next generation CAKUT and VACTERL association. Kidney Int.
sequencing strategy. J Med Genet. 2011;48:105–16. 2014;85:1310–7.
177. Halbritter J, Porath JD, Diaz KA, Braun DA, Kohl S, 180. Gee HY, Ashraf S, Wan X, Vega-Warner V, Esteve-
Chaki M, Allen SJ, Soliman NA, Hildebrandt F, Rudd J, Lovric S, Fang H, Hurd TW, Sadowski CE,
Otto EA. Identification of 99 novel mutations in a Allen SJ, Otto EA, Korkmaz E, Washburn J, Levy S,
worldwide cohort of 1,056 patients with a Williams DS, Bakkaloglu SA, Zolotnitskaya A,
nephronophthisis-related ciliopathy. Hum Genet. Ozaltin F, Zhou W, Hildebrandt F. Mutations in
2013;132:865–84. EMP2 cause childhood-onset nephrotic syndrome.
178. Saisawat P, Tasic V, Vega-Warner V, Kehinde EO, Am J Hum Genet. 2014;94:884–90.
Gunther B, Airik R, Innis JW, Hoskins BE, 181. Malone AF, Phelan PJ, Hall G, Cetincelik U,
Hoefele J, Otto EA, Hildebrandt F. Identification of Homstad A, Alonso AS, Jiang R, Lindsey TB,
two novel CAKUT-causing genes by massively par- Wu G, Sparks MA, Smith SR, Webb NJ, Kalra PA,
allel exon resequencing of candidate genes in patients Adeyemo AA, Shaw AS, Conlon PJ, Jennette JC,
with unilateral renal agenesis. Kidney Int. 2012; Howell DN, Winn MP, Gbadegesin RA. Rare hered-
81:196–200. itary COL4A3/COL4A4 variants may be mistaken for
179. Saisawat P, Kohl S, Hilger AC, Hwang DY, Yung familial focal segmental glomerulosclerosis. Kidney
Gee H, Dworschak GC, Tasic V, Pennimpede T, Int. 2014;86:1253–9.
Translational Research Methods:
Renal Stem Cells 18
Kenji Osafune

Contents Clinical Application and Practical Use


of Renal Stem Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 557
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 526
Renal Stem Cell Research: Future Directions . . . . 558
Embryonic Renal Stem/Progenitor Cells . . . . . . . . . 527
Kidney Development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 527 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 559
Identification of Embryonic Renal
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 560
Stem/Progenitor Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 530
Embryonic Renal Stem/Progenitor Cells
Differentiated from Pluripotent Stem Cells . . . . . . 533
Pluripotent Stem Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 533
Mechanisms of Cues for Directed
Differentiation of Pluripotent Stem Cells . . . . . . . . . . . 534
Directed Differentiation of Mouse ESCs/iPSCs
into Embryonic Renal Stem/Progenitor Cells . . . . . . . 535
Directed Differentiation of Human ESCs/iPSCs
into Embryonic Renal Stem/Progenitor Cells . . . . . . . 540
Embryonic Renal Stem/Progenitor Cells
Reprogrammed from Adult Renal Cells . . . . . . . . . . . . . 545
Adult Renal Stem/Progenitor Cells . . . . . . . . . . . . . . . 545
Stem Cells of Extra-Renal Sources . . . . . . . . . . . . . . . . . . 545
Sources of Adult Renal Cells Responsible for
In Vivo Regeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 546
Adult Renal Stem/Progenitor Cells Expanded
Ex Vivo . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 547
Stem Cell Niche in Adult Kidney . . . . . . . . . . . . . . . . . . . 554
Cancer Stem Cells in Kidney . . . . . . . . . . . . . . . . . . . . . . 555
Cancer Stem Cells in Adult Renal Cell
Carcinoma (RCC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 555
Cancer Stem Cells in Wilms’ Tumor . . . . . . . . . . . . . . . . 556

K. Osafune (*)
Center for iPS Cell Research and Application (CiRA),
Kyoto University, Sakyo-ku, Kyoto, Japan
e-mail: osafu@cira.kyoto-u.ac.jp

# Springer-Verlag Berlin Heidelberg 2016 525


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_16
526 K. Osafune

Introduction two cell types specific for kidneys will be described


as one entity, “renal stem/progenitor cells” or “renal
The kidney is the main excretory organ in the progenitors,” in this chapter.
body and performs essential functions, such as With regard to usage for regenerative medicine,
the excretion of waste products from the blood, stem cells are largely classified into two major
the regulation of water volume, electrolyte bal- categories: tissue-specific or somatic stem cells
ance and pH levels, and the maintenance of blood and pluripotent stem cells (Fig. 1c, d). Tissue-
pressure, erythropoiesis and bone metabolism. In specific stem cells, such as hematopoietic stem
humans, these functions are carried out by the cells in bone marrow [9] and neural stem cells in
combined action of approximately two million nervous systems [10], have been identified in most
nephrons, which are the functional units of the organs or tissues of our body, including renal stem
kidney. Any pathologic process that leads to a cells. Tissue-specific stem cells generally show
loss of nephrons may eventually cause decreased relatively limited proliferation capability on culture
renal function and perturbs the physiological dishes and limited differentiation potential into a
homeostasis maintained by the kidney, which is restricted number of cell types constituting the
a process collectively termed chronic kidney dis- original organ or tissue, while pluripotent stem
ease (CKD) [1–3]. CKD gradually progress to end cells, such as embryonic stem cells (ESCs)
stage renal disease (ESRD). At present, ESRD [11–14] and induced pluripotent stem cells
patients have only two therapeutic options: renal (iPSCs) [15–17], have virtually unlimited replica-
transplantation and dialysis therapy. Renal trans- tive capacity on culture dishes and are theoretically
plantation is a radical treatment, but hampered by able to give rise to any cell type in the body. These
serious problems, including the shortage of donor stem cells have been increasingly used as sources
organs and chronic use of immunosuppressive for cell replacement therapy. Like the generation of
drugs. Dialysis therapy can substitute for a part other clinically useful cell types, vigorous efforts to
of renal function, but it eventually causes systemic differentiate renal lineage cells from ESCs/iPSCs
complications in ESRD patients [3–6]. The prev- have already been carried out [18–24].
alence of CKD is increasing worldwide, mainly Recently, there have been an increasing num-
due to the increased incidence of diabetes mellitus ber of reports that describe the successful genera-
and hypertension. Moreover, CKD is associated tion of the desired cell types by manipulating core
with elevated morbidity and mortality due to the transcriptional factors to convert fully-
increased risk of cardiovascular diseases, which differentiated cells to a pluripotent state or from
gives growing economic burden to health care one fully-differentiated cell type to another
systems [7]. Since CKD represents a gradual [25–27]. This experimental strategy, cellular
loss of nephron number, substantial efforts have reprogramming, has already been utilized to gen-
sought cell sources that can compensate the loss. erate renal progenitors [28].
One option might be the supplementation of renal To date, a growing body of evidence has
stem cells [8]. supported the idea that malignant tumors are initi-
Stem cells are defined as cells that have self- ated and maintained by a population of tumor cells
renewal capacity and can give rise to multiple that share similar biologic properties with normal
terminally-differentiated cell types constituting the stem cells [29, 30]. This cancer stem cell model is
body (Fig. 1a). Progenitor cells that differentiate based on the observation that tumors arise from
from stem cells are more developmentally commit- stem cells that show the ability to self-renew as
ted to some cell lineages than stem cells, but are well as give rise to differentiated tissue cells. Can-
nevertheless undifferentiated or immature com- cer stem cells are usually thought to occur by the
pared with fully-differentiated and matured cells transformation of normal embryonic or adult stem
(Fig. 1b). In fact, it is sometimes difficult to clearly cells in their own tissues. While most knowledge
distinguish progenitor cells from stem cells, and on cancer stem cells had initially come from leuke-
both cell types are often equated. Therefore, the mia stem cells that were prospectively isolated by
18 Translational Research Methods: Renal Stem Cells 527

a Stem cell b

Unlimited Progenitor cell


self-renewal
capacity

Limited
Limited differentiation
Multipotency
self-renewal potential
capacity

ESC
HSC

c d

Red blood Platelet White blood


cell cell Neuron Blood cell Hepatocyte

Fig. 1 Stem cell and progenitor cell. (a, b) Stem cells (a) stem cells (HSCs; c) differentiate into cell types constitut-
show unlimited self-renewal capacity and multipotent dif- ing their original tissue, while pluripotent stem cells, such
ferentiation potentials, while progenitor cells (b) exhibit as embryonic stem cells (ESCs; d) differentiate into any
more restricted proliferation and differentiation capability. cell type in the body
(c, d) Tissue-specific stem cells, such as hematopoietic

flow cytometry sorting and transplanted into immu- renal stem/progenitor cells by the directed differ-
nocompromised mice to examine their tumorigenic entiation of pluripotent stem cells or by the cellu-
potentials, similar techniques have recently been lar reprogramming of other cell types. Present
used to identify cancer stem cells from solid knowledge on cancer stem cells of kidney-related
tumors, such as breast carcinomas and brain tumors malignancies will also be described (Fig. 2).
[31, 32]. The elucidation of the biological proper-
ties of cancer stem cells would allow the develop-
ment of therapies that target the stem cell Embryonic Renal Stem/Progenitor
populations. However, due to differences between Cells
tumors derived from other organs, only a small
number of studies have focused on the identifica- Kidney Development
tion of cancer stem cells in kidney [33].
This chapter will summarize the current status To understand the entity of embryonic renal stem/
of research on the identification and isolation of progenitor cells, this section starts by briefly sum-
renal stem/progenitor cells from embryonic or marizing mammalian kidney development (Fig. 3;
adult kidneys and on the generation of embryonic [34–39]). See also ▶ Chap. 1, “Embryonic
528 K. Osafune

Embryo Cancer Adult


patient

Isolation
Isolation
Isolation

Transformation? Transformation?
Embryonic Adult
stem cell stem cell
Cancer
stem cell
Directed
differentiation Cellular
reprogramming

ESC/iPSC Fully differentiated


renal cell

Fig. 2 Renal stem cell. Three types of renal stem cells Adult renal stem cells can be isolated from adult kidneys
have been reported: embryonic renal stem cells, adult renal by various methods, but are not necessarily equivalent to
stem cells and renal cancer stem cells. Embryonic renal embryonic renal stem cells. Renal cancer stem cells are
stem cells can be isolated from embryonic kidney, or isolated from tumor tissues and may arise from the trans-
generated from pluripotent stem cells by directed differen- formation of embryonic or adult renal stem cell
tiation or from adult renal cells by cellular reprogramming. populations

Development of the Kidney” in this text. The high BMP4 induce lateral plate mesoderm, and
kidneys are derived from an early embryonic the antagonism of BMP signals is required for
germ layer, the intermediate mesoderm (IM); paraxial mesoderm differentiation [40].
[40–42], which is located between the lateral and In vertebrates, the IM successively develops
paraxial mesoderms and derived from the primi- three types of kidneys: the pronephros, mesoneph-
tive streak, which is the progenitor population for ros and metanephros. The three kidneys are similar
both the mesoderm and endoderm [43, 44]. A in that they consist of a basic functional unit, the
number of factors, such as Wnt, bone morphoge- nephron, although the number of nephrons differs
netic proteins (BMPs), fibroblast growth factors among the three. Mesonephros is the adult kidney
(FGFs), Nodal/activin and retinoic acid of fish and amphibians, while metanephros is the
(RA) signals, contribute to the cell fate commit- functional kidney of adult amniotes, reptiles, birds
ment of primitive streak or mesodermal lineage, and mammals. The mammalian adult kidney, the
including the IM [40, 45–50]. Especially, BMP metanephros, is formed by reciprocally inductive
signalling is the key morphogen regulating interactions between two embryonic precursor tis-
lateral-medial patterning of early mesoderm sues derived from the IM: the metanephric mesen-
development. Low BMP4 induces IM, while chyme (MM) and the ureteric bud (UB), a
18 Translational Research Methods: Renal Stem Cells 529

Nephron Podocyte
progenitor
(Six2, Sall1)
Proximal
tubule
Metanephric
mesenchyme Distal
Posterior (Hox11, Wt1, tubule
intermediate Six1, Eya1) Stromal
Primitive mesoderm progenitor
streak (Osr1, Eya1) (Foxd1, Pbx1) Interstitium
Fertilized (Brachyury,
egg Mixl1) ?
Mesangium

Vascular progenitor (Flk1)

Collecting
Anterior
duct
intermediate
mesoderm
Ureteric bud Lower urinary
(Osr1, Lhx1,
(Ret, Hoxb7, tract
Pax2)
Spry1)

Fig. 3 Developmental process and lineage commitment progenitor cells, nephron progenitors, stromal precursors
of kidney. Each developmental stage and its specific and ureteric bud cells, are highlighted by boxes
marker gene are shown. Three kinds of embryonic renal

derivative of the nephric (Wolffian) duct. The UB [56, 57], which is a chemotactic factor for
induces the MM to differentiate into epithelialized UB. GDNF drives the outgrowth of the UB from
nephrons and interstitium, while the MM directs within the Wolffian duct and the extension of the
the UB to elaborate the lower urinary tract from the UB toward the MM. The attracted UB in turn
collecting ducts through the renal pelvis and ureter secretes Wnt9b, which is the primary inducer for
to a part of the urinary bladder [51]. Recently, the MM [58] to induce the adjacent MM to con-
Taguchi et al. redefined the origin of the MM and dense around the UB tip, thereby forming the
elucidated the lineage commitment from the CM. Thereafter, Wnt4 secreted from the induced
nascent mesoderm through the IM into either the CM epithelializes the CM itself in an autocrine
MM or UB [52]. They demonstrated that the MM is fashion [59]. During the process of mesenchymal-
generated from the posterior IM, which is distinct epithelial transition (MET), the CM cells sequen-
from the anterior UB precursor IM population. tially go through specific developmental stages,
A portion of MM cells condenses around the tips such as renal vesicles, pre-tubular aggregates, and
of the UB and differentiates into structures termed C- and S-shaped bodies, to eventually differenti-
cap mesenchyme (CM). CM cells self-renew at the ate into all nephron segments ranging from glo-
UB tips and give rise to various cell types comprising merular podocytes to distal renal tubules.
the nephron. Thus, the CM has been considered Among the key molecules involved in the pro-
multipotent nephron stem/progenitor cells, which cess of kidney development, a homeodomain
can self-renew and differentiate into different types transcriptional factor, Six2, which serves as the
of nephron epithelia [53, 54]. Additionally, CM cells most specific marker of CM cells, has been
express a unique combination of transcription factors, shown to play a central role in the regulation of
such as the Hox11 paralogs, Osr1, Pax2, Eya1, Wt1, renal progenitor CM cells, particularly on the
Six2, Sall1 and Cited1, before UB induction [55]. ability of CM cells to continuously self-renew
The MM produces the secreted neurotrophin, throughout nephrogenesis [53, 54]. It was
glial cell line-derived neurotrophic factor (GDNF; reported that FGF9 and FGF20 are required for
530 K. Osafune

the formation and maintenance of the CM pro- stages, but thereafter a strict nephron and stromal
genitors, respectively [60]. MM survival in vitro lineage boundary are formed between the nephron
also depends on FGF2 and BMP7 [61]. Urbach and stromal progenitor compartments.
et al. have recently demonstrated that a Despite the evidence described above, the origin
RNA-binding protein, Lin28, sustains early renal of renal stromal cells remains unknown. A fate
progenitors by preventing the final wave of mapping study by Guillaume et al. demonstrated
nephrogenesis during kidney development in that large populations of renal stromal cells origi-
mice [62]. Interestingly, the overexpression of nate from a different part of mesoderm, the paraxial
Lin28 markedly expands nephrogenic progeni- mesoderm, which gives rise to mesenchymal tis-
tors, resulting in a pathology highly reminiscent sues, such as bones, skeletal muscle and tendon
of Wilms’ tumor, a pediatric kidney cancer that [74]. Moreover, some studies have indicated that
evolves from the failure of terminal differentiation an embryonic ectodermal tissue, the neural crest,
of the embryonic kidney. may contribute to renal stroma [75–79]. In addition
Upon completion of the kidney development, to stromal progenitors, embryonic kidneys have
transcription factors expressed in the CM, includ- been shown to harbor resident Flk1(+) vascular
ing Six2, are no longer expressed and the embry- progenitors, which give rise to renal vessels
onic nephron progenitors disappear, which leads to [80–82], although cross-species transplantation
no formation of new nephrons. Embryonic nephron studies have suggested that extrarenal cells also
progenitors do not exist in adult mice even after contribute to the glomerular vasculature [83]. There-
acute kidney injury (AKI) in ischemia-reperfusion fore, details of the developmental origin of stroma
(I/R) models, which was elegantly demonstrated by and vasculatures in kidney remain to be elucidated.
a lineage tracing study for Six2 [63]. In contrast to Most knowledge on kidney development has
mammals, which lose the renal progenitor pool been obtained from genetic approaches on experi-
around birth, a self-renewing nephron stem cell mental animal models (see also ▶ Chap. 15, Trans-
population has been shown to exist in the adult lational Research Methods: The Value of Animal
kidneys of lower organisms, such as zebrafish and Models in Renal Research in this text), including
drosophila, and to form new nephrons throughout fish, frog, chick and mice [34–39]. Previous works
adult life, contributing to regenerating nephrons de have also examined embryonic kidneys in human
novo after injury [64, 65]. and nonhuman primates, such as rhesus monkey
In addition to the Six2-expressing CM nephron [84–86]. Humans, nonhuman primates and mice
progenitors that give rise to several epithelial cell share a number of molecules and corresponding
types constituting nephrons, such as glomerular spatial expression patterns that act as developmen-
podocytes and renal tubular epithelia, the devel- tal markers, although major differences have
oping kidney also contains progenitor cell types of been found in the temporal patterns of kidney
other lineages. These include Foxd1(+) stromal development. Nephrogenesis starts in human and
progenitors in the MM, which differentiate into nonhuman primates at the end of the first trimester
renal interstitial cells, mesangial cells and vascu- and continues throughout the mid-third trimester
lar smooth muscle cells and pericytes in adult with a term of 165  10 days, while mouse
kidney [66–72]. Kobayashi et al. have recently nephrogenesis begins in mid-gestation and com-
demonstrated by performing lineage tracing pletes postnatally with a term of 20 days [85, 87].
experiments that the Foxd1-expressing cortical
stroma represents a distinct multipotent self-
renewing progenitor population that gives rise to Identification of Embryonic Renal
the adult renal cell types described above through- Stem/Progenitor Cells
out kidney organogenesis [73]. Interestingly, it
was demonstrated that a small subset of As described above, the metanephric kidney con-
Foxd1(+) stromal cells contributes to Six2(+) tains several stem/progenitor cell populations
nephron progenitors at early developmental required for the formation of a fully functional
18 Translational Research Methods: Renal Stem Cells 531

kidney. This property has also been demonstrated kidney and cannot be used to expand undiffer-
by the transplantation of whole embryonic kid- entiated nephron progenitors.
ney rudiments into extrarenal space of host ani- Other approaches to isolate progenitor cells
mals. Hammerman et al. [88, 89] and Dekel include the formation of spheres based on the
et al. [90, 91] demonstrated the formation of ability of progenitor cells grown in
vascularized renal tissues from transplanted low-attachment conditions. Lusis et al.
metanephroi inside the rodent body. Notably, established nephrospheres from murine embry-
when mouse, rat, porcine or even human early- onic kidneys, which, although robustly propa-
gestational metanephroi are transplanted into the gated in vitro, displayed limited nephrogenic
kidney subcapsule or the abdominal cavity of capacity [96]. To date, expansion culture of these
immunodeficient mice, they survive, grow, murine renal progenitors in vitro has not been
undergo complete nephrogenesis and form func- successful, underscoring the major role of the
tional organs that are able to produce diluted in vivo stem cell niche in supporting the growth
urine by integrating with the host vasculature of these cells. Therefore, the detailed mechanisms
and forming vascularized glomeruli [91]. regulating progenitor cell proliferation by the
In addition to the transplantation of whole corresponding niche need to be elucidated.
metanephric rudiments, Kim et al. reported that Methods to prospectively isolate fetal kidney
transplantation of a heterogeneous population of progenitor cells using surrogate surface markers
dissociated embryonic day (E) 14.5 and E17.5 rat of intra-cellular renal transcription factor expres-
fetal kidney cells under the renal capsule of a 5/6 sion have also been examined to supply an unlim-
nephrectomy kidney injury model resulted in the ited source of nephron progenitors, which in turn
formation of renal structures and improved renal can be used to regenerate damaged epithelial cells
functions [92]. However, the research group later within the nephron by combining culture condi-
demonstrated that rat fetal kidney cells experi- tions for expanding nephron progenitors.
enced proliferation arrest and lost in vivo regen- By using a previously-reported isolation
erative potential after repeated passages of in vitro method for the putative progenitor population in
expansion culture [93]. adult kidney [97], Lazzeri et al. isolated a similar
The selection or isolation of specific lineage of CD24(+)CD133(+) cell population from human
progenitor cells included in embryonic kidneys fetal kidney [98]. They reported that the CD24
have also been performed either by sorting based (+)CD133(+) population represents 35–50 % of
on the expression of specific markers or by selec- embryonic kidney cells, but gradually decreases in
tive in vitro assays. By creating a clonogenic proportion and becomes restricted to the urinary
assay system which uses stimulation of Wnt4, an pole of the Bowman’s capsule. This population
epithelializing factor for MM in embryonic devel- may possibly persist into adulthood as progeni-
opment, Osafune et al. previously demonstrated tors. Importantly, when isolated and injected into
that the MM contains multipotent nephron pro- immunodeficient mice with AKI by glycerol-
genitors that can give rise to several kinds of induced rhabdomyolysis, CD24(+)CD133(+)
epithelial cells, such as glomerular podocytes cells regenerated different portions of the nephron,
and the epithelia of the proximal and distal renal reduced tissue necrosis and fibrosis, and signifi-
tubules and Henle’s loop [94]. The progenitor cantly improved renal function without tumor for-
cells are contained only in the cell population mation [98]. The authors concluded that CD24(+)
that strongly expresses Sall1, a zinc finger tran- CD133(+) cells represent a subset of multipotent
scription factor that is essential for kidney devel- embryonic progenitors that persist in human kid-
opment [95], and can reconstitute three- neys from the early stages of nephrogenesis.
dimensional kidney structures in an organ culture A group headed by Benjamin Dekel employed
setting that contain glomeruli- and tubule-like a unique stepwise strategy in order to identify cell
components (Fig. 4). This clonogenic assay, how- surface markers for isolation of human embryonic
ever, has not yet been applied to human fetal renal stem/progenitor cells [27]. To identify
532 K. Osafune

a b
Day3 Day20 PNA LTL E-cad LTL

c d e
Day3 Day7 Wt1 LTL
g

t t
g

f Renal Colony Renal Glomerulus


progenitors formation progenitors
Proximal tubule

Henle`s loop

Distal tubule

Fig. 4 Multipotent nephron progenitors in mouse meta- organ culture setting. In contrast, metanephric cells weakly
nephric kidney (Ref. [94]). (a) In vitro colony formation expressing Sall1 (Sall1low) or not expressing Sall1 (Sall1)
from a single progenitor cell within metanephric mesen- disappear without differentiation (data not shown). (d)
chyme on NIH3T3 feeder cells expressing Wnt4. (b) A Hematoxylin-eosin staining of sections of Sall1high aggre-
single progenitor cell is seen to divide and differentiate into gates at day 10 of the organ culture shown in (c). Tubule-
two renal lineages: (1) podocytes (red: peanut agglutinin; (t) and glomerulus-like structures (g) are seen. (e) Double
PNA) and proximal tubular epithelia (green: Lotus staining of a renal tissues section formed from Sall1high
Tetragonobulus lectin; LTL) in the left panel, and (2) prox- cells with the podocyte marker Wt1 (red) and the proximal
imal (green: LTL) and distal tubular epithelia (red: tubule marker LTL (green). Bars: (a, b) 50 μm, (c) 500 μm,
E-cadherin) in the right panel. (c) A cell population (d, e) 25 μm. (f) Schematic drawing of multipotent nephron
strongly expressing Sall1 (Sall1high) and containing neph- progenitors in a colony-forming assay. Several kinds of
ron progenitors differentiates into renal structures in an epithelia constituting nephron arise from the progenitors

candidate markers that are highly expressed in (e.g., SIX2, OSR1 and PAX2) and surface markers
renal progenitor-rich tissues compared with dif- (NCAM1, PSA-NCAM, FZD7, FZD2, DLK1,
ferentiated renal tissues, the group compared the ACVRIIb and NTRK2) [100]. The group narrowed
global gene expressions of human embryonic kid- the candidate surface molecules by observing
ney and Wilms’ tumor to human adult kidney by exclusive expression in undifferentiated cortical
oligonucleotide microarray [99]. Since Wilms’ areas of embryonic kidneys and the possession of
tumor specimens frequently harbor cells that have characteristic traits of tissue-specific stem cells
differentiated into renal tubular epithelia or stroma, determined by a series of assays, such as over-
in addition to uninduced metanephric blastema, the expression of renal progenitor genes, self-renewal
group serially propagated Wilms’ tumor xenografts capacity, colony formation capability and renal dif-
in mice to purify blastema tissues at the expense of ferentiation potential [101]. They eventually iden-
differentiated elements for the analysis. This anal- tified NCAM1 (CD56) as a potential marker for
ysis uncovered a large number of renal “stemness” human embryonic renal progenitors that fulfils all
genes, including nephron progenitor markers these criteria [100, 102].
18 Translational Research Methods: Renal Stem Cells 533

NCAM1(+) cells isolated from human embry- can be broken down into various types, including
onic kidney cells and cultured in serum-free the aforementioned ESCs [11–14] and iPSCs
medium showed significant enrichment of nephron [15–17], embryonal carcinoma (EC) cells [104],
progenitor genes, such as SIX2, OSR1, PAX2, embryonic germ (EG) cells [105, 106] and
SALL1, CITED1 and WT1, compared to NCAM1 multipotent germ stem (mGS) cells [107]. EC
() cells [102]. The renal differentiation potential of cells are the first derived pluripotent stem cells
NCAM1(+) cells was demonstrated using a chorio- and come from teratocarcinomas, a type of tumor
allantoic membrane (CAM) transplantation model, that consists of three germ layer components
in which cells are grafted onto a highly vascularized [104]. EG cells are generated from primordial
niche, rich in developmental cues [103]. The model germ cells, which are embryonic cells that give
showed that NCAM1(+) cells generated well- rise to germ cells, sperm and oocytes [105,
developed epithelial tubular structures expressing 106]. Finally, mGS cells occur as a rare cell popu-
markers for the proximal tubule, distal tubule and lation in the culture of sperm stem cells from mouse
loop of Henle. Harari-Steinberg et al. evaluated the testis [107]. However, when it comes to directed
regenerative potential of the NCAM1(+) population differentiation strategies for regenerative medicine,
in 5/6 nephrectomy CKD models. They demon- ESCs and iPSCs receive the most attention.
strated that NCAM1(+) nephron progenitors from ESCs are pluripotent stem cells derived from
human embryos engrafted and integrated into dis- the inner cell mass of fertilized eggs in mammals
eased murine kidneys after transplantation, and that ([11–14]; Fig. 5a–c), while iPSCs are generated
consecutive intra-parenchymal administration from somatic cells by the transduction of
halted the progression of renal disease, as reprogramming factors, such as most famously
manifested by the increased creatinine clearance the combination of Oct3/4, Sox2, Klf4 and
throughout 12 weeks of treatment [102]. Therefore, c-Myc, and closely resemble ESCs ([15–17];
it was concluded that NCAM1(+) human nephron Fig. 5d–f). The successful derivation of human
progenitors are a pool of mitotically competent yet ESCs has stimulated regenerative medicine strat-
strongly nephron-generating-biased cells. egies that aim to recover dysfunctional organs by
Although the embryonic kidney progenitors transplanting organ-specific cell types generated
represent a promising cell source for renal regen- from ESCs in vitro. iPSCs are appealing because
eration, the ethical problems arising from the use they offer the same attractive features of ESCs
of human embryos as well as the allogeneic nature while potentially overcoming two drawbacks
of the human cells limit the use of these cells. associated with the use of human ESCs: ethical
Therefore, efforts to identify a nephrogenic cell problems related to the use of human fertilized
population in adult kidneys or to generate the eggs and the potential for immune rejection after
embryonic renal progenitors from autologous transplantation due to antigenic differences
stem cells, such as iPSCs, are required to acceler- between the ESCs and patients.
ate research towards the clinical application of Although iPSCs were originally derived from
transplantation therapy for kidney diseases. fibroblasts, the stem cell lines have since been
generated from terminally-differentiated somatic
cells, such as adult liver and stomach cells [108],
Embryonic Renal Stem/Progenitor keratinocytes [109], hematopoietic cells [110,
Cells Differentiated from Pluripotent 111] and pancreatic β cells [112]. In fact, it
Stem Cells appears that reprogramming potential exists in
almost any cell type, including the cells that con-
Pluripotent Stem Cells stitute adult kidneys [113–116], although the effi-
ciency of the iPSC derivation differs among
Pluripotent stem cells are characterized by their original cell types.
high proliferation potential and multipotent differ- Despite the excitement and potential of iPSCs
entiation ability into any cell type of the body. They for medicine, they have the risk of cancer
534 K. Osafune

a b c

Inner cell mass

Fertilized egg ESC

d e f
Oct4
Somatic Sox2
cells Klf4
c-Myc

iPSC

Fig. 5 Embryonic stem cell (ESC) and induced pluripo- somatic cells by introducing reprogramming transcrip-
tent stem cell (iPSC). (a) ESCs are generated from the inner tional factors. (e) Mouse iPSCs (178B5). (f) Human
cell mass of fertilized eggs. (b) Mouse ESCs (D3). (c) iPSCs (201B7). Bars: 100 μm
Human ESCs (khES-3). (d) iPSCs are derived from

development. iPSCs were originally derived using and vascular cells, hematopoietic cells, pancreatic
genome-integrating vectors, such as retroviral and β cell-like cells and hepatocyte-like cells
lentiviral vectors, to introduce the reprogramming [18–21]. However, methods for the directed dif-
factors into somatic cells [15–17]. To solve the ferentiation of ESCs/iPSCs into renal lineage cells
mutagenesis problem, integration-free and safer have not been fully established. Most reports have
methods to generate iPSCs have been reported, described the differentiation of mouse ESCs into
including (1) non-genome integrating vectors, renal lineages, but few have examined human
such as adenoviruses, Sendai viruses, plasmids ESCs or iPSCs. However, several research groups
and episomal plasmid vectors, (2) genetic manip- have more recently published methods to induce
ulation, such as transposons and Cre-loxP sys- human ESCs/iPSCs into kidney lineages [22–24].
tems, (3) the introduction of mRNA and proteins
of reprogramming transcription factors and
(4) treatment with chemical compounds [19, 20]. Mechanisms of Cues for Directed
For clinical application, xeno-free and defined Differentiation of Pluripotent Stem
culture conditions have been developed to main- Cells
tain and differentiate ESC/iPSC cultures
[117]. Clinical trials using human ESC-derived There are two common differentiation formats of
somatic cell types have been already performed pluripotent stem cells, EB formation and mono-
to treat spinal cord injury with oligodendrocytes layer or adherent cultures. Conventional differen-
[118] and retinal degenerative disorders with ret- tiation protocols developed by Doetschman
inal pigmented epithelial cells [119]. Recently, the et al. have used multicellular aggregations, termed
first clinical trial using human iPSC-derived reti- embryoid bodies (EBs), which constitute three-
nal pigmented epithelial sheets was performed for dimensional cultures mimicking developing
a patient suffering from age-related macular embryos, although the three body axes of the
degeneration [120]. embryo, i.e., anterior-posterior, dorsal-ventral
Already, many cell types have been generated and left-right, are randomly formed [121]. In
from ESCs/iPSCs, including neural cells, cardiac these systems, complex intercellular signals
18 Translational Research Methods: Renal Stem Cells 535

within three-dimensional structures that resemble that it can help identify small molecule inducers
those in developing embryos can be reproduced to for renal lineage differentiation, which will also
generate target cell types from ESCs/iPSCs. On help elucidate the mechanisms underlying kidney
the other hand, Nishikawa et al. established development and differentiation.
two-dimensional monolayer differentiation of Substantial differences exist in the differentia-
mouse ESCs into vascular endothelia tion potential among human ESC/iPSC lines,
[122]. Here, exogenous inducing factors can uni- although only marginal differences have been
formly affect almost all cells to result in a higher observed in the undifferentiated state
induction rate of the target cell types than EB [131–133]. Some human ESC/iPSC lines even
formation does. Two-dimensional cultures also have a propensity to differentiate into certain lin-
have the advantage of simplicity and are easy to eages or cell types. Moreover, it has been shown
use for mechanistic analyses. Therefore, an that iPSCs harbor residual DNA methylation sig-
increasing number of studies for the directed dif- natures characteristic of their original somatic
ferentiation of ESCs/iPSCs have adopted mono- cells. Such “epigenetic memory” of the parental
layer culture formats [123, 124]. tissue may predispose the differentiation into lin-
Regarding the inducers used to direct the dif- eages related to the parental cells, a property
ferentiation of ESCs/iPSCs into specific lineages observed in multiple iPSC lines including those
by mimicking the signals involved in normal from mouse blood cells [134, 135] and human
development, combinatorial treatments have pancreatic β cells [112]. These findings indicate
been developed. These treatments combine dif- that iPSCs derived from renal lineage cells might
fusible factors (growth factors, cytokines and favour differentiation into kidney lineages.
chemical compounds), cell-cell interactions Indeed, human iPSCs have been derived from
(co-culture with cell lines) and positional cues mesangial cells [113] and renal tubular cells
(extracellular matrix) that ultimately regulate the dropped into urine [114, 115]. In addition,
gene expression, which together confer a specific mouse iPSCs were also generated from renal
cellular identity and function. Genetic and epige- tubular epithelial cells [116]. These findings
netic manipulations using the overexpression of underscore the importance of using suitable cell
cDNA, small interfering RNA (siRNA), short lines for renal lineage differentiation.
hairpin RNA (shRNA) and microRNA (miRNA)
have also been utilized to induce specific cell fates
[19, 20, 22–24]. Directed Differentiation of Mouse
Some reports have demonstrated that high- ESCs/iPSCs into Embryonic Renal Stem/
throughput screening (HTS) of low-molecular- Progenitor Cells
weight chemical compounds, including synthetic
and natural compounds, can be used to identify By recapitulating the signals involved in the dif-
small molecules that have the potential to induce ferentiation steps during normal embryonic devel-
the differentiation of ESCs/iPSCs into specific opment, step-by-step protocols have been
lineages, such as cardiomyocytes, pancreatic pro- explored to directly differentiate ESCs/PSCs into
genitor cells, definitive endoderm and pancreatic the specific cell types included in adult organs,
endocrine cells [125–129]. Araoka et al. has used such as pancreas and liver [123, 124]. For exam-
this approach to investigate directed differentia- ple, pancreatic β cells have been induced by dif-
tion into kidney lineages and recently identified ferentiating ESCs/iPSCs into the definitive
two retinoids, AM580 and TTNPB, that effi- endoderm, then the foregut endoderm, followed
ciently induce human ESCs/iPSCs into a by pancreatic progenitors and eventually into
nephrogenic IM [130]. These IM cells induced insulin-producing cells [123]. Most studies
with chemicals give rise to renal lineage cells aiming to generate renal cells from ESCs/iPSCs
that have the developmental potential to form have so far adopted a similar strategy of recapitu-
renal tubular structures. The appeal of HTS is lating kidney development [22–24].
536 K. Osafune

The in vitro injection of ESCs into immunode- ROSA26 locus (LacZ/T/GFP) for cell selection
ficient mice can produce a teratoma, which is a and lineage tracing [141]. They further optimized
tumor that contains the derivatives of all three the differentiation conditions using only activin A
embryonic germ layers: the ectoderm, mesoderm treatment for renal progenitors and used the
and endoderm. Yamamoto et al. reported that glo- expression of Cadherin11, Wt1, Pax2 and Wnt4
merulus- and renal tubule-like structures can be as markers of differentiation. The renal progeni-
formed in teratomas generated from mouse ESCs, tors, isolated via flow cytometry, were injected
indicating that mouse ESCs have the developmen- into developing live-newborn mouse kidneys
tal potential to differentiate into renal cells and stably integrated into the proximal tubules
[136]. Steenhard et al. demonstrated that undiffer- for 7 months without teratoma formation. It was
entiated mouse ESCs incorporate into the concluded that the defined differentiation of
renal tubules and glomeruli of host kidneys by mouse ESCs via EB formation and with activin
responding to developmental signals within A treatment generated proximal renal tubular pro-
the host kidney microenvironment when genitors that may have adequate safety for regen-
microinjected into E12.5-13.5 mouse kidneys in erative therapies.
organ culture settings [137]. Bruce et al. induced mouse EBs into the meso-
Moreover, substantial efforts have been made derm by culturing with serum or serum-free cul-
to selectively or specifically induce renal progen- tures supplemented with BMP4 and detected the
itors and fully-differentiated renal cell types gene expression of kidney lineage markers by
in vitro from mouse ESCs (Table 1). Kramer real-time RT-PCR analysis [142]. The generation
et al. reported that the differentiation cultures of of urogenital Pax2(+) cells was demonstrated by
mouse ESCs with EB formation in the presence of immunostaining and flow cytometry using
serum alone produced cells expressing renal line- reporter mouse ESC lines containing either a
age markers [140]. modified Pax2 promoter-lacZ or bacterial artificial
Kobayashi et al. generated mouse ESC lines chromosome (BAC)-GFP transgene. This
that stably express Wnt4, a renal-epithelializing approach enabled direct monitoring of renal rather
factor [138]. They induced Wnt4-transformed than the mid-hindbrain Pax2 expression. In addi-
mouse ESCs into renal tubular cells that expressed tion, three BMPs, BMP2, BMP4 and BMP7, were
Aquaporin-2 (AQP2) and three-dimensional tubu- compared in EB systems to evaluate the specifi-
lar structures in vitro by EB formation and treat- cation of the mesoderm. It was found that a low
ment with hepatocyte growth factor (HGF) and concentration of BMP4 activates ventral (blood)
activin A in addition to Wnt4. The transplantation and intermediate (kidney) mesoderm gene expres-
of the treated EBs into mice renal cortices also sion, while BMP2 and BMP7 promote only kid-
resulted in the formation of teratomas, some of ney lineage.
which contained AQP2(+) tubular structures. Nakane et al. generated mouse ESC lines using
Kim and Dressler reported that EBs formed a tetracycline-inducible gene expression of Pax2
from mouse ESCs can be induced into the IM and examined the effects of Pax2 overexpression
in vitro by a combinatorial treatment with on renal lineage differentiation, since Pax2
nephrogenic growth factors, such as RA, activin expression was shown to be sufficient to induce
A and BMP7 [139]. These EBs developed renal the ectopic formation of nephric structures in
tubule-like structures by co-culturing with embry- chick [143]. They found that Pax2 overexpression
onic spinal cord, which is a classical inducer for the in undifferentiated ESCs failed to induce the
MM, and the induced cells were integrated into the expression of renal lineage markers, while its
proximal tubules when microinjected into E12.5 overexpression in EBs led to the expression of
mouse kidney rudiments in organ culture settings. two renal lineage markers, integrin α8 and Aqp1,
Vigneau et al. generated mouse ESC lines with suggesting that along with Pax2, additional fac-
green fluorescent protein (GFP) knocked into the tors induced in EBs are required for renal lineage
Brachyury (T) locus as well as lacZ into the differentiation.
18

Table 1 Summary of reports on the directed differentiation of mouse ESCs/iPSCs into kidney lineages
Primary
Author Cell line Format Medium Inducer target cell Functional examination References
Kobayashi T BL6 EB IMEM/15 % Wnt4 overexpression, 10 ng/mL HGF NP (MM) Tubule formation by three- [138]
et al. 2005 FCS and 10 ng/mL activin A for 5–20 days dimensional culture in Matrigel.
Formation of teratoma that contain
tubules by injection into renal
cortex of immunodeficient mice
Kim D R26 EB DMEM/10 % 0.1 μM RA, 10 ng/mL activin A, NP (MM) Tubule formation by co-culture [139]
et al. 2005 FCS 50 ng/mL BMP4 or BMP7 for 5–7 with embryonic mouse spinal cord.
days Integration into proximal tubules
by injection into mouse
metanephric rudiments
Kramer J D3 EB DMEM/20 % Spontaneous differentiation: 20 % Renal Formation of ring-like glomerular [140]
et al. 2006 FCS FCS for up to 40 days. Induction using lineage structures consisting of podocyte-
recombinant proteins: 100 ng/mL cell like cells in EB outgrowths
FGF2 for 5 days followed by
Translational Research Methods: Renal Stem Cells

100 ng/mL FGF2 in combination with


50 ng/mL LIF for an additional 7 days
Vigneau C E14.1,129/ EB 50 % 2–100 ng/mL activin A for 1–8 days NP (MM) Integration into nephrogenic zone [141]
et al. 2007 Ola Neurobasal/50 by injection into mouse
% DMEM/F12 metanephric rudiments.
Integration into proximal tubule by
injection into live newborn mice
kidneys
Bruce SJ W9.5 EB DMEM/10 % Spontaneous differentiation: 10 % Renal NA [142]
et al. 2007 FCS or serum- FCS or 2 ng/mL BMP4 and 1 U/mL lineage
free chemically LIF for 19 days. Induction using cell
defined media recombinant proteins: 2 ng/mL BMP4
and 1 U/mL LIF for 4 days followed
by 7 days with BMP2, 4 or 7
Nakane A MGZRTcH2 EB GMEM/10 % 15 % FCS for 5 days followed by Pax2 Renal NA [143]
et al. 2009 cell FCS overexpression lineage
(MG1.19) cell
Morizane R EB3, EB DMEM/10 % 10 ng/mL activin A for 4 days Renal NA [144]
et al. 2009 MEF-Nanog- FCS followed by 150 ng/mL GDNF and lineage
38C (iPSC) 15 ng/mL BMP7 for an additional cell
537

14 days
(continued)
538

Table 1 (continued)
Primary
Author Cell line Format Medium Inducer target cell Functional examination References
Ren X R1 EB DMEM/15 % 10 ng/mL activin A and 0.1 μM RA Renal NA [145]
et al. 2010 FCS for 8 days followed by 10 days with lineage
conditioned medium from E13.5 mice cell
ureteric bud
Mae S 129/sv Monolayer DMEM/F12/2 1 μM JAK inhibitor, 5 μM LY294002, IM NA [146]
et al. 2010 % FCS 1 μM CCG1423 and 0.1 μM RA for
8 days
Nishikawa M J1 Monolayer DMEM/10 % Stage 1: 10 ng/mL activin A NP NA [147]
et al. 2012 FCS (A) during days 0–2, A plus 50 ng/mL (MM) and
BMP4 during days 2–4, A4 plus UB
10 mM lithium chloride (L) during
days 4–6; Stage 2: A4L plus 0.1 μM
RA during days 6–8; Stage 3:
conditioned media from UB or MM
Oeda S D3 Monolayer ESF medium/ 10 ng/mL activin A and 0.1 μM RA IM Integration into renal tubules by [148]
et al. 2013 0.5 mg/mL BSA for 6 days injection into mouse metanephric
rudiments
Taguchi A E14.1 (Osr1- EB 75 % IMDM/25 Stage 1: 0.5–1 ng/mL activin A for NP Formation of glomeruli and renal [52]
et al. 2014 GFP), % Ham’s F12 1 day; Stage 2: 1 ng/mL BMP4, 10 μM tubules by co-culture with
EB3-DsRed) with N2 and CHIR99021 for 1.5 days; Stage 3: embryonic mice spinal cord
B27 1 ng/mL BMP4, 10 μM CHIR99021
supplements for 1 day; Stage 4: 10 ng/mL
and 0.05 % BSA activin A, 3 ng/mL BMP4, 3 μM
CHIR99021, 0.1 μM RA, 10 μM
Y27632 for 1 day; Stage 5: 1 μM
CHIR99021, 5 ng/mL FGF9, 10 μM
Y27632 for 2 days
EB embryoid body, NP nephron progenitor, MM metanephric mesenchyme, NA not applicable, IM intermediate mesoderm, UB ureteric bud
K. Osafune
18 Translational Research Methods: Renal Stem Cells 539

Ren et al. used the factors secreted from plus BMP4 during days 2–4 and A4 plus lithium
embryonic renal cells to differentiate mouse (L), a surrogate for Wnt signalling, during days
ESCs into renal precursor cells [145]. They first 4–6. For Stage 2, RA was added to A4L during
generated mesodermal populations expressing days 6–8. For Stage 3, conditioned media derived
Pax2 and Brachyury by treating mice EB with from a mouse UB cell line, CMUB-1 [152], and a
activin A and RA. The mesodermal cells were mouse undifferentiated MM cell line, MK3 [153],
then treated with conditioned medium collected were used to induce MM and UB, respectively.
from the cultures of UB cells from E13.5 mice The group also found a gradual increase in the cell
embryos, which resulted in the generation of dif- population expressing CD24 from Stages 1–3 and
ferentiated cells expressing marker genes charac- accordingly proposed CD24 as a cell surface
teristic of the initiation of nephrogenesis (Wt1 and marker that can be used for the purification of
Pax2) and terminally-differentiated renal cell IM and MM cells in the culture when combined
types (POD1 and E-cadherin). with other surface markers.
Mae et al. reported the differentiation of mouse Oeda et al. established a differentiation proto-
ESCs into IM cells in monolayer culture with col from mouse ESCs into IM cells by treatment
combinatorial treatments of small molecules with activin A and RA under defined, serum-free,
[146]. They established the requirement for a adherent, monolayer culture conditions
combination of four small molecules: (1) Janus- [148]. These mesoderm cells integrated into lam-
associated tyrosine kinase (JAK) inhibitor 1, an inin(+) tubular cells and Pax2(+) renal cells when
inducer of differentiation from ESCs into epi- co-cultured with embryonic kidneys in organ cul-
blasts by inhibiting leukemia inhibitory factor tures. The group also demonstrated that signals
(LIF) signals, to maintain an undifferentiated initiated through RA receptors (RARs), but not
state of mouse ESCs [149]; (2) LY294002, an retinoid X receptors (RXRs), are primary in the
inhibitor of phosphatidylinositol 3-kinase (PI3K) differentiation of the IM.
to promote differentiation into the mesendoderm Viñas et al. used microarray analysis to show
[150]; (3) CCG1423, an inhibitor of RhoA tran- that miRNA let-7e was the most upregulated
scriptional signalling to promote the epithelial- miRNA in mouse EBs treated with activin A and
mesenchymal transition (EMT) during gastrula- RA [154]. Furthermore, the expression of several
tion by breaking down the basement membrane marker genes for IM and MM, such as Pax2, Wt1,
[151]; and (4) RA, to upregulate the expression of Wnt9b, Wnt4 and Notch2, had increased. The
IM marker genes in mouse ESC differentiation group demonstrated that miRNA let-7e is
when combined with activin A treatment involved in renal lineage differentiation by stim-
[139]. The combination of JAK inhibitor ulating the Wnt pathway, which suppresses the
1, LY294002 and CCG1423 resulted in the gen- phosphorylation of glycogen synthase kinase
eration of BMP7(+) mesoderm cells, and treat- (GSK)3β and subsequently stabilizes β-catenin
ment with all four factors efficiently induced by inhibiting protein kinase C (PKC) β.
mouse ESCs into IM cells. Morizane et al. examined the renal lineage
Nishikawa et al. reported a stepwise renal lin- differentiation from both mouse ESCs and iPSCs
eage differentiation from mouse ESCs by tracing by combinatorial treatment of EBs with activin A,
in vivo development, which consisted of three BMP7 and GDNF [144]. Using the expression
stages performed in monolayer cultures: the analysis of several renal marker genes, including
induction of a mesoderm that expresses Pax2, Wt1 and Kidney-specific protein (Ksp),
Brachyury (Stage 1), an IM that expresses Pax2 both mouse ESCs and iPSCs could be differenti-
and Lim1 (Stage 2) and MM cells that express ated into renal lineages, although differences in
Wt1, Gdnf and Cadherin 11 along with UB cells the gene expression levels between the two stem
that express Hoxb7, Ret and Wnt11 (Stage 3; cells were found.
[147]). For Stage 1, three factors were sequen- In some reports using mouse ESCs or iPSCs,
tially added: activin A (A) during days 0–2, A the induced renal lineage cells were shown to
540 K. Osafune

form tubule-like structures in organ cultures [139] lineages [13, 155]. Schuldiner et al. examined
or to be integrated into developing renal tubules of the effects of eight growth factors (nerve growth
mouse kidneys [141]. Notably, Taguchi factor (NGF), HGF, epidermal growth factor
et al. recently established a multistep differentia- (EGF), RA, bFGF, BMP4, transforming growth
tion protocol from mouse ESCs into nephron pro- factor (TGF)-β1 and activin A) on the differenti-
genitors that could form glomerulus-like ation of human ESCs, showing that six (RA, NGF,
structures in addition to renal tubules [52]. As bFGF, activin A, HGF and EGF) could induce the
described above, the group demonstrated that expression of two kidney markers, RENIN and
nephron progenitors are derived from the poste- KALLIKREIN, according to RT-PCR
rior mesoderm and identified the optimal combi- analysis [156].
nations of inducers that promote differentiation That report was followed by studies on selec-
from the posterior mesoderm into nephron pro- tive differentiation methods for kidney lineages
genitors. They established a stepwise differentia- from human ESCs/iPSCs (Table 2). Batchelder
tion protocol from mouse ESCs through the et al. spontaneously differentiated human ESCs
epiblast, nascent mesoderm, posterior nascent via EB formation with 10 % serum and used real-
mesoderm and posterior IM into nephron progen- time RT-PCR to detect the expression of several
itors. The induced nephron progenitors could marker genes for the IM (OSR1, PAX2, SIX2 and
form three-dimensional kidney structures WT1), early kidney precursors (CD24, CDH11,
containing both proximal and distal renal tubules EYA1, LIM1 and SIX1), the developing kidney
as well as numerous glomeruli by co-culturing (GDNF, RET and WNT4) and more mature renal
with embryonic spinal cords. The researchers cells (AQP2, CDH16, CLCN5, CYP27, NPHS1,
even developed vascularized glomeruli when NPHS2, PODXL and SLC12A1; [157]). Directed
integrated with the host vasculature after trans- differentiation of these ESCs using activin A, RA,
plantation under the kidney capsule of BMP4 and BMP7, which are the same
immunodeficient mice. nephrogenic factors used in mouse ESCs [139],
In summary, nephron progenitors can be gen- showed that marker gene expression for kidney
erated from mouse ESCs/iPSCs through the IM, lineages was highest when human EBs attached to
and renal tissues containing glomeruli and renal gelatin-coated plates [157]. Additionally, the
tubules can be also induced in vitro from the immunostaining patterns of PAX2 and vimentin
induced nephron progenitors. Moreover, in human ESC differentiation cultures were simi-
transplanted mouse ESC/iPSC-derived nephron lar to those in human fetal kidneys and embryonic
progenitors are capable of integrating into the rhesus monkey kidneys.
proximal renal tubules of developing mouse Lin et al. differentiated human ESCs with
kidney. reduced serum concentration and a reduced den-
sity of feeder cells [155]. MM cells were then
selected via flow cytometry with two surface
Directed Differentiation of Human markers, CD24 and PODOCALYXIN [36], but
ESCs/iPSCs into Embryonic Renal Stem/ lacking the well-characterized marker for
Progenitor Cells undifferentiated ESCs, GCTM2 [13]. Microarray
analyses of the purified CD24(+)
In contrast to the works using mouse ESCs/iPSCs, PODOCALYXIN(+)GCTM2() cell population
a small number of reports have described the and comparison with GUDMAP, a transcriptome
directed differentiation of the renal lineages from profile database for urogenital development [166],
human ESCs or iPSCs [22–24]. Like mouse showed that the top 200 differentially
ESCs, the presence of renal cells and tissues was up-regulated genes in the cell fraction clustered
demonstrated in teratomas generated from human into a group of genes associated with the MM at
ESCs, which indicated that human ESCs also E11.5 and the cortico-nephrogenic interstitium at
have the potential to differentiate into kidney E15.5 of murine kidney development [155].
18

Table 2 Summary of reports on the directed differentiation of human ESCs/iPSCs into kidney lineages
Primary
Author Cell line Format Medium Inducer target cell Functional examination References
Batchelder HSF-6 EB αMEM/10 % Spontaneous differentiation: 10 % FCS for Renal NA [157]
CA FCS, High- up to 22 days. Directed differentiation: 0.1 lineage
et al. 2009 glucose DMEM/ μM RA, 10 ng/mL activin A, 50 ng/mL cell
10 % FCS BMP4 or BMP7 for 6–8 days
Lin SA HES-2, Monolayer DMEM/20 % FCS Reduced density (2  104 cells/cm2) of Renal NA [155]
et al. 2010 HES-3, HES-4 feeder cells with 20 % FCS for 2 days and lineage
with 5 % FCS for an additional 12 days cell
Song B iPSCs derived EB DMEM/F12/2.5 10 ng/mL activin A, 15 ng/mL BMP7 and 0.1 Podocyte Podocytes showing a contractile response to [158]
et al. 2012 from normal % FCS μM RA for 10 days. the addition of angiotensin II (AII) and the
human kidney uptake of albumin. Integration into glomeruli
mesangial by co-culture with mouse metanephric cells
cells (NKMC)
Mae S hiPSCs Monolayer DMEM/F12/2 % Stage 1: 100 ng/mL activin A and 3 μM IM Formation of renal tubular structures by [159]
et al. 2013 (201B7, FCS or B27 for CHIR99021 for 2 days; Stage 2: 100 ng/mL co-culture with mouse metanephric cells
OSR1-GFP) Stage 1, DMEM/ BMP7 and 3 μM CHIR99021 for 4–20 days
F12/10 % KSR or
Translational Research Methods: Renal Stem Cells

B27 for Stage 2


Narayanan K HUES7 Monolayer REGM medium/ 10 ng/mL activin A, 0.1 μM RA, 10 ng/mL Proximal Proximal tubular epithelia showing increase [160]
et al. 2013 0.5 % FCS BMP2 and 2.5 ng/mL BMP7 for 20 days renal in cAMP levels in response to PTH, GGT
tubular activity, ammonia production and water
epithelial transport. Integration into tubules by
cell injection into newborn mice kidneys.
Formation of tubular structures by
subcutaneous injection into immunodeficient
mice
Xia Y H1, human Monolayer DMEM/F12/17.5 Stage 1: 30 ng/mL BMP4 and 50 ng/mL UB Integration into ureteric bud structures by [161]
et al. 2013 fibroblast mg/mL BSA bFGF for 2 days; Stage 2: 1 μM RA, co-culture with mouse metanephric cells
episomal- 10 ng/mL activin A and 100 ng/mL BMP2
derived iPSCs for 2 days
Taguchi A 201B7 EB DMEM/F12/B27 Stage 0: 0.5 ng/mL BMP4, 10 μM Y27632 NP Formation of glomeruli and renal tubules by [52]
et al. 2014 for 1 day; Stage 1: 1 ng/mL activin A, co-culture with embryonic spinal cord
20 ng/mL FGF2 for 2 days; Stage 2: 1 ng/mL
BMP4, 10 μM CHIR99021 for 2 days; Stage
3: 1 ng/mL BMP4, 10 μM CHIR99021 for
4 days; Stage 4: 10 ng/mL activin A, 3 ng/mL
BMP4, 3 μM CHIR99021, 0.1 μM RA,
10 μM Y27632 for 2 days; Stage 5: 1 μM
CHIR99021, 5 ng/mL FGF9, 10 μM Y27632
for 3 days
541

(continued)
542

Table 2 (continued)
Primary
Author Cell line Format Medium Inducer target cell Functional examination References
Takasato M HES3, H9, Monolayer Serum-free APEL Stage 1: 30 ng/mL BMP4 and 10 ng/mL MM and Integration into ureteric epithelia, renal [162]
et al. 2014 CRL2429C11- mediuma activin A or 8 μM CHIR99021 only for UB vesicles, and nephron progenitors by
hiPSC 2 days; Stage 2: 200 ng/mL FGF9 for 4 days; co-culture with mouse metanephric cells.
Stage 3: 200 ng/mL FGF9, 50 ng/mL BMP7 Formation of ureteric epithelia, renal vesicles,
and 0.1 μM RA and nephron progenitors in an organ culture
Lam AQ H1,H9,CHB8- Monolayer Advaced RPMI Stage 1: 5 μM CHIR99021 for 24–48 h; IM and Integration into renal tubules by co-culture [163]
et al. 2014 H2B-GFP Stage 2: 100 ng/mL bFGF, 1 μM RA for NP with mouse metanephric cells. In vitro
hESCs, dermal 36 h–3 days; Stage 3: 100 ng/mL FGF9 and formation of renal tubular structures
fibroblast- 10 ng/mL activin A for 3 days
derived
hiPSCs
Araoka T 201B7 Monolayer DMEM/F12/2 % Stage 1: 3 μM CHIR99021 and 1 μM AM580 IM Formation of renal tubular structures by [130]
et al. 2014 (OSR1-GFP) FCS or B27 for or TTNPB for 2 days; Stage 2: 1 μM AM580 co-culture with mouse metanephric cells
Stage 1, DMEM/ or TTNPB for 3–12 days
F12/10 % KSR or
B27 for Stage 2
Kang M H9, Monolayer RPMI/B27 Stage 1: 100 ng/mL activin A and 100 ng/mL NP In vitro differentiation into renal tubular [164]
et al. 2014 CRL-iPSCs Wnt3a for 1 day; Stage 2: 20 ng/mL BMP4 epithelia and glomerular podocytes. Tubule
and 10 ng/mL bFGF for 2 days; Stage 3: formation in collagen type I gel
10 μM RA, 50 ng/mL BMP7 and 10 ng/mL
bFGF for 6 (hESCs) and 8 days (hiPSCs);
Stage 4: 150 ng/mL BMP7 and 50 ng/mL
bFGF for 15 days
EB embryoid body, NA not applicable, IM intermediate mesoderm, cAMP cyclic adenosine monophosphate, PTH parathyroid hormone, GGT γ-glutamyl transferase, UB ureteric bud, NP nephron
progenitor, MM metanephric mesenchyme
a
See Ref. [165]
K. Osafune
18 Translational Research Methods: Renal Stem Cells 543

a b e
PNA Nuclei LTL Nuclei

hMito LTL Nuclei

c d
AQP1 HuNu DBA Nuclei

hMito LAMININ Nuclei

Fig. 6 Renal cells and tissues differentiated from human tetragonolobus lectin (LTL) and AQUAPORIN (AQP) 1:
iPSC (Ref. [159]). (a, b) Adult renal cells differentiated markers for the proximal renal tubule; Dolichos biflorus
from human iPSC-derived intermediate mesoderm (IM) aggulutinin (DBA): a marker for the collecting duct; HuNu:
cells in vitro. Glomerular podocytes (a) and proximal human nuclear antigen. (e) Three-dimensional renal tubu-
tubular cells (b). (c, d) Adult renal cells differentiated lar structure formed from human iPSC-derived IM cells by
from human iPSC-derived IM cells in vivo. Proximal tubu- co-culturing with mouse metanephric cells in an organ
lar cells (c) and collecting duct cells (d). Peanut agglutinin culture setting. hMito human mitochondria. Bars: 50 μm
(PNA): a marker for glomerular podocytes; Lotus

More recent research has established differen- structures in organ culture settings when com-
tiation methods from human ESCs/iPSCs into bined with dissociated mouse E11.5 metanephric
renal lineage cells that indicate developmental cells (Fig. 6e).
potential as well as marker gene expression In another study, Xia et al. published the gen-
[167]. Mae et al. used BAC-based targeting vec- eration of UB-like progenitors from human ESCs/
tors to induce homologous recombination in iPSCs [161]. They established a 4-day monolayer
human iPSCs [159]. This approach successfully culture protocol for human ESCs/iPSCs that
generated reporter human iPSC lines in which the included a combinatorial treatment with BMP4
GFP reporter gene was knocked into the locus of and FGF2 for the first 2 days in order to induce
an IM marker, OSR1. These cell lines allow for mesoderm-committed cells, followed by an addi-
quantitative evaluation of the IM cell induction tional 2-day treatment with RA, activin A and
rate. The researchers found that combinatorial BMP2 to generate the IM. They found that marker
treatment with activin A, BMP7 and genes for UB, including HOXB7, RET and
CHIR99021 in two-dimensional monolayer cul- GFRA1, were expressed in the culture rather than
tures can achieve an induction rate of over 90 % marker genes for the MM. When human IM cells
for OSR1(+) cells. These human mesoderm cells were co-cultured with dissociated mouse E11.5
have the developmental potential to further differ- metanephric cells in organ cultures, the differenti-
entiate into renal lineage cells both in vitro and ated cells were integrated only into UB structures
in vivo, such as glomerular podocytes and renal and no chimaeric MM structures were formed.
tubular cells (Fig. 6a–d). Furthermore, the cells These observations suggest the generation of UB
can form three-dimensional renal tubular progenitor-like cells from human ESCs/iPSCs.
544 K. Osafune

Taguchi et al. applied the differentiation proto- Lam et al. established a simple, rapid and effi-
col that induces mouse ESCs into nephron pro- cient differentiation protocol from human iPSCs/
genitors described above to human iPSC ESCs into the IM [163]. They first induced human
differentiation cultures by modifying the early ESCs/iPSCs into a BRACHYURY(+)MIXL1(+)
steps for mesoderm induction and adjusting the mesendoderm with CHIR99021 and then into a
culture periods of the later steps [52]. The induced PAX2(+)LHX1(+) IM with bFGF and RA with
human cells expressed several nephron progenitor 70–80 % efficiency. These IM cells gave rise to
markers, including WT1, PAX2, SALL1 and SIX2, apically ciliated tubular structures expressing
and formed three-dimensional kidney structures proximal tubule markers, such as Lotus
that consisted of WT1(+)NEPHRIN(+) glomeruli, tetragonolobus lectin (LTL), N-CADHERIN and
CADHERIN6(+) proximal tubules and KSP, and partially integrated into embryonic kid-
E-CADHERIN(+) distal tubules when ney explant cultures. In addition, the IM cells
co-cultured with mouse embryonic spinal cords. were further differentiated into cells expressing
The report was the first to show that kidney tissues markers for nephron progenitors following addi-
containing glomerular structures can be generated tional treatment with FGF9 and activin A.
from mouse ESCs and human iPSCs. Araoka et al. aimed to replace growth factors
Takasato et al. reported the generation of “self- with low-molecular-weight compounds for the
organizing kidneys” from human ESCs that were induction of the IM [130]. High-throughput chem-
synchronously formed from derivative tissues of ical screening identified two retinoids, AM580 and
both the MM and UB [162]. They treated human TTNPB, as potent inducers for the IM from human
ESCs in monolayer cultures with a combination of iPSCs/ESCs. The combination of either AM580 or
high BMP4 and low activin A or with only high TTNPB with CHIR99021 efficiently induced
CHIR99021 to induce the posterior primitive OSR1(+) IM cells that have the developmental
streak and then included FGF9 treatment to potential to differentiate into renal lineage cells
induce the IM. The resulting IM cells were treated and form three-dimensional renal tubular structures.
with a combination of FGF9, BMP7 and RA, Kang and Han published a stepwise differenti-
which caused the expression of marker genes for ation method from human ESCs/iPSCs into neph-
the MM, UB and several adult renal cell types, ron progenitors through the primitive streak and
including glomerular podocytes and epithelial IM in a serum- and feeder-free two-dimensional
cells in the proximal renal tubule and collecting culture with the combination of bFGF, activin A,
duct. Additionally, the formation of Wnt3a, BMP4, RA and BMP7 [164]. These
E-CADHERIN(+)PAX2(+) ureteric epithelia human nephron progenitors further differentiated
surrounded by WT1(+)SIX2(+) MM cells and into cells that expressed markers for podocytes
even luminal connections between ureteric epithe- (SYNAPTOPODIN and PODOCALYXIN) and
lia and renal vesicles were observed in these cul- renal tubules (AQP1, CD13 and MUC1) and
tures. When co-cultured with mouse E12.5 formed three-dimensional tubule-like structures.
metanephric cells in organ cultures, the induced Selective differentiation methods for the gener-
cells were integrated into ureteric epithelia, early ation of adult renal cell types from human iPSCs
nephron/renal vesicles and clusters of nephron into glomerular podocytes were reported by Song
progenitors. Furthermore, when the induced cells et al. [158]. They formed EB-like cellular aggre-
were reaggregated to pellets and cultured in organ gates from human iPSCs in suspension cultures
cultures with 10 % serum, the resulting cellular treated with activin A, BMP7 and RA, and attached
organoids spontaneously developed into renal to gelatin-coated plates. After 10 days, the cultures
structures, such as E-CADHERIN(+)PAX2(+) gave rise to podocytes that showed typical mor-
AQP2(+) ureteric epithelia that were surrounded phology, marker expression (SYNAPTOPODIN,
by WT1(+)PAX2(+) MM, AQP1(+)SLC3A1(+) NEPHRIN and PODOCIN) and function, includ-
proximal tubules and JAG1(+)E-CADHERIN ing a contractile response to angiotensin II (AII)
(+) renal vesicles. and the endocytic uptake of albumin.
18 Translational Research Methods: Renal Stem Cells 545

A specific differentiation protocol from human resulted in the activation of a network of genes
ESCs into proximal renal tubular epithelia was consistent with nephron progenitors and that the
also reported by Narayanan et al. in which cultur- induced cells showed the ability to contribute to
ing undifferentiated human ESCs in renal epithe- the Six2(+) nephron progenitor fields of an
lial growth medium (REGM) with a combination embryonic kidney explant.
of activin A, RA, BMP2 and BMP7 generated In addition, Omer et al. showed that treating
AQP1-expressing cells at an induction rate of mature human adult kidney epithelial cells with
around 40 % [160]. In addition to the expected valproic acid, a histone deacetylase (HDAC)
marker gene expression and morphology of prox- inhibitor, results in significant activation or
imal renal tubular epithelia, these cells showed embryonic renal progenitor genes, such as SIX2,
physiological functions, such as an increase in OSR1, SALL1, NCAM, and PSA-NCAM
the intracellular levels of cyclic adenosine [168]. However, the same treatment inhibits cell
monophosphate (cAMP) in response to parathy- de-differentiation towards a more replicative mes-
roid hormone (PTH), γ-glutamyl transferase enchymal state at the same time, which is required
(GGT) activity, ammonia production and water for the clonogenic expansion and acquisition of
transport. Moreover, these cells retained their stem cell characteristics.
functional characteristics under conditions in Research on direct cellular reprogramming
bioartificial kidneys. into renal cell types is still nascent, and further
In summary, human ESCs/iPSCs can be differ- studies will be required to establish methods for
entiated into both nephron progenitors and ure- generating renal stem/progenitor cells. For exam-
teric bud cells through the IM, and nephron ple, the in vitro reprogramming of somatic cell
structures containing glomeruli and renal tubules types other than renal cells, such as fibroblasts,
and ureteric bud-like tubular structures can be into renal progenitors remains to be achieved.
formed in vitro from induced human embryonic
renal progenitors. Selective differentiation into
glomerular podocytes and proximal renal tubular Adult Renal Stem/Progenitor Cells
epithelia from human ESCs/iPSCs has also been
achieved. Stem Cells of Extra-Renal Sources

Two types of tissue-specific stem cells identified


Embryonic Renal Stem/Progenitor Cells in bone marrow have been reported to contribute
Reprogrammed from Adult Renal Cells to renal tissues: hematopoietic stem cells [9] and
mesenchymal stem cells or multipotent marrow
The success of direct reprogramming from one stromal cells (MSCs: [169]). MSCs were hypoth-
terminally-differentiated cell type into another esized to be circulating from bone marrow and
has been shown in several cell types including responsible for the homeostasis, regeneration and
pancreatic exocrine cells into β cells [25] and repair of adult mesenchymal tissues [170], but are
fibroblasts into neurons [26]. In these reports, the now considered a subpopulation of perivascular
transduction of a combination of three or more cells or pericytes that reside in almost all tissues of
transcription factors essential for the development our body [171, 172]. MSCs are currently thought
or differentiation of the target cell types were to be recruited from their perivascular niche in
utilized. This strategy was successfully applied each tissue to sites of tissue injury, where they
to the artificial generation of embryonic nephron establish a regenerative microenvironment to
progenitors from fully-differentiated adult renal facilitate tissue regeneration by secreting various
cell types [28]. Hendry et al. reported that the bioactive molecules [173].
forced expression of six transcription factors Early reports described that bone marrow-
(SIX1, SIX2, OSR1, EYA1, HOXA11 and derived cells can differentiate into multiple
SNAI2) in an adult proximal tubule cell line somatic cell types including renal tubular cells,
546 K. Osafune

indicating the “plasticity” of stem cells to transdif- to the regeneration of renal tissues is not because
ferentiate into cells belonging to other organs, of their differentiation into tubular cells in vivo
including organs derived from other germ layers Humphreys et al. elegantly demonstrated this con-
[174–179]. However, since those reports, sponta- cept by using mice lineage tracing experiments
neous cell fusion was demonstrated between cells [63]. They demonstrated that the cells responsible
from mouse brain and mouse ESCs and between for tubular regeneration after AKI induced by I/R
mouse bone marrow cells and mouse ESCs [180, injury originate from remaining renal tubular
181]. These findings suggested that at least part of cells, thereby excluding any possible contribution
the transdifferentiation of bone marrow stem cells of extra-renal cells to regenerating kidney.
could be explained by spontaneous cell fusion Instead, MSCs seem to contribute to kidney repair
between transplanted bone marrow cells and after damage by secreting trophic factors, such as
recipient host tissue cells. vascular endothelial growth factor (VEGF),
It has also been reported that bone marrow- bFGF, monocyte chemoattractant protein-1
derived cells contribute to renal regeneration (MCP-1), HGF and insulin-like growth factor
after injury by transdifferentiating into tubular 1 (IGF1), and immunomodulators, such as
cells, suggesting the renal regeneration potential TGF-β and prostaglandin E2 (PGE2)
of exogenously administered bone marrow- [200]. While MSCs have consistently been
derived cells [182–187]. Other reports showed shown to exert renoprotective effects in animal
that hematopoietic stem cells and MSCs can dif- models of AKI [190–194], the therapeutic effec-
ferentiate into glomerular mesangial cells in vivo tiveness of MSCs on CKD is controversial
[184, 188]. Yokoo et al. demonstrated the ability [201–203]. Indeed, phase-I clinical trials testing
of human MSCs to differentiate and contribute to MSC transplantation in patients at high risk of
nephrogenesis during development in whole developing AKI following cardiac surgery are
rodent embryo cultures [189]. The group injected currently being carried out [204].
human MSCs that were engineered to produce
GDNF and lacZ using a replication-defective ade-
noviral construct into the nephrogenic IM region Sources of Adult Renal Cells
of E9.5 and 11.5 rodent embryos, which gives rise Responsible for In Vivo Regeneration
to the developing metanephros. After ex vivo
whole-embryo culture for up to 48 h, these cells As mentioned above, the lineage tracing experi-
showed a contribution to the developing rodent ments done by Humphreys et al. demonstrated
kidneys. that the cells responsible for tubular regeneration
In addition, the injection of bone marrow- after I/R injury are of tubular origin, not extra-
derived cells, especially MSCs, has repeatedly renal cells [63]. The study also demonstrated that
been shown to improve renal function in AKI adult kidneys do not contain embryonic nephron
induced by toxins (cisplatin and glycerol) or I/R progenitor-like cells expressing Six2, and that the
injury [190–194]. Similar beneficial effects on reactivation of this cell population does not occur
renal function may be obtained by administering even after ischemia damage. It was therefore indi-
growth factors, such as granulocyte colony- cated that the regeneration of new tubular cells
forming factor (G-CSF), granulocyte/monocyte originates from intrinsic epithelial cells in adult
colony-forming factor (GM-CSF), monocyte kidneys. However, it remains unclear whether
colony-forming factor (M-CSF), or stem cell fac- these regenerating cells are epithelial progenitors
tor, to mobilize a patient’s bone marrow cells. with more restricted differentiation potential than
Several studies have indicated such administra- Six2(+) embryonic progenitors or are fully-
tion improves renal function [195–198], but a differentiated tubular cells that can expand after
counter observation was also reported [199]. ischemia damage.
Current data suggest that any contribution Kusaba et al. answered this question by genet-
made by non-renal stem cells, including MSCs, ically labelling fully-differentiated proximal
18 Translational Research Methods: Renal Stem Cells 547

tubule cells using mice in which a CreER regenerating proximal tubular cells [208]. They
(T2) cassette was knocked into the sodium- found that tubular regeneration after I/R injury
dependent inorganic phosphate transporter occurred from any surviving tubular cells rather
SLC34a1 locus, which is expressed only in than from fixed progenitor cells.
fully-differentiated proximal tubules, and exam- Rinkevich et al. examined the mechanisms and
ined the cells’ contribution to regenerating renal magnitude by which the mammalian kidney gen-
epithelia after ischemia injury [205]. After injury, erates and maintains tubular cells by using multi-
all tubular cells remained labelled, indicating that colored “Rainbow” mice, in which every cell is
new regenerating tubular cells could originate randomly marked by one of four fluorescent
from the proliferation of surviving differentiated colors, allowing long term lineage tracing and
tubular epithelia. Interestingly, injury to labelled clonal analysis of new tubular cells [209]. It was
proximal tubule epithelia induced a certain degree demonstrated that the adult mammalian kidney
of de-differentiation as manifested by the marker undergoes tubulogenesis during homeostasis and
expression of putative epithelial stem cells, such after damage by clonal expansion of lineage- or
as CD24, CD133, vimentin, and kidney-injury fate-restricted precursor cells that give rise only to
molecule-1 (KIM-1). Their results provided no cells of the same nephron segment, and that
evidence for an intratubular stem cell population, the adult renal clones are derived from
but rather indicated that terminally-differentiated Wnt-responsive precursors. They concluded that
tubular epithelia de-differentiate to some extent fate-restricted precursors functioning as unipotent
by re-expressing stem cell markers and repair the progenitors continuously maintain and self-
damaged tubules [205]. preserve the mouse kidney throughout life.
Consistent with these results, Vogetseder et al. In summary, the origin of regenerating tubular
investigated the contribution of stem cells to the cells is fully-differentiated tubular epithelia, but at
growth of proximal tubular S3 segments in healthy the same time these cells may acquire some features
rats by examining cell cycle status with the applica- of unipotent epithelial stem-like cells that can clon-
tion of thymidine analog and bromo-deoxyuridine ally expand under specific proliferation stimuli.
(BrdU) and the expression of cell cycle markers, However, most studies examining the origin of
such as the G(1)-phase marker cyclin D1 tubular regeneration have been performed in AKI
[206, 207]. It was shown that cycling and mice models induced by I/R injury. It is known that
non-cycling tubular cells have the same degree of different regenerative mechanisms can take place
differentiation, that the transit amplifying cells, usu- in different disease status in other organ systems.
ally thought to be the descendent of stem cells, were For example, liver normally regenerates from
absent and that most tubular cells in the S3 segment injury, such as partial hepatectomy, by simply
divided or entered the cell cycle in response to a expanding the remaining hepatocytes, whereas
proliferative stimulus. These results indicate that some stem/progenitor populations, known as oval
tubular cells originate from differentiated tubular cells, are responsible for the regeneration after
cells, not from stem cells [206]. The group also specific types of insults, such as toxic drugs
demonstrated that around 40 % of S3 tubular cells [210, 211]. These oval cells are termed facultative
are in G(1) phase and that a strong mitotic stimulus stem/progenitor cells. It is therefore possible that
shortens the period of quiescence following cell different regenerative mechanisms may be
division, suggesting that the capacity of the proxi- involved in other kidney injury models too.
mal tubule to rapidly recruit cells into division
depends on a large reserve pool of G(1) cells and
on the shortening of the quiescence period [207]. Adult Renal Stem/Progenitor Cells
Similar results were demonstrated by Berger Expanded Ex Vivo
et al. using the transgenic PEC (parietal epithelial
cell)-reverse-tetracycline trans-activator (rtTA) Many studies have already characterized adult
mouse, which allowed cell fate tracing of renal stem/progenitor cells by mainly using
548 K. Osafune

Glomerulus Juxtaglomerular
Proximal compartment
tubule ・Sagrinati et al., ・Bruno et al.,
2006, CD24(+) 2009, resident ・Pippin et al., 2013,
CD133(+) cell MSC Renin(+) cell
・Maeshima et al.,
2003, LRC
・Kitamura et al., Distal
2005, Cloned cell tubule
・Challen et al., 2006,
SP cell
・Gupta et al., 2006,
MRPC
・Buzhor et al., 2013,
NCAM1(+) cell

Interstitium

・Oliver et al.,2004,
LRC
・Bussolati et al.,
2005, CD133(+) cell
・Hishikawa et al., Henle’s
2005, SP cell loop
Collecting
・Dekel et al., 2006,
duct
Sca1(+)Lin(-) cell
・Lee et al., 2010, ・Li et al., 2014,
MKPC MSC-like cell

Fig. 7 Adult renal stem/progenitor cells isolated from various locations by different methods

in vitro cultures. These cell populations might clonogenic assays [222, 223, 229], sphere or
result from cell-culture–induced changes in mor- spheroid assays [230, 231], flow cytometry using
phology, behavior, and marker expression, but cell surface marker [97, 224, 225], lineage tracing
still show the possibility of clinical usefulness. [227] and side population (SP) assays ([216,
The establishment of methods to expand 232–234]; Fig. 7, Table 3).
nephron-forming cells from adult kidney while The label-retaining cells (LRCs) examined by
preserving their stem/progenitor function would in vivo BrdU labeling exhibited a characteristic
supply a source for cell therapy. slow cycling of tissue-specific stem cells.
Renal stem/progenitor cells have been reported Maeshima et al. showed that LRCs were found
to exist in or were isolated from various locations in renal epithelial tubular cells of rat kidney and
in adult kidneys [212, 213], including the renal that they reentered mitosis and differentiated into
papilla [214, 215], interstitium [216–218], tubular tubular epithelia in response to renal ischemia,
epithelial cells [219–223], Bowman’s capsule thus contributing to tubular regeneration after
[97, 224, 225], decapsulated glomeruli [226], damage [219]. The group also reported that these
juxtaglomerular apparatus [227] and collecting tubular LRCs proliferate, migrate and transdif-
ducts [228], by various identification methods, ferentiate into fibroblast-like cells during renal
such as label-retaining assays [214, 219–221], fibrosis [220]. In addition, tubular LRCs isolated
18

Table 3 Summary of reports on the isolation or identification of adult renal stem/progenitor cells
Location or
Author Assay Species origin in kidney Medium and main factor Functional examination References
Maeshima A LRC Rat Proximal tubule DMEM/F12/5 % FCS with 200 ng/mL EGF, Formation of tubular or tubulocystic structures [219–221]
et al. 2003 200 ng/mL TGF-α, 200 ng/mL IGF-I, or in collagen gel. Integration into proximal
100 ng/mL HGF tubules and collecting ducts by injection into
metanephros in an organ culture
Oliver JA LRC Rat, Interstitium DMEM/F12/10 % FCS with 5 % chicken In vitro differentiation into mesenchymal, [214]
et al. 2004 mouse (Papilla) embryo extract and 5 % rat serum epithelial and neural cells and myofibroblasts,
and sphere formation. Integration into renal
parenchyma by injection into the renal
subcapsule of transient ischemic rat
Bussolati B Surface Human Intestitium 60 % LG-DMEM/40 % MCDB-201 with In vitro differentiation into tubular epithelia [217]
et al. 2005 markers (Cortex) 10 ng/mL EGF, 10 ng/mL PDGF-BB and 2 % and endothelia. Formation of tubules and
CD133(+) FCS vessels by subcutaneous injection into
immunodeficient mouse and integration into
Translational Research Methods: Renal Stem Cells

tubules of AKI models by glycerol-induced


rhabdomyolysis
Hishikawa K SP Mouse Intestitium NA Integration into interstitium and therapeutic [216]
et al. 2005 effects by injection into cisplatin-induced AKI
models
Kitamura S Clonogenic Rat Proximal tubule 1:1 mixture of culture supernatant of mouse In vitro differentiation into proximal and distal [222, 223]
et al. 2005 culture (S3 segment) mesenchymal cells in DMEM/10 % FCS and renal tubular cells and epithelia of Henle’s
modified K1 medium (1:1 DMEM and Ham’s loop. Formation of tubular structures and
F12)/10 % FCS with 25 ng/mL HGF integration into tubules and therapeutic effects
on cisplatin-induced and I/R injury AKI
models. Responsiveness to PTH and AVP
Challen GA SP Mouse Mainly proximal DMEM/10 % FCS In vitro differentiation into osteocytes and [234]
et al. 2006 tubule adipocytes and integration into metanephric
mesenchyme- and ureteric bud-derived
structures by injection into mouse
metanephros. In vivo integration into
glomeruli, proximal and distal tubules and
collecting ducts at low frequency. Therapeutic
effects on adriamycin-induced acute
glomerular injury models
549

(continued)
550

Table 3 (continued)
Location or
Author Assay Species origin in kidney Medium and main factor Functional examination References
Sagrinati C Surface Human Bowman’s Endothelial growth medium-microvascular In vitro differentiation into proximal and distal [97]
et al. 2006 markers capsule (EGM-MV)/20 % FCS tubular epithelia, osteocytes, adipocytes and
CD24(+) neural cells. Formation of tubular structures of
CD133(+) different nephron segments with therapeutic
effects by intravenous injection into AKI
models by glycerol-induced rhabdomyolysis
Gupta S Low Rat Proximal tubule 60 % DMEM-LG/40 % MCDB-201 with In vitro differentiation into cells expressing [235]
et al. 2006 density 1 mg/mL LA-BSA, 2 % FCS, 10 ng/mL EGF, endothelial, hepatocyte and neural markers.
culture 10 ng/mL PDGF-BB and 10 ng/mL LIF Integration into proximal and distal tubules by
injection into I/R injury AKI models without
therapeutic benefits
Dekel B Surface Mouse Intestitium DMEM 1.0 g/mL glucose/10 % FCS In vitro differentiation into myocytes, [215]
et al. 2006 markers (Papilla) osteocytes, adipocytes and neural cells.
Sca1(+)Lin Integration into tubules by injection into I/R
() injury AKI models
Bruno S Surface Human Decapsulated RPMI/10 % FCS In vitro differentiation into osteocytes, [226]
et al. 2009 markers glomerulus adipocytes, chondrocytes, endothelia,
CD133() podocytes and mesangial cells
CD146(+)
Lee PT Myh9(+) Mouse Interstitium LG-DMEM/10 % charcoal-stripped calf In vitro differentiation into endothelia and [218]
et al. 2010 cells from (Medulla and serum (CCS) osteoblasts. Integration into endothelia and
Myh9-GFP papilla) tubular epithelia and formation of blood
mice vessels by injection into normal mice.
Therapeutic effects on I/R injury AKI models
with improved mortality
K. Osafune
18

Buzhor E Spheroid Human Proximal and Serum-containing medium (SCM): IMDM/10 In vitro differentiation into tubular structures [229]
et al. 2011 culture distal renal % FCS with 50 ng/mL bFGF, 50 ng/mL EGF of different nephron segments by grafting onto
tubules and and 5 ng/mL SCF. Serum-free medium the chorioallantoic membrane (CAM) of chick
collecting duct (SFM): DMEM/F12 with B27 and N2 embryo
supplements, 10 ng/mL bFGF and 20 ng/mL
EGF
Pippin JW Lineage Mouse Juxtaglomerular NA In vivo differentiation into glomerular [227]
et al. 2013 tracing compartment podocytes and parietal epithelial cells
(Renin(+) cell)
Bombelli S Sphere Human ND DMEM/F12 with B27 supplement, 20 ng/mL In vitro and in vivo formation of 3D tubular [231]
et al. 2013 EGF and 20 ng/mL bFGF structures. In vitro differentiation into
proximal and distal tubular epithelia,
podocytes and endothelia
Buzhor E Clonogenic Human Proximal tubule Serum-containing medium (SCM): IMDM/10 In vitro differentiation into osteoblasts and [230]
et al. 2013 culture (NCAM1(+) % FCS with 50 ng/mL bFGF, 50 ng/mL EGF adipocytes, and formation of 3D kidney
cell) and 5 ng/mL SCF. Serum-free medium spheroid and tubular structures by grafting
(SFM): DMEM/F12 with B27 and N2 onto CAM of chick embryo. Formation of
supplements, 10 ng/mL bFGF and 20 ng/mL tubular structures by injection into
Translational Research Methods: Renal Stem Cells

EGF subcutaneous space of mice


Li J Clonogenic Mouse Collecting duct α-MEM/20 % FCS In vitro differentiation into adipocytes, [228]
et al. 2014 culture chondrocytes and osteocytes and formation of
tubular structures in collagen gel. Integration
into medullary collecting ducts by injection
into live newborn mice kidneys
LRC label-retaining cell, SP side population, NA not applicable, I/R ischemia/reperfusion, PTH parathyroid hormone, AVP arginine vasopressin, ND not determined
551
552 K. Osafune

by flowcytometry sorting formed three- CD44(+) phenotype. When grafted in low cell
dimensional tubular structures in vitro by cultur- numbers onto the CAM of chick embryos, the
ing in collagen gel and integrated into proximal spheroid cells exclusively reconstituted renal
tubules and collecting ducts when transplanted tubular epithelia of different nephron segments.
into the metanephric kidney [221]. Bombelli et al. reported a modified sphere-
Oliver et al. identified a population of LRCs in formation protocol to generate nephrospheres
the interstitium or upper papilla of adult rodent from normal human adult kidneys [231]. The cul-
kidneys [214]. These LRCs can be cultured under tured nephrospheres activated stem cell pathways,
hypoxic conditions to form aggregates of cells contained cells at different levels of maturation
expressing Nestin, a marker in other stem cell and generated three-dimensional tubular struc-
types. Single cell-derived clones of these cells tures both in vitro and in vivo. The group identi-
co-expressed mesenchymal and epithelial fied a quiescent PKHhigh stem cell population
markers, and gave rise to myofibroblasts and within the spheres that demonstrated in vitro
cells expressing neuronal markers. They also self-renewal capacity and differentiation potential
formed a compartment of rapidly proliferating into epithelial, endothelial and podocytic
cells in response to ischemic insult, suggesting progenies.
that they may play a role after acute injury. Regarding isolation using the combination of
However, the LRCs from these reports might cell surface markers, Bussolati et al. first used this
represent transiently amplifying cells rather than methodology to isolate tubule cells expressing a
stem cells. Proof of the existence of a pluripotent hematopoietic stem cell marker, CD133, from
adult renal stem cell population with long-term human renal cortex [217]. These cells expanded,
self-renewal capacity and clonogenicity remains differentiated in vitro into epithelial or endothelial
lacking. cells and showed developmental potential to form
Kitamura et al. screened for stem cells in var- tubule-like structures and functional vessels. They
ious regions of the postnatal rodent kidney by the were also incorporated into tubules when intrave-
dissection of various nephron segments and cul- nously injected into immunodeficient mice with
ture after dissociation to single cells [222]. They AKI induced by glycerol-induced rhabdomyolysis.
established clonally expanded cells from the S3 Dekel et al. used a stem cell marker, Sca1, and
segment of rat proximal renal tubules, which are identified non-tubular Sca1(+) progenitor cells in
known to be most susceptible to injury and to have the papillary interstitial space of the adult mouse
the highest proliferation rate in nephron segments. kidney [215]. The cloned renal Sca1(+)Lin()
The established cell line showed features of stem cells showed in vitro differentiation potential for
cells, could be maintained long term without myogenic, osteogenic, adipogenic and neural lin-
transformation, and expressed Pax2, Wnt4 and eages, and newly isolated Sca1(+)Lin() cells
Wt1. The transplantation of these progenitors adopted a tubular phenotype when injected
resulted in the contribution to renal tubules and directly into the renal parenchyma of AKI mice
significantly improved the renal function in both by I/R injury. These cells displayed features
cisplatin-induced and I/R injury AKI overlapping those of the papillary LRCs described
models [223]. above [214].
Buzhor et al. established three-dimensional A group headed by Paola Romagnani has
spheroid cultures of human kidney epithelial reported that a population of CD24(+)CD133(+)
cells retrieved from human nephrectomy tissue cells localized at the urinary pole of the Bowman’s
samples by suspension cultures in capsule in adult human, mouse or rat kidneys
low-attachment conditions [229]. The human kid- represents renal progenitors that give rise to both
ney spheroids upregulated certain renal develop- glomerular podocytes and renal tubular epithelia.
mental and “stemness” markers, including PAX2, Clonally-expanded CD24(+)CD133(+) cells also
SALL1, SIX2, WT1, NANOG and GPC3, and indi- contributed to the regeneration of tubular struc-
cated mostly an EpCAM(+)CD24(+)CD133(+) tures and significantly ameliorated functional
18 Translational Research Methods: Renal Stem Cells 553

kidney damage after their intravenous injection adult progenitor cells [236]. MRPCs expressed
into immunodeficient mice with AKI induced by Oct4 and Pax2 and differentiated into tubular-
rhabdomyolysis [97, 224]. It has also been structures in vitro and in vivo. However, these
reported that these progenitor cells compose the cells displayed spindle-shaped morphology,
hyperplastic glomerular lesion in several human suggesting non-tubular and possibly extra-renal
proliferative glomerulonephritis [225]. origin.
Bruno et al. found that human glomeruli Pippin et al. demonstrated by performing line-
deprived of Bowman’s capsule contain a popula- age tracing experiments that cells of renin lineage
tion of CD133()CD146(+) cells that express in the juxtaglomerular compartment serve as pro-
MSC markers and renal stem cell markers, genitors of glomerular podocytes and parietal epi-
CD24 and PAX2 [226]. These MSCs exhibit thelial cells in experimental glomerular disease
self-renewal capability, clonogenicity and [227]. Renal cells expressing renin were geneti-
multipotency capable of osteogenic, adipogenic cally fate-mapped in adult Ren1cCreER x
and chondrogenic differentiation. In addition, Rs-tdTomato-R, Ren1cCre x Rs-ZsGreen-R- and
they are able to differentiate into endothelia, epi- Ren1dCre x Z/EG reporter mice, and podocyte
thelial cells expressing podocyte markers and depletion was induced in these mice by cytotoxic
mesangial marker-expressing cells. The expres- anti-podocyte antibody. After a decrease in
sion of the kidney-specific PAX2 gene and deter- podocyte number, a significant increase in the
mination of donor sex identity when the MSCs number of labelled cells of renin lineage was
were isolated from glomeruli of a renal allograft observed in glomeruli in a focal distribution
suggested these cells are a tissue-resident along Bowman’s capsule, within the glomerular
population. tuft, or in both locations, and the labelled cells
Buzhor et al. established a two-dimensional and expressed markers for glomerular parietal cells or
low-density clonal culture of human kidney epithe- podocytes.
lial cells from adult kidney tissue of SP cells are defined as a cell population that
nephrectomized patients, in which the reactivation can rapidly efflux DNA-binding dyes, such as
of gene expression of an embryonic mesenchymal Hoechst 33342 and Rhodamine 123, and may
marker, NCAM1, was observed in a specific cell exhibit stem cell-like characteristics, including
population that attained a stem/progenitor state the expression of specific membrane transporters
[230]. The isolated NCAM1(+) cells showed prox- associated with stemness [237]. SP cells in the
imal tubular origin, expressed early nephron pro- bone marrow overlap hematopoietic stem cells,
genitor markers, such as PAX2, SALL1, SIX2 and and the cell type has also been isolated from
WT1, and acquired a mesenchymal fate, indicated other tissues, including the adult kidney, to exam-
by high VIMENTIN and reduced E-CADHERIN ine their characteristics as tissue-specific stem
expression levels. The cell population showed cells. Several groups have reported the existence
clonogenic potential by single cell cultures, gener- of SP cells in adult rodent kidney [216,
ated three-dimensional kidney spheroids, and dif- 232–234]. However, the relative size, origin and
ferentiated into tubular structures when grafted differentiation ability of these cells have not been
onto the CAM of chick or into the subcutaneous consistent. Iwatani et al. showed no evidence for a
space of immunodeficient mice. capacity to transdifferentiate into renal cells
Gupta et al. reported the isolation of in vivo [233]. Hishikawa et al. reported that
multipotent renal progenitor cells (MRPCs) from Musculin/MyoR was a marker of renal SP cells,
rat kidneys by modifying the culture conditions of and these cells were located in the interstitium of
renal cells to preserve only progenitor cells the recipient kidney when administered into a
[235]. MRPCs were isolated from adult rat kid- cisplatin-induced AKI mouse model, but showed
neys using low density culture conditions for 4–6 little evidence for transdifferentiation [216].
weeks that were similar to those used for the Challen et al. using more stringent isolation pro-
culture of bone marrow-derived multipotent cedures, demonstrated that renal SP cells
554 K. Osafune

represent 0.1 % of the kidney [234]. These cells mesenchymal transition to form clonogenic,
showed multilineage differentiation potential into long-term self-renewal and retains a capacity to
osteocytes and adipocytes in vitro, and into sev- form epithelial structures in vitro and in vivo
eral nephron segments in vivo at low efficiency [228]. After extensive passage, these kidney
after their injection into an adriamycin-induced MSC-like cells selectively integrated into the
acute glomerular injury model. However, despite AQP2(+) medullary collecting duct when
stringent selection, the cell population remains microinjected into the kidneys of neonatal mice.
heterogeneous, including a monocytic fraction, Interestingly, examination of the origin of kidney
which may result in the apparent phenotypic plas- MSC-like cells revealed that a Wnt4-expressing
ticity. In both studies, the injection of adult kidney interstitial population intercalates into the
SP cells did not show significant tubular integra- collecting duct in neonatal kidney, giving rise to
tion in acute renal damage models, although the the MSC-like cells.
treatment reduced renal damage. While SP cells in Various types of adult renal stem/progenitor
bone marrow overlap the hematopoietic stem cells cells have been identified by different methods.
isolated by flowcytometry sorting using cell sur- Although these adult stem/progenitor cells have
face markers, the population derived from other been reported to contribute to the epithelia of
organs does not always show characteristics of glomeruli and renal tubules like embryonic pro-
tissue-specific stem cells. This finding may also genitors, they may represent a different cell pop-
apply to renal SP cells. Nevertheless, these results ulation from embryonic nephron progenitors and
suggested that humoral factors from SP cells may possess different gene expression profiles and cel-
directly ameliorate renal injury rather than con- lular origins. Detailed characterization of these
tributing to the host tissues. Isolation techniques cells and examination of the therapeutic potential
for renal SP cells with high reproducibility could of each stem/progenitor cell population are there-
solve the problem of heterogeneity and facilitate fore required.
further analyses.
Lee et al. isolated mouse kidney progenitor
cells (MKPCs) with MSC properties that reside Stem Cell Niche in Adult Kidney
in the mouse kidney interstitium of the medulla
and papilla [218]. MKPCs displayed features Stem cells are usually located in specific anatomic
including spindle-shaped morphology, self- locations termed “niches,” which regulate how
renewal of more than 100 passages without evi- stem cells participate in tissue generation, main-
dence of senescence, and expression of Oct4, tenance and repair [238]. Stem cells are located in
Pax2, Wnt4, Wt1, vimentin, α-smooth muscle deep, narrow tissue niches, while transiently
actin, CD29 and S100A4. MKPC exhibited plas- amplifying precursor cells and differentiated
ticity and multipotent differentiation potential, as cells at various stages of maturation lie in the
demonstrated by the ability to differentiate into progressively more distal cell layers. In addition
endothelial cells and osteoblasts in vitro and endo- to heterologous cellular components, any given
thelial and tubular epithelial cells in vivo. Impor- niche is thought to consist of an extracellular
tantly, intrarenal injection of MKPCs in matrix and other non-cellular constituents. Both
immunodeficient mice with I/R injury rescued paracrine and metabolic factors could also serve
renal damage both histologically and functionally, as a niche, since they respond to exogenous sig-
and some MKPCs formed vessels with red blood nals via a sensing system consisting of the ner-
cells inside and incorporated into renal tubules. In vous and/or circulatory system. Accordingly, the
addition, MKPC treatment reduced the mortality perivascular site is considered a stem cell niche
of mice after I/R injury. [171, 239, 240].
Li et al. has recently identified a MSC-like Other studies have indicated that the MSC
population from the collecting duct within adult compartment, which resides in the connective tis-
mouse kidney that undergoes an epithelial-to- sue of most organs and is capable of
18 Translational Research Methods: Renal Stem Cells 555

differentiating into osteoblasts, adipocytes and up the majority of cells in a tumor


chondrocytes, extends throughout the body, [29, 30]. Their ability to initiate tumor growth
including the kidneys, as a result of its when grafted into immunocompromised mice
perivascular location. Based on similar marker demonstrates that cancer stem cells exist in a
genes, such as CD146, 3G5 [241], and differenti- variety of human tumors [31]. Specific signalling
ation potential into osteoblasts, adipocytes and pathways play a functional role in self-renewal
chondrocytes, it has been hypothesized that vas- and/or differentiation of cancer stem cells,
cular pericytes may represent the MSC population suggesting targeted therapies are possible. In
within any given organ. Indeed, pericytes are qui- addition, the identification of markers that enable
escent, slow-cycling cells, but in the angiogenic the prospective isolation of cancer stem cells,
stage they show highly-proliferative potential, which are a minority population of tumor-
capacity for self-renewal, and the ability to gener- constituting cells, from whole tumor tissues
ate osteoblasts, chondroblasts, fibroblasts and should help clarify the biological properties of
pre-adipocytes in vitro, indicating adult stem cell cancer stem cells [32]. To date, only a small num-
characters [240]. ber of studies have reported the identification of
The microvasculature of the renal papilla is renal cancer stem cells [33].
particularly rich in pericytes that regulate micro-
vascular integrity in the perivascular capillary net-
work and give the descending vasa recta, the Cancer Stem Cells in Adult Renal Cell
arteriolar segments supplying blood to the Carcinoma (RCC)
medulla, their contractile function. Therefore,
the MSC compartment residing in the renal papilla Renal cell carcinoma (RCC) is a common form of
might be linked to the perivascular niche. Consis- urologic tumors, accounting for approximately
tent with this hypothesis, Lee et al. [218] identified 3.8 % of total adult human malignancies with an
MKPCs with MSC properties that reside in the annual increasing incidence [242]. Histologic
mouse kidney interstitium of the medulla and subtypes of RCC include the clear cell carcinoma,
papilla in close association with endothelial cells. which is the most common form of RCC, papil-
Also consistent are reports by Oliver et al. [214], lary carcinoma, chromophobe carcinoma,
who showed LRCs with several characteristics of collecting duct carcinoma, and unclassified carci-
adult stem cells in the papillary interstitium and nomas [243]. Although the prognosis of RCC has
collecting duct, and Dekel et al. [215], who dem- recently improved due to earlier detection and
onstrated that non-tubular Sca1(+) multipotent pro- increased treatment options, including tyrosine
genitor cells are found mainly in the papillary kinase inhibitors and mTor inhibitors, RCC
interstitial space of adult mouse kidney. remains a tumor of unpredictable presentation and
To date, the renal papilla has been a candidate poor clinical outcome [244]. RCC is considered to
stem cell niche in kidney. However, there are originate from a mutated tubular renal population
relatively little data on kidney stem cell niches with stem/progenitor cell properties, much like in
compared with other organs, meaning there is other organs [245]. Renal cancer stem cells have
still much to learn about the identify of these been isolated from RCC using different stem cell
niches and their mechanisms for regulating renal markers or by functional approaches of stem cell
stem cells. isolation by sphere-formation or SP assays.
Bussolati et al. identified a population of
tumor-initiating cells from renal clear cell and
Cancer Stem Cells in Kidney undifferentiated carcinomas by means of cell
sorting with a mesenchymal marker, CD105
Cancer stem cells drive tumor growth by their [246]. These cells were cloned in stringent culture
self-renewal, as well as giving rise to a large conditions and displayed tumor-initiating ability,
population of differentiated progeny that make generating serially transplantable carcinomas.
556 K. Osafune

Functionally, CD105(+) cells are clonogenic and cells, Wilms’ tumor is propagated by an equiva-
able to generate spheres. Moreover, these cells lent transformed population, termed Wilms’
differentiate into epithelial cells in vitro and tumor cancer stem cells [250]. It was reported
in vivo to recapitulate the histological pattern of that the nephrogenic process in the tumor was
the tumor. Interestingly, CD105(+) cells do not deranged, as the transcription factors that specify
express CD133, which is a candidate marker for renal stem/progenitor cells (Six2, Wt1 and Pax2)
adult renal progenitors and also expressed in can- were epigenetically dysregulated in the Wilms’
cer stem cells of tumors in other organs, such as tumor blastema with aberrant DNA methylation
the nervous system, colon, prostate, pancreas and at the promoter regions [251] and chromatin mod-
lungs [32]. ifications [252]. These changes may lead to an
Other functional assays commonly used to iden- enhanced activation and accumulation of early
tify stem/progenitor cells have been used to isolate stem/progenitor cells instead of undergoing the
renal cancer stem cells. Zhong et al. selected cells kidney-specific differentiation program.
from a renal cancer cell line, SK-RC-42, that have Pode-Shakked et al. reported that NCAM1, a
the ability to form spheres in serum-free medium marker for normal embryonic renal progenitors, is
supplemented with EGF and bFGF [247]. The cell also a putative cancer stem cell marker in Wilms’
population expressed several stemness genes, tumor based on microarray data [253]. It was
including Oct4, BMI, β-catenin and Nanog, further found that NCAM1(+) cells showed the
showed resistance to chemotherapeutic agents and expression of a “Wilms’ tumor-stem” signature
irradiations and have higher in vivo tumorigenicity gene set and clonogenicity. The group also
than monolayer adherent cells. performed serial transplantation of a NCAM1(+)
Addla et al. used the SP assay to identify renal cell fraction within Wilms’ tumor into immuno-
cancer stem cells [248]. SP cells isolated from deficient mice, demonstrating that NCAM1(+)
renal carcinomas expressed mesenchymal cells give rise to new tumors as well as
markers including CD105, but not CD133, and NCAM1() differentiated cells. Therefore, the
showed high proliferative potential and an ability NCAM1(+) population fits within the criteria of
to give rise to differentiated spheroids. However, cancer stem cells, as it can self-renew and has
the characteristics and roles of this cell population multipotency in vivo [254].
in carcinogenesis are unknown. Shukrun et al. screened to purify the cancer stem
cell population in Wilms’ tumor and revealed
that NCAM1(+) aldehyde dehydrogenase 1
Cancer Stem Cells in Wilms’ Tumor (ALDH1)(+) cells exclusively localized within the
blastemal compartment of the tumor, but that
Among pediatric malignancies, Wilms’ tumor NCAM1(+)ALDH() or NCAM1()ALDH1(+)
(nephroblastoma) is the most common and is cells did not, and that the population had tumori-
rated fourth in overall incidence among childhood genic potential [255]. Surprisingly, they showed
cancers [249]. Wilms’ tumor is typically com- that ALDH1(+) Wilms’ tumor cancer stem cells
posed of three elements: undifferentiated blas- are not the most primitive cell population in
tema, stroma and tubular structures at varing Wilms’ tumor blastema, but correspond to a more
levels of differentiation, which combined resem- committed epithelial progenitor that is manifested
ble the nephrogenic zone of developing kidney by a lower expression of several renal progenitor
[33, 250]. Therefore, it is widely accepted that markers including SIX2, OSR1, PAX2, CITED1,
Wilms’ tumor arises from transformed renal WT1 and SALL1, and a relatively high expression
stem cells, which maintain themselves as the of epithelial markers. Thus, it can be deduced that
undifferentiated blastemal component of the Wilms’ tumor cancer stem cells, which sustain
tumor as well as maldifferentiate to give rise to tumor growth, both de-differentiated toward less
tubular elements [250]. Thus, while embryonic differentiated SIX2high blastemal cells and differ-
nephrogenesis is driven by normal renal stem entiated into mature epithelial structures.
18 Translational Research Methods: Renal Stem Cells 557

In summary, both RCC and Wilms’ tumor pos- human renal cells in vitro, renal lineage cells
sess cancer stem cells with high tumorigenic abil- generated from human ESCs/iPSCs might
ity and resistance to chemo and radio therapy, thus become an important source for the clinical appli-
providing a rationale for new therapies targeting cation of cell therapy approaches.
these stem cell populations. Other research areas that utilize ESC/iPSC-
derived renal lineage cells for clinical application
or practical use include disease modelling,
Clinical Application and Practical Use drug discovery and toxicology screening
of Renal Stem Cells [19, 20]. iPSC technology enables the creation
of patient- or disease-specific iPSC lines that
In order to develop regenerative therapies for kid- carry disease genotypes. Differentiation of these
ney diseases, substantial efforts have already been iPSCs into the cell types injured by the disorder
made to examine the therapeutic effectiveness of can be used as in vitro models for the study of
transplanting renal stem/progenitor cells. Most disease mechanisms and for the identification of
studies have examined stem/progenitor cells new drug compounds [256–258]. As an example,
from adult kidneys, such as LRCs, cloned cell Xia et al. reported that human iPSCs generated
lines, isolated cells with surface markers and SP from somatic cells of a patient with autosomal
cells, by using acute renal damage models, includ- dominant polycystic kidney disease (ADPKD)
ing I/R injury-, rhabdomyolysis- and cisplatin- could be differentiated into collecting duct cells,
induced tubular injury models, and an one of the renal cell types injured by ADPKD,
adriamycin-induced glomerular injury model using their differentiation method, but no results
(Table 3). On the other hand, few studies have on the mechanistic analysis of the disorder were
used embryonic renal stem/progenitor cells or shown [161]. Regarding toxicology screening, Li
chronic renal injury models. Lazzeri et al. reported et al. reported that renal proximal tubule-like cells
that CD24(+)CD133(+) cell populations isolated differentiated from human ESCs could be used to
from human embryonic kidney regenerated differ- examine the nephrotoxicity of drug
ent portions of the nephron, reduced tissue necro- compounds [259].
sis and fibrosis, and significantly improved renal Research aiming to generate entire kidneys has
function when injected into AKI mice by been conducted using ESCs/iPSCs or renal lineage
glycerol-induced rhabdomyolysis [98]. Of note, cells derived from ESCs/iPSCs [260]. One
Harari-Steinberg et al. demonstrated that the approach to generate entire kidneys would be the
transplantation of NCAM1(+) nephron progeni- use of a biological scaffold that forms a three-
tors derived from aborted human embryos have dimensional architecture. Ross et al. decellularized
therapeutic benefits when transplanted into a 5/6 rat kidneys by detergent perfusion and reseeded
resection mice model for chronic renal them with undifferentiated mouse ESCs, resulting
failure [102]. in the differentiation of mouse ESCs into renal cells
These reports suggest the possibility that in the scaffold [261]. Song et al. published a report
embryonic nephron progenitors differentiated showing that decellularized rat kidneys were
from human ESCs/iPSCs or reprogrammed from repopulated with rat neonatal kidney cells and
adult renal cells could be used to develop cell HUVECs, and that the regenerated kidneys showed
therapies for kidney diseases. However, no reports urine production both in vitro and after transplan-
describe the transplantation of renal lineage cells, tation in vivo [262]. Decellularizing patient-
including embryonic renal progenitors, generated derived kidneys and refilling them with the renal
from human ESCs/iPSCs or reprogrammed from precursors and vascular progenitors differentiated
adult renal cells, into renal disease models. Since from the patient’s own iPSCs might lead to the
it is difficult to obtain a sufficient number of renal reconstruction of three-dimensional whole kidneys
cell samples from embryonic or adult kidneys, that could be used for autologous renal
especially human embryos, or to expand the transplantation.
558 K. Osafune

On the other hand, Kobayashi et al. demon- embryonic renal progenitors, while knowledge
strated that entire organs can be generated using a on nephron progenitors has been increasing, a
method involving interspecific blastocyst comple- relatively small number of studies have examined
mentation [263]. They injected mouse and rat other precursor populations, such as ureteric bud
ESCs/iPSCs into Pdx1 (/) (pancreatogenesis- and stromal progenitors.
disabled) rat and mouse blastocysts, respectively, In addition to the works using mouse ESCs,
and generated an almost entirely ESC/iPSC- recent advances in the directed differentiation of
derived pancreas. Similar experiments have human ESCs/iPSCs into kidney lineages make it
already been performed to generate entire kidneys possible to generate embryonic renal progenitors
by injecting mouse ESCs/iPSCs into the blasto- and immature kidney tissues in vitro. However,
cysts of Sall1(/) mice, which lose the parts of further improvement in the differentiation methods
their renal tissues derived from the metanephric as well as more detailed characterization of the
mesenchyme and die of renal insufficiency after induced renal cells and tissues based on compari-
birth [264]. The resultant kidneys showed a chi- sons with their in vivo counterparts are needed. It
merism of donor-derived mesenchyme- and remains to be answered whether the renal tissues
stroma-lineage renal cells and host-derived vascu- formed from ESC/iPSC-derived renal progenitors
lar and ureteric bud-lineage cells. These can exert proper renal physiological functions, such
approaches hold tremendous promise for generat- as glomerular filtration, tubular reabsorption and
ing functional whole kidneys. The generation of secretion, and hormone synthesis and secretion.
genetically engineered host animals that lack all There are currently few functional assay systems,
the components of kidneys, including the vascu- however, that can evaluate the developmental or
lature, is required for further analyses. physiological functions of induced renal lineage
cells. Further studies will also be required to estab-
lish selective and efficient differentiation methods
Renal Stem Cell Research: Future for adult renal cell types from ESCs/iPSCs through
Directions embryonic renal progenitors, such as glomerular
podocytes, renal tubular epithelia and collecting
Regarding kidney development, further efforts to duct cells. A stable supply of these cell types will
identify specific marker genes for the IM and renal accelerate research for disease modelling, drug dis-
lineage cells and to elucidate the developmental covery and toxicology screening.
mechanisms of mesoderm formation and the cell Various types of adult renal progenitors have
fate commitment into renal lineages and fully- been identified by different methods, some of
differentiated adult renal cell types, including which have been shown to have therapeutic ben-
those that regulate how nephron progenitors give efits on renal disease models. The detailed char-
rise to several types of nephron-forming epithelia, acterization of these progenitor cells and the
such as glomerular podocytes and renal tubular comparison of the therapeutic potential among
epithelia, should be made. These investigations different progenitor populations are required. To
are necessary to establish methods that can manip- conduct these characterizations will require the
ulate isolated embryonic renal stem/progenitor development of in vitro expansion methods and
cells, including a method for their in vitro expan- therapeutically effective transplantation methods.
sion, and methods that can induce the population Regarding renal cancer stem cells, a definitive
by the directed differentiation of pluripotent stem selective marker for the isolation and targeting of
cells or by cellular reprogramming from other stem cells is still lacking. Future studies to identify
adult cell types. Furthermore, investigations on these selection markers and to understand the
directed differentiation or cellular reprogramming intracellular pathways that regulate renal cancer
to generate renal progenitors as a model for kid- stem cell maintenance and differentiation should
ney development will facilitate our understanding contribute to the development of new therapeutic
of kidney lineage specification. Regarding strategies.
18 Translational Research Methods: Renal Stem Cells 559

Conclusion administration of embryonic or adult stem/pro-


genitor cells can be therapeutically beneficial in
Because of the recent advances in elucidating the kidney injury models, suggesting this strategy
mechanisms of kidney development, the biology should be further examined for clinical
of embryonic renal stem/progenitor cells has been application.
increasingly clarified. The CM population in Cancer stem cells, which exhibit tumor-
metanephros, which is marked by Six2 expres- initiating capability and pluripotency, have been
sion, represents a pool of embryonic renal pro- isolated in renal carcinomas, although no specific
genitors, and it is currently accepted that the markers for the cell population have been identi-
post-natal mammalian kidney does not harbor fied and thus their characterization is mainly
a progenitor population equivalent to the based on functional assays. As in other tumors,
CM. NCAM1(+) progenitors in human embryonic however, renal cancer stem cells show a high
kidney might also be nephron-forming and clini- tumorigenic ability and are resistant to chemo
cally useful cells, although ethical issues and the and radio therapy, providing a rationale for
allogenetic nature may limit the use of these cells. targeting these stem cell populations with new
The generation of embryonic renal progenitors therapies. A pediatric malignancy, Wilms’ tumor,
from ESCs/iPSCs by directed differentiation or has been thought to arise from a transformed
from adult renal cells by cellular reprogramming embryonic renal progenitor. Cancer stem cells
would also provide a potential source for the have been isolated using several selection
clinical application of renal progenitors. Directed markers, including NCAM1 and ALDH. Since
differentiation methods from mouse and human there exists significant relevance between kidney
ESCs/iPSCs into embryonic renal progenitors, development and Wilms’ tumor formation, a
such as nephron progenitors and ureteric bud, tumor formation model could contribute to the
have been increasingly reported, and renal struc- understanding of kidney development in addition
tures, including glomeruli, tubules and collecting to establishing anti-cancer therapeutic strategies.
ducts, can be generated in vitro from the progen- Continued efforts to isolate and characterize
itor cells, although the differentiation methods the renal stem/progenitor cells in embryonic or
needed for such strategies have not yet been adult kidneys as well as in renal tumors, and to
fully established. generate renal progenitors from ESCs/iPSCs by
Several recent studies have revealed the source directed differentiation or from other cell types by
of the newly-formed regenerating renal tubular cellular reprogramming, are required. Because of
epithelia after acute injury, demonstrating that the annual increase in the number of patients with
the repair of injured renal epithelia starts from CKD around the world, regenerative medicine
mature tubular cells, rather than extra-renal or strategies using renal stem/progenitor cells are
resident stem cells. In this process, mature tubular being aggressively sought.
cells show a marked plasticity, entering into an
activated proliferative state characterized by the
Acknowledgements The author would like to thank all
reactivation of mesenchymal markers detectable the members of CiRA, Kyoto University, especially
during nephrogenesis. Dr. Peter Karagiannis for critically reading and revising
Adult renal stem/progenitor cells contain sev- the manuscript, Dr. Kazutoshi Takahashi for providing the
eral different cell populations, although they may photos of iPSCs, and apologize to authors whose studies
could not be cited owing to space limitations. The research
not contribute to in vivo tubular regeneration of the author is supported by the Japan Society for the
after acute injury. These adult progenitor cells Promotion of Science (JSPS) through its “Funding Pro-
may not be equivalent to those in embryonic gram for World-Leading Innovative R&D on Science and
kidneys. The renal papilla contains a population Technology (FIRST Program),” and by the Japan Science
and Technology Agency (JST) through its research grant
of adult stem cells with MSC-like characters and “Core Center for iPS Cell Research and Technological
might represent the perivascular renal stem cell Development, Research Center Network for Realization
niche. Several studies have shown that the of Regenerative Medicine.”
560 K. Osafune

References 15. Takahashi K, Yamanaka S. Induction of pluripotent


stem cells from mouse embryonic and adult fibroblast
1. Coresh J, Selvin E, Stevens LA, Manzi J, Kusek JW, cultures by defined factors. Cell. 2006;126:663–76.
Eggers P, et al. Prevalence of chronic kidney disease 16. Takahashi K, Tanabe K, Ohnuki M, Narita M,
in the United States. JAMA. 2007;298:2038–47. Ichisaka T, Tomoda K, et al. Induction of pluripotent
2. Lysaght MJ. Maintenance dialysis population dynam- stem cells from adult human fibroblasts by defined
ics: current trends and long-term implications. J Am factors. Cell. 2007;131:861–72.
Soc Nephrol. 2002;13 Suppl 1:S37–40. 17. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-
3. Grimm Jr RH, Svendsen KH, Kasiske B, Keane WF, Bourget J, Frane JL, Tian S, et al. Induced pluripotent
Wahi MM. Proteinuria is a risk factor for mortality stem cell lines derived from human somatic cells.
over 10 years of follow-up. MRFIT Research Group. Science. 2007;318:1917–20.
Multiple Risk Factor Intervention Trial. Kidney Int 18. Murry CE, Keller G. Differentiation of embryonic
Suppl. 1997;63:S10–4. stem cells to clinically relevant populations: lessons
4. Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, from embryonic development. Cell. 2008;132:
Culleton B, Hamm LL, et al. Kidney disease as a risk 661–80.
factor for development of cardiovascular disease: a 19. Lau F, Ahfeldt T, Osafune K, Akutsu H, Cowan
statement from the American Heart Association CA. Induced pluripotent stem (iPS) cells: an up-to-
Councils on Kidney in Cardiovascular Disease, the-minute review. F1000 Biol Rep. 2009;1:84.
High Blood Pressure Research, Clinical Cardiology, 20. Karagiannis P, Yamanaka S. The fate of cell
and Epidemiology and Prevention. Circulation. reprogramming. Nat Methods. 2014;11(10):1006–8.
2003;108:2154–69. 21. Kiskinis E, Eggan K. Progress toward the clinical
5. Tonelli M, Wiebe N, Culleton B, House A, Rabbat C, application of patient-specific pluripotent stem cells.
Fok M, et al. Chronic kidney disease and mortality J Clin Invest. 2010;120:51–9.
risk: a systematic review. J Am Soc Nephrol. 22. Osafune K. In vitro regeneration of kidney from plu-
2006;17:2034–47. ripotent stem cells. Exp Cell Res. 2010;316:2571–7.
6. Matsushita K, van der Velde M, Astor BC, 23. Osafune K. iPS cell technology-based research for the
Woodward M, Levey AS, de Jong PE, et al. Associ- treatment of diabetic nephropathy. Semin Nephrol.
ation of estimated glomerular filtration rate and albu- 2012;32:479–85.
minuria with all-cause and cardiovascular mortality in 24. Takasato M, Maier B, Little MH. Recreating kidney
general population cohorts: a collaborative meta- progenitors from pluripotent cells. Pediatr Nephrol.
analysis. Lancet. 2010;375:2073–81. 2014;29:543–52.
7. Trivedi H. Cost implications of caring for chronic 25. Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton
kidney disease: are interventions cost-effective? Adv DA. In vivo reprogramming of adult pancreatic exo-
Chronic Kidney Dis. 2010;17(3):265–70. crine cells to beta-cells. Nature. 2008;455:627–32.
8. Little MH. Regrow or repair: potential regenerative 26. Panq ZP, Yang N, Vierbuchen T, Ostermeier A, Fuen-
therapies for the kidney. J Am Soc Nephrol. 2006;17 tes DR, Yang TQ, et al. Induction of human neuronal
(9):2390–401. cells by defined transcription factors. Nature.
9. Till JE, McCulloch EA, Siminovitch L. A stochastic 2011;476:220–3.
model of stem cell proliferation, based on the growth 27. Harari-Steinberg O, Pleniceanu O, Dekel B. Selecting
of spleen colony-forming cells. Proc Natl Acad Sci the optimal cell for kidney regeneration: fetal, adult or
USA. 1964;51:29–36. reprogrammed stem cells. Organogenesis. 2011;7:
10. Reynolds BA, Weiss S. Generation of neurons and 123–34.
astrocytes from isolated cells of the adult mammalian 28. Hendry CE, Vanslambrouck JM, Ineson J, Suhaimi N,
central nervous system. Science. 1992;255:1707–10. Takasato M, Rae F, et al. Direct transcriptional
11. Evans MJ, Kaufman MH. Establishment in culture of reprogramming of adult cells to embryonic nephron
pluripotential cells from mouse embryos. Nature. progenitors. J Am Soc Nephrol. 2013;24:1424–34.
1981;292:154–6. 29. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T,
12. Martin GR. Isolation of a pluripotent cell line from Caceres-Cortes J, et al. A cell initiating human acute
early mouse embryos cultured in medium conditioned myeloid leukaemia after transplantation into SCID
by teratocarcinoma stem cells. Proc Natl Acad Sci mice. Nature. 1994;367(6464):645–8.
USA. 1981;78:7634–8. 30. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem
13. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz cells, cancer, and cancer stem cells. Nature. 2001;414
MA, Swiergiel JJ, Marshall VS, et al. Embryonic stem (6859):105–11.
cell lines derived from human blastocysts. Science. 31. Al-Hajj M, Wicha MS, Benito-Hernandez
1998;282:1145–7. A. Prospective identification of tumorigenic breast
14. Reubinoff BE, Pera MF, Fong CY, Trounson A, cancer cells. Proc Natl Acad Sci USA. 2003;100:
Bongso A. Embryonic stem cell lines from human 3983–8.
blastocysts: somatic differentiation in vitro. Nat 32. Ailles LE, Weissman IL. Cancer stem cells in solid
Biotechnol. 2000;18:399–404. tumors. Curr Opin Biotechnol. 2007;18(5):460–6.
18 Translational Research Methods: Renal Stem Cells 561

33. Bussolati B, Dekel B, Azzarone B, Camussi G. 48. Kattman SJ, Witty AD, Gagliardi M, Dubois NC,
Human renal cancer stem cells. Cancer Lett. 2013; Niapour M, Hotta A, et al. Stage-specific optimization
338(1):141–6. of activin/nodal and BMP signaling promotes cardiac
34. Saxen L. Organogenesis of the kidney. Cambridge: differentiation of mouse and human pluripotent stem
Cambridge University Press; 1987. cell lines. Cell Stem Cell. 2011;8:228–40.
35. Burrow CR. Regulatory molecules in kidney devel- 49. Bernardo AS, Faial T, Gardner L, Niakan KK,
opment. Pediatr Nephrol. 2000;14:240–53. Ortmann D, Senner CE, et al. BRACHYURY and
36. Challen GA, Martinez G, Davis MJ, Taylor DF, CDX2 mediate BMP-induced differentiation of
Crowe M, Teasdale RD, et al. Identifying the molec- human and mouse pluripotent stem cells into embry-
ular phenotype of renal progenitor cells. J Am Soc onic and extraembryonic lineages. Cell Stem Cell.
Nephrol. 2004;15:2344–57. 2011;9:144–55.
37. Dressler GR. The cellular basis of kidney develop- 50. Fleming BM, Yelin R, James RG, Schultheiss TM. A
ment. Annu Rev Cell Dev Biol. 2006;22:509–29. role for Vg1/Nodal signaling in specification of the
38. Dressler GR. Advances in early kidney specification, intermediate mesoderm. Development.
development and patterning. Development. 2009; 2013;140:1819–29.
136:3863–74. 51. Costantini F, Kopan R. Patterning a complex organ:
39. Little MH, McMahon AP. Mammalian kidney devel- branching morphogenesis and nephron segmentation
opment: principles, progress, and projections. Cold in kidney development. Dev Cell. 2010;18:698–712.
Spring Harb Perspect Biol. 2012;4(5):0008300. 52. Taguchi A, Kaku Y, Ohmori T, Sharmin S, Ogawa M,
40. James RG, Schultheiss TM. Bmp signaling promotes Sasaki H, et al. Redefining the in vivo origin of
intermediate mesoderm gene expression in a dose- metanephric nephron progenitors enables generation
dependent, cell-autonomous and translation- of complex kidney structures from pluripotent stem
dependent manner. Dev Biol. 2005;288:113–25. cells. Cell Stem Cell. 2014;14:53–67.
41. James RG, Kamei CN, Wang Q, Jiang R, Schultheiss 53. Self M, Lagutin OV, Bowling B, Hendrix J, Cai Y,
TM. Odd-skipped related 1 is required for develop- Dressler GR, et al. Six2 is required for suppression of
ment of the metanephric kidney and regulates forma- nephrogenesis and progenitor renewal in the develop-
tion and differentiation of kidney precursor cells. ing kidney. EMBO J. 2006;25(21):5214–28.
Development. 2006;133:2995–3004. 54. Kobayashi A, Valerius MT, Mugford JW, Carroll TJ,
42. Mugford JW, Sipilä P, McMahon JA, McMahon AP. Self M, Oliver G, et al. Six2 defines and regulates a
Osr1 expression demarcates a multi-potent population multipotent self-renewing nephron progenitor popu-
of intermediate mesoderm that undergoes progressive lation throughout mammalian kidney development.
restriction to an Osr1-dependent nephron progenitor Cell Stem Cell. 2008;8:169–81.
compartment within the mammalian kidney. Dev 55. Reidy KJ, Rosenblum ND. Cell and molecular biol-
Biol. 2008;324:88–98. ogy of kidney development. Semin Nephrol. 2009;29
43. Kinder SJ, Tsang TE, Wakamiya M, Sasaki H, (4):321–37.
Behringer RR, Nagy A, et al. The organizer of the 56. Shakya R, Watanabe T, Costantini F. The role of
mouse gastrula is composed of a dynamic population GDNF/Ret signaling in ureteric bud cell fate and
of progenitor cells for the axial mesoderm. Develop- branching morphogenesis. Dev Cell. 2005;8:65–74.
ment. 2001;128:3623–34. 57. Skinner MA, Safford SD, Reeves JG, Jackson ME,
44. Tada S, Era T, Furusawa C, Sakurai H, Nishikawa S, Freemerman AJ. Renal aplasia in humans is associ-
Kinoshita M, et al. Characterization of mesendoderm: ated with RET mutations. Am J Hum Genet.
a diverging point of the definitive endoderm and 2008;82:344–51.
mesoderm in embryonic stem cell differentiation cul- 58. Carroll TJ, Park JS, Hayashi S, Majumdar A,
ture. Development. 2005;132:4363–74. McMahon AP. Wnt9b plays a central role in the
45. Lu CC, Robertson EJ. Multiple roles for Nodal in the regulation of mesenchymal to epithelial transitions
epiblast of the mouse embryo in the establishment of underlying organogenesis of the mammalian urogen-
anterior-posterior patterning. Dev Biol. 2004;273: ital system. Dev Cell. 2005;9:283–92.
149–59. 59. Stark K, Vainio S, Vassileva G, McMahon
46. Gadue P, Huber TL, Paddison PJ, Keller GM. Wnt AP. Epithelial transformation of metanephric mesen-
and TGFbeta signaling are required for the induction chyme in the developing kidney regulated by Wnt-4.
of an in vitro model of primitive streak formation Nature. 1994;372:679–83.
using embryonic stem cells. Proc Natl Acad Sci 60. Barak H, Huh SH, Chen S, Jeanpierre C,
USA. 2006;103:16806–11. Martinovic J, Parisot M, et al. FGF9 and FGF20
47. Sumi T, Tsuneyoshi N, Nakatsuji N, Suemori H. maintain the stemness of nephron progenitors in
Defining early lineage specification of human embry- mice and man. Dev Cell. 2012;22:1191–207.
onic stem cells by the orchestrated balance 61. Poladia DP, Kish K, Kutay B, Hains D, Kegg H,
of canonical Wnt/beta-catenin, Activin/Nodal Zhao H, et al. Role of fibroblast growth factor recep-
and BMP signaling. Development. 2008;135: tors 1 and 2 in the metanephric mesenchyme. Dev
2969–79. Biol. 2006;291:325–39.
562 K. Osafune

62. Urbach A, Yermalovich A, Zhang J, Spina CS, Zhu H, 77. Bronner-Fraser M, Fraser SE. Cell lineage analysis
Perez-Atayde AR, et al. Lin28 sustains early renal reveals multipotency of some avian neural crest cells.
progenitors and induces Wilms tumor. Genes Dev. Nature. 1988;335(6186):161–4.
2014;28(9):971–82. 78. Sainio K, Nonclercq D, Saarma M, Palgi J, Saxen L,
63. Humphreys BD, Valerius MT, Kobayashi A, Mugford Sariola H. Neuronal characteristics in embryonic
JW, Soeung S, Duffield JS, et al. Intrinsic epithelial renal stroma. Int J Dev Biol. 1994;38(1):77–84.
cells repair the kidney after injury. Cell Stem Cell. 79. Asada N, Takase M, Nakamura J, Oguchi A,
2008;2:284–91. Asada M, Suzuki N, et al. Dysfunction of fibroblasts
64. Diep CQ, Ma D, Deo RC, Holm TM, Naylor RW, of extrarenal origin underlies renal fibrosis and renal
Arora N, et al. Identification of adult nephron pro- anemia in mice. J Clin Invest. 2011;121(10):3981–90.
genitors capable of kidney regeneration in zebrafish. 80. Dekel B, Hochman E, Sanchez MJ, Maharshak N,
Nature. 2011;470:95–100. Amariglio N, Green AR, et al. Kidney, blood, and
65. Singh SR, Liu W, Hou SX. The adult Drosophila endothelium: developmental expression of stem cell
malpighian tubules are main-tained by multipotent leukemia during nephrogenesis. Kidney Int. 2004;65:
stem cells. Cell Stem Cell. 2007;1:191–203. 1162–9.
66. Hendry C, Rumballe B, Moritz K, Little 81. Quaggin SE, Kreidberg JA. Development of the renal
MH. Defining and redefining the nephron progenitor glomerulus: good neighbors and good fences. Devel-
population. Pediatr Nephrol. 2011;26:1395–406. opment. 2008;135:609–20.
67. Duffield JS, Humphreys BD. Origin of new cells in 82. Sequeira Lopez ML, Gomez RA. Development of the
the adult kidney: results from genetic labeling tech- renal arterioles. J Am Soc Nephrol. 2011;22:
niques. Kidney Int. 2011;79:494–501. 2156–65.
68. Hatini V, Huh SO, Herzlinger D, Soares VC, Lai 83. Vaughan MR, Quaggin SE. How do mesangial and
E. Essential role of stromal mesenchyme in kidney endothelial cells form the glomerular tuft? J Am Soc
morphogenesis revealed by targeted disruption of Nephrol. 2008;19(1):24–33.
Winged Helix transcription factor BF-2. Genes Dev. 84. Rodriguez CG, Makori N, Cukierski MA, Hendrickx
1996;10:1467–78. AG. Development of the definitive kidney in the
69. Mendelsohn C, Batourina E, Fung S, Gilbert T, Dodd cynomolgus monkey (Macaca fascicularis). J Med
J. Stromal cells mediate retinoid-dependent functions Primatol. 1996;25:122–32.
essential for renal development. Development. 85. Batchelder CA, Lee CC, Martinez ML, Tarantal
1999;126:1139–48. AF. Ontogeny of the kidney and renal developmental
70. Dudley AT, Godin RE, Robertson EJ. Interaction markers in the rhesus monkey (Macaca mulatta).
between FGF and BMP signaling pathways regulates Anat Rec (Hoboken). 2010;293:1971–83.
development of metanephric mesenchyme. Genes 86. Metsuyanim S, Harari-Steinberg O, Buzhor E,
Dev. 1999;13:1601–13. Omer D, Pode-Shakked N, Ben-Hur H,
71. Schnabel CA, Godin RE, Cleary ML. Pbx1 regulates et al. Expression of stem cell markers in the human
nephrogenesis and ureteric branching in the develop- fetal kidney. PLoS One. 2009;4:e6709.
ing kidney. Dev Biol. 2003;254:262–76. 87. Hartman HA, Lai HL, Patterson LT. Cessation
72. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, of renal morphogenesis in mice. Dev Biol. 2007;
Nowlin BT, Bonventre JV, et al. Fate tracing reveals 310(2):379–87.
the pericyte and not epithelial origin of 88. Rogers SA, Lowell JA, Hammerman NA,
myofibroblasts in kidney fibrosis. Am J Pathol. Hammerman MR. Transplantation of developing
2010;176:85–97. metanephroi into adult rats. Kidney Int. 1998;54(1):
73. Kobayashi A, Mugford JW, Krautzberger AM, 27–37.
Naiman N, Liao J, McMahon AP. Identification of a 89. Hammerman MR. Transplantation of renal precursor
multipotent self-renewing stromal progenitor popula- cells: a new therapeutic approach. Pediatr Nephrol.
tion during mammalian kidney organogenesis. Stem 2000;14:513–7.
Cell Reports. 2014;3(4):650–62. 90. Dekel B, Amariglio N, Kaminski N, Schwartz A,
74. Guillaume R, Bressan M, Herzlinger D. Paraxial Goshen E, Arditti FD, et al. Engraftment and differ-
mesoderm contributes stromal cells to the developing entiation of human metanephroi into functional
kidney. Dev Biol. 2009;329(2):169–75. mature nephrons after transplantation into mice is
75. Le Douarin NM, Teillet MA. Experimental analysis accompanied by a profile of gene expression similar
of the migration and differentiation of neuroblasts of to normal human kidney development. J Am Soc
the autonomic nervous system and of neurectodermal Nephrol. 2002;13:977–90.
mesenchymal derivatives, using a biological cell 91. Dekel B, Burakova T, Arditti FD, Reich-Zeliger S,
marking technique. Dev Biol. 1974;41(1):162–84. Milstein O, Aviel-Ronen S, et al. Human and porcine
76. Le Douarin NM. The avian embryo as a model to early kidney precursors as a new source for transplan-
study the development of the neural crest: a long tation. Nat Med. 2003;9:53–60.
and still ongoing story. Mech Dev. 2004;121(9): 92. Kim SS, Park HJ, Han J, Gwak SJ, Park MH,
1089–102. Song KW, et al. Improvement of kidney failure with
18 Translational Research Methods: Renal Stem Cells 563

fetal kidney precursor cell transplantation. Transplan- 106. Resnick JL, Bixler LS, Cheng L, Donovan PJ. Long-
tation. 2007;83(9):1249–58. term proliferation of mouse primordial germ cells in
93. Kim SS, Gwak SJ, Han J, Park MH, Song KW, Kim culture. Nature. 1992;359:550–1.
BS. Regeneration of kidney tissue using in vitro cul- 107. Kanatsu-Shinohara M, Inoue K, Lee J, Yoshimoto M,
tured fetal kidney cells. Exp Mol Med. 2008; Ogonuki N, Miki H, et al. Generation of pluripotent
40:361–9. stem cells from neonatal mouse testis. Cell.
94. Osafune K, Takasato M, Kispert A, Asashima M, 2004;119:1001–12.
Nishinakamura R. Identification of multipotent pro- 108. Aoi T, Yae K, Nakagawa M, Ichisaka T, Okita K,
genitors in the embryonic mouse kidney by a novel Takahashi K, et al. Generation of pluripotent stem
colony-forming assay. Development. 2006;133: cells from adult mouse liver and stomach cells. Sci-
151–61. ence. 2008;321:699–702.
95. Nishinakamura R, Matsumoto Y, Nakao K, 109. Aasen T, Raya A, Barrero MJ, Garreta E, Consiglio A,
Nakamura K, Sato A, Copeland NG, et al. Murine Gonzalez F, et al. Efficient and rapid generation of
homolog of SALL1 is essential for ureteric bud inva- induced pluripotent stem cells from human
sion in kidney development. Development. 2001; keratinocytes. Nat Biotechnol. 2008;26:1276–84.
128(16):3105–15. 110. Hanna J, Markoulaki S, Schorderet P, Carey BW,
96. Lusis M, Li J, Ineson J, Christensen ME, Rice A, Beard C, Wernig M, et al. Direct reprogramming of
Little MH. Isolation of clono-genic, long-term self terminally differentiated mature B lymphocytes to
renewing embryonic renal stem cells. Stem Cell Res. pluripotency. Cell. 2008;133:250–64.
2010;5:23–39. 111. Loh YH, Agarwal S, Park IH, Urbach A, Huo H,
97. Sagrinati C, Netti GS, Mazzinghi B, Lazzeri E, Heffner GC, et al. Generation of induced pluripotent
Liotta F, Frosali F, et al. Isolation and characterization stem cells from human blood. Blood. 2009;113:5476–9.
of multipotent progenitor cells from the Bowman’s 112. Bar-Nur O, Russ HA, Efrat S, Benvenisty
capsule of adult human kidneys. J Am Soc Nephrol. N. Epigenetic memory and preferential lineage-
2006;17(9):2443–56. specific differentiation in induced pluripotent stem
98. Lazzeri E, Crescioli C, Ronconi E, Mazzinghi B, cells derived from human pancreatic islet beta cells.
Sagrinati C, Netti GS, et al. Regenerative potential Cell Stem Cell. 2011;9:17–23.
of embryonic renal multipotent progenitors in acute 113. Song B, Niclis JC, Alikhan MA, Sakkal S, Sylvain A,
renal failure. J Am Soc Nephrol. 2007;18(12): Kerr PG, et al. Generation of induced pluripotent stem
3128–38. cells from human kidney mesangial cells. J Am Soc
99. Dekel B, Metsuyanim S, Schmidt-Ott KM, Nephrol. 2011;22:1213–20.
Fridman E, Jacob-Hirsch J, Simon A, et al. Multiple 114. Zhou T, Benda C, Duzinger S, Huang Y, Li X, Li Y,
imprinted and stemness genes provide a link between et al. Generation of induced pluripotent stem cells
normal and tumor progenitor cells of the developing from urine. J Am Soc Nephrol. 2011;22:1221–8.
human kidney. Cancer Res. 2006;66:6040–9. 115. Zhou T, Benda C, Dunzinger S, Huang Y, Ho JC,
100. Pode-Shakked N, Metsuyanim S, Rom-Gross E, Yang J, et al. Generation of human induced pluripo-
Mor Y, Fridman E, Goldstein I, et al. Developmental tent stem cells from urine samples. Nat Protoc.
tumourigenesis: NCAM as a putative marker for the 2012;7:2080–9.
malignant renal stem/progenitor cell population. J 116. Wang WW, Wang W, Jiang Y, Han GF, Lu S, Li G,
Cell Mol Med. 2009;13(8B):1792–808. et al. Reprogramming of mouse renal tubular epithe-
101. Pleniceanu O, Harari-Steinberg O, Dekel B. lial cells to induced pluripotent stem cells.
Concise review: kidney stem/progenitor cells: differ- Cytotherapy. 2013;15:578–85.
entiate, sort out, or reprogram? Stem Cells. 2010;28: 117. Nakagawa M, Taniguchi Y, Senda S, Takizawa N,
1649–60. Ichisaka T, Asano K, et al. A novel efficient feeder-
102. Harari-Steinberg O, Metsuyanim S, Omer D, free culture system for the derivation of human
Gnatek Y, Gershon R, Pri-Chen S, et al. Identification induced pluripotent stem cells. Sci Rep. 2014;4:3594.
of human nephron progenitors capable of generation 118. Alper J. Geron gets green light for human trial of ES
of kidney structures and functional repair of chronic cell-derived product. Nat Biotechnol. 2009;27:213–4.
renal disease. EMBO Mol Med. 2013;5:1556–68. 119. Schwartz SD, Hubschman JP, Heilwell G, Franco-
103. Noiman T, Buzhor E, Metsuyanim S, Harari- Cardenas V, Pan CK, Ostrick RM, et al. Embryonic
Steinberg O, Morgenshtern C, Dekel B, et al. A stem cell trials for macular degeneration: a prelimi-
rapid in vivo assay system for analyzing the organo- nary report. Lancet. 2012;379:713–20.
genetic capacity of human kidney cells. Organogene- 120. Reardon S, Cyranoski D. Japan stem-cell trial stirs
sis. 2011;7(2):140–4. envy. Nature. 2014;513(7518):287–8.
104. Martin GR. Teratocarcinomas and mammalian 121. Doetschman TC, Eistetter H, Katz M, Schmidt W,
embryogenesis. Science. 1980;209:768–76. Kemler R. The in vitro development of blastocyst-
105. Matsui Y, Zsebo K, Hogan BL. Derivation of derived embryonic stem cell lines: formation of vis-
pluripotential embryonic stem cells from murine pri- ceral yolk sac, blood islands and myocardium. J
mordial germ cells in culture. Cell. 1992;70:841–7. Embryol Exp Morphol. 1985;87:27–45.
564 K. Osafune

122. Nishikawa SI, Nishikawa S, Hirashima M, pluripotent stem cells. Nat Biotechnol. 2010;28:
Matsuyoshi N, Kodama H. Progressive lineage anal- 848–55.
ysis by cell sorting and culture identifies FLK1 + 136. Yamamoto M, Cui L, Johkura K, Asanuma K,
VE-cadherin + cells at a diverging point of endothelial Okouchi Y, Ogiwara N, et al. Branching ducts similar
and hemopoietic lineages. Development. 1998;125: to mesonephric ducts or ureteric buds in teratomas
1747–57. originating from mouse embryonic stem cells. Am J
123. D’Amour KA, Bang AG, Eliazer S, Kelly OG, Physiol Renal Physiol. 2006;290:F52–60.
Agulnick AD, Smart NG, et al. Production of pancre- 137. Steenhard BM, Isom KS, Cazcarro P, Dunmore JH,
atic hormone-expressing endocrine cells from human Godwin AR, St John PL, et al. Integration of embry-
embryonic stem cells. Nat Biotechnol. 2006;24: onic stem cells in metanephric kidney organ culture.
1392–401. J Am Soc Nephrol. 2005;16:1623–31.
124. Hay DC, Zhao D, Fletcher J, Hewitt ZA, McLean D, 138. Kobayashi T, Tanaka H, Kuwana H, Inoshita S,
Urruticoechea-Uriguen A, et al. Efficient differentia- Teraoka H, Sasaki S, et al. Wnt4-transformed mouse
tion of hepatocytes from human embryonic stem cells embryonic stem cells differentiate into renal tubular
exhibiting markers recapitulating liver development cells. Biochem Biophys Res Commun. 2005;336:
in vivo. Stem Cells. 2008;26:894–902. 3527–34.
125. Xu Y, Shi Y, Ding S. A chemical approach to stem- 139. Kim D, Dressler GR. Nephrogenic factors promote
cell biology and regenerative medicine. Nature. differentiation of mouse embryonic stem cells into
2008;453:338–44. renal epithelia. J Am Soc Nephrol. 2005;16:3527–34.
126. Wu X, Ding S, Ding Q, Gray NS, Schultz PG. Small 140. Kramer J, Steinhoff J, Klinger M, Fricke L, Rohwedel
molecules that induce cardiomyogenesis in embry- J. Cells differentiated from mouse embryonic stem
onic stem cells. J Am Chem Soc. 2004;126:1590–1. cells via embryoid bodies express renal marker mol-
127. Chen S, Borowiak M, Fox JL, Maehr R, Osafune K, ecules. Differentiation. 2006;74:91–104.
Davidow L, et al. A small molecule that directs dif- 141. Vigneau C, Polgar K, Striker G, Elliott J, Hyink D,
ferentiation of human ESCs into the pancreatic line- Weber O, et al. Mouse embryonic stem cell-derived
age. Nat Chem Biol. 2009;5:258–65. embryoid bodies generate progenitors that integrate
128. Borowiak M, Maehr R, Chen S, Chen AE, Tang W, long term into renal proximal tubules in vivo. J Am
Fox JL, et al. Small molecules efficiently direct endo- Soc Nephrol. 2007;18:1709–20.
dermal differentiation of mouse and human embry- 142. Bruce SJ, Rea RW, Steptoe AL, Busslinger M,
onic stem cells. Cell Stem Cell. 2009;4:348–58. Bertram JF, Perkins AC. In vitro differentiation of
129. Sakano D, Shiraki N, Kikawa K, Yamazoe T, murine embryonic stem cells toward a renal lineage.
Kataoka M, Umeda K, et al. VMAT2 identified as a Differentiation. 2007;75:337–49.
regulator of late-stage β-cell differentiation. Nat 143. Nakane A, Kojima Y, Hayashi Y, Kohri K, Masui S,
Chem Biol. 2014;10:141–8. Nishinakamura R. Pax2 overexpression in embryoid
130. Araoka T, Mae S, Kurose Y, Uesugi M, Ohta A, bodies induces upregulation of integrin alpha8 and
Yamanaka S, et al. Efficient and rapid induction of aquaporin-1. In Vitro Cell Dev Biol Anim. 2009;45:
human iPSCs/ESCs into nephrogenic intermediate 62–8.
mesoderm using small molecule-based differentiation 144. Morizane R, Monkawa T, Itoh H. Differentiation of
methods. PLoS One. 2014;9:e84881. murine embryonic stem cell and induced pluripotent
131. Adewumi O, Aflatoonian B, Ahrlund-Richter L, stem cells to renal lineage in vitro. Biochem Biophys
Amit M, Andrews PW, Beighton G, Res Commun. 2009;390:1334–9.
et al. Characterization of human embryonic stem cell 145. Ren X, Zhang J, Gong X, Niu X, Zhang X, Chen P,
lines by the International Stem Cell Initiative. Nat et al. Differentiation of murine embryonic stem cells
Biotechnol. 2007;25:803–16. toward renal lineages by conditioned medium from
132. Osafune K, Caron L, Borowiak M, Martinez RJ, Fitz- ureteric bud cells in vitro. Acta Biochim Biophys Sin
Gerald CS, Sato Y, et al. Marked differences in dif- (Shanghai). 2010;42:464–71.
ferentiation propensity among human embryonic 146. Mae S, Shirasawa S, Yoshie S, Sato F, Kanoh Y,
stem cell lines. Nat Biotechnol. 2008;26:313–5. Ichikawa H, et al. Combination of small molecules
133. Kajiwara M, Aoi T, Okita K, Takahashi R, Inoue H, enhances differentiation of mouse embryonic stem
Takayama N, et al. Donor-dependent variations in cells into intermediate mesoderm through BMP7-
hepatic differentiation from human-induced pluripo- positive cells. Biochem Biophys Res Commun.
tent stem cells. Proc Natl Acad Sci U S A. 2012;109: 2010;393:877–82.
12538–43. 147. Nishikawa M, Yanagawa N, Kojima N, Yuri S,
134. Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, Hauser PV, Jo OD, et al. Stepwise renal lineage dif-
et al. Epigenetic memory in induced pluripotent ferentiation of mouse embryonic stem cells tracing
stem cells. Nature. 2010;467:285–90. in vivo development. Biochem Biophys Res
135. Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S, Commun. 2012;417:897–902.
Tan KY, et al. Cell type of origin influences the 148. Oeda S, Hayashi Y, Chan T, Takasato M, Aihara Y,
molecular and functional properties of mouse induced Okabayashi K, et al. Induction of intermediate
18 Translational Research Methods: Renal Stem Cells 565

mesoderm by retinoic acid receptor signaling from 161. Xia Y, Nivet E, Sancho-Martinez I, Gallegos T,
differentiating mouse embryonic stem cells. Int J Suzuki K, Okamura D, et al. Directed differentiation
Dev Biol. 2013;57:383–9. of human pluripotent cells to ureteric bud kidney
149. Niwa H, Ogawa K, Shimosato D, Adachi K. A paral- progenitor-like cells. Nat Cell Biol. 2013;15:
lel circuit of LIF signalling pathways maintains 1507–15.
pluripotency of mouse ES cells. Nature. 2009;460: 162. Takasato M, Er PX, Becroft M, Vanslambrouck JM,
118–22. Stanley EG, Elefanty AG, et al. Directing human
150. McLean AB, D’Amour KA, Jones KL, embryonic stem cell differentiation towards a renal
Krishnamoorthy M, Kulik MJ, Reynolds DM, lineage generates a self-organizing kidney. Nat Cell
et al. Activin a efficiently specifies definitive endo- Biol. 2014;16:118–26.
derm from human embryonic stem cells only when 163. Lam AQ, Freedman BS, Morizane R, Lerou PH,
phosphatidylinositol 3-kinase signaling is suppressed. Valerius MT, Bonventre JV. Rapid and efficient dif-
Stem Cells. 2007;25:29–38. ferentiation of human pluripotent stem cells into inter-
151. Nakaya Y, Sukowati EW, Wu Y, Sheng G. RhoA and mediate mesoderm that forms tubules expressing
microtubule dynamics control cell-basement mem- kidney proximal tubular markers. J Am Soc Nephrol.
brane interaction in EMT during gastrulation. Nat 2014;25:1211–25.
Cell Biol. 2008;10:765–75. 164. Kang M, Han YM. Differentiation of human pluripo-
152. Ye P, Habib SL, Ricono JM, Kim NH, Choudhury tent stem cells into nephron progenitor cells in a serum
GG, Barnes JL, et al. Fibronectin induces ureteric bud and feeder free system. PLoS One. 2014;9:e94888.
cells branching and cellular cord and tubule forma- 165. Ng ES, Davis R, Stanley EG, Elefanty AG. A protocol
tion. Kidney Int. 2004;66:1356–64. describing the use of a recombinant protein-based,
153. Valeriusa MT, Pattersonb LT, Wittec DP, Potter animal product-free medium (APEL) for human
SS. Microarray analysis of novel cell lines embryonic stem cell differentiation as spin embryoid
representing two stages of metanephric mesenchyme bodies. Nat Protoc. 2008;3:768–76.
differentiation. Mech Dev. 2002;112:219–32. 166. Brunskill EW, Aronow BJ, Georgas K, Rumballe B,
154. Viñas JL, Ventayol M, Br€ une B, Jung M, Sola A, Pi F, Valerius MT, Aronow J, et al. Atlas of gene expres-
et al. miRNA let-7e modulates the Wnt pathway and sion in the developing kidney at microanatomic reso-
early nephrogenic markers in mouse embryonic stem lution. Dev Cell. 2008;15:781–91.
cell differentiation. PLoS One. 2013;8:e60937. 167. Gouon-Evans V. The race for regeneration: pluripo-
155. Lin SA, Kolle G, Grimmond SM, Zhou Q, Doust E, tent-stem-cell-derived 3D kidney structures. Cell
Little MH, et al. Subfractionation of differentiating Stem Cell. 2014;14:5–6.
human embryonic stem cell populations allows the 168. Omer D, Harari-Steinberg O, Buzhor E,
isolation of a mesodermal population enriched for Metsuyanim S, Pleniceanu O, Zundelevich A,
intermediate mesoderm and putative renal progeni- et al. Chromatin-modifying agents reactivate embry-
tors. Stem Cells Dev. 2010;19:1637–48. onic renal stem/progenitor genes in human adult kid-
156. Schuldiner M, Yanuka O, Itskovitz-Eldor J, Melton ney epithelial cells but abrogate dedifferentiation and
DA, Benvenisty N. Effects of eight growth factors on stemness. Cell Reprogram. 2013;15(4):281–92.
the differentiation of cells derived from human 169. Friedenstein AJ, Piatetzky-Shapiro II, Petrakova
embryonic stem cells. Proc Natl Acad Sci USA. KV. Osteogenesis in transplants of bone marrow
2000;97:11307–12. cells. J Embryol Exp Morphol. 1966;16(3):381–90.
157. Batchelder CA, Lee CI, Matsell DG, Yoder MC, 170. Caplan AI. Review: mesenchymal stem cells: cell-
Tarantal AF. Renal ontogeny in the rhesus monkey based reconstructive therapy in orthopedics. Tissue
(Macaca mulatta) and directed differentiation of Eng. 2005;11:1198–211.
human embryonic stem cells towards kidney precur- 171. Crisan M, Yap S, Casteilla L, Chen CW, Corselli M,
sors. Differentiation. 2009;78:45–56. Park TS, et al. A perivascular origin for mesenchymal
158. Song B, Smink AM, Jones CV, Callaghan JM, Firth stem cells in multiple human organs. Cell Stem Cell.
SD, Bernard CA, et al. The directed differentiation of 2008;3:301–13.
human iPS cells into kidney podocytes. PLoS One. 172. da Silva ML, Caplan AI, Nardi NB. In search of the
2012;7:e46453. in vivo identity of mesenchymal stem cells. Stem
159. Mae S, Shono A, Shiota F, Yasuno T, Kajiwara M, Cells. 2008;26:2287–99.
Gotoda-Nishimura N, et al. Monitoring and robust 173. Caplan AI, Correa D. The MSC: an injury drugstore.
induction of nephrogenic intermediate mesoderm Cell Stem Cell. 2011;9:11–5.
from human pluripotent stem cells. Nat Commun. 174. Bjornson CR, Rietze RL, Reynolds BA, Magli MC,
2013;4:1367. Vescovi AL. Turning brain into blood: a hematopoi-
160. Narayanan K, Schumacher KM, Tasnim F, etic fate adopted by adult neural stem cells in vivo.
Kandasamy K, Schumacher A, Ni M, et al. Human Science. 1999;283:534–7.
embryonic stem cells differentiate into functional 175. Brazelton TR, Rossi FM, Keshet GI, Blau HM. From
renal proximal tubular-like cells. Kidney Int. 2013; marrow to brain: expression of neuronal phenotypes
83:593–603. in adult mice. Science. 2000;290:1775–9.
566 K. Osafune

176. Mezey E, Chandross KJ, Harta G, Maki RA, 191. Herrera MB, Bussolati B, Bruno S, Morando L,
McKercher SR. Turning blood into brain: cells bear- Mauriello-Romanazzi G, Sanavio F,
ing neuronal antigens generated in vivo from bone et al. Exogenous mesenchymal stem cells localize to
marrow. Science. 2000;290:1779–82. the kidney by means of CD44 following acute tubular
177. Lagasse E, Connors H, Al-Dhalimy M, Reitsma M, injury. Kidney Int. 2007;72:430–41.
Dohse M, Osborne L, et al. Purified hematopoietic 192. Lange C, Togel F, Ittrich H, Clayton F, Nolte-
stem cells can differentiate into hepatocytes in vivo. Ernsting C, Zander AR, et al. Administered mesen-
Nat Med. 2000;6(11):1229–34. chymal stem cells enhance recovery from ischemia/
178. Clarke DL, Johansson CB, Wilbertz J, Veress B, reperfusion-induced acute renal failure in rats. Kidney
Nilsson E, Karlström H, et al. Generalized potential Int. 2005;68:1613–7.
of adult neural stem cells. Science. 2000;288(5471): 193. Cheng K, Rai P, Plagov A, Lan X, Kumar D,
1660–3. Salhan D, et al. Transplantation of bone marrow-
179. Krause DS, Theise ND, Collector MI, Henegariu O, derived MSCs improves cisplatinum-induced renal
Hwang S, Gardner R, et al. Multi-organ, multi-lineage injury through paracrine mechanisms. Exp Mol
engraftment by a single bone marrow-derived stem Pathol. 2013;94:466–73.
cell. Cell. 2001;105(3):369–77. 194. Bi B, Schmitt R, Israilova M, Nishio H, Cantley
180. Terada N, Hamazaki T, Oka M, Hoki M, Mastalerz LG. Stromal cells protect against acute tubular injury
DM, Nakano Y, et al. Bone marrow cells adopt the via an endocrine effect. J Am Soc Nephrol.
phenotype of other cells by spontaneous cell fusion. 2007;18:2486–96.
Nature. 2002;416(6880):542–5. 195. Fang TC, Alison MR, Cook HT, Jeffery R, Wright
181. Ying QL, Nichols J, Evans EP, Smith AG. Changing NA, Poulsom R. Proliferation of bone marrow-
potency by spontaneous fusion. Nature. 2002;416 derived cells contributes to regeneration after folic
(6880):545–8. acid-induced acute tubular injury. J Am Soc Nephrol.
182. Herrera MB, Bussolati B, Bruno S, Fonsato V, 2005;16:1723–32.
Romanazzi GM, Camussi G. Mesenchymal stem 196. Stokman G, Leemans JC, Claessen N, Weening JJ,
cells contribute to the renal repair of acute tubular Florquin S. Hematopoietic stem cell mobilization
epithelial injury. Int J Mol Med. 2004;14:1035–41. therapy accelerates recovery of renal function inde-
183. Behr L, Hekmati M, Fromont G, Borenstein N, Noel pendent of stem cell contribution. J Am Soc Nephrol.
LH, Lelievre-Pegorier M, et al. Intra renal arterial 2005;16:1684–92.
injection of autologous mesenchymal stem cells in 197. Nishida M, Fujimoto S, Toiyama K, Sato H, Hamaoka
an ovine model in the post ischemic kidney. Nephron. K. Effect of hematopoietic cytokines on renal func-
2007;107:65–76. tion in cisplatin-induced ARF in mice. Biochem
184. Wong CY, Cheong SK, Mok PL, Leong CF. Differ- Biophys Res Commun. 2004;324:341–7.
entiation of human mesenchymal stem cells into 198. Iwasaki M, Adachi Y, Minamino K, Suzuki Y,
mesangial cells in post-glomerular injury murine Zhang Y, Okigaki M, et al. Mobilization of bone
model. Pathology. 2008;40:52–7. marrow cells by G-CSF rescues mice from cisplatin-
185. Humphreys BD, Bonventre JV. Mesenchymal stem induced renal failure, and M-CSF enhances the effects
cells in acute kidney injury. Annu Rev Med. of G-CSF. J Am Soc Nephrol. 2005;16:658–66.
2008;59:311–25. 199. Togel F, Isaac J, Westenfelder C. Hematopoietic stem
186. Morigi M, Benigni A. Mesenchymal stem cells and cell mobilization-associated granulocytosis severely
kidney repair. Nephrol Dial Transplant. 2013;28: worsens acute renal failure. J Am Soc Nephrol.
788–93. 2004;15:1261–7.
187. Roufosse C, Cook HT. Stem cells and renal regener- 200. Imberti B, Morigi M, Tomasoni S, Rota C, Corna D,
ation. Nephron Exp Nephrol. 2008;109:e39–45. Longaretti L, et al. Insulin-like growth factor-1 sus-
188. Masuya M, Drake CJ, Fleming PA, Reilly CM, tains stem cell mediated renal repair. J Am Soc
Zeng H, Hill WD, et al. Hematopoietic origin of Nephrol. 2007;18:2921–8.
glomerular mesangial cells. Blood. 2003;101(6): 201. Ninichuk V, Gross O, Segerer S, Hoffmann R,
2215–8. Radomska E, Buchstaller A, et al. Multipotent mes-
189. Yokoo T, Ohashi T, Shen JS, Sakurai K, Miyazaki Y, enchymal stem cells reduce interstitial fibrosis but do
Utsunomiya Y, et al. Human mesenchymal stem cells not delay progression of chronic kidney disease in
in rodent whole-embryo culture are reprogrammed collagen4A3-deficient mice. Kidney Int. 2006;70:
to contribute to kidney tissues. Proc Natl Acad Sci 121–9.
U S A. 2005;102(9):3296–300. 202. Semedo P, Correa-Costa M, Antonio Cenedeze M,
190. Togel F, Hu Z, Weiss K, Isaac J, Lange C, Maria Avancini Costa Mal-heiros D, Antonia dos
Westenfelder C. Administered mesenchymal stem Reis M, Shimizu MH, et al. Mesenchymal stem
cells protect against ischemic acute renal failure cellsattenuate renal fibrosis through immune modula-
through differentiation-independent mechanisms. tion and remodeling properties in a rat remnant kidney
Am J Physiol Renal Physiol. 2005;289:F31–42. model. Stem Cells. 2009;27:3063–73.
18 Translational Research Methods: Renal Stem Cells 567

203. Choi S, Park M, Kim J, Hwang S, Park S, Lee Y. The 218. Lee PT, Lin HH, Jiang ST, Lu PJ, Chou KJ, Fang HC,
role of mesenchymal stem cells in the functional et al. Mouse kidney progenitor cells accelerate renal
improvement of chronic renal failure. Stem Cells regeneration and prolong survival after ischemic
Dev. 2009;18:521–9. injury. Stem Cells. 2010;28(3):573–84.
204. Togel FE, Westenfelder C. Mesenchymal stem cells: a 219. Maeshima A, Yamashita S, Nojima Y. Identification
new therapeutic tool for AKI. Nat Rev Nephrol. of renal progenitor-like tubular cells that participate in
2010;6:179–83. the regeneration processes of the kidney. J Am Soc
205. Kusaba T, Lalli M, Kramann R, Kobayashi A, Nephrol. 2003;14:3138–46.
Humphreys BD. Differentiated kidney epithelial 220. Yamashita S, Maeshima A, Nojima Y. Involvement of
cells repair injured proximal tubule. Proc Natl Acad renal progenitor tubular cells in epithelial-to-mesen-
Sci U S A. 2014;111:1527–32. chymal transition in fibrotic rat kidneys. J Am Soc
206. Vogetseder A, Picard N, Gaspert A, Walch M, Nephrol. 2005;16(7):2044–51.
Kaissling B, Le Hir M. Proliferation capacity of the 221. Maeshima A, Sakurai H, Nigam SK. Adult kidney
renal proximal tubule involves the bulk of differenti- tubular cell population showing phenotypic plasticity,
ated epithelial cells. Am J Physiol Cell Physiol. tubulogenic capacity, and integration capability into
2008;294(1):C22–8. developing kidney. J Am Soc Nephrol. 2006;17
207. Vogetseder A, Palan T, Bacic D, Kaissling B, Le Hir (1):188–98.
M. Proximal tubular epithelial cells are generated by 222. Kitamura S, Yamasaki Y, Kinomura M, Sugaya T,
division of differentiated cells in the healthy kidney. Sugiyama H, Maeshima Y, et al. Establishment and
Am J Physiol Cell Physiol. 2007;292(2):C807–13. characterization of renal progenitor like cells from S3
208. Berger K, Bangen JM, Hammerich L, Liedtke C, segment of nephron in rat adult kidney. FASEB
Floege J, Smeets B, et al. Origin of regenerating J. 2005;19(13):1789–97.
tubular cells after acute kidney injury. Proc Natl 223. Kinomura M, Kitamura S, Tanabe K, Ichinose K,
Acad Sci U S A. 2014;111(4):1533–8. Hirokoshi K, Takazawa Y, et al. Amelioration of
209. Rinkevich Y, Montoro DT, Contreras-Trujillo H, cisplatin-induced acute renal injury by renal
Harari-Steinberg O, Newman AM, Tsai JM, et al. In progenitor-like cells derived from the adult rat kidney.
vivo clonal analysis reveals lineage-restricted progen- Cell Transplant. 2008;17:143–58.
itor characteristics in mammalian kidney develop- 224. Romagnani P, Remuzzi G. Renal progenitors in
ment, maintenance, and regeneration. Cell Rep. non-diabetic and diabetic nephropathies. Trends
2014;7(4):1270–83. Endocrinol Metab. 2013;24:13–20.
210. Miyajima A, Tanaka M, Itoh T. Stem/progenitor cells 225. Rizzo P, Perico N, Gagliardini E, Novelli R, Alison
in liver development, homeostasis, regeneration, and MR, Remuzzi G, et al. Nature and mediators of pari-
reprogramming. Cell Stem Cell. 2014;14(5):561–74. etal epithelial cell activation in glomerulonephritides
211. Grompe M. Liver stem cells, where art thou? Cell of human and rat. Am J Pathol. 2013;183(6):
Stem Cell. 2014;15(3):257–8. 1769–78.
212. Little MH, Bertram JF. Is there such a thing as a renal 226. Bruno S, Bussolati B, Grange C, Collino F, di
stem cell? J Am Soc Nephrol. 2009;20(10):2112–7. Cantogno LV, Herrera MB, et al. Isolation and char-
213. Anglani F, Mezzabotta F, Ceol M, Cristofaro R, Del acterization of resident mesenchymal stem cells in
Prete D, D’Angelo A. The regenerative potential of human glomeruli. Stem Cells Dev. 2009;18(6):
the kidney: what can we learn from developmental 867–80.
biology? Stem Cell Rev. 2010;6:650–7. 227. Pippin JW, Sparks MA, Glenn ST, Buitrago S,
214. Oliver JA, Maarouf O, Cheema FH, Martens TP, Coffman TM, Duffield JS, et al. Cells of renin lineage
Al-Awqati Q. The renal papilla is a niche for adult are progenitors of podocytes and parietal epithelial
kidney stem cells. J Clin Invest. 2004;114:795–804. cells in experimental glomerular disease. Am J
215. Dekel B, Zangi L, Shezen E, Reich-Zeliger S, Pathol. 2013;183(2):542–57.
Eventov-Friedman S, Katchman H, et al. Isolation 228. Li J, Ariunbold U, Suhaimi N, Sunn N, Guo J,
and characterization of nontubular sca-1 + lin- McMahon JA, et al. Collecting duct-derived cells
multipotent stem/progenitor cells from adult mouse display mesenchymal stem cell properties and retain
kidney. J Am Soc Nephrol. 2006;17(12):3300–14. selective in vitro and in vivo epithelial capacity. J Am
216. Hishikawa K, Marumo T, Miura S, Nakanishi A, Soc Nephrol. 2015;23(1):81–94.
Matsuzaki Y, Shibata K, et al. Musculin/MyoR is 229. Buzhor E, Harari-Steinberg O, Omer D,
expressed in kidney side population cells and can Metsuyanim S, Jacob-Hirsch J, Noiman T,
regulate their function. J Cell Biol. 2005;169(6): et al. Kidney spheroids recapitulate tubular organoids
921–8. leading to enhanced tubulogenic potency of human
217. Bussolati B, Bruno S, Grange C, Buttiglieri S, kidney-derived cells. Tissue Eng A. 2011;17:
Deregibus MC, Cantino D, et al. Isolation of renal 2305–19.
progenitor cells from adult human kidney. Am J 230. Buzhor E, Omer D, Harari-Steinberg O, Dotan Z,
Pathol. 2005;166(2):545–55. Vax E, Pri-Chen S, et al. Reactivation of NCAM1
568 K. Osafune

defines a subpopulation of human adult kidney epi- (RCC): a cochrane systematic review of published
thelial cells with clonogenic and stem/progenitor randomized trials. BJU Int. 2011;108:1556–63.
properties. Am J Pathol. 2013;183(5):1621–33. 245. Sell S. On the stem cell origin of cancer. Am J Pathol.
231. Bombelli S, Zipeto MA, Torsello B, Bovo G, Di 2010;176:2584–94.
Stefano V, Bugarin C, et al. PKH (high) cells within 246. Bussolati B, Bruno S, Grange C, Ferrando U,
clonal human nephrospheres provide a purified adult Camussi G. Identification of a tumor-initiating stem
renal stem cell population. Stem Cell Res. 2013; cell population in human renal carcinomas. FASEB
11(3):1163–77. J. 2008;22:3696–705.
232. Asakura A, Rudnicki MA. Side population cells from 247. Zhong Y, Guan K, Guo S, Zhou C, Wang D, Ma W,
diverse adult tissues are capable of in vitro hemato- et al. Spheres derived from the human SK-RC-42
poietic differentiation. Exp Hematol. 2002;30(11): renal cell carcinoma cell line are enriched in cancer
1339–45. stem cells. Cancer Lett. 2010;299(2):150–60.
233. Iwatani H, Ito T, Imai E, Matsuzaki Y, Suzuki A, 248. Addla SK, Brown MD, Hart CA, Ramani VA, Clarke
Yamato M, et al. Hematopoietic and NW. Characterization of the Hoechst 33342 side pop-
nonhematopoietic potentials of Hoechst(low)/side ulation from normal and malignant human renal epi-
population cells isolated from adult rat kidney. Kid- thelial cells. Am J Physiol Renal Physiol. 2008;295
ney Int. 2004;65(5):1604–14. (3):F680–7.
234. Challen GA, Bertoncello I, Deane JA, Ricardo SD, 249. Coppes MJ, Pritchard-Jones K. Principles of Wilms’
Little MH. Kidney side population reveals tumor biology. Urol Clin North Am. 2000;27(3):
multilineage potential and renal functional capacity 423–33.
but also cellular heterogeneity. J Am Soc Nephrol. 250. Dénes FT, Duarte RJ, Cristófani LM, Lopes RI.
2006;17(7):1896–912. Pediatric genitourinary oncology. Front Pediatr.
235. Gupta S, Verfaillie C, Chmielewski D, Kren S, 2013;1:48.
Eidman K, Connaire J, et al. Isolation and character- 251. Metsuyanim S, Pode-Shakked N, Schmidt-Ott KM,
ization of kidney-derived stem cells. J Am Soc Keshet G, Rechavi G, Blumental D,
Nephrol. 2006;17(11):3028–40. et al. Accumulation of malignant renal stem cells is
236. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, associated with epigenetic changes in normal renal
Keene CD, Ortiz-Gonzalez XR, et al. Pluripotency of progenitor genes. Stem Cells. 2008;26(7):1808–17.
mesenchymal stem cells derived from adult marrow. 252. Aiden AP, Rivera MN, Rheinbay E, Ku M, Coffman
Nature. 2002;418:41–9. EJ, Truong TT, et al. Wilms tumor chromatin profiles
237. Goodell MA, Brose K, Paradis G, Conner AS, Mulli- highlight stem cell properties and a renal develop-
gan RC. Isolation and functional properties of murine mental network. Cell Stem Cell. 2010;6(6):591–602.
hematopoietic stem cells that are replicating in vivo. J 253. Pode-Shakked N, Dekel B. Wilms tumor–a renal stem
Exp Med. 1996;183(4):1797–806. cell malignancy? Pediatr Nephrol. 2011;26(9):1535–43.
238. Scadden DT. The stem-cell niche as an entity of 254. Pode-Shakked N, Shukrun R, Mark-Danieli M,
action. Nature. 2006;441(7097):1075–9. Tsvetkov P, Bahar S, Pri-Chen S, et al. The isolation
239. da Silva ML, Chagastelles PC, Nardi NB. Mesenchy- and characterization of renal cancer initiating cells
mal stem cells reside in virtually all post-natal organs from human Wilms’ tumour xenografts unveils new
and tissues. J Cell Sci. 2006;119(Pt 11):2204–13. therapeutic targets. EMBO Mol Med. 2013;5(1):
240. Díaz-Flores L, Gutiérrez R, Madrid JF, Varela H, 18–37.
Valladares F, Acosta E, et al. Pericytes. 255. Shukrun R, Pode-Shakked N, Pleniceanu O, Omer D,
Morphofunction, interactions and pathology in a qui- Vax E, Peer E, et al. Wilms’ tumor blastemal stem
escent and activated mesenchymal cell niche. Histol cells dedifferentiate to propagate the tumor bulk.
Histopathol. 2009;24(7):909–69. Stem Cell Reports. 2014;3(1):24–33.
241. Covas DT, Panepucci RA, Fontes AM, Silva Jr WA, 256. Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM,
Orellana MD, Freitas MC, et al. Multipotent mesen- Mitsumoto H, Chung W, et al. Induced pluripotent
chymal stromal cells obtained from diverse human stem cells generated from patients with ALS can be
tissues share functional properties and gene- differentiated into motor neurons. Science. 2008;321:
expression profile with CD146+ perivascular cells 1218–21.
and fibroblasts. Exp Hematol. 2008;36(5):642–54. 257. Park IH, Arora N, Huo H, Maherali N, Ahfeldt T,
242. Koul H, Huh JS, Rove KO, Crompton L, Koul S, Shimamura A, et al. Disease-specific induced plurip-
Meacham RB, et al. Molecular aspects of renal cell otent stem cells. Cell. 2008;134:877–86.
carcinoma: a review. Am J Cancer Res. 2011;1(2): 258. Inoue H, Nagata N, Kurokawa H, Yamanaka S. iPS
240–54. cells: a game changer for future medicine. EMBO
243. Kovacs G, Akhtar M, Beckwith BJ, Bugert P, Cooper J. 2014;33(5):409–17.
CS, Delahunt B, et al. The Heidelberg classification of 259. Li Y, Kandasamy K, Chuah JK, Lam YN, Toh WS,
renal cell tumors. J Pathol. 1997;183:131–6. Oo ZY, et al. Identification of nephrotoxic compounds
244. Coppin C, Kollmannsberger C, Le L, Porzsolt F, Wilt with embryonic stem-cell-derived human renal prox-
TJ. Targeted therapy for advanced renal cell cancer imal tubular-like cells. Mol Pharm. 2014;11:1982–90.
18 Translational Research Methods: Renal Stem Cells 569

260. Osafune K. Will it be possible to generate kidney 263. Kobayashi T, Yamaguchi T, Hamanaka S, Kato-
tissue from induced pluripotent stem cells for regen- Itoh M, Yamazaki Y, Ibata M, et al. Generation of
erative therapy? Regen Med. 2014;9:9–12. rat pancreas in mouse by interspecific blastocyst
261. Ross EA, Williams MJ, Hamazaki T, Terada N, Clapp injection of pluripotent stem cells. Cell. 2010;142:
WL, Adin C, et al. Embryonic stem cells proliferate 787–99.
and differentiate when seeded into kidney scaffolds. 264. Usui J, Kobayashi T, Yamaguchi T, Knisely AS,
J Am Soc Nephrol. 2009;20:2338–47. Nishinakamura R, Nakauchi H. Generation of kidney
262. Song JJ, Guyette JP, Gilpin SE, Gonzalez G, Vacanti from pluripotent stem cells via blastocyst comple-
JP, Ott HC. Regeneration and experimental orthotopic mentation. Am J Pathol. 2012;180:2417–26.
transplantation of a bioengineered kidney. Nat Med.
2013;19:646–51.
Translational Research Methods:
Tissue Engineering of the Kidney 19
and Urinary Tract

Austin G. Hester and Anthony Atala

Contents Introduction
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
Patients suffering from diseased or injured geni-
The Basic Components of Tissue Engineering . . . 572
Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572
tourinary organs are often treated with reconstruc-
Biomaterials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 576 tive surgery or transplants, but there is a severe
shortage of donor tissue and organs. This shortage
Engineering Specific Tissues and Organs . . . . . . . . 578
Urethra . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578 worsens yearly as modern medicine increases the
Bladder . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579 human lifespan. The aging population grows, and
Kidney . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581 the need for organs grows with it. Physicians and
Summary and Conclusions . . . . . . . . . . . . . . . . . . . . . . . . 588 scientists have begun to look to the fields of
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 588
regenerative medicine and tissue engineering to
provide new options for these patients. These
fields apply the principles of cell transplantation,
material science, and bioengineering to construct
biological substitutes that can significantly
improve the quality of life of the urologic patient
by eliminating the need for intensive grafting pro-
cedures or transplant surgery.
Tissue engineering, one of the major compo-
nents of regenerative medicine, follows the prin-
ciples of cell transplantation, materials science,
and engineering to develop biological substitutes
that can restore and maintain normal organ func-
tion. Tissue engineering strategies generally fall
into two categories: the use of acellular matrices
designed to direct the body’s natural ability to use
its own cells to regenerate damaged tissue and the
use of matrices seeded with cells in the laboratory
to produce novel tissues and organs. Acellular
A.G. Hester • A. Atala (*) tissue matrices are usually prepared by
School of Medicine, Department of Urology, Wake Forest
manufacturing artificial scaffolds or by removing
Institute for Regenerative Medicine, Wake Forest
University, Winston Salem, NC, USA cellular components from donor tissues via
e-mail: ahester@wakehealth.edu; aatala@wfubmc.edu mechanical and chemical manipulation to
# Springer-Verlag Berlin Heidelberg 2016 571
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_17
572 A.G. Hester and A. Atala

produce collagen-rich matrices [1–4]. These This chapter will review the major components
matrices slowly degrade after implantation and of most tissue engineering techniques and will
are replaced by the extracellular matrix (ECM) describe how these techniques are being applied
proteins secreted by the ingrowing cells. Cells to the reconstruction and regeneration of the gen-
themselves can also be used for therapy via injec- itourinary system.
tion, either with carriers such as hydrogels or
alone.
The most common way to use cells in tissue The Basic Components of Tissue
engineering is to obtain a small piece of donor tissue Engineering
and dissociate it into individual cells in the labora-
tory. These cells are either implanted directly into Cells
the host or are expanded in culture and attached to a
support matrix. The cell/matrix construct is then Native Cells
reimplanted into the host. The source of the donor In the past, one of the limitations of applying cell-
tissue can be heterologous (such as bovine), alloge- based regenerative medicine techniques to organ
neic (same species, different individual), or autolo- replacement was the inherent difficulty of grow-
gous. Ideally, autologous cells are used, because in ing certain cell types in large quantities. Even
this case, both structural and functional tissue when some organs, such as the liver, have a high
replacement will usually occur with minimal com- regenerative capacity in vivo, cell growth and
plications. To accomplish this, a biopsy of tissue is expansion in vitro can be difficult. By studying
obtained from a host, the cells are dissociated and the privileged sites for committed precursor cells
expanded in culture, and the expanded cells are in these organs, as well as by exploring the con-
implanted back into the same host [2, 5–12]. The ditions that promote differentiation and/or self-
use of autologous cells, although it may cause an renewal of these cells, it has been possible to
inflammatory response, avoids rejection, and thus, overcome some of the obstacles that limit cell
the deleterious side effects of lifelong immunosup- expansion in vitro. One example is the urothelial
pression can be avoided. cell. Typically, in the past, intestinal segments
However, for many patients with extensive have been used to replace urinary structures in
end-stage organ failure, a tissue biopsy may not patients with congenital or acquired defects. How-
yield enough normal cells for expansion and ever, this comes with a series of complications,
transplantation. In other instances, primary autol- including metabolic imbalances, urinary stone
ogous cells cannot be expanded from a particular formation, hypocontractility, and neoplasia, to
organ, such as the pancreas. In these situations, name a few [13]. Several protocols have been
stem cells are envisioned as an alternative source developed that identify the undifferentiated cells
of cells from which the desired tissue can be in a mixed culture of cells isolated from the uri-
derived. Stem cells can be derived from discarded nary tract and keep them undifferentiated during
human embryos (human embryonic stem cells), their growth phase, which now allows expansion
from fetal tissue, or from adult sources (bone of a urothelial culture that initially covered a sur-
marrow, fat, skin). However, there are ethical face area of 1 cm2 to one covering a surface area
issues involved in the use of embryonic stem of 4202 m2 (the equivalent of one football field)
cells, and most human applications are currently within 8 weeks [11, 14–17]. More recent studies
banned in the United States. Despite this, the field have developed methods of isolating these cells
of stem cell biology is advancing rapidly, and from urine samples and bladder washings rather
cutting-edge techniques such as therapeutic clon- than from more invasive endoscopic and surgical
ing and somatic cell reprogramming circumvent techniques [18, 19]. More studies have indicated
some of the ethical questions and offer a poten- that it is possible to collect autologous bladder
tially limitless source of these cells for tissue cells from human patients, expand them in cul-
engineering applications. ture, and return them to the donor in sufficient
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 573

quantities for reconstructive purposes [11, 15–17, demonstrated the ability to differentiate bone mar-
20–22]. In fact, seven pediatric patients requiring row mesenchymal stem cells into smooth muscle
cystoplasty secondary to a myelomeningocele and urothelium-like cells, resulting in another
were implanted with bladders grown from autol- potential cellular basis for bladder tissue engineer-
ogous urothelial and muscle cells [23]. Major ing [36]. Additionally, another study indicated
advances in cell culture techniques have been that urine-derived stem cells demonstrate even
made within the past decade, and these techniques more growth potential than bone marrow mesen-
make use of autologous cells possible for clinical chymal stem cells, thus producing yet another
application. cellular background. These cells have demon-
strated the ability to expand in culture and to
Embryonic Stem Cells differentiate into multiple cell lines [37].
In 1981, pluripotent cells were found in the inner However, in addition to the ethical dilemma
cell mass of the human embryo, and the term surrounding the use of embryonic stem cells,
“human embryonic stem cell” was coined their clinical application is limited because they
[24]. These cells are able to differentiate into all represent an allogeneic resource and thus have the
cells of the human body, excluding placental cells potential to evoke an immune response. New stem
(only cells from the morula are totipotent, i.e., cell technologies (such as somatic cell nuclear
able to develop into all cells of the human body). transfer and reprogramming) promise to over-
These cells have great therapeutic potential, but come this limitation.
their use is limited by both biological and ethical
factors. Therapeutic Cloning (Somatic Cell
The political controversy surrounding stem Nuclear Transfer)
cells began in 1998 with the creation of human Somatic cell nuclear transfer (SCNT), or thera-
embryonic stem (hES) cells derived from peutic cloning, entails the removal of an oocyte
discarded embryos. hES were isolated from the nucleus in culture, followed by its replacement
inner cell mass of a blastocyst (an embryo 5 days with a nucleus derived from a somatic cell
postfertilization) using an immunosurgical tech- obtained from a patient. Activation with
nique. Given that some cells cannot be expanded chemicals or electricity stimulates cell division
ex vivo, ES cells could be the ideal resource for up to the blastocyst stage.
tissue engineering because of their fundamental At this point, it is extremely important to dif-
properties: the ability to self-renew indefinitely ferentiate between the two types of cloning that
and the ability to differentiate into cells from all exist – reproductive cloning and therapeutic clon-
three embryonic germ layers. Skin and neurons ing. Both involve the insertion of donor DNA into
have been formed, indicating ectodermal an enucleated oocyte to generate an embryo that
differentiation [25–28]. Blood, cardiac cells, car- has identical genetic material to its DNA source.
tilage, endothelial cells, and muscle have been However, the similarities end there. In reproduc-
formed, indicating mesodermal differentiation tive cloning, the embryo is then implanted into the
[29–31]. Pancreatic cells have been formed, indi- uterus of a pseudopregnant female to produce an
cating endodermal differentiation [32]. In addi- infant that is a clone of the donor. A world-famous
tion, as further evidence of their pluripotency, example of this type of cloning resulted in the
embryonic stem cells can form embryoid bodies, birth of a sheep named Dolly in 1997 [38]. How-
which are cell aggregations that contain all three ever, there are many ethical concerns surrounding
embryonic germ layers while in culture, and can such practices, and as a result, reproductive clon-
form teratomas in vivo [33]. These cells have ing has been banned in most countries.
demonstrated longevity in culture and can main- While therapeutic cloning also produces an
tain their undifferentiated state for at least 80 pas- embryo that is genetically identical to the donor,
sages when grown using current published this process is used to generate blastocysts that are
protocols [34, 35]. Recent studies have explanted and grown in culture, rather than in
574 A.G. Hester and A. Atala

Fig. 1 Strategies for therapeutic cloning in regenerative medicine

utero. Embryonic stem cell lines can then be fibroblast into an enucleated oocyte [43].
derived from these blastocysts, which are only Although the blastocyst was implanted (reproduc-
allowed to grow up to a 100-cell stage. At this tive cloning), the purpose was to generate renal,
time, the inner cell mass is isolated and cultured, cardiac, and skeletal muscle cells, which were
resulting in ES cells that are genetically identical then harvested, expanded in vitro, and seeded
to the patient. This process is detailed in Fig. 1. onto biodegradable scaffolds. These scaffolds
It has been shown that nuclear-transferred ES cells were then implanted into the donor steer from
derived from fibroblasts, lymphocytes, and olfac- whom the cells were cloned to determine if cells
tory neurons are pluripotent and can generate live were histocompatible. Analysis revealed that
pups after injection into blastocysts. This shows cloned renal cells showed no evidence of T-cell
that cells generated by SCNT have the same response, suggesting that rejection will not neces-
developmental potential as blastocysts that are sarily occur in the presence of oocyte-derived
fertilized and produced naturally [39–42]. In addi- mtDNA. This finding represents a step forward
tion, the ES cells generated by SCNT are perfectly in overcoming the histocompatibility problem of
matched to the patient’s immune system and no stem cell therapy.
immunosuppressants would be required to pre- Although promising, SCNT has certain limita-
vent rejection should these cells be used in tissue tions that require further improvement before its
engineering applications. clinical application, in addition to the ethical con-
Although ES cells derived from SCNT contain siderations regarding the potential of the resulting
the nuclear genome of the donor cells, mitochon- embryos to develop into cloned embryos if
drial DNA (mtDNA) contained in the implanted into a uterus. In addition, this technique
oocyte could lead to immunogenicity after trans- has not been shown to work in humans. The initial
plantation. To assess the histocompatibility of failures and fraudulent reports of nuclear transfer
tissue generated using SCNT, Lanza in humans reduced enthusiasm for human appli-
et al. microinjected the nucleus of a bovine skin cations [44–46], although it was recently reported
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 575

that nonhuman primate ES cell lines were gener- they possessed a distinct gene expression signa-
ated by SCNT of nuclei from adult skin fibroblasts ture that was different from that of ES cells.
[47, 48]. In addition, the epigenetic state of the iPS cells
Before SCNT-derived ES cells can be used as was somewhere between that found in somatic
clinical therapy, careful assessment of quality of cells and that found in ES cells, suggesting that
the lines must be determined. For example, some the reprogramming was incomplete.
cell lines generated by SCNT have contained These results were improved significantly by
chromosomal translocations, and it is not known Wernig and Jaenisch in July 2007 [52]. In this
whether these abnormalities originated from study, DNA methylation, gene expression pro-
aneuploid embryos or if they occurred during ES files, and the chromatin state of the reprogrammed
cell isolation and culture. In addition, the low cells were similar to those of ES cells. Teratomas
efficiency of SNCT (0.7 %) and the inadequate induced by these cells contained differentiated
supply of human oocytes further hinder the ther- cell types representing all three embryonic germ
apeutic potential of this technique, as well as the layers. Most importantly, the reprogrammed cells
continued need for cell cycle synchronization from this experiment were able to form viable
between the donor cells and recipient oocytes chimeras and contribute to the germ line like ES
[49, 50]. Still, these studies renew the hope that cells, suggesting that these iPS cells were
ES cell lines could one day be generated from completely reprogrammed.
human cells to produce patient-specific stem It has recently been shown that reprogramming
cells with the potential to cure many human dis- of human cells is possible [53, 54]. Yamanaka
eases that are currently untreatable. generated human iPS cells that are similar to
hES cells in terms of morphology, proliferation,
Reprogrammed Somatic Cells gene expression, surface markers, and teratoma
Recently, exciting reports of the successful trans- formation. Thompson’s group showed that retro-
formation of adult cells into pluripotent stem cells viral transduction of the stem cell markers OCT4,
through a type of genetic “reprogramming” have SOX2, NANOG, and LIN28 could generate plu-
been published. Reprogramming is a technique ripotent stem cells. However, in both studies, the
that involves dedifferentiation of adult somatic human iPS cells were similar but not identical to
cells to produce patient-specific pluripotent stem hES cells. The major concern with this technique
cells, eliminating the need to create embryos. is that these retroviral genes could increase tumor-
Cells generated by reprogramming would be igenesis [49]. A study by Okita in 2007 demon-
genetically identical to the somatic cells (and strated 20 % of subjects generated from Nanog-
thus, the patient who donated these cells) and iPS cells developed tumors secondary to c-myc
would not be rejected. Yamanaka was the first to retroviral expression [49, 55]. However, this study
discover that mouse embryonic fibroblasts along with another study has indicated that by
(MEFs) and adult mouse fibroblasts could be using transient expression, there is silencing of
reprogrammed into an “induced pluripotent state the viral transcripts and thus can be used only
(iPS)” [51]. These iPS cells possessed the immor- for induction of pluripotency rather than mainte-
tal growth characteristics of self-renewing ES nance [49, 55, 56]. The study by Meissner has also
cells, expressed genes specific for ES cells, and indicated that iPS cells do not necessarily require
generated embryoid bodies in vitro and teratomas transgenic donor cells but can in fact be formed
in vivo. When iPS cells were injected into mouse from non-transgenic donor cells [56].
blastocysts, they contributed to a variety of cell Even more recently, studies are now indicating
types. However, although iPS cells selected in this there is a more effective manner to reprogram
way were pluripotent, they were not identical to somatic cells using a technique called stimulus-
ES cells. Unlike ES cells, chimeras made from iPS triggered acquisition of pluripotency (STAP),
cells did not result in full-term pregnancies. Gene which uses external stimuli to induce pluripotency
expression profiles of the iPS cells showed that in somatic cells as opposed to nuclear
576 A.G. Hester and A. Atala

manipulation [57, 58]. In particular, Obokata human fetal somatic cells from the chorion at the
noted that cells treated in a low-pH environment time of birth and thus stir less of an ethical debate
(pH 5.4–5.8) were able to be reprogrammed to [60, 61]. They could be preserved for self-use and
express the gene Oct4-GFP, a marker of used without rejection, or they could be banked.
pluripotency, with considerable decreases in the A bank of 100,000 specimens could potentially
methylation patterns in the regulatory regions. supply 99 % of the US population with a perfect
In addition, cells produced via the STAP method genetic match for transplantation. Such a bank
have been shown to contain both embryonic and may be easier to create than with other cell
placental tissues [57, 58]. While this technique sources, since there are approximately 4.5 million
still requires further exploration and many ques- births per year in the United States [59].
tions remain, including why this mechanism
exists, this is certainly a promising path for tissue
engineering. Biomaterials

Placental and Amniotic Fluid Stem Cells In the most common tissue engineering proce-
Recently, it has been shown that pluripotent cells dures, isolated cells are seeded onto a scaffold
may be derived from the amniotic fluid and pla- composed of an appropriate biomaterial. These
centa. Both amniotic fluid and placenta are known biomaterials replicate the biologic and mechanical
to contain multiple partially differentiated cell function of the native extracellular matrix (ECM)
types derived from the developing fetus. Stem found in tissues in the body by serving as an
cell populations have been isolated from these artificial ECM. Biomaterials provide a three-
sources. Called amniotic fluid and placental stem dimensional space for the cells to develop into
cells (AFPSC), they express embryonic and adult new tissues with appropriate structure and func-
stem cell markers [59]. The undifferentiated stem tion. They can also allow delivery of appropriate
cells expand extensively without a feeder cell bioactive factors (e.g., cell adhesion peptides,
layer and double every 36 h. Unlike human growth factors) to the developing tissue [62] to
embryonic stem cells, the AFPSC do not form help regulate cellular function. As the majority of
tumors in vivo. Lines maintained for over mammalian cell types are anchorage-dependent
250 population doublings retained long telomeres and will die if no cell adhesion substrate is avail-
and a normal complement of chromosomes. AFS able, biomaterials provide this substrate that can
cells are broadly multipotent, and human lines can deliver cells to specific sites in the body with high
be induced to differentiate into cell types loading efficiency. Biomaterials can also provide
representing each embryonic germ layer, includ- mechanical support against in vivo forces so that
ing cells of adipogenic, osteogenic, myogenic, the predefined three-dimensional structure of the
endothelial, neuronal, and hepatic lineages. engineered implant is maintained during tissue
Examples of differentiated cells derived from development.
AFS cells and displaying specialized functions The ideal biomaterial should be biodegradable
include neuronal lineage secreting the neurotrans- and bioresorbable and support the replacement of
mitter L-glutamate or expressing G-protein-gated normal tissue without inducing inflammation.
inwardly rectifying potassium (GIRK) channels, Incompatible materials are destined for an inflam-
hepatic lineage cells producing urea, and osteo- matory or foreign-body response that eventually
genic lineage cells forming tissue-engineered leads to rejection and/or necrosis. Degradation
bone. In this respect, they meet a commonly products, if produced, should be removed from
accepted criterion for pluripotent stem cells, with- the body via metabolic pathways at an adequate
out implying that they can generate every adult rate so that the concentration of these degradation
tissue. The cells could be obtained either from products in the tissues remains at a tolerable level
amniocentesis or chorionic villous sampling in [63]. The biomaterial should also provide an envi-
the developing fetus or from the placenta or ronment in which appropriate regulation of cell
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 577

behavior (adhesion, proliferation, migration, and implant and the extent of intermolecular cross-
differentiation) can occur. Cell behavior in the linking – the lower the density, the greater the
newly formed tissue has been shown to be regu- space between collagen fibers and the larger the
lated by multiple interactions of the cells with pores for cell infiltration, leading to a higher rate
their microenvironment, including interactions of implant degradation. Collagen contains cell
with cell-adhesion ligands [64] and with soluble adhesion domain sequences (e.g., RGD) that
growth factors. Since biomaterials provide tem- may assist to retain the phenotype and activity of
porary mechanical support while the cells many types of cells, including fibroblasts [69] and
undergo spatial reorganization into the tissue, the chondrocytes [70]. Additionally, recent studies
properly chosen biomaterial should allow the have employed the usage of chitin as an adhesion
engineered tissue to maintain sufficient mechani- molecule in the collagen-based scaffolds [71].
cal integrity to support itself in early development, Alginate, a polysaccharide isolated from sea-
while in late development, it should begin to weed, has been used as an injectable cell delivery
degrade so that it does not hinder further tissue vehicle [72] and a cell immobilization matrix [73]
growth [62]. owing to its gentle gelling properties in the pres-
Generally, three classes of biomaterials have ence of divalent ions such as calcium. Alginate is
been utilized for engineering tissues: naturally relatively biocompatible and is approved by the
derived materials (e.g., collagen and alginate), Food and Drug Administration (FDA) for human
acellular tissue matrices (e.g., bladder submucosa use as a wound dressing material. Alginate is a
and small intestinal submucosa), and synthetic family of copolymers of D-mannuronate and
polymers such as polyglycolic acid (PGA), L-guluronate. The physical and mechanical prop-
polylactic acid (PLA), and poly(lactic-co-glycolic erties of alginate gel are strongly correlated with
acid) (PLGA). These classes of biomaterials have the proportion and length of polyglucuronic block
been tested in respect to their biocompatibility in the alginate chains [72].
[65, 66]. Naturally derived materials and acellular
tissue matrices have the potential advantage of Acellular Tissue Matrices
biological recognition. However, synthetic poly- Acellular tissue matrices are collagen-rich matri-
mers can be produced reproducibly on a large ces prepared by removing cellular components
scale with controlled properties such as strength, from tissues. The matrices are often prepared by
degradation rate, and microstructure, which mechanical and chemical manipulation of a seg-
would aid in the preparation of easily used, “off- ment of tissue [1–4]. These matrices slowly
the-shelf” scaffold material. degrade upon implantation and are replaced and
remodeled by ECM proteins synthesized and
Naturally Derived Materials secreted by transplanted or in growing cells. The
Collagen is the most abundant and ubiquitous most commonly used acellular tissue matrices are
structural protein in the body and may be readily small intestinal submucosa (SIS) and bladder sub-
purified from both animal and human tissues with mucosa (BSM) [49].
an enzyme treatment and salt/acid extraction
[67]. Collagen implants, under normal conditions, Synthetic Polymers
degrade through a process involving phagocytosis Polyesters of naturally occurring a-hydroxy acids,
of collagen fibrils by fibroblasts [68]. This is including PGA, PLA, and PLGA, are widely used
followed by sequential attack by lysosomal in tissue engineering. These polymers are
enzymes including cathepsins B1 and D. Under FDA-approved for a variety of applications,
inflammatory conditions, the implants can be rap- including sutures [74]. The ester bonds in these
idly degraded largely by matrix metalloproteins polymers are hydrolytically labile, and they
(MMPs) and collagenases [68]. However, the degrade by nonenzymatic hydrolysis. The degra-
in vivo resorption rate of a collagen implant can dation products of PGA, PLA, and PLGA are
be regulated by controlling the density of the nontoxic natural metabolites and are eventually
578 A.G. Hester and A. Atala

eliminated from the body in the form of carbon experimentally and clinically. In animal studies,
dioxide and water [74]. The degradation rate of segments of the urethra were resected and
these polymers can be tailored to the application replaced with acellular matrix grafts in an onlay
by altering crystallinity, initial molecular weight, fashion. Histological examination showed com-
and the copolymer ratio of lactic to glycolic acid. plete epithelialization and progressive vessel and
Generally, the optimal degradation time ranges muscle infiltration, and the animals were able to
from several weeks to several years. Since these void through the neourethras [4]. These results
polymers are thermoplastics, they can be easily were confirmed in a clinical study of patients
formed into a three-dimensional scaffold with a with hypospadias and urethral stricture disease
desired microstructure, gross shape, and dimen- [85] (Fig. 2). Decellularized cadaveric bladder
sion by various techniques, including molding, submucosa was used as an onlay matrix for ure-
extrusion, solvent casting [75], phase separation thral repair in patients with stricture disease and
techniques, and gas-foaming techniques hypospadias. Patent, functional neourethras were
[76]. Many applications in tissue engineering noted in these patients with up to a 7-year follow-
often require a scaffold with high porosity and up. The use of an off-the-shelf matrix appears to
ratio of surface area to volume. Other biodegrad- be beneficial for patients with abnormal urethral
able synthetic polymers, including poly(anhy- conditions and obviates the need for obtaining
drides) and poly(ortho esters), can also be used autologous grafts, thus decreasing operative time
to fabricate scaffolds for tissue engineering with and eliminating donor site morbidity.
controlled properties [77]. Unfortunately, the above techniques are not
applicable for tubularized urethral repairs. The
collagen matrices are able to replace urethral seg-
Engineering Specific Tissues ments only when used in an onlay fashion. How-
and Organs ever, if a tubularized repair is needed, the collagen
matrices should be seeded with autologous cells to
Investigators around the world, including our lab- avoid the risk of stricture formation and poor
oratory, have been working toward the develop- tissue development [83]. Therefore, tubularized
ment of several cell types, tissues, and organs for collagen matrices seeded with autologous cells
clinical application. The following sections will can be used successfully for total penile urethra
describe this research in detail. replacement. An initial study in 2008 with 5 adult
patients requiring tubularized repairs secondary to
lichen sclerosis was somewhat successful. These
Urethra patients had grafts engineered from the buccal
mucosa, but 2 of the 5 patients required excision
Various biomaterials without cells, such as PGA secondary to fibrosis and contraction and the
and acellular collagen-based matrices derived remaining 3 required some form of instrumenta-
from decellularized small intestine and bladder, tion [86]. We performed a study in the past few
have been used in animal models for the regener- years where five boys were successfully
ation of urethral tissue [4, 78–82]. Some of these implanted with urethras engineered from autolo-
biomaterials, like acellular collagen matrices gous cells for posterior urethral defects. Upon
derived from bladder submucosa, have also been biopsy following only 3 months from implanta-
seeded with autologous cells for urethral recon- tion, these grafts were found to contain normal
struction. Our laboratory has been able to replace urethral architecture, and at a median follow-up of
tubularized urethral segments with cell-seeded 71 months, these patients all remained patent and
collagen matrices [83, 84]. functional [87]. Another study performed with
Acellular collagen matrices derived from blad- children, this time with hypospadias, demon-
der submucosa by our laboratory have been used strated similar results. These children had
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 579

Fig. 2 Urethral repair using a collagen matrix. (a) Repre- the area of stricture in the urethra. On the right, the arrow
sentative case of a patient with a bulbar stricture. (b) indicates the repaired stricture. Note that the engineered
During surgery, strictured tissue is excised, preserving the tissue now obscures the native white urethral tissue in an
urethral plate on the left side, and the matrix is anasto- onlay fashion in the right photograph. (c) Urethrogram
mosed to the urethral plate in an onlay fashion on the right. 6 months after repair. (d) Cystoscopic view of urethra
The boxes in both photos indicate the area of interest, before surgery on the left side and 4 months after repair
including the urethra, which appears white in the left on the right side
photograph. In the left photograph, the arrow indicates

autologous urothelial cells isolated from bladder Bladder


washes, and urethral implants were engineered.
Most of these children had minor complications Currently, gastrointestinal segments are com-
over the following 3–5 years, but only one monly used as tissues for bladder replacement or
required a urethrotomy for an obstruction. The repair. However, gastrointestinal tissues are
remaining children were able to void in a standing designed to absorb specific solutes, whereas blad-
position [13, 88]. der tissue is designed for the excretion of solutes.
Despite the success of these techniques, there Due to the problems encountered with the use of
are still the continued complications of synthetic gastrointestinal segments, numerous investigators
materials used and implanted into a human subject have attempted alternative materials and tissues
serving as a nidus for fibrosis and stricture forma- for bladder replacement or repair.
tion. A more recent technique has been more The success of cell transplantation strategies
successful in animal models [13]. In this model, for bladder reconstruction depends on the ability
the graft and scaffolding are created from human to use donor tissue efficiently and to provide the
cells without relying on any exogenous materials right conditions for long-term survival, differenti-
and by using a self-assembly method [89]. In fact, ation, and growth. Urothelial and muscle cells can
an additional study has shown how using endo- be expanded in vitro, seeded onto polymer scaf-
thelial cells in the process of growing these grafts folds, and allowed to attach and form sheets of
produces earlier vascularization of the cells [91]. These principles were applied in the
material [90]. creation of tissue-engineered bladders in an
580 A.G. Hester and A. Atala

Fig. 3 Construction of engineered bladder. (a) Scaffold running 4–0 polyglycolic sutures. (c) Implant covered
material seeded with cells for use in bladder repair. (b) The with fibrin glue and omentum
seeded scaffold is anastomosed to native bladder with

animal model that required a subtotal cystectomy interactions and potentially allow studying of the
with subsequent replacement with a tissue- cell growth and development process in these
engineered organ in beagle dogs [12]. Urothelial engineered organs [93, 95].
and muscle cells were separately expanded from A clinical experience involving engineered
an autologous bladder biopsy and seeded onto a bladder tissue for cystoplasty reconstruction was
bladder-shaped biodegradable polymer scaffold. conducted starting in 1999. A small pilot study of
The results from this study showed that it is pos- seven patients was reported, using a collagen
sible to tissue engineer bladders that are anatom- scaffold seeded with cells either with or without
ically and functionally normal. In subsequent omentum coverage or a combined PGA-collagen
years, these findings have been replicated, and scaffold seeded with cells and omental coverage
the subjects have been followed for longer periods (Fig. 3). The patients reconstructed with the
of time, indicating native histology, good capac- engineered bladder tissue created with the
ity, and excellent compliance [92, 93]. Clinical PGA-collagen cell-seeded scaffolds showed
trials for the application of this technology are increased compliance, decreased end-filling pres-
currently being conducted. sures, increased capacities, and longer dry periods
Despite the success of the previously men- [23] (Fig. 4). Although the experience is promis-
tioned studies for subtotal cystectomy, there ing in terms of showing that engineered tissues
have been subsequent reports of difficulties can be implanted safely, it is just a start in terms of
while attempting to engineer larger sections of accomplishing the goal of engineering fully func-
the bladder. In 2008, a study, again in canines, tional bladders. Since this time, two multicenter
compared the results of seeded versus Phase II clinical trials have been performed, one in
unseeded small intestinal segments for large- pediatric patients and one in adult patients, in
portion (90 %) bladder replacements. Many of order to further test the safety of engineered
the subjects in this study demonstrated graft Neo-Bladder Augments™, produced by Tengion,
shrinkage and adhesions [94]. Additionally, there Inc. These studies had good results but did find
has been some recent interest in using bioreactors that the engineered bladder augments were more
to mechanically stimulate the engineered likely to succeed in patients who still retained
bladder. Theoretically, the mechanical stimulation natural bladder cycling function, further indica-
provided by the bioreactor allows for functional tion to pursue bioreactors for mechanical stimula-
filling and emptying of the tissue which tion [13, 93]. Further experimental and clinical
would provide an increase in cell-scaffold work is being conducted.
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 581

Fig. 4 Cystograms and urodynamic studies of a patient the cystogram and abnormal bladder pressures as the blad-
before and after implantation of the tissue-engineered blad- der is filled via urodynamic study. (b) Postoperatively,
der. (a) Preoperative results indicate an irregular bladder in findings are significantly improved

Kidney functional nephron units should be used. Optimal


culture conditions to nurture renal cells have been
Renal tissue is arguably one of the most difficult extensively studied and cells grown under these
tissues to replicate in the laboratory. The kidney is conditions have been reported to maintain their
a complex organ and the unique structural and cellular characteristics [107]. Moreover, renal
cellular heterogeneity present within this cells placed in a three-dimensional culture envi-
organ creates many challenges. The system of ronment are able to reconstitute into renal
nephrons and collecting ducts within the kidney structures.
is composed of multiple functionally and Recent investigative efforts in the search for a
morphologically distinct segments. For this reliable cell source have been expanded to stem
reason, appropriate conditions must be provided and progenitor cells. The use of these cells for
to ensure the long-term survival, differentiation, tissue regeneration is attractive due to their ability
and growth of many types of cells. Recent efforts to differentiate and mature into specific cell types
in the area of kidney tissue regeneration have needed. This is particularly useful in instances
focused on the development of a reliable cell where primary renal cells are unavailable due to
source [96–101]. Moreover, optimal growth con- extensive tissue damage. Bone marrow-derived
ditions have been extensively investigated to pro- human mesenchymal stem cells have been
vide adequate enrichment to achieve stable renal shown to be a potential source due to their
cell expansion systems [102–106]. ability to differentiate into several cell lineages
Isolation of particular cell types that produce [96, 97, 100]. These cells have been shown to
specific factors may be a good approach for selec- participate in the kidney development when they
tive cell therapies. For example, cells that produce are placed in a rat embryonic niche that allows for
erythropoietin could be isolated in culture, and continued exposure to a repertoire of nephrogenic
these cells could eventually be used to treat ane- signals [101]. These cells, however, were found to
mia that results from end-stage renal failure. How- contribute mainly to regeneration of damaged
ever, total renal function would not be achieved glomerular endothelial cells after injury. In addi-
using this approach. To create kidney tissue that tion, the major cell source of kidney regeneration
would deliver full renal function, a culture was found to originate from intrarenal cells in an
containing all of the cell types comprising the ischemic renal injury model [96, 99]. Another
582 A.G. Hester and A. Atala

potential cell source for kidney regeneration is Developmental Approaches to Kidney


circulating stem cells, which have been shown to Regeneration
transform into tubular and glomerular epithelial Transplantation of a kidney precursor, such as the
cells, podocytes, mesangial cells, and interstitial metanephros, into a diseased kidney has been
cells after renal injury [97, 98, 108–112]. These proposed as a possible method for functional res-
observations suggest that controlling stem and toration. In an animal study, human embryonic
progenitor cell differentiation may lead to suc- metanephroi, transplanted into the kidneys of an
cessful regeneration of kidney tissues. immune-deficient mouse model, has developed
Although isolated renal cells are able to retain into mature kidneys [124]. The transplanted meta-
their phenotypic and functional characteristics in nephroi which produced urine-like fluid, how-
culture, transplantation of these cells in vivo may ever, failed to develop ureters. This study
not result in structural remodeling. In addition, suggests that development of an in vitro system
cell or tissue components cannot be implanted in in which metanephroi could be grown may lead to
large volumes due to limited diffusion of transplant techniques that could produce a small
oxygen and nutrients [113]. Thus, a cell- replacement kidney within the host. In another
support matrix, preferably one that encourages study, the metanephros was divided into mesen-
angiogenesis, is necessary to allow diffusion chymal tissue and ureteral buds, and each of the
across the entire implant. A variety of synthetic tissue segments was cultured in vitro [125]. After
and naturally derived materials has been exam- 8 days in culture, each portion of the mesenchy-
ined in order to determine the ideal support mal tissues had grown to the original size.
structures for the regeneration [12, 23, 85, A similar method was used for ureteral buds,
114, 115]. Biodegradable synthetic materials, which also propagated. These studies indicate
such as polylactic and polyglycolic acid poly- that if the mesenchyme and ureteral buds were
mers, have been used to provide structural support placed together and cultured in vitro, a
for cells. Synthetic materials can be easily fabri- metanephros-like structure would develop and
cated and configured in a controlled manner, suggest that the metanephros could be propagated
which makes them attractive options for tissue under an optimal condition.
engineering. However, naturally derived mate- In another study, transplantation of metaneph-
rials, such as collagen, laminin, and fibronectin, roi into a non-immunosuppressed rat omentum
are much more biocompatible and provide a sim- showed that the implanted metanephroi are able
ilar extracellular matrix environment to normal to undergo differentiation and growth that is not
tissue. For this reason, collagen-based scaffolds confined by a tight organ capsule [126]. When the
have been used increasingly in many applications metanephroi with an intact ureteric bud were
[116–119]. implanted, the metanephroi are able to enlarge
Over the years, the engineered scaffoldings and become a kidney-shaped tissue within
have continued to have only partial success with 3 weeks. The metanephroi transplanted into the
angiogenesis. However, in recent years, the con- omentum were able to develop into a kidney tis-
cept of whole-organ decellularization has come to sue structure with a well-defined cortex and
fruition. By using a natural acellular organ bed, medulla. Mature nephrons and collecting system
the internal vascular system and microarchitecture structures are shown to be indistinguishable from
can be maintained while generating simple tissue those of normal kidneys by light or electron
matrices for production of heart valves and intes- microscopy [127, 128]. Moreover, these struc-
tinal segments [120]. However, this technology tures become vascularized via arteries that origi-
suggests that more complex organ structures can nate at the superior mesenteric artery of the host
be produced in a similar manner. These scaffolds [127, 128]. It has been demonstrated that the
can then be recellularized and have been studied metanephroi transplanted into the omentum sur-
in animal models [121–123], with the potential for vive for up to 32 weeks postimplantation
future clinical studies [120]. [129]. These studies show that developmental
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 583

approach may be a viable option for regenerating monoclonal anti-BrdU antibodies. These results
renal tissue for functional restoration. demonstrated that renal-specific cells can be suc-
Currently, there is a lot of interest in branching cessfully harvested and cultured and can subse-
morphogenesis and its impact on tissue regenera- quently attach to artificial biodegradable
tion for the kidney. A recent review discussed how polymers. The renal cell-polymer scaffolds can
the epithelial buds in certain tissues, including the be implanted into host animals where the cells
kidney, form into iterative tip-stalk generators replicate and organize into nephron segments, as
(ITSG) which are able to form the necessary the polymer, which serves as a cell delivery vehi-
ducts and tubules needed for tissue development. cle, undergoes biodegradation.
Recent studies have demonstrated how this Initial experiments showed that implanted cell-
branching strategy can be used to form polymer scaffolds gave rise to renal tubular struc-
3-dimensional scaffolds for tissue engineering tures. However, it was unclear whether the tubular
[130–132]. structures reconstituted de novo from dispersed
renal elements, or if they merely represented frag-
Tissue Engineering Approaches ments of donor tubules which survived the origi-
to Kidney Regeneration nal dissociation and culture processes intact.
The ability to grow and expand renal cells is one Further investigation was conducted in order to
of the essential requirements in engineering tis- examine the process [133]. Mouse renal cells were
sues. The feasibility of achieving renal cell harvested and expanded in culture. Subsequently,
growth, expansion, and in vivo reconstitution single isolated cells were seeded on biodegradable
using tissue engineering techniques was investi- polymers and implanted into immune-competent
gated [114]. Donor rabbit kidneys were removed syngeneic hosts. Renal epithelial cells were
and perfused with a non-oxide solution which observed to reconstitute into tubular structures
promoted iron particle entrapment in the glomer- in vivo. Sequential analyses of the retrieved
uli. Homogenization of the renal cortex and frac- implants over time demonstrated that renal epithe-
tionation in 83–210 μm sieves with subsequent lial cells first organized into a cord-like structure
magnetic extraction yielded three separate puri- with a solid center. Subsequent canalization into a
fied suspensions of distal tubules, glomeruli, and hollow tube could be seen by 2 weeks. Histolog-
proximal tubules. The cells were plated separately ical examination with nephron segment-specific
in vitro and after expansion, were seeded onto lactins showed successful reconstitution of prox-
biodegradable polyglycolic acid scaffolds and imal tubules, distal tubules, loop of Henle,
implanted subcutaneously into host athymic collecting tubules, and collecting ducts. These
mice. This included implants of proximal tubular results showed that single suspended cells are
cells, glomeruli, distal tubular cells, and a mixture capable of reconstituting into tubular structures,
of all three cell types. Animals were sacrificed at with homogeneous cell types within each tubule.
one week, two weeks, and one month after Although these studies demonstrated that renal
implantation, and the retrieved implants were ana- cells seeded on biodegradable polymer scaffolds
lyzed. An acute inflammatory phase and a chronic are able to form some renal structures in vivo,
foreign-body reaction were seen, accompanied by complete renal function could not be achieved in
vascular ingrowth by 7 days after implantation. these studies. In a subsequent study, we sought to
Histological examination demonstrated progres- create a functional artificial renal unit which could
sive formation and organization of the nephron produce urine [134]. Mouse renal cells were
segments within the polymer fibers with time. harvested, expanded in culture, and seeded onto
Renal cell proliferation in the cell-polymer scaf- a tubular device constructed from polycarbonate
folds was detected by in vivo labeling of replicat- [119]. The tubular device was connected at one
ing cells with the thymidine analog end to a Silastic catheter which terminated into a
bromodeoxyuridine [99]. BrdU incorporation reservoir. The device was implanted subcutane-
into renal cell DNA was confirmed using ously in athymic mice. The implanted devices
584 A.G. Hester and A. Atala

Fig. 5 Combining therapeutic cloning and tissue engi- of T cells that secrete IFNγ after stimulation with alloge-
neering to produce kidney tissue. (a) Illustration of the neic renal cells, cloned renal cells, or nuclear donor fibro-
tissue-engineered renal unit. (b) Renal unit seeded with blasts. Cloned renal cells produce fewer IFNγ spots than
cloned cells, 3 months after implantation, showing the the allogeneic cells, indicating that the rejection response
accumulation of urine-like fluid. (c) Clear unidirectional to cloned cells is diminished. The presented wells are
continuity between the mature glomeruli, their tubules, and single representatives of duplicate wells
Silastic catheter. (d) ELISPOT analyses of the frequencies

were retrieved and examined histologically and collected fluid showed an 8.2-fold increase in
immunocytochemically at 1, 2, 3, 4, and 8 weeks concentration, as compared to serum. These
after implantation. Fluid was collected from results demonstrated that single cells form
inside the implant, and uric acid and creatinine multicellular structures can become organized
levels were determined. into functional renal units that are able to excrete
Histological examination of the implanted high levels of solutes through a urine-like
device demonstrated extensive vascularization as fluid [134].
well as formation of glomeruli and highly orga- To determine whether renal tissue could be
nized tubule-like structures. Immunocytochemi- formed using an alternative cell source, nuclear
cal staining with anti-osteopontin antibody, transplantation (therapeutic cloning) was
which is secreted by proximal and distal tubular performed to generate histocompatible tissues,
cells and the cells of the thin ascending loop of and the feasibility of engineering syngeneic renal
Henle, stained the tubular sections. Immunohisto- tissues in vivo using these cloned cells was
chemical staining for alkaline phosphatase stained investigated [107]. Nuclear material from bovine
proximal tubule-like structures. Uniform staining dermal fibroblasts was transferred into
for fibronectin in the extracellular matrix of newly unfertilized enucleated donor bovine eggs. Renal
formed tubes was observed. The fluid collected cells from the cloned embryos were harvested,
from the reservoir was yellow and contained expanded in vitro, and seeded onto three-
66 mg/dl uric acid (as compared to 2 mg/dl in dimensional renal devices (Fig. 5a). The devices
plasma) suggesting that these tubules are capable were implanted into the back of the same steer
of unidirectional secretion and concentration of from which the cells were cloned and were
uric acid. The creatinine assay performed on the retrieved 12 weeks later.
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 585

This process produced functioning renal However, a naturally derived tissue matrix
units. Urine production and viability were with existing three-dimensional kidney architec-
demonstrated after transplantation back into the ture would be preferable to the artificial matrix
nuclear donor animal (Fig. 5b). Chemical used in these experiments, because it would allow
analysis suggested unidirectional secretion and for transplantation of a larger number of cells,
concentration of urea nitrogen and creatinine. resulting in greater renal tissue volumes. Thus,
Microscopic analysis revealed formation of orga- we developed an acellular collagen-based kidney
nized glomeruli and tubular structures (Fig. 5c). matrix, which is identical to the native renal archi-
Immunohistochemical and RT-PCR analysis con- tecture. In a subsequent study, we investigated
firmed the expression of renal mRNA and whether these collagen-based matrices could
proteins. accommodate large volumes of renal cells and
Since previous studies have shown that bovine form kidney structures in vivo [141].
clones harbor the oocyte mitochondrial DNA Acellular collagen matrices, derived from por-
[135–137], the donor egg’s mitochondrial DNA cine kidneys, were obtained through a multistep
(mtDNA) was thought to be a potential source of decellularization process. During this process,
immunologic incompatibility. Differences in serial evaluation of the matrix for cellular rem-
mtDNA-encoded proteins expressed by cloned nants was performed using histochemistry, scan-
cells could stimulate a T-cell response specific ning electron microscopy (SEM), and RT-PCR.
for mtDNA-encoded minor histocompatibility Mouse renal cells were harvested, grown, and
antigens when the cloned cells are implanted seeded on 80 of the decellularized collagen matri-
back into the original nuclear donor [138]. Mater- ces at a concentration of 30  106 cells/ml. Forty
nally transmitted minor histocompatibility anti- cell-matrix constructs grown in vitro were ana-
gens in mice have been shown to stimulate both lyzed 3 days and 1, 2, 4, and 6 weeks after
skin allograft rejection in vivo and cytotoxic seeding. The remaining 40 cell-containing matri-
T-lymphocytes expansion in vitro [138] that ces were implanted in the subcutaneous space of
could prevent the use of these cloned constructs 20 athymic mice. The animals were sacrificed
in patients with chronic rejection of major histo- 3 days and 1, 2, 4, 8, and 24 weeks after implan-
compatibility matched human renal transplants tation for analyses. Gross, SEM, histochemical,
[139, 140]. We tested for a possible T-cell immunocytochemical, and biochemical analyses
response to the cloned renal devices using were performed.
delayed-type hypersensitivity testing in vivo and Scanning electron microscopy and histological
ELISPOT analysis of interferon-γ-secreting T examination confirmed the acellularity of the
cells in vitro. Both analyses revealed that the processed matrix. RT-PCR performed on the kid-
cloned renal cells showed no evidence of a T-cell ney matrices demonstrated the absence of any
response, suggesting that rejection will not neces- RNA residues. Renal cells seeded on the matrix
sarily occur in the presence of oocyte-derived adhered to the inner surface and proliferated to
mtDNA (Fig. 5d). This finding may represent a confluency 7 days after seeding, as demonstrated
step forward in overcoming the histocompatibility by SEM. Histochemical and immunocytochemi-
problem of stem cell therapy [140]. cal analyses performed using H&E, periodic acid-
These studies demonstrated that cells derived Schiff, alkaline phosphatase, anti-osteopontin,
from nuclear transfer can be successfully and anti-CD-31 identified stromal, endothelial,
harvested, expanded in culture, and transplanted and tubular epithelial cell phenotypes within the
in vivo with the use of biodegradable scaffolds on matrix. Renal tubular and glomerulus-like struc-
which the single suspended cells can organize into tures were observed 8 weeks after implantation.
tissue structures that are genetically identical to MTT proliferation and titrated thymidine incorpo-
that of the host. These studies were the first dem- ration assays performed 6 weeks after cell seeding
onstration of the use of therapeutic cloning for demonstrated a population increase of 116 % and
regeneration of tissues in vivo. 92 %, respectively, as compared to the 2-week
586 A.G. Hester and A. Atala

time points. This study demonstrates that renal implication of this study is that under the correct
cells are able to adhere to and proliferate on the conditions, it is possible to rapidly decellularize
collagen-based kidney matrices. The renal cells and recellularize a 3-dimensional scaffold while
reconstitute renal tubular and glomeruli-like maintaining vascular integrity and differentiation
structures in the kidney-shaped matrix. The of the cells.
collagen-based kidney matrix system seeded
with renal cells may be useful in the future for Genital Tissues
augmenting renal function. Reconstructive surgery is required for a wide vari-
We also investigated the feasibility of creating ety of pathologic penile conditions, such as penile
three-dimensional renal structures for in situ carcinoma, trauma, severe erectile dysfunction,
implantation within the native kidney tissue. Pri- and congenital conditions such as ambiguous gen-
mary renal cells from 4-week-old mice were italia, hypospadias, and epispadias. One of the
grown and expanded in culture. These renal cells major limitations of phallic reconstructive surgery
were labeled with fluorescent markers and is the scarcity of sufficient autologous tissue.
injected into mouse kidneys in a collagen gel for The major components of the phallus are cor-
in vivo formation of renal tissues. Collagen injec- poral smooth muscle and endothelial cells. The
tion without cells and sham-operated animals creation of autologous functional and structural
served as controls. In vitro reconstituted renal corporal tissue de novo would be beneficial.
structures and in vivo implanted cells were Autologous cavernosal smooth muscle and endo-
retrieved and analyzed. thelial cells were harvested, expanded, and seeded
The implanted renal cells formed tubular and on acellular collagen matrices and implanted in a
glomerular structures within the kidney tissue, as rabbit model [143, 144]. Histological examination
confirmed by the fluorescent markers. There was confirmed the appropriate organization of
no evidence of renal tissue formation in the con- penile tissue phenotypes, and structural and
trol and the sham-operated groups. These results functional studies, including cavernosography,
demonstrate that single renal cells are able to cavernosometry, and mating studies, demon-
reconstitute kidney structures when placed in a strated that it is possible to engineer autologous
collagen-based scaffolding system. The implanted functional penile tissue. Corporal body grafts
renal cells are able to self-assemble into tubular have been successfully formed using small intes-
and glomerular structures within the kidney tis- tinal submucosa (SIS), but results do seem to vary,
sue. These findings suggest that this system may with a single layer SIS resulting in fewer compli-
be the preferred approach to engineer functional cations than the four-layer SIS [145]. Multiple
kidney tissues for the treatment of end-stage renal studies have reiterated this point and have
disease. established this technique as the standard for cor-
More recent studies have explored ways to poral body grafting [146–149]. Congenital
optimize the decellularization process as well as malformations of the uterus may have profound
the recellularization process, with the intention of implications clinically. Patients with cloacal
decreasing processing times and increasing vas- exstrophy and intersex disorders may not have
cular perfusion within the grafts [142]. Bonandrini sufficient uterine tissue present for future repro-
and colleagues successfully decellularized rat kid- duction. We investigated the possibility of engi-
neys following a 17 h perfusion of sodium neering functional uterine tissue using autologous
dodecyl sulfate while still maintaining the basic cells [150]. Autologous rabbit uterine smooth
microarchitecture, including glomeruli and vascu- muscle and epithelial cells were harvested and
lar structures. The scaffold was recellularized with then grown and expanded in culture. These cells
murine embryonic stem cells through the renal were seeded onto preconfigured uterine-shaped
artery while using pressure-controlled perfusion biodegradable polymer scaffolds, which were
for 24–72 h, and initial cellular differentiation was then used for subtotal uterine tissue replacement
noted at 24 h and marked at 72 h. The major in the corresponding autologous animals.
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 587

Upon retrieval 6 months after implantation, histo- cells of female rabbits were harvested, grown, and
logical, immunocytochemical, and Western blot expanded in culture. These cells were seeded onto
analyses confirmed the presence of normal uterine biodegradable polymer scaffolds, and the cell-
tissue components. Biomechanical analyses and seeded constructs were then implanted into nude
organ bath studies showed that the functional mice for up to 6 weeks. Immunocytochemical,
characteristics of these tissues were similar to histological, and Western blot analyses confirmed
those of normal uterine tissue. Breeding studies the presence of vaginal tissue phenotypes. Electri-
using these engineered uteri are currently being cal field stimulation studies in the tissue-
performed. A recent study of regenerated endo- engineered constructs showed similar functional
metrial tissue in mice has demonstrated the ability properties to those of normal vaginal tissue.
to implant functional endometrial tissue in vivo When these constructs were used for autologous
[151]. Additionally, in vitro studies have demon- total vaginal replacement, patent vaginal structures
strated the ability of tissue-engineered endome- were noted in the tissue-engineered specimens,
trial tissues to respond to cues allowing for normal while the non-cell-seeded structures were noted to
uterine activity such as menstruation [152] be stenotic [153]. A prospective study performed in
(Fig. 6). 2013 in patients with Mayer-Rokitansky-K€uster-
Similarly, several pathologic conditions, Hauser syndrome undergoing vaginoplasty dem-
including congenital malformations and malig- onstrated positive results. These patients received
nancy, can adversely affect normal vaginal devel- tissue-engineered acellular dermal matrix and were
opment or anatomy. Vaginal reconstruction has advised to dilate the vagina postoperatively to
traditionally been challenging due to the paucity avoid contraction of the graft. At a mean follow-
of available native tissue. The feasibility of up of 21.1 months, all patients had patent vaginal
engineering vaginal tissue in vivo was investi- grafts with a similar Female Sexual Function Index
gated [153]. Vaginal epithelial and smooth muscle to their age-matched controls [154].

Fig. 6 Autologous chondrocytes for the treatment of autologous chondrocytes. A catheter was inserted into the
vesicoureteral reflux. (a) Preoperative voiding cystoure- bladder via the urethra, and a radioactive solution was
throgram of a patient with bilateral reflux. A catheter was inserted into the bladder. The bladder was scanned during
inserted into the bladder via the urethra, and contrast mate- filling and emptying phases. This panel includes sequential
rial was instilled intravesically. Here, contrast material can images of the bladder as it was filled and emptied. This
be seen within both ureters and within the kidneys, indi- shows a normal, round bladder that fills and empties prop-
cating reflux is present. (b) Postoperative radionuclide erly. If reflux had been present, the ureters would have been
cystogram of the same patient 6 months after injection of visible in the scan above the round bladder
588 A.G. Hester and A. Atala

Summary and Conclusions 11. Cilento BG, et al. Phenotypic and cytogenetic char-
acterization of human bladder urothelia expanded
in vitro. J Urol. 1994;152(2 Pt 2):665–70.
Regenerative medicine efforts are currently 12. Oberpenning F, et al. De novo reconstitution of a
underway experimentally for virtually every type functional mammalian urinary bladder by tissue engi-
of tissue and organ within the human body. neering. [see comment]. Nat Biotechnol. 1999;17
As regenerative medicine incorporates the fields (2):149–55.
13. Orabi H, Bouhout S, Morissette A, Rousseau A,
of tissue engineering, cell biology, nuclear trans- Chabaud S, Bolduc S. Tissue engineering of urinary
fer, and materials science, personnel who have bladder and urethra: advances from bench to patients.
mastered the techniques of cell harvest, culture, Sci World J. 2013;2013.
expansion, transplantation, and polymer design 14. Scriven SD, et al. Reconstitution of human
urothelium from monolayer cultures. J Urol.
are essential for the successful application of 1997;158(3 Pt 2):1147–52.
these technologies to extend human life. Various 15. Liebert M, et al. Expression of mal is associated with
tissues are at different stages of development, with urothelial differentiation in vitro: identification by
some already being used clinically, a few in pre- differential display reverse-transcriptase polymerase
chain reaction. Differentiation. 1997;61(3):177–85.
clinical trials, and some in the discovery stage. 16. Liebert M, et al. Stimulated urothelial cells produce
Recent progress suggests that engineered tissues cytokines and express an activated cell surface anti-
may have an expanded clinical applicability in the genic phenotype. Semin Urol. 1991;9(2):124–30.
future and may represent a viable therapeutic 17. Puthenveettil JA, Burger MS, Reznikoff
CA. Replicative senescence in human uroepithelial
option for those who would benefit from the life- cells. Adv Exp Med Biol. 1999;462:83–91.
extending benefits of tissue replacement or repair. 18. Nagele U, Maurer S, Feil G, Bock C, Krug J, Sievert
KD, Stenzl A. In vitro investigations of tissue-
engineered multilayered urothelium established
from bladder washings. Eur Urol. 2008;54
References (6):1414–22.
19. Zhang Y, McNeill E, Tian H, Soker S, Andersson KE,
1. Dahms SE, et al. Composition and biomechanical Yoo JJ, Atala A. Urine derived cells are a potential
properties of the bladder acellular matrix graft: com- source for urological tissue reconstruction. J Urol.
parative analysis in rat, pig and human. Br J Urol. 2008;180(5):2226–33.
1998;82(3):411–19. 20. Freeman MR, et al. Heparin-binding EGF-like growth
2. Yoo JJ, et al. Bladder augmentation using allogenic factor is an autocrine growth factor for human
bladder submucosa seeded with cells. Urology. urothelial cells and is synthesized by epithelial and
1998;51(2):221–5. smooth muscle cells in the human bladder. J Clin
3. Piechota HJ, et al. In vitro functional properties of the Invest. 1997;99(5):1028–36.
rat bladder regenerated by the bladder acellular matrix 21. Nguyen HT, et al. Cell-specific activation of the
graft. J Urol. 1998;159(5):1717–24. HB-EGF and ErbB1 genes by stretch in primary
4. Chen F, Yoo JJ, Atala A. Acellular collagen matrix as human bladder cells. In Vitro Cell Dev Biol Anim.
a possible “off the shelf” biomaterial for urethral 1999;35(7):371–5.
repair. Urology. 1999;54(3):407–10. 22. Harriss DR. Smooth muscle cell culture: a new
5. Amiel GE, Atala A. Current and future modalities for approach to the study of human detrusor physiology
functional renal replacement. Urol Clin N and pathophysiology. Br J Urol. 1995;75 Suppl
Am. 1999;26(1):235–46. 1:18–26.
6. Amiel GE, et al. Engineering of blood vessels from 23. Atala A, et al. Tissue-engineered autologous bladders
acellular collagen matrices coated with human endo- for patients needing cystoplasty. Lancet. 2006;367
thelial cells. Tissue Eng. 2006;12(8):2355–65. (9518):1241–6.
7. Yoo JJ, et al. Autologous engineered cartilage rods for 24. Martin GR. Isolation of a pluripotent cell line from
penile reconstruction. J Urol. 1999;162(3 Pt early mouse embryos cultured in medium conditioned
2):1119–21. by teratocarcinoma stem cells. Proc Natl Acad Sci
8. Atala A. Autologous cell transplantation for urologic U S A. 1981;78(12):7634–8.
reconstruction. J Urol. 1998;159(1):2–3. 25. Reubinoff BE, et al. Neural progenitors from human
9. Atala A. Bladder regeneration by tissue engineering. embryonic stem cells. Nat Biotechnol. 2001;19
[see comment]. BJU Int. 2001;88(7):765–70. (12):1134–40 [see comment].
10. Atala A. Creation of bladder tissue in vitro and 26. Schuldiner M, et al. Induced neuronal differentiation
in vivo. A system for organ replacement. Adv Exp of human embryonic stem cells. Brain Res. 2001;913
Med Biol. 1999;462:31–42. (2):201–5.
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 589

27. Schuldiner M, et al. Effects of eight growth factors on 42. Eggan K, et al. Mice cloned from olfactory sensory
the differentiation of cells derived from human neurons 1. Nature. 2004;428(6978):44–9.
embryonic stem cells. Proc Natl Acad Sci 43. Lanza RP, et al. Generation of histocompatible tissues
U S A. 2000;97(21):11307–12. using nuclear transplantation 1. Nat Biotechnol.
28. Zhang SC, et al. In vitro differentiation of transplant- 2002;20(7):689–96.
able neural precursors from human embryonic stem 44. Hwang WS, et al. Patient-specific embryonic stem
cells. Nat Biotechnol. 2001;19(12):1129–33 [see cells derived from human SCNT blastocysts 1. Sci-
comment]. ence. 2005;308(5729):1777–83.
29. Kaufman DS, et al. Hematopoietic colony- 45. Simerly C, et al. Molecular correlates of primate
forming cells derived from human embryonic stem nuclear transfer failures 2. Science. 2003;300
cells. Proc Natl Acad Sci U S A. 2001;98 (5617):297.
(19):10716–21. 46. Hwang WS, et al. Evidence of a pluripotent human
30. Kehat I, et al. Human embryonic stem cells can dif- embryonic stem cell line derived from a cloned blas-
ferentiate into myocytes with structural and functional tocyst 1. Science. 2004;303(5664):1669–74.
properties of cardiomyocytes. J Clin Invest. 2001;108 47. Byrne J et al. Producing primate embryonic stem cells
(3):407–14 [see comment]. by somatic cell nuclear transfer 1. Nature. 2007;450
31. Levenberg S, et al. Endothelial cells derived from (7169):497–502.
human embryonic stem cells. Proc Natl Acad Sci 48. Mitalipov S. Reprogramming following somatic cell
U S A. 2002;99(7):4391–6. nuclear transfer in primates is dependent upon nuclear
32. Assady S, et al. Insulin production by human embry- remodeling. Hum Reprod. 2007;22:2232–42.
onic stem cells. Diabetes. 2001;50(8):1691–7. 49. Atala A. Tissue engineering of reproductive tissues
33. Itskovitz-Eldor J, et al. Differentiation of human and organs. Fertil Steril. 2012;98(1):21–9.
embryonic stem cells into embryoid bodies 50. Dinnyés A, De Sousa P, King T, Wilmut I. Somatic
compromising the three embryonic germ layers. Mol cell nuclear transfer: recent progress and challenges.
Med. 2000;6(2):88–95. Cloning Stem Cells. 2002;4(1):81–90.
34. Reubinoff BE, et al. Embryonic stem cell lines from 51. Takahashi K, Yamanaka S. Induction of pluripotent
human blastocysts: somatic differentiation in vitro. stem cells from mouse embryonic and adult fibroblast
Nature Biotechnol. 2000;18(4):399–404 [see com- cultures by defined factors 2. Cell. 2006;126
ment] [erratum appears in Nat Biotechnol (4):663–76.
2000 May;18(5):559]. 52. Wernig M, et al. In vitro reprogramming of fibroblasts
35. Thomson JA, et al. Embryonic stem cell lines derived into a pluripotent ES-cell-like state 1. Nature.
from human blastocysts. Science. 1998;282 2007;448(7151):318–24.
(5391):1145–7 [see comment] [erratum appears in 53. Takahashi K, et al. Induction of pluripotent stem cells
Science 1998 Dec 4;282(5395):1827]. from adult human fibroblasts by defined factors. Cell.
36. Tian H, Bharadwaj S, Liu Y, Ma PX, Atala A, Zhang 2007;131(5):861–72.
Y. Differentiation of human bone marrow mesenchy- 54. Yu J et al. Induced pluripotent stem cell lines derived
mal stem cells into bladder cells: potential for uro- from human somatic cells. Science 2007; 318
logical tissue engineering. Tissue Eng Part (5858):1917–1920.
A. 2010;16(5):1769–79. 55. Okita K, Ichisaka T, Yamanaka S. Generation of
37. Bharadwaj S, Liu G, Shi Y, Wu R, Yang B, He T, germline-competent induced pluripotent stem cells.
Fan Y, Lu X, Zhou X, Liu H, Atala A, Rohozinski J, Nature. 2007;448(7151):313–17.
Zhang Y. Multipotential differentiation of human 56. Meissner A, Wernig M, Jaenisch R. Direct
urine-derived stem cells: potential for therapeutic reprogramming of genetically unmodified fibroblasts
applications in urology. Stem Cells. 2013;31 into pluripotent stem cells. Nat Biotechnol. 2007;25
(9):1840–56. (10):1177–81.
38. Wilmut I, et al. Viable offspring derived from fetal 57. Obokata H, Wakayama T, Sasai Y, Kojima K, Vacanti
and adult mammalian cells. Nature. 1997;385 MP, Niwa H, Yamato M, Vacanti CA. Stimulus-
(6619):810–13 [see comment] [erratum appears in triggered fate conversion of somatic cells into
Nature 1997 Mar 13;386(6621):200]. pluripotency. Nature. 2014;505(7485):641–7.
39. Brambrink T, et al. ES cells derived from cloned and 58. Obokata H, Sasai Y, Niwa H, Kadota M, Andrabi M,
fertilized blastocysts are transcriptionally and func- Takata N, Tokoro M, Terashita Y, Yonemura S,
tionally indistinguishable 1. Proc Natl Acad Sci Vacanti CA, Wakayama T. Bidirectional developmen-
U S A. 2006;103(4):933–8. tal potential in reprogrammed cells with acquired
40. Rideout III WM, et al. Correction of a genetic defect pluripotency. Nature. 2014;505(7485):676–80.
by nuclear transplantation and combined cell and 59. De Coppi P, et al. Isolation of amniotic stem cell lines
gene therapy. Cell. 2002;109(1):17–27. with potential for therapy.[see comment]. Nat
41. Hochedlinger K, Jaenisch R. Monoclonal mice gen- Biotechnol. 2007;25(1):100–6.
erated by nuclear transfer from mature B and T donor 60. Jiang G, Di Bernardo J, Delong CJ, Monteiro da
cells. Nature. 2002;415(6875):1035–8. Rocha A, O’Shea KS, Kunisaki SM, Induced
590 A.G. Hester and A. Atala

Pluripotent Stem Cells from Human Placental 76. Harris LD, Kim BS, Mooney DJ. Open pore biode-
Chorion for Perinatal Tissue Engineering Applica- gradable matrices formed with gas foaming. J Biomed
tions. Tissue Eng Part C Methods. 2014;20 Mater Res. 1998;42(3):396–402.
(9):731–40. 77. Peppas NA, Langer R. New challenges in biomate-
61. Zhong ZN, Zhu SF, Yuan AD, Lu GH, He ZY, Fa ZQ, rials. Science. 1994;263(5154):1715–20 [see
Li WH. Potential of placenta-derived mesenchymal comment].
stem cells as seed cells for bone tissue engineering: 78. Chen F, Yoo JJ, Atala A. Experimental and clinical
preliminary study of osteoblastic differentiation and experience using tissue regeneration for urethral
immunogenicity. Orthopedics. 2012;35(9):779–88. reconstruction. World J Urol. 2000;18(1):67–70.
62. Kim BS, Mooney DJ. Development of biocompatible 79. Atala A, et al. Formation of urothelial structures
synthetic extracellular matrices for tissue engineering. in vivo from dissociated cells attached to biodegrad-
Trends Biotechnol. 1998;16(5):224–30. able polymer scaffolds in vitro. J Urol. 1992;148(2 Pt
63. Bergsma JE, et al. In vivo degradation and biocom- 2):658–62.
patibility study of in vitro pre-degraded 80. Olsen L, et al. Urethral reconstruction with a new
as-polymerized polyactide particles. Biomaterials. synthetic absorbable device. An experimental study.
1995;16(4):267–74 [see comment]. Scand J Urol Nephrol. 1992;26(4):323–6.
64. Hynes RO. Integrins: versatility, modulation, and sig- 81. Kropp BP, et al. Rabbit urethral regeneration using
naling in cell adhesion. Cell. 1992;69(1):11–25. small intestinal submucosa onlay grafts. Urology.
65. Pariente JL, Kim BS, Atala A. In vitro biocompatibil- 1998;52(1):138–42.
ity assessment of naturally derived and synthetic bio- 82. Sievert KD, et al. Homologous acellular matrix graft
materials using normal human urothelial cells. for urethral reconstruction in the rabbit: histological
J Biomed Mater Res. 2001;55(1):33–9. and functional evaluation. J Urol. 2000;163
66. Pariente JL, Kim BS, Atala A. In vitro biocompatibil- (6):1958–65.
ity evaluation of naturally derived and synthetic bio- 83. De Filippo RE, Yoo JJ, Atala A. Urethral replacement
materials using normal human bladder smooth muscle using cell seeded tubularized collagen matrices.
cells. J Urol. 2002;167(4):1867–71. J Urol. 2002;168(4 Pt 2):1789–92; discussion 1792–3.
67. Li ST. Biologic biomaterials: tissue derived biomate- 84. Atala A. Experimental and clinical experience with
rials (collagen). In: JD B, editor. The biomedical tissue engineering techniques for urethral reconstruc-
engineering handbook. Boca Raton: CRS Press; tion. Urol Clin N A. 2002;29(2):485–92.
1995. p. 627–47. 85. El-Kassaby AW, et al. Urethral stricture repair with an
68. Arora PD, et al. A novel model system for character- off-the-shelf collagen matrix. J Urol. 2003;169
ization of phagosomal maturation, acidification, and (1):170–3; discussion 173.
intracellular collagen degradation in fibroblasts. 86. Bhargava S, Patterson JM, Inman RD, MacNeil S,
J Biol Chem. 2000;275(45):35432–41. Chapple CR. Tissue-engineered buccal mucosa
69. Silver FH, Pins G. Cell growth on collagen: a review urethroplasty-clinical outcomes. Eur Urol. 2008;53
of tissue engineering using scaffolds containing extra- (6):1263–9.
cellular matrix. J Long Term Eff Med Implants. 87. Raya-Rivera A, Esquiliano DR, Yoo JJ, Lopez-
1992;2(1):67–80. Bayghen E, Soker S, Atala A. Tissue-engineered
70. Sams AE, Nixon AJ. Chondrocyte-laden collagen autologous urethras for patients who need reconstruc-
scaffolds for resurfacing extensive articular cartilage tion: an observational study. Lancet. 2011;377
defects. Osteoarthritis Cartilage. 1995;3(1):47–59. (9772):1175–82.
71. Shen Y, Dai L, Li X, Liang R, Guan G, Zhang Z, 88. Fossum M, Svensson J, Kratz G, Nordenskjold
Cao W, Liu Z, Mei S, Liang W, Qin S, Xu J, Chen A. Autologous in vitro cultured urothelium in hypo-
H. Epidermal stem cells cultured on collagen- spadias repair. J Pediatr Urol. 2007;3(1):10–8.
modified chitin membrane induce in situ tissue regen- 89. Cattan V, Bernard G, Rousseau A, Bouhout S,
eration of full-thickness skin defects in mice. PLoS Chabaud S, Auger FA, Bolduc S. Mechanical
One. 2014;9(2). stimuli-induced urothelial differentiation in a human
72. Smidsrod O, Skjak-Braek G. Alginate as immobiliza- tissue-engineered tubular genitourinary graft. Eur
tion matrix for cells. Trends Biotechnol. 1990;8 Urol. 2011;60(6):1291–8.
(3):71–8. 90. Imbeault A, Bernard G, Rousseau A, Morissette A,
73. Lim F, Sun AM. Microencapsulated islets as Chabaud S, Bouhout S, Bolduc S. An endothelialized
bioartificial endocrine pancreas. Science. 1980;210 urothelial cell-seeded tubular graft for urethral
(4472):908–10. replacement. Can Urol Assoc J. 2013;7(1–2):E4–9.
74. Gilding D. Biodegradable polymers. In: Williams D, 91. Atala A, et al. Implantation in vivo and retrieval of
editor. Biocompatibility of clinical implant materials. artificial structures consisting of rabbit and human
Boca Raton: CRC Press; 1981. p. 209–32. urothelium and human bladder muscle. J Urol.
75. Mikos AG, et al. Wetting of poly(L-lactic acid) and 1993;150(2 Pt 2):608–12.
poly(DL-lactic-co-glycolic acid) foams for tissue cul- 92. Jayo MJ, Jain D, Ludlow JW, Payne R, Wagner BJ,
ture. Biomaterials. 1994;15(1):55–8. McLorie G, Bertram TA. Long-term durability, tissue
19 Translational Research Methods: Tissue Engineering of the Kidney and Urinary Tract 591

regeneration and neo-organ growth during skeletal 109. Ito T, et al. Bone marrow is a reservoir of repopulating
maturation with a neo-bladder augmentation con- mesangial cells during glomerular remodeling. J Am
struct. Regen Med. 2008;3(5):671–82. Soc Nephrol. 2001;12(12):2625–35.
93. Yoo JJ, Olson J, Atala A, Kim B. Regenerative med- 110. Iwano M, et al. Evidence that fibroblasts derive from
icine strategies for treating neurogenic bladder. Int epithelium during tissue fibrosis. J Clin Invest.
Neurourol J. 2011;15(3):109–19. 2002;110(3):341–50.
94. Zhang Y, Frimberger D, Cheng EY, Lin HK, Kropp 111. Poulsom R, et al. Bone marrow contributes to renal
BP. Challenges in a larger bladder replacement with parenchymal turnover and regeneration. J Pathol.
cell-seeded and unseeded small intestinal submucosa 2001;195(2):229–35.
grafts in a subtotal cystectomy model. BJU Int. 112. Rookmaaker MB, et al. Bone-marrow-derived cells
2006;98(5):1100–5. contribute to glomerular endothelial repair in experi-
95. Farhat WA, Yeger H. Does mechanical stimulation mental glomerulonephritis. Am J Pathol. 2003;163
have any role in urinary bladder tissue engineering? (2):553–62.
World J Urol. 2008;26(4):301–5. 113. Folkman J, Hochberg M. Self-regulation of
96. Ikarashi K, et al. Bone marrow cells contribute to growth in three dimensions. J Exp Med. 1973;138
regeneration of damaged glomerular endothelial (4):745–53.
cells. Kidney Int. 2005;67(5):1925–33. 114. Atala A, Schlussel RN, Retik AB. Renal cell growth
97. Kale S, et al. Bone marrow stem cells contribute to in vivo after attachment to biodegradable polymer
repair of the ischemically injured renal tubule. J Clin scaffolds. J Urol. 1995;153(4):10.
Invest. 2003;112(1):42–9. 115. Tachibana M, Nagamatsu GR, Addonizio JC. Ureteral
98. Lin F, et al. Hematopoietic stem cells contribute to the replacement using collagen sponge tube grafts. J Urol.
regeneration of renal tubules after renal ischemia- 1985;133(5):866–9.
reperfusion injury in mice. J Am Soc Nephrol. 116. Freed LE, et al. Biodegradable polymer scaffolds for
2003;14(5):1188–99. tissue engineering. Bio/Technology. 1994;12
99. Lin F, Moran A, Igarashi P. Intrarenal cells, not bone (7):689–93.
marrow-derived cells, are the major source for regen- 117. Hubbell JA, et al. Endothelial cell-selective materials
eration in postischemic kidney. J Clin Invest. for tissue engineering in the vascular graft via a new
2005;115(7):1756–64. receptor. Bio/Technology. 1991;9(6):568–72.
100. Prockop DJ. Marrow stromal cells as stem cells for 118. Mooney DJ, et al. Stabilized polyglycolic acid fibre-
nonhematopoietic tissues. Science. 1997;276 based tubes for tissue engineering. Biomaterials.
(5309):71–4. 1996;17(2):115–24.
101. Yokoo T, et al. Human mesenchymal stem cells in 119. Wald HL, et al. Cell seeding in porous transplantation
rodent whole-embryo culture are reprogrammed to devices. Biomaterials. 1993;14(4):270–8.
contribute to kidney tissues. Proc Natl Acad Sci 120. Arenas-Herrera JE, Ko IK, Atala A, Yoo
U S A. 2005;102(9):3296–300. JJ. Decellularization for whole organ bioengineering.
102. Carley WW, Milici AJ, Madri JA. Extracellular Biomed Mater. 2013;8(1).
matrix specificity for the differentiation of 121. Ross EA, Williams MJ, Hamazaki T, Terada N, Clapp
capillary endothelial cells. Exp Cell Res. 1988;178 WL, Adin C, Ellison GW, Jorgensen M, Batich
(2):426–34. CD. Embryonic stem cells proliferate and differenti-
103. Horikoshi S, Koide H, Shirai T. Monoclonal anti- ate when seeded into kidney scaffolds. J Am Soc
bodies against laminin A chain and B chain in the Nephrol. 2009;20(11):2338–47.
human and mouse kidneys. Lab Invest. 1988;58 122. Nakayama KH, Batchelder CA, Lee CI, Tarantal
(5):532–8. AF. Decellularized rhesus monkey kidney as a three-
104. Humes HD, Cieslinski DA. Interaction between dimensional scaffold for renal tissue engineering. Tis-
growth factors and retinoic acid in the induction of sue Eng Part A. 2010;16(7):2207–16.
kidney tubulogenesis in tissue culture. Exp Cell Res. 123. Baptista PM, Orlando G, Mirmalek-Sani SH,
1992;201(1):8–15. Siddigui M, Atala A, Soker S. Whole organ
105. Milici AJ, Furie MB, Carley WW. The formation of decellularization – a tool for bioscaffold fabrication
fenestrations and channels by capillary endothelium and organ bioengineering. Conf Proc IEEE Eng Med
in vitro. Proc Natl Acad Sci U S A. 1985;82 Biol Soc. 2009;2009:6526–9.
(18):6181–5. 124. Dekel B, et al. Human and porcine early kidney pre-
106. Schena FP. Role of growth factors in acute renal cursors as a new source for transplantation. Nat Med.
failure. Kidney Int Suppl. 1998;66:S11–15. 2003;9(1):53–60.
107. Lanza RP, et al. Generation of histocompatible tissues 125. Steer DL, et al. A strategy for in vitro propagation of
using nuclear transplantation. Nat Biotechnol. rat nephrons. Kidney Int. 2002;62(6):1958–65.
2002;20(7):689–96. 126. Rogers SA, et al. Transplantation of developing meta-
108. Gupta S, et al. A role for extrarenal cells in the nephroi into adult rats. Kidney Int. 1998;54(1):27–37.
regeneration following acute renal failure. Kidney 127. Hammerman MR. Transplantation of embryonic kid-
Int. 2002;62(4):1285–90. neys. Clin Sci (Lond). 2002;103(6):599–612.
592 A.G. Hester and A. Atala

128. Hammerman MR. Xenotransplantation of developing Benigni A, Remuzzi A, Remuzzi G. Recellularization


kidneys. Am J Physiol Renal Physiol. 2002;283(4): of well-preserved acellular kidney scaffold using
F601–6. embryonic stem cells. Tissue Eng Part A. 2014;20
129. Rogers SA, Powell-Braxton L, Hammerman (9–10):1486–98.
MR. Insulin-like growth factor I regulates renal devel- 143. Kershen RT, et al. Reconstitution of human corpus
opment in rodents. Dev Genet. 1999;24(3–4):293–8. cavernosum smooth muscle in vitro and in vivo. Tis-
130. Nigam SK. Concise review: can the intrinsic power of sue Eng. 2002;8(3):515–24.
branching morphogenesis be used for engineering 144. Kwon TG, Yoo JJ, Atala A. Autologous penile cor-
epithelial tissues and organs? Stem Cells Transl pora cavernosa replacement using tissue engineering
Med. 2013;2(12):993–1000. techniques. J Urol. 2002;168(4 Pt 2):1754–8.
131. Rosines E, Johkura K, Zhang X, Schmidt HJ, 145. Palmer BW, Kropp BP. Update on tissue engineering
Decambre M, Bush KT, Nigam SK. Constructing in pediatric urology. Curr Urol Rep. 2013;14
kidney-like tissues from cells based on programs for (4):327–32.
organ development: toward a method of in vitro tissue 146. Elmore JM, Kirsch AJ, Scherz HC, Smith EA. Small
engineering of the kidney. Tissue Eng Part A. 2010;16 intestinal submucosa for corporeal body grafting in
(8):2441–55. severe hypospadias requiring division of the urethral
132. Basu J, Ludlow JW. Developmental engineering the plate. J Urol. 2007;178(4 Pt 2):1698–701.
kidney: leveraging principles of morphogenesis for 147. Leslie JA, Cain MP, Kaefer M, Meldrum KK,
renal regeneration. Birth Defects Res C Embryo Misseri R, Rink RC. Corporeal grafting for severe
Today. 2012;96(1):30–8. hypospadias: a single institution experience with
133. Fung LCT, Elenius K, Freeman M, Donovan MJ, 3 techniques. J Urol. 2008;180(4 Suppl):1749–52.
Atala A. Reconstitution of poor EGFr-poor renal epi- 148. Hayn MH, Bellinger MF, Schneck FX. Small intes-
thelial cells into tubular structures on biodegradable tine submucosa as a corporal body graft in the repair
polymer scaffold. Pediatrics. 1996;98(Suppl):S631. of severe chordee. Urology. 2009;73(2):277–9.
134. Yoo J, Ashkar S, Atala A. Creation of functional 149. Castellan M, Gosalbez R, Devendra J, Bar-Yosef Y,
kidney structures with excretion of kidney-like fluid Labbie A. Ventral corporal body grafting for
in vivo. Pediatrics. 1996;98:S605. correcting severe penile curvature associated with
135. Evans MJ, et al. Mitochondrial DNA genotypes in single or two-stage hypospadias repair. J Pediatr
nuclear transfer-derived cloned sheep. Nat Genet. Urol. 2011;7(3):289–93.
1999;23(1):90–3. 150. Wang T, Koh C, Yoo JJ. Creation of an engineered
136. Hiendleder S, et al. Transmitochondrial differences uterus for surgical reconstruction. New Orleans:
and varying levels of heteroplasmy in nuclear transfer American Academy of Pediatrics Section on Urology;
cloned cattle. Mol Reprod Dev. 1999;54(1):24–31. 2003.
137. Steinborn R, et al. Mitochondrial DNA heteroplasmy 151. Takagi S, Shimizu T, Kuramoto G, Ishitani K,
in cloned cattle produced by fetal and adult cell clon- Matsui H, Yamato M, Okano T. Reconstruction of
ing. Nat Genet. 2000;25(3):255–7. functional endometrium-like tissue in vitro and
138. Fischer Lindahl K, et al. Maternally transmitted anti- in vivo using cell sheet engineering. Biochem
gen of mice: a model transplantation antigen. Annu Biophys Res Commun. 2014;446(1):335–40.
Rev Immunol. 1991;9:351–72. 152. Schutte SC, Taylor RN. A tissue-engineered human
139. Hadley GA, Linders B, Mohanakumar endometrial stroma that responds to cues for secretory
T. Immunogenicity of MHC class I alloantigens differentiation, decidualization, and menstruation.
expressed on parenchymal cells in the human kidney. Fertil Steril. 2012;97(4):997–1003.
Transplantation. 1992;54(3):537–42. 153. De Filippo RE, Yoo JJ, Atala A. Engineering of
140. Yard BA, et al. Analysis of T cell lines from rejecting vaginal tissue in vivo. Tissue Eng. 2003;9(2):301–6.
renal allografts. Kidney Int Suppl. 1993;39:S133–8. 154. Zhu L, Zhou H, Sun Z, Lou W, Lang J. Anatomic and
141. Yoo JJ, Atala A. A novel gene delivery system using sexual outcomes after vaginoplasty using tissue-
urothelial tissue engineered neo-organs. J Urol. engineered biomaterial graft in patients with Mayer-
1997;158(3 Pt 2):1066–70. Rokitansky-K€ uster-Hauser syndrome: a new mini-
142. Bonandrini B, Figliuzzi M, Papadimou E, Morigi M, mally invasive and effective surgery. J Sex Med.
Perico N, Casiraghi F, Dipl C, Sangalli F, Conti S, 2013;10(6):1652–8.
Part IV
Clinical Approach to the Child with
Suspected Renal Disease
Clinical Evaluation of the Child with
Suspected Renal Disease 20
Mohan A. Shenoy and Nicholas J. A. Webb

Contents Clinical Examination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 602


General Assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 602
Presentation of Renal Disease in Children . . . . . . . 596 Growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 603
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 596 Nutrition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
Antenatal Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 596 Hydration/Volume Status . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
Abnormalities of Appearance of Urine . . . . . . . . . . . . . . 596 Cardiovascular System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
Abnormalities of Volume of Urine . . . . . . . . . . . . . . . . . . 598 Respiratory System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 605
Wetting and Abnormalities of the Passage of Urine 598 Abdomen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 605
Edema . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 599 Nervous System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 606
Asymptomatic Presentation Following Screening or Skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 606
Routine Assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 599 Face . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 607
Urinary Tract Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 600 Eyes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 607
Clinical History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 600 Ears . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 607
Antenatal History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 600 Musculoskeletal System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 608
Birth History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 601 Examination of the Urine . . . . . . . . . . . . . . . . . . . . . . . . . . 608
Family History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 601 Macroscopic Examination . . . . . . . . . . . . . . . . . . . . . . . . . . . 608
General Medical History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 601 Dipstick Examination of Urine . . . . . . . . . . . . . . . . . . . . . . 608
Urine and Micturition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 602
Dietary History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 602 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 611
Psychosocial History . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 602

M.A. Shenoy • N.J.A. Webb (*)


Department of Pediatric Nephrology, Royal Manchester
Children’s Hospital, Manchester, UK
e-mail: nicholas.webb@cmft.nhs.uk

# Springer-Verlag Berlin Heidelberg 2016 595


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_18
596 M.A. Shenoy and N.J.A. Webb

Presentation of Renal Disease kidney (MCDK), pelvi-ureteric junction obstruc-


in Children tion, and the polycystic kidney diseases. In the
case of posterior urethral valves, this has resulted
Introduction in the possibility of antenatal therapy, though the
efficacy and safety of this require further confir-
Renal disease in children may present with overt mation in clinical trials. These may be difficult to
symptoms clearly associated with the urinary recruit to: the UK PLUTO clinical trial investigat-
tract, such as the development of macroscopic ing vesico-amniotic shunting had to close early
hematuria or profound oliguria. However, in because of suboptimal recruitment [6]. Large kid-
many instances, symptoms may be very neys on antenatal imaging may suggest
nonspecific or seemingly mild. Children with hydronephrosis, polycystic kidney disease
chronic kidney disease present with a wide variety (PKD), MCDK, congenital nephrotic syndrome,
of symptoms including enuresis, failure to thrive, or rarely a renal tumor, whereas small kidneys
short stature, lethargy, and pallor. The onset may may point to the presence of renal dysplasia or
be silent and the progress insidious, with symp- hypoplasia. PKD, cystic dysplasia, and
toms only developing late in the disease course. glomerulocystic disease are common causes of
Urinary tract infection in infants and small chil- echobright kidneys. TCF2 gene mutations are
dren may, in contrast to older children, present found in almost one third of the children with
with nonspecific manifestations including poor antenatally diagnosed bilateral echogenic kidneys
feeding, vomiting, irritability, abdominal pain, [7]; however, counseling families with this muta-
failure to thrive, lethargy, and restlessness. The tion is difficult as the phenotype is extremely
possibility of renal disease should therefore be variable, even within a single family. The pres-
considered in the differential diagnosis of any ence of renal macrocysts in the antenatal period
child presenting to the hospital with acute or should alert the physician towards a diagnosis of
chronic symptoms. autosomal dominant PKD, PKD associated with
tuberous sclerosis, MCDK, or cystic dysplasia.
The sensitivity of ultrasound imaging does,
Antenatal Imaging however, result in the detection of many
minor abnormalities, particularly unilateral renal
Since the first report of renal abnormalities being pelvic dilatation, which appear to resolve
detected by antenatal ultrasonography in the spontaneously in later pregnancy or after birth
1970s, there have been major developments in and are of no long-term significance. Their detec-
the antenatal assessment of the urinary tract tion may be a source of significant parental anxi-
[1–4]. In most countries, pregnant women ety and multiple invasive investigations in the
undergo routine ultrasound assessment of the neonatal period where appropriate protocols are
fetus at various stages throughout pregnancy, not in operation.
including a detailed scan performed at around
20 weeks of gestation. Ultrasound is particularly
informative with regard to renal abnormalities, Abnormalities of Appearance of Urine
which account for around 20 % of all significant
fetal abnormalities detected during gestation Red or Dark Urine
[5]. Recent developments including 3D ultra- The presence of blood in the urine causes it to
sound and magnetic resonance imaging of the develop a pink to red color. Only a relatively small
fetus allow these anomalies to be studied in fur- amount of blood is necessary to produce discolor-
ther detail. This has resulted in many significant ation. Prolonged contact between blood in the
congenital and inherited abnormalities of the uri- urinary tract and acidic urine causes the heme
nary tract being detected antenatally, notably pos- pigment to become oxidized to a methaem deriv-
terior urethral valves, the multicystic dysplastic ative, giving the urine a brown color. In general,
20 Clinical Evaluation of the Child with Suspected Renal Disease 597

Table 1 False-positive hematuria; alternative causes of urinary red cells for differences in blood group
red urine type or, if required, DNA profiling.
Foods, e.g., beetroot, berries containing anthocyanins, Dark urine associated with jaundice in the neo-
and food dyes nate or older child should alert the physician to the
Hemoglobinuria, e.g., in intravascular hemolysis presence of conjugated hyperbilirubinemia, indic-
Myoglobinuria, e.g., in rhabdomyolysis
ative of significant liver or biliary tract disease.
Urate crystals (a cause of pink discoloration of nappies)
Bilirubin and urobilinogen will be detected on
Drugs, e.g., rifampicin, phenothiazines, desferrioxamine,
phenindione dipstick testing the urine in such cases.
Inborn errors of metabolism, e.g., porphyria and
alkaptonuria Cloudy Urine
The urine may become cloudy secondary to the
presence of white blood cells (pyuria) associated
the longer the contact and the more acidic the with urinary infection, calcium phosphate crys-
urine, the darker brown the urine becomes. tals, or a combination of calcium salts, uric acid,
Contamination with blood from menstruation oxalate, cystine, or struvite. These substances are
in older girls needs ruling out. There are a number normally present in the urine, though may be
of causes of false-positive hematuria, where alter- present in excess in various disease states. The
native substances produce red discoloration precipitation of phosphates and urates is enhanced
(Table 1). One of the most common of these is by refrigeration of the urine sample.
the pink discoloration seen in nappies caused by
the precipitation of urate crystals. Urine micros- Frothy Urine
copy is therefore mandatory following the detec- The presence of significant quantities of protein in
tion of red or dark urine. It is important that this is the urine will result in it becoming frothy. Presen-
performed on a fresh sample as red cell lysis tation with frothy urine should therefore prompt
occurs where samples are allowed to stand for the physician to perform dipstick analysis
prolonged periods prior to examination. followed by formal quantification of protein
The passage of fresh red blood in the urine, content.
with or without clots, is most likely to originate
from the lower urinary tract, particularly where Offensive or Unusual-Smelling Urine
this is most marked at the beginning or the end of A number of factors may result in an alteration in
the urinary stream. This highlights the importance the smell of urine. Infection with urea-splitting
of obtaining a comprehensive clinical history in organisms including Ureaplasma urealyticum, Pro-
such children. Causes include urethritis, trauma, teus, Staphylococcus, Klebsiella, Providentia, and
bladder calculus, and schistosomiasis. Urological Pseudomonas species may generate an ammonia-
assessment and cystoscopy are indicated in like smell, though the positive predictive value of
most such cases. More uncommon causes of offensive urine for bona fide urinary infection is
bleeding from the upper tracts include trauma, very low. A number of the inborn errors of metab-
arteriovenous malformations, tumors, and olism, including maple syrup urine disease, phenyl-
angiomyolipomas associated with tuberous ketonuria, and isovaleric acidemia, also produce
sclerosis. characteristic odors. The change in urine odor fol-
Fabricated and induced illness by proxy, pre- lowing the ingestion of asparagus was first reported
viously termed Munchausen syndrome by proxy, in the eighteenth century by a physician of the
is a very rare disorder, though falsification of urine French royal family [9]. This is thought to be due
samples by contamination with maternal or other to the presence of S-methyl-thioacrylate and
blood is one of the more common modes of pre- S-methyl-3-(methylthio)thiopropionate [10],
sentation [8]. A high index of suspicion needs to though a combination of methyl mercaptan,
be maintained to detect such cases. Absolute con- dimethyl sulfide, and small amounts of sulfur-
firmation of contamination requires analysis of oxidized compounds could also be responsible [11].
598 M.A. Shenoy and N.J.A. Webb

Passage of Gravel or Stones Table 2 Causes of polyuria in children


The passage of gravel or stones in the urine is an Increased fluid intake
unusual though recognized mode of presentation Psychogenic/behavioral polydipsia
of urinary tract calculi. Stones are either metabolic Hypothalamic polydipsia
or infective in origin. Hyperreninemia including Wilms tumor
Increased osmotic load
Recovery from acute kidney injury
Abnormalities of Volume of Urine Diabetes mellitus and other causes of hyperglycemia
Chronic kidney disease
Oliguria Following mannitol infusion
The otherwise healthy neonate is oliguric for the Failure of ADH production
Cranial diabetes insipidus
first 2–3 days of life until the onset of the postnatal
Basal skull fracture
diuresis. Ninety-two percent of neonates will pass
Cranial tumors
urine within the first 24 h of life, and almost all
Post hypophysectomy
newborns will do so in the first 48 h [12]. Beyond
Infection; encephalitis, meningitis, tuberculosis
the immediate neonatal period, oliguria is defined Vascular aneurysm or thrombosis
as urine output of less than 500 ml/24 h/1.73 m2, Failure of renal response to ADH
which is sufficient to maintain homeostasis. The Nephrogenic diabetes insipidus (X-linked)
most common cause of oliguria in children is Acquired unresponsiveness to ADH
intravascular volume depletion. Where this has Obstructive uropathy
been excluded or adequately treated and volume Hypokalemia
depletion persists, a search for intrinsic or obstruc- Hypercalcemia
tive renal disease should be commenced. Sickle cell disease
Drugs, e.g., amphotericin, methoxyflurane,
Polyuria and Polydipsia tetracycline
Of those children presenting to medical practi-
tioners with symptoms of polyuria, only a very
small proportion will have a significant renal or Wetting and Abnormalities
other pathology. A strict definition of polyuria of the Passage of Urine
does not exist, though a daily urine output exceed-
ing 2 l in school-aged children is unusual. It is Wetting is the most common urinary tract disorder
important to distinguish polyuria from frequency of childhood, though in most cases, there is no
of micturition where small volumes of urine are organic pathology. The majority of children will
passed frequently though the 24 h urine output is achieve daytime bladder control between 3 and
within normal limits. 4 years of age [13]. The prevalence of nocturnal
Significant pathologies which result in poly- enuresis is 15–20 % at age 5 years and up to 3 % in
uria are shown in Table 2 and include excessive young adults [14]. Nocturnal enuresis has a strong
fluid intake, increased osmotic load, failure to genetic predisposition and on its own, is a benign
produce or release antidiuretic hormone (ADH), self-limiting condition. Daytime wetting, which
and resistance to the actions of ADH in the kidney. can occur as a result of a number of disorders
The distinction between primary polydipsia and including detrusor overactivity, dysfunctional
primary polyuria may be inferred from the relative voiding, and, rarely, giggle incontinence, which
osmolality of plasma and urine and the response to is more common in girls and usually resolves by
controlled water deprivation. The distinction the age of 9 years. Secondary enuresis, which is
between pituitary and renal causes of polyuria defined as enuresis that develops after at least
may be made by the plasma ADH concentration 6 months of dryness, raises the possibility of an
and the urinary response to D-amino-arginine organic cause (e.g., neuropathic bladder) espe-
vasopressin. cially in boys; however, it is commonly associated
20 Clinical Evaluation of the Child with Suspected Renal Disease 599

with psychosocial disturbances such as parental in a gravity-dependent distribution, the ankle and
separation, the birth of a new child, or a death in sacral areas being the most severely affected. This
the family. Diabetes mellitus should always be may be associated with abdominal distension sec-
excluded. A careful voiding and wetting history ondary to ascites.
along with a focused physical examination will
help to identify any organic pathology. The pres-
ence of dysuria and frequency might suggest a Asymptomatic Presentation Following
diagnosis of urinary tract infection, whereas poor Screening or Routine Assessment
urinary stream and an enlarged bladder would
point to a diagnosis of a neuropathic bladder or While the universal routine testing of children’s
an obstructive pathology such as posterior ure- urine by dipstick examination is not performed in
thral valves. Examination of the urine for infec- the large majority of countries, this has been
tion is essential in all children with wetting. In a undertaken in Japan, having been introduced by
child with persistent daytime wetting, a pre- and the Ministry of Education in 1973 with the aim of
post-void ultrasound of the renal tract is useful to the early detection of asymptomatic renal disease
screen for urinary tract anomalies and to assess [15]. Outside of universal screening programs,
bladder emptying. Urodynamic assessment to children may have their urine tested at the time
identify abnormalities of bladder and sphincter of attendance at hospital emergency departments
function is indicated in children with a neuro- and outpatient clinics or as part of a routine med-
pathic bladder and in some children with persis- ical assessment for insurance or immigration pur-
tent and troublesome wetting who do not poses. Such testing may detect abnormalities in
demonstrate a good response to conservative the urine (commonly microscopic hematuria),
management. which results in referral for further assessment.
Pollakiuria, a benign, self-limiting condition, It is estimated that 5 % of children have hyper-
is characterized by the very frequent (every 5–20 tension [16]. Children may have hypertension
min) passage of urine during the daytime hours. It detected following a routine medical examination
most commonly presents in preschool-age chil- performed at school or for participation in partic-
dren and is not associated with wetting. ular sporting activities. In such instances, it is
firstly essential to rule out erroneously high
blood pressure, which has occurred as a result of
Edema the use of incorrect measuring equipment or white
coat hypertension. While renal disease is widely
Children with renal disease may present with reported to be the commonest cause of hyperten-
edema, a major clinical manifestation of ECF sion in children, the rising tide of childhood obe-
volume expansion. Edema may occur in the sity may soon see this become the predominant
acute nephritic syndrome and other causes of cause [17]. There is an ongoing debate as to
acute renal impairment as a result of failure of whether children ought to be screened for hyper-
salt and water excretion. Here, peripheral edema tension, especially since up to 40 % of children
is accompanied by intravascular volume expan- will have end-organ damage at the time of diag-
sion with hypertension and pulmonary edema. In nosis of hypertension [18].
the nephrotic syndrome, loss of plasma oncotic The screening of asymptomatic siblings of
pressure secondary to hypoproteinemia and index cases of renal disease is a difficult area.
increased vascular permeability result in the loss Clearly where the detection of significant pathol-
of fluid from the intravascular into the extravas- ogy necessitating the commencement of specific
cular space. Here, edema is initially first evident as therapy is possible, e.g., cystinosis, hemolytic
swelling of the periorbital region, which is often uremic syndrome associated with complement
most marked in the morning. With progressive abnormalities, and Alport syndrome, then appro-
fluid retention, more generalized edema develops priate testing should be performed. Siblings and
600 M.A. Shenoy and N.J.A. Webb

close contacts of children presenting with STEC- Diagnosis of urinary tract infection is impor-
associated hemolytic uremic syndrome are not tant; as aside from the acute morbidity associated
infrequently found to have evidence of varying with infection, the development of infection is an
degrees of renal impairment and anemia in the important marker of underlying congenital abnor-
absence of any significant symptoms, and such malities of the kidney and urinary tract (CAKUT),
detection will allow the commencement of in particular, vesicoureteric reflux. Furthermore,
appropriate therapy and follow-up. In contrast, infection itself is an important cause of renal cor-
there is no consensus as yet regarding screening tical scarring in those predisposed to this. There-
of asymptomatic siblings of children with fore, a diagnosis of urinary tract infection should
autosomal dominant PKD. Many authorities always result in consideration being given to
would recommend that ultrasound or genetic radiological assessment of the urinary tract. Ultra-
screening does not take place in these children as sonography of the kidneys and bladder should be
there is no proven treatment as yet. Potentially performed to detect anatomic abnormalities. Pre-
affected individuals could undergo an annual vious guidelines recommended that all children be
check of blood pressure and urinalysis until they investigated after a single infection, though more
are able to make a fully informed decision about recent guidelines, for instance, those produced by
the relative merits of presymptomatic detection, the UK National Institute for Health and Clinical
bearing in mind the implications that this may Excellence and the American Academy of Pedi-
have. However, preventative treatment for atrics, have recommended that further radiologi-
ADPKD may not be far off, and there may be cal investigations are only performed on the very
advantages in early identification of the condition. young and those with recurrent or difficult-to-treat
There are a number of situations where medical infections (http://www.nice.org.uk/guidance/
opinion is divided, for instance, whether index.jsp?action=byID&o=11819, http://pediat
newborn siblings of children with vesicoureteric rics.aappublications.org/content/early/2011/08/
reflux should undergo radiological assessment; 24/peds.2011-1330).
definitive answers to these questions are only
likely to be obtained through the enrolment of
such children in prospective randomized con- Clinical History
trolled trials.
Antenatal History

Urinary Tract Infection A record should be made of the results of antenatal


imaging studies, particularly the detailed scan
Urinary tract infection is common. Swedish data performed at around 20 weeks of gestation.
report a cumulative incidence by 2 years of age of Abnormalities of liquor volume should be
2.2 % in boys and 2.1 % in girls [19]. In the recorded. In the absence of rupture of the amniotic
northern United Kingdom, cumulative referral membranes, oligohydramnios or anhydramnios
rates by 7 years of age are as high as 2.8 % for after 16 weeks of gestation represents failure of
boys and 8.2 % for girls, rising to 3.6 and 11.3 %, urine excretion due to obstruction to urine flow or
respectively, by 16 years of age [20]. While older inability of the kidneys to produce urine. Polyuric
children may present with classical symptoms of states including neonatal Bartter syndrome and
either lower urinary tract infection (dysuria, fre- congenital nephrotic syndrome are associated
quency, wetting) or upper urinary tract infection with polyhydramnios; it should be remembered,
(systemic upset, fever, loin pain), these features however, that only a small proportion of cases of
are much less pronounced in the younger child; polyhydramnios are caused by renal disease.
here, nonspecific manifestations may include poor A raised maternal serum alpha-fetoprotein
feeding, vomiting, irritability, failure to thrive, level between 15 and 20 weeks of gestation is
lethargy, and abdominal pain. associated with spinal cord defects and the
20 Clinical Evaluation of the Child with Suspected Renal Disease 601

congenital nephrotic syndromes and is an indica- considered good practice to document the
tion for a detailed antenatal ultrasound scan and genogram in the case records. A variety of renal
amniocentesis. diseases including PKD, familial glomerulone-
Close attention should be paid to drug history. phritis, and tubular disorders, e.g., Bartter and
Maternal intake of medications such as COX-2 Gitelman syndromes, are inherited. A family his-
inhibitors, angiotensin-converting enzyme inhibi- tory of epilepsy or learning difficulties should
tors (ACEis), or angiotensin receptor blockers alert the clinician to the possibility of tuberous
(ARBs) in the second and third trimester is asso- sclerosis in a child with polycystic kidneys. An
ciated with renal failure in the neonatal period. increasing number of hereditary renal disorders
First-trimester exposure to ACEis or ARBs can can now be diagnosed by DNA analysis, and it
lead to major cardiovascular, central nervous sys- is therefore essential that the family is provided
tem, and renal malformations [21]. with detailed counseling with regard to the poten-
Other aspects of maternal medical history tial future implications.
should be noted. Previous recurrent miscarriages Parental consanguinity is common in a number
should alert the physician to the possibility of a of societies including the Muslim Middle Eastern
chromosomal abnormality. Maternal gestational countries and the UK Pakistani community, where
diabetes and obesity are associated with increased up to 55 % are married to a first cousin [25]. This
risk of CAKUT. Maternal obesity is also associ- should raise the possibility of conditions which
ated with increased risk of neural tube are transmitted in an autosomal recessive manner
defects [22]. such as autosomal recessive PKD, juvenile
nephronophthisis, and cystinosis. End-stage
renal failure of all causes is also more prevalent
Birth History in this population [26].
Deafness and renal failure in a male relative
An enlarged placenta (greater than 25 % of the may suggest a diagnosis of Alport syndrome,
infant’s birth weight) is a feature of congenital which is most commonly inherited in an
nephrotic syndrome. The presence of hematuria X-linked manner. The inheritance of isolated
following perinatal asphyxia may be due to renal vesicoureteric reflux is thought to be consistent
venous thrombosis. Umbilical arterial catheteriza- with multifactorial or autosomal dominant with
tion is associated with vascular damage and is the reduced penetrance and has a sibling recurrence
commonest cause of hypertension in the neonatal rate of 30–50 % [27].
period [23]. The presence of a single umbilical
artery in an infant is associated with an increased
risk of a variety of renal anomalies including General Medical History
vesicoureteric reflux, MCDK, and renal aplasia
and dysplasia [24]. It is debatable as to whether Renal disease may be a feature of a number of
these infants merit further imaging unless the childhood disorders. A pediatric nephrologist
antenatal scan has demonstrated any abnormality; working in a tertiary center will often be asked
in such cases, an ultrasound scan of the renal tract to see children under the care of other specialists
will provide useful initial information and guide and must therefore be familiar with the renal man-
further evaluation. ifestations of systemic disorders and iatrogenic
problems. A number of drugs including nonste-
roidal anti-inflammatory drugs (NSAID),
Family History aminoglycosides, and acicyclovir are associated
with AKI. Renal involvement may be a part of a
A detailed family history forms an essential com- multisystem disorder such as Henoch-Schönlein
ponent of the clinical evaluation and may often purpura or systemic lupus erythematosus.
provide important clues to the diagnosis. It is Hyponatremia due to cerebral salt wasting or
602 M.A. Shenoy and N.J.A. Webb

syndrome of inappropriate ADH secretion is a that these children are often malnourished, and
common problem on the neurosurgical ward. input from an experienced pediatric dietician is
A diagnosis of retinitis pigmentosa should alert mandatory.
to the possibility of juvenile nephronophthisis. One of the consequences of the obesity epi-
Tubulopathy is often seen in children on the pedi- demic has been the increasing numbers of over-
atric oncology ward following chemotherapy with weight children with hypertension, disturbed
agents such as adriamycin or ifosfamide. Atypical glucose metabolism, and hyperlipidemia
hemolytic uremic syndrome is known to be asso- [29]. Excessive salt intake may be associated
ciated with cisplatin, the oral contraceptive pill, with hypercalciuria.
and tacrolimus. Asymptomatic renal calculi are
known to occur after a short course of ceftriaxone,
and nephrocalcinosis is associated with the Psychosocial History
prolonged use of furosemide.
The impact of the disease on the child, for exam-
ple, the amount of school missed, body image,
Urine and Micturition limitations on lifestyle, self-esteem, and peer reac-
tions, should be assessed. There is a wealth of
The clinical history should record information evidence supporting the view that chronic physi-
regarding daytime and nighttime wetting (see cal illness and disability are risk factors for mental
above) and abnormalities of micturition, includ- health problems in children. Social disadvantage,
ing the nature of the urinary stream (e.g., hesi- poverty, poor housing, educational failure, and
tancy, staccato micturition, terminal dribbling, the family instability are other risk factors. Caring
need to sit to void, etc.). Poor urinary stream for a chronically ill child is a huge challenge for
should raise the possibility of posterior urethral any family, and parents often have to give up
valves in a male infant, though following the employment in order to achieve this. Impact on
introduction of routine ultrasound scanning, the siblings should be given consideration. A skilled
majority of such cases would be detected in the social worker and child psychiatrist or psycholo-
antenatal period. By 3 years of age, most children gist are therefore essential members of a multipro-
have some conscious control over micturition and fessional pediatric nephrology team. The impact
achieve daytime control, but enuresis and daytime of the disease on the child’s intellectual, emo-
accidents can continue to occur. In an older child, tional, and social progress must be assessed. An
the voiding frequency and an estimate of the uri- awareness of the risk factors will help the physi-
nary volume should be documented [28]. cian identify those children who are at particularly
high risk of developing mental health problems.
Adherence with prescribed medications is a par-
Dietary History ticular problem during adolescence and is the
commonest cause of rejection and graft loss in
The fluid and dietary restrictions in renal disease this age group [30].
depend on the type and stage of disease. For
instance, while salt and fluid restriction is not
necessary in polyuric states such as proximal Clinical Examination
renal tubular disorders and Bartter syndrome,
anephric children on dialysis will require tight General Assessment
control of their salt and fluid intake. Most children
with advanced renal failure will be on a restricted An initial rapid assessment of the level of con-
potassium, phosphorus, and protein diet. sciousness, airway, breathing, and circulation
Anorexia associated with renal failure means should be performed to determine how ill the
20 Clinical Evaluation of the Child with Suspected Renal Disease 603

1. Airway 6. Abdomen
Breathing Scars, PD catheter
Circulation Distension, peritonitis
III or well child Renal mass
Bowel sounds
2. General assessment
Transplant kidney-
Hydration
tendemess/bruit
Growth - centile charts
Genitalia, urethral opening
Nutrition
Anus
Dysmorphic features
7. Central nervous system
Tanner staging
Mental status
3. Skin Cranial nerves
Sallow Tone, power, reflexes
Pallor Gait, coordination
Jaundice
Oedema 8. Musculoskeletal system
Cushingold features Muscle wasting, weakness
Scratch marks Hemihypertrophy
Rash Joint swelling
Central venous/ Spine, sacrum
haemodialysis catheter Linb deformity
Wrist swelling
Eyes 9. Urine analysis
Face and oral cavity Microscopy
Ear deformity Dipstick for blood, protein,
nitrite leucocytes and glucose
4. Cardiovascular system
Pulse including femorals
BP
JVP
AV fistula
Cardiomegaly
Murmur
Bruits
5. Chest
Kussmaul breathing
Rib rosary
Harrison’s sulcus
Pleural effusion/pulmonary oedema

Fig. 1 Assessment of the child with renal disease

child is (Fig. 1). Any obvious dysmorphic features Growth


should be recorded. Pain associated with renal
disease is often confused with back pain and Growth is an invaluable measure of health during
other causes of abdominal pain in childhood. childhood, and accurate measurement of height
The state of the child (clothing and hygiene) and weight along with the head circumference in
should be noted. A record of the mood and younger children is therefore essential. Obtaining
demeanor of the child together with the family parental height is important to calculate the target
interaction is informative. height centile for the child. Most growth charts
604 M.A. Shenoy and N.J.A. Webb

express anthropometric measures in terms of pulse, hepatomegaly, and the development of pul-
percentiles; however, not infrequently children monary edema. Sometimes, assessment of hydra-
with renal disease are significantly below the tion state may be difficult. In this situation, rather
lowest percentile lines, and therefore, expressing than administering a large-volume fluid bolus
height, weight, and body mass index in terms of such as 20 ml/kg, it might be more appropriate
standard deviation score (SDS or z-score) is to administer a smaller volume such as 5–10
sometimes more informative. Pubertal develop- ml/kg and then to re-evaluate the child.
ment (Tanner staging) should also be formally
assessed.
Cardiovascular System

Nutrition Pulse
The rate, rhythm, and volume of the pulse should
Assessment of nutrition in children with renal be recorded. Tachycardia with a low-volume
disease can be a challenge; fluid overload and pulse is a feature of intravascular volume
abnormalities in the distribution of fat and lean depletion. The presence of a gallop rhythm sug-
body tissue can lead to misinterpretation of the gests congestive cardiac failure. The femoral
nutritional anthropometric measures [31]. The pulses should always be examined in a child
commonly used nutritional markers are dietary with hypertension; coarctation of the aorta may
assessment, height, estimated dry weight, body present with hypertension and cardiac failure. The
mass index, serum albumin, and skin fold thick- presence of bruits on auscultation over major
ness. Tools for assessing body composition such arteries such as the carotid or renal arteries sug-
as dual energy x-ray absorptiometry and bioelec- gests extensive vascular disease or renal artery
trical impedance analysis can sometimes provide stenosis.
useful information.
Blood Pressure
Measurement of the blood pressure is an integral
Hydration/Volume Status part of the assessment of the child with renal
disease. It is important to ensure that the right
Assessment of the state of hydration and circula- equipment and standards are used to ensure that
tory status is an important skill for all physicians hypertension is correctly diagnosed; a wrong
dealing with children with kidney disease. An diagnosis due to incorrect measurement can lead
understanding of total body water and its distribu- to a battery of unnecessary invasive tests. There is
tion between the intravascular (plasma) and extra- an increasing use of ambulatory blood pressure
cellular fluid (interstitial) compartments is monitoring in children, especially to diagnose
essential. A child with nephrotic syndrome usu- essential and white coat hypertension [32].
ally presents with a recent increase in weight and The investigation of hypertension is discussed
peripheral edema, reflecting an increase in ECF in ▶ Chap. 61, “Evaluation of Hypertension in
volume, though at the same time may exhibit Childhood Diseases”.
signs of intravascular volume depletion such as
reduced capillary refill time, cold extremities, Precordium
tachycardia, and oliguria. Occasionally, clinical Fluid overload, long-standing hypertension, and
signs may be subtle, and measurement of urinary cardiomyopathy will give rise to cardiomegaly.
sodium, fractional excretion of sodium (FeNa), Pericarditis with a rub may be a feature of severe
and urine osmolality may provide additional uremia. A heart murmur may suggest a structural
information. In contrast, expansion of the intra- anomaly but can also be a feature of infective
vascular volume, such as in nephritic states, endocarditis or a hyperdynamic state as occurs in
results in hypertension, raised jugular venous anemia.
20 Clinical Evaluation of the Child with Suspected Renal Disease 605

Table 3 Causes of an abdominal mass of renal origin


Abnormality Causes if unilateral Causes if bilateral
Hydronephrosis (obstructive and PUJ obstruction Causes of bladder outlet obstruction including
nonobstructive) posterior urethral valves
VUJ obstruction Eagle-Barrett syndrome
Primary megaureter
Cystic mass Multicystic dysplastic PKD (dominant and recessive)
kidney
Simple cyst Cystic disease associated with syndromal
diagnosis
Cystic dysplasia Cystic dysplasia
Infection Acute pyelonephritis
Xanthogranulomatous
pyelonephritis
Perinephric abscess
Vascular Renal venous thrombosis
Arterial thrombosis
Acute cortical necrosis
Tumors Wilms tumor
Mesoblastic nephroma
Leukemic infiltrate
Miscellaneous Compensatory Beckwith-Wiedemann syndrome
hypertrophy
Duplex kidney Acute renal failure
Fused crossed ectopia Storage disorders
Congenital nephrotic syndrome

Respiratory System aware of the possibility of adhesions and obstruc-


tion presenting as an acute surgical abdomen.
Hyperventilation with Kussmaul respiration sug- Tenderness with guarding and rigidity is highly
gests the presence of metabolic acidosis. Rachitic suggestive of peritonitis, and in these circum-
deformity of the ribs can be seen in long-standing stances, the opinion of a pediatric surgeon should
chronic kidney disease. Dullness on percussion be sought.
over the lung fields might suggest pleural effusion
in a child with nephrotic syndrome or a peritino- Abdominal Mass
pleural leak in a patient on peritoneal dialysis. Prior to the onset of routine antenatal imaging, the
Fine crepitations over the lung bases may indicate detection of an abdominal mass was a reasonably
the presence of pulmonary edema. frequent mode of presentation of a variety of renal
and urological disorders, including the MCDK,
pelvi-ureteric junction obstruction, and other
Abdomen causes of hydronephrosis. In the neonatal period,
55 % of abdominal masses are renal in origin
Abdominal Distension (hydronephrosis 25 % and MCDK 15 %). In
Abdominal distension in renal disease may be due later infancy and childhood, the proportion of
to ascites, bladder enlargement due to urinary abdominal masses which are renal in origin
retention and mass (see below), and the presence increases, largely due to the increased rate of
of peritoneal dialysis fluid. In children who have malignant tumors in the age group [33].
undergone previous abdominal surgery, for exam- Renal causes of an abdominal mass are shown
ple, urinary diversion procedures, one should be in Table 3.
606 M.A. Shenoy and N.J.A. Webb

Features in the clinical history may help in gynecological problem in prepubertal girls caus-
ascertaining the likely cause of the mass. In the ing dysuria, should be noted. In male children, the
newborn period, the large majority of these state of the foreskin and the position of the ure-
lesions will have been identified antenatally, thral meatus should be recorded. Hypospadias is a
though additional clinical information may common problem with an incidence of up to 1:130
inform subsequent investigation and likely diag- live births; parents should be made aware that
nosis, i.e., the presence of oligohydramnios or circumcision is contraindicated where this is pre-
polyhydramnios. Beyond the newborn period, a sent. Eagle-Barrett syndrome is associated with
family history of severe hypertension may point to bilateral cryptorchidism. The presence of male
a diagnosis of PKD. Presentation with symptoms pseudohermaphroditism in association with pro-
and signs of urinary tract infection may point to teinuria and renal impairment in an infant is sug-
many of the anomalies associated with urinary gestive of Denys-Drash syndrome.
obstruction or stasis. Children on peritoneal dial-
ysis are at increased risk of developing inguinal Anus
and abdominal herniae. The presence of an imperforate anus in a neonate
should alert the physician towards other associ-
Hepatosplenomegaly ated anomalies such as sacral dysplasia, spinal
Liver enlargement in association with renal dis- dysraphism, and vesicoureteric reflux.
ease can be seen in a number of conditions includ-
ing glycogen storage disease type 1, tyrosinemia,
and Fanconi-Bickel syndrome. Splenomegaly in a Nervous System
child with hematuria might suggest infective
endocarditis. Autosomal recessive PKD may Whilst a detailed neurological examination is
cause hypersplenism as a consequence of hepatic often not required, it is important to record the
fibrosis. Hepatosplenomegaly in association with level of consciousness and mental status and
renal disease reflects a multisystem disorder, examine the cranial nerves and muscle tone,
which can be inflammatory such as systemic power, and reflexes along with the gait and coor-
lupus erythematosus or a neoplastic process, for dination. Hypertension can present with
instance, lymphoma presenting with AKI second- papilledema and is the commonest cause of bilat-
ary to uric acid nephropathy. eral Bell’s palsy. Seizures are associated with
electrolyte abnormalities, uremia, hypertensive
Urinary Bladder encephalopathy, vasculitis, and hemolytic uremic
An enlarged urinary bladder can be detected by syndrome. Blindness can result from rapid lower-
palpation and percussion and is characteristically ing of blood pressure in severe hypertension. Pos-
associated with suprapubic pain. Gross constipa- terior reversible encephalopathy syndrome is
tion and local genital inflammation are the com- known to occur in association with the use of
moner causes of acute urinary retention in immunosuppressive agents including methylpred-
children and rarely require urethral catheteriza- nisolone [34] and tacrolimus following renal
tion. Lower urinary tract obstruction due to uro- transplantation [35]. In a child with continence
logical causes such as posterior urethral valves, issues, careful examination of the peripheral sen-
pelvic tumors, and the neuropathic bladder should sory and motor functions together with the assess-
be considered an emergency, immediate decom- ment of the anal tone is essential.
pression of the urinary tract being indicated.

Genitalia Skin
Clinical evaluation of the renal patient is incom-
plete without examination of the genitalia. The Itching is a feature of severe uremia. SLE can
presence of vulvovaginitis, a common present with a characteristic fixed erythematosus
20 Clinical Evaluation of the Child with Suspected Renal Disease 607

malar rash. The presence of a palpable purpura phosphate, and hydroxyapatite in the conjunctiva
over the extensor surfaces and buttocks suggests [37]. Slit lamp examination may reveal character-
Henoch-Schönlein purpura. Multiple café au lait istic crystal deposits in the cornea in cystinosis. In
spots are seen in neurofibromatosis, which can a child with unexplained acute kidney injury,
give rise to hypertension secondary to renal artery examination of the eyes may clinch the diagnosis,
stenosis. Tuberous sclerosis is characterized by for example, in tubulointerstitial nephritis and
typical dermatological lesions including facial uveitis (TINU) syndrome. Uveitis is also a feature
adenofibromas (adenoma sebaceum), shagreen of systemic diseases such as juvenile rheumatoid
patches over the lower back, and hypopigmented arthritis. Aniridia is associated with an increased
ash leaf macules which may be present anywhere risk of Wilms tumor. Presence of anterior
in the body. Ichthyosis is a recognized feature of lenticonus, which is a conical protrusion in the
the arthrogryposis, renal Fanconi syndrome, and anterior aspect of the lens (seen almost exclu-
cholestasis (ARC) syndrome. Dystrophic nails sively in males) along with macular changes, is a
should raise the possibility of nail patella characteristic feature of Alport syndrome. Cata-
syndrome. racts are seen in Lowe syndrome and galacto-
semia but can also be a consequence of
corticosteroid therapy.
Face Not infrequently, a child with severe hyperten-
sion is referred by an optician with papilledema
Examination of the face can provide valuable and retinal hemorrhages and exudates. Diabetic
clues to the underlying clinical problem. Any retinopathy, which parallels nephropathy, is rare
obvious dysmorphic features such as Down or in childhood and correlates with the duration and
William syndrome should be noted. Long-term control of disease. Nephronophthisis is associated
ciclosporin use is associated with hypertrichosis, with several characteristic eye lesions: retinitis
particularly in the Asian population [36]. Hirsut- pigmentosa in Senior-Loken syndrome,
ism is a well-recognized feature of steroid toxicity tapetoretinal degeneration in juvenile
and Cushing syndrome. Nephrotic syndrome nephronophthisis, and oculomotor apraxia in chil-
often presents with early morning periorbital dren with Cogan syndrome. Retinitis pigmentosa
swelling and is frequently misdiagnosed as aller- is a feature of Bardet-Biedl syndrome.
gic rhinitis. Examination of the oral cavity is
important and may reveal gingival overgrowth as
a consequence of ciclosporin therapy. Asking the Ears
child to smile may reveal a facial expression
resembling crying in the child with Ochoa syn- Minor external ear malformations such as
drome, which is associated with the presence of a preauricular skin tags and pits are found in
neuropathic bladder. 0.5–1 % of newborns and do not warrant renal
evaluation unless accompanied by other systemic
malformations [38]. Branchial fistulae are a feature
Eyes of branchio-oto-renal syndrome. Alport syndrome
is characterized by the insidious onset of high-tone
The eyes are affected in a number of renal dis- sensorineural deafness. Sensorineural deafness is
eases, and therefore input from an experienced also a feature of type 4 Bartter syndrome which is
pediatric ophthalmologist is invaluable. the most severe phenotype presenting in the neo-
Coloboma of the eyelid is seen in Goldenhar natal period with life-threatening volume depletion
syndrome. Uncontrolled renal osteodystrophy and chronic kidney disease. Deafness can be a
can lead to scleral calcification. The presence of consequence of aminoglycoside toxicity and is
hypercalcemia may cause redness of the eyes sec- also seen following rapid administration of a large
ondary to the metastatic crystallization of calcium, dose of furosemide.
608 M.A. Shenoy and N.J.A. Webb

Musculoskeletal System half of children with sarcoidosis. Metabolic disor-


ders such as Lowe syndrome and Lesch-Nyhan
Muscles syndrome may present with arthropathy.
Muscle wasting is a feature of chronic uremia. Although hyperuricemia is often seen in children
Myopathy can also be secondary to rickets and with HNF1 beta mutation and pediatric transplant
renal osteodystrophy, which may improve follow- recipients, gouty arthritis is rare.
ing vitamin D therapy. One of the recognized side
effects of corticosteroid therapy is proximal
myopathy, demonstrated by eliciting Gower’s Examination of the Urine
maneuver. Up to half of children with mitochon-
drial disorders will have renal involvement with Macroscopic Examination
tubular dysfunction being the commonest
pathology. The usual yellow color of urine is due to the
presence of a number of pigments, some of
Skeletal System which are derived from food (i.e., riboflavin) and
Florid skeletal deformities of the weight-bearing others produced endogenously. The intensity of
limbs, such as genu varum or valgum, can be seen the coloration depends upon the urinary flow rate;
in infants with rickets. Rachitic rosary and swell- where large quantities of urine are produced, e.g.,
ing of the wrists and ankles due to epiphyseal following high fluid intake, the urine is paler in
swelling are other well-recognized features of color though darker in color at times of reduced
rickets. Slipped femoral capital epiphysis is a urine production.
feature of renal osteodystrophy, and avascular Where the urine is abnormally red or dark in
necrosis of the head of the femur may complicate color, dipstick testing and microscopy examina-
corticosteroid therapy, particularly following tion of the sample should be performed.
renal transplantation.
Whilst polydactyly is a recognized feature of
disorders such as Bardet-Biedl and Meckel- Dipstick Examination of Urine
Gruber syndrome, isolated polydactyly is usually
associated with a favorable outcome. Dipsticks
Hemihypertrophy and Beckwith-Wiedemann It is important that urine dipsticks are kept dry in
syndrome are associated with an increased risk their container with the lid on and that the manu-
of Wilms tumor. Spinal dysraphism should be facturer’s expiry date is adhered to. A number of
suspected in infants with a lower midline back automated devices are available. These ensure
lesion such as a subcutaneous mass, dermal vas- that the stick is read at the correct time, thus
cular malformation, hypertrichosis, a midline reducing inter-observer variability. Most will pro-
dimple or sinus tract, a skin tag, or an asymmetric duce a printout, which can be attached to the
gluteal cleft. Sacral agenesis is seen in infants of patient’s medical record.
insulin-dependent diabetic mothers but may also
be part of the familial Currarino triad syndrome Specific Gravity
(presacral mass, sacral agenesis, and anorectal Most modern urine dipsticks measure urinary spe-
malformation) [39]. cific gravity, though this information is rarely
utilized in clinical practice. Specific gravity is a
Joints measure of the mass of individual solutes present
Arthropathy is a characteristic feature of Henoch- per unit volume of urine, as opposed to osmolal-
Schönlein purpura and SLE. It is also seen in ity, which is the measure of the total particle
systemic onset juvenile idiopathic arthritis, concentrations, irrespective of the mass of the
which if poorly controlled can lead to renal amy- individual particles. Low values of specific grav-
loidosis. Arthralgia and arthritis affect almost one ity (less than 1.010) are found at times of maximal
20 Clinical Evaluation of the Child with Suspected Renal Disease 609

water excretion and high values (greater than Table 4 Causes of false-positive and false-negative pro-
1.025) at time of maximal urinary concentration. teinuria on dipstick testing
False-negative
pH False-positive proteinuria proteinuria
The pH of healthy urine varies between 4.5 and Concentrated urine Dilute urine
8.0. Fasting produces low values, and the highest Alkaline urine Acidic urine
Gross hematuria, pyuria,
pH measurements are seen following meals. pH
bacteriuria
values are low where acidemia is present, except Dipstick left in urine too long or
for where this is secondary to renal tubular delay in reading
acidosis. Contamination and drugs
Antiseptics: chlorhexidine,
Blood benzalkonium
Urine dipsticks detect the presence of hemoglobin
in the urine through its ability to catalyze a reac-
tion between hydrogen peroxide and o-tolidine. fever, or acute illness and is of no significance
Spotted positivity indicates intact red cells, with regard to long-term renal outcome.
whereas uniform positivity indicates free hemo-
globin (e.g., in intravascular hemolysis or red cell Glucose
lysis in the urinary tract). Glucose is a small molecule, which is freely fil-
There are a number of causes of false positiv- tered in the glomerulus. In health, the proximal
ity, including the presence of myoglobinuria, tubule reabsorbs all of the filtered glucose by an
oxidizing agents in the urine, and heavy bacterial active process, which has an upper rate limit or
contamination. The presence of reducing agents, transport maximum (TM). Certain normal individ-
such as ascorbic acid in the urine, may cause a uals have a lower TM for glucose and will have
false-negative result. These highlight the glycosuria at normal or only slightly increased
importance of confirmation of the presence of plasma glucose concentrations (so-called renal
red blood cells by microscopy of a fresh sample glycosuria). Glycosuria also occurs where overt
of urine. hyperglycemia is present, e.g., in diabetes mellitus
and in a number of tubular abnormalities, includ-
Protein ing Fanconi syndrome. The lower limit of detec-
Urine dipsticks undergo color change from yellow tion for glucose in the urine is 0.5 mmol/l.
to green following binding with proteins. Albu-
min is better detected than other urinary proteins Leukocytes
including globulins, tubular proteins, etc. There A number of routinely available urine testing
are a number of causes of false-negative and false- sticks will detect the presence of leukocyte ester-
positive results (Table 4). Dipstick analysis is not ase, indicating the presence of pyuria. Where test-
a good quantitative test because of the effect of ing is positive, urine microscopy should be used to
urinary concentration; more concentrated urine confirm this finding.
will show higher protein content, and where pro-
teinuria is detected, formal quantification with a Nitrites
urinary protein/creatinine or albumin/creatinine A number of routinely available urine testing
ratio is indicated. sticks will also detect the presence of nitrites.
It is well recognized that urinary protein excre- These are produced by the majority of pathogenic
tion increases throughout the daytime with bacteria, and their detection provides further
prolonged duration in the upright position, and screening data to assist in the diagnosis of urinary
first morning samples should be assessed to rule tract infection. The test has a high specificity but a
out any element of orthostatic proteinuria. Tran- low sensitivity for the diagnosis of UTI. As such,
sient proteinuria may occur following exertion, as a standalone test, it is of limited usefulness.
610 M.A. Shenoy and N.J.A. Webb

Where UTI is suspected or needs to be excluded, a of lower urinary tract origin from dysmorphic
urine culture is necessary to determine the bacte- RBCs of glomerular origin, which have been
riological cause and antibiotic sensitivities or to distorted during their passage through the filtra-
confidently rule out UTI. tion barrier. This is best performed using phase
contract microscopy, though possible with ordi-
Microscopy of Urine nary light microscopy. In its purest form, the dis-
There are many who advocate the routine bedside tinction is useful, though often a mixture of
microscopy of urine in all children who present abnormal and normal cells is present and interpre-
with suspected renal disease or urinary tract infec- tation is difficult. Acanthocytes are red blood cells
tion. This is not, however, widely performed for a with thorn-like projections of varying lengths dis-
variety of reasons including unfamiliarity with the tributed over the surface due to an increase in
technique and availability of appropriate micro- membrane cholesterol:lethicin ratio. Where
scopes in clinical areas. acanthocytes constitute greater than 5 % of the
urinary red cell population, this may indicate the
Casts presence of a glomerulonephritis.
Casts are produced by the aggregation of Tamm-
Horsfall protein with cells or cellular debris in the White Blood Cells
renal tubule. They are best seen in unspun urine as The presence of greater than 10 white cells/mm3
centrifugation may damage casts. Where urine has in a midstream sample of urine is considered
been centrifuged for other reasons, casts are most abnormal. Neutrophils are detected in urinary
frequently seen at the edge of the coverslip. tract infection but are also seen in contaminated
Hyaline casts are present in proteinuric states, urine samples, proliferative glomerulonephritis,
though may be found in concentrated specimens and interstitial nephritis. The presence of eosino-
of urine from normal individuals. These may phils in the urine is a sensitive and specific sign of
appear waxy if lipid droplets are present. acute interstitial nephritis.
Cellular casts contain cellular material, the
source of which may provide some clues regard- Bacteria and Other Organisms
ing the underlying pathology. Red blood cell casts Urinary bacteria may be clearly visible without
are always pathological and indicate glomerular Gram staining. Their detection may be enhanced
bleeding. White blood cell casts indicate renal by the use of phase contrast microscopy. Fungi, e.
inflammation secondary to pyelonephritis or g., Candida and Schistosoma species (a rare cause
immunologically mediated disease. Epithelial of hematuria), may also be detected.
cell casts (often present with red and white blood
cell casts) are produced from shed tubular epithe- Epithelial Cells
lial cells and may be seen during recovery from The presence of epithelial cells in the urine
acute tubular necrosis. may represent desquamation from the urinary
tract. Tubular cells may be seen following
Red Blood Cells tubular injury (acute tubular necrosis, acute trans-
The excretion of a small number of red cells is a plant rejection). Squamous cells are commonly
normal phenomenon, which increases with exfoliated from the urethra and are a normal
increasing age and following vigorous exercise. finding.
Children with febrile illness may develop a tran-
sient increase in red cell excretion. However, the Crystals
persistent presence of greater than 5 red cells/mm3 Normal urine contains crystals of calcium phos-
in uncentrifuged urine is abnormal. Urine micros- phate and oxalate. Other crystals may be cystine,
copy can distinguish anatomically normal RBCs uric acid, or dihydroxyadenine.
20 Clinical Evaluation of the Child with Suspected Renal Disease 611

References 18. Brady TM, Redwine KM, Flynn JT. Screening blood
pressure measurement in children: are we saving lives?
1. Garrett WJ, Grunwald G, Robinson DE. Prenatal diag- Pediatr Nephrol. 2014;29:947–50.
nosis of fetal polycystic kidney by ultrasound. Aust NZ 19. Jakobbson B, Esbjorner E, Hansson S. Minimum inci-
J Obst Gynaecol. 1970;10:7–9. dence and diagnostic rate of first urinary tract infection.
2. De Bruyn R, Marks SD. Postnatal investigation of fetal Pediatrics. 1999;104:222–6.
renal disease. Semin Fetal Neonatal Med. 20. Coulthard MG, Lambert H, Keir M. Occurrence of
2008;13:133–41. renal scars in children after their first referral for urinary
3. Helin I, Persson PH. Prenatal diagnosis of urinary tract tract infection. BMJ. 1997;315:918–9.
abnormalities by ultrasound. Pediatrics. 1986;78 21. Cooper WO, Hernandez-Diaz S, Arbogast PG,
(5):879–83. et al. Major congenital malformations after first-
4. Livera LN, Brookfield DS, Egginton JA, trimester exposure to ACE inhibitors. N Engl J Med.
et al. Antenatal ultrasonography to detect fetal renal 2006;354(23):2443–51.
abnormalities: a prospective screening programme. 22. Rasmussen SA, Chu SY, Kim SY, et al. Maternal obe-
BMJ. 1989;298(6685):1421–3. sity and risk of neural tube defects: a metaanalysis. Am
5. Smith NC, Hau C. A six year study of the antenatal J Obstet Gynecol. 2008;198(6):611–9.
detection of fetal abnormality in six Scottish health 23. Singh HP, Hurley RM, Myers TF. Neonatal hyperten-
boards. Br J Obstet Gynaecol. 1999;106:206–12. sion: incidence and risk factors. Am J Hypertens.
6. Morris RK, Malin GL, Quinlan-Jones E, 1992;5:51–5.
et al. Percutaneous vesicoamniotic shunting versus 24. Thummala MR, Raju TN, Langenberg P. Isolated sin-
conservative management for fetal lower urinary tract gle umbilical artery anomaly and the risk for congenital
obstruction (PLUTO): a randomised trial. Lancet. malformations: a meta-analysis. J Pediatr Surg.
2013;382(9903):1496–506. 1998;33:580–5.
7. DeCramer S, Parant O, Beaufils S, et al. Anomalies of 25. Darr A, Modell B. The frequency of consanguineous
the TCF2 gene are the main cause of fetal marriage among British Pakistanis. J Med Genet.
bilateral hyperechogenic kidneys. J Am Soc Nephrol. 1988;25:186–90.
2007;18(3):923–33. 26. Moghal NE, Milford DV, Hulton SA, Taylor CM. The
8. Meadow R. Munchausen syndrome by proxy. Arch Dis prevalence and treatment of end stage renal disease in
Child. 1982;57:92–8. an Asian child population. Nephrol Dial Transpl.
9. McGee H. On food and cooking: the science and lore of 1997;12(12):2517–20.
the kitchen. New York: Simon and Schuster; 1997. p. 194. 27. Connolly LP, Treves ST, Connolly SA,
10. White RH. Occurrence of S-methyl thioesters in urine et al. Vesicoureteric reflux in children:
of humans after they have eaten asparagus. Science. incidence and severity in siblings. J Urol.
1975;189:810–1. 1997;157(6):2287–90.
11. Waring RH, Mitchell SC, Fenwick GR. The chemical 28. Nash D. The development of micturition control with
nature of the urinary odour produced by man special reference to enuresis. Ann R Coll Surg Eng.
after asparagus ingestion. Xenobiotica. 1949;5:318–44.
1987;17:1363–71. 29. Flynn JT, Alderman MH. Characteristics of children
12. Sherry SN, Kramer I. The time of passage of the first with primary hypertension seen at a referral center.
urine by the newborn infant. J Pediatr. 1955;46:158. Pediatr Nephrol. 2005;20(7):961–6.
13. Muellner S. Development of urinary control in chil- 30. Rianthovorn P, Ettenger RB. Medication
dren. JAMA. 1960;172:1256–61. non-adherence in the adolescent renal transplant
14. Forsythe WI, Redmond A. Enuresis and spontaneous recipient: a clinician’s viewpoint. Pediatr Transplant.
cure rate. A study of 1129 enuretics. Arch Dis Child. 2005;9(3):398–407.
1974;49(4):259–63. 31. Foster BJ, Leonard MB. Nutrition in children with
15. Kitagawa T. Lessons learned from the Japanese kidney disease: pitfalls of popular assessment methods.
nephritis screening study. Pediat Nephrol. Perit Dial Int. 2005;25 Suppl 3:S143–6.
1988;2:256–63. 32. Gavrilovici C, Goldsmith DJ, Reid C. What is the role
16. National High Blood Pressure Education Program of ambulatory BP monitoring in pediatric nephrology?
Working Group on High Blood Pressure in Children J Nephrol. 2004;17(5):642–52.
and Adolescents. The fourth report on the diagnosis, 33. Merten DF, Kirks DR. Diagnostic imaging of paediat-
evaluation and treatment of high blood pressure in ric abdominal masses. Pediatr Clin North
children and adolescents. Pediatrics 2004;114(2 Suppl Am. 1985;32:1397–425.
4th Report):555–76. 34. Ikeda M, Ito S, Hataya H, et al. Reversible posterior
17. Sinha MD, Reid CJ. Evaluation of blood pressure in leukoencephalopathy in a patient with minimal change
children. Curr Opin Nephrol Hypertens. nephrotic syndrome. Am J Kidney Dis. 2001;37(4):
2007;16:577–84. E30.
612 M.A. Shenoy and N.J.A. Webb

35. Ishikura M, Ikeda M, Hamasaki Y, et al. Posterior 37. Berlyne GM, Shaw AB. Red eyes in renal failure.
reversible leukoencephalopathy syndrome in children: Lancet. 1967;i:4–7.
its high prevalence and more extensive imaging find- 38. Deshpande SA, Watson H. Renal ultrasonography not
ings. Am J Kidney Dis. 2006;48(2):231–8. required in babies with isolated minor ear anomalies.
36. Webb NJA, Coulthard MG, Trompeter RS, Arch Dis Child Fetal Neonatal Ed. 2006;91(1):F29–30.
et al. Correlation between finger-prick and venous 39. Currarino G, Coln D, Votteler G. Triad of anorectal,
ciclosporin levels: association with gingival over- sacral and presacral anomalies. AJR Am J Roentgenol.
growth and hypertrichosis. Pediatr Nephrol. 1981;137(2):395–8.
2007;22:2111–8.
Laboratory Investigation of the Child
with Suspected Renal Disease 21
George van der Watt, Fierdoz Omar, Anita Brink, and
Mignon McCulloch

Contents Calcium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 626


Magnesium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 626
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 614 Phosphate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 627
Glomerular Function and Injury . . . . . . . . . . . . . . . . . . 614 Glucose . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 628
Use of Endogenous Substrates in Clinical Urine Amino Acid Analysis . . . . . . . . . . . . . . . . . . . . . . . . . 628
Medicine for the Estimation of GFR . . . . . . . . . . . . . . . . 615 Potassium and Transtubular Gradient of
Use of Exogenous Substrates for Measurement Potassium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 628
of GFR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 618 Assessment of Renal Acid/Base Homeostasis . . . . . . 629
Urate, Oxalate, Citrate . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 630
Proteinuria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621 Renal Tubular Concentrating Ability . . . . . . . . . . . . . . . . 631
Measurement of Urinary Protein . . . . . . . . . . . . . . . . . . . . 622
Renal Biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 624 Parathyroid Hormone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 632

Urinalysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 624 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 633

Tubular Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 625


Fractional Excretion (FE) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 626
Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 626
Chloride . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 626

G.Watt (*) • F. Omar


Chemical Pathology, University of Cape Town and
National Health Laboratory Service, Red Cross Children’s
Hospital and Groote Schuur Hospital, Cape Town,
South Africa
e-mail: George.vanderwatt@uct.ac.za;
Fierdoz.omar@uct.ac.za
A. Brink
Department of Pediatrics and Child Health (Nuclear
Medicine), University of Cape Town, Red Cross War
Memorial Children’s Hospital, Cape Town, South Africa
e-mail: anita.brink@uct.ac.za
M. McCulloch
Department of Paediatric Intensive Care/Nephrology,
University of Cape Town, Red Cross War Memorial
Children’s Hospital, Cape Town, Western Cape,
South Africa
e-mail: mignon.mcculloch@uct.ac.za;
mignonmcculloch@yahoo.co.uk

# Springer-Verlag Berlin Heidelberg 2016 613


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_19
614 G. Watt et al.

Introduction secreted by the tubules, and not have inherent


toxicity or affect renal function. Thus, the sub-
The kidney can be injured by a variety of different stance is neither added to urine nor removed from
mechanisms. Investigating the type and assessing urine during its path through the tubule.
the degree of injury and its progression involves Compounds used to measure clearance are
laboratory assessment and often tissue sampling. either exogenous or endogenous. Exogenous
This chapter will discuss laboratory assessment and compounds are nonphysiological substances that
investigation with emphasis on the use of blood and are infused and measured in plasma and/or urine
urine samples to investigate renal function. over time to calculate GFR, while endogenous
There are a number of terms frequently used in compounds are produced by the body at a rela-
laboratory assessment that are now more precisely tively constant rate and maintain a stable plasma
defined and help us think about how we use labora- concentration at a specific GFR.
tory assessment. In general, laboratory studies are Creatinine clearance is arguably still consid-
used to help with diagnosis, prognosis, follow-up, ered the most convenient endogenous estimate
or effect of therapy. Usually, the specific laboratory of GFR despite the fact that it does not fulfill all
study performed fits the definition of a biomarker – a the criteria for an ideal substance, whereas inulin
characteristic that is measured and evaluated objec- clearance (a fructose polymer) is considered the
tively as an indicator of normal biologic or patho- “gold exogenous standard” as it fulfills all the
genic processes or pharmacologic responses to a ideal criteria. In clinical practice, however, less
therapeutic intervention. This chapter will discuss ideal radiolabeled agents such as technetium-
those measurements used primarily in patient care. 99m-labeled diethylenetriamine pentaacetic acid
(99mTc-DTPA), chromium-51-labeled ethylene-
diaminetetraacetic acid (51Cr-EDTA), and
Glomerular Function and Injury iodine-125-labeled iothalamate (125I-iothalamate)
clearances are usually employed due to the fact
The glomerular filtration rate (GFR) is considered that routine measurement is less onerous than
to be the most accurate measure of renal func- inulin. Briefly stated, 51Cr-EDTA and 99mTc-
tional capacity and reliably reflects the number DTPA clearances correlate well with inulin clear-
of functional nephrons. As a physiological mea- ance [2, 3], whereas 125I-iothalamate’s correlation
surement, it has high sensitivity and specificity for to inulin clearance is only fair [4]. In general,
99m
changes in renal function and by convention is Tc-DTPA and 51Cr-EDTA do not perform
expressed in mL/min/1.73 m2. The GFR is a well in neonates, 51Cr-EDTA appears to have an
dynamic measurement and is defined as the vol- extrarenal source of elimination, and 99mTc may
ume of plasma that can be cleared of an ideal dissociate from DTPA during clearance studies,
substance per unit of time, corrected to body sur- whereas iothalamate appears to undergo some
face area. Clearance of an ideal substance is there- tubular secretion and reabsorption [2]. Further-
fore considered to be equal to the GFR where more, poor correlation exists between techniques
clearance = [U]/[P]  V where [U] is the urine and the various radionuclide agents, and as such,
concentration of the substance, [P] the plasma it is recommended that technique-specific refer-
concentration, and [V] the urine flow rate in ence intervals are used [5].
mL/min. Body surface area is usually calculated Iohexol is increasingly being used as an alterna-
according to the formula: BSA (m2) = 0.024265 tive to inulin. Iohexol is a nonionic, nonradioactive,
 weight (kg)0.5378  height (cm)0.3964 [1]. X-ray contrast medium of low molecular weight
For a clearance measurement to be equal to the that is eliminated from plasma solely by glomerular
GFR, the substance to be measured should fulfill filtration [6]. It is easily measured by HPLC using
the so-called “ideal” criteria, namely, be present at very small sample volumes, increasing its useful-
a stable plasma concentration, be freely filterable ness in children, and clearance correlates well with
through the glomerulus, not be reabsorbed or inulin clearance [7, 8].
21 Laboratory Investigation of the Child with Suspected Renal Disease 615

Table 1 Plasma creatinine reference intervals (2.5–97.5th percentiles) [9]


Enzymatic creatinine Jaffe creatinine
Age group mg/dL umol/L Age group mg/dL umol/L
0–14 days 0.32–0.92 28–81 0–14 days 0.42–1.05 37–93
15 days to <2 years 0.1–0.36 9–32 15 days to <1 year 0.31–0.53 27–47
2 to <5 years 0.2–0.43 18–38 1 to <4 years 0.39–0.55 34–49
5 to <12 years 0.31–0.61 27–54 4 to <7 years 0.44–0.65 39–57
12 to <15 years 0.45–0.81 40–72 7 to <12 years 0.52–0.69 46–61
15–19 years male 0.62–1.08 55–95 12 to 15 years 0.57–0.80 50–71
15 to <19 years female 0.49–0.84 43.3–74 15 to <17 years male 0.65–1.04 57–92
15 to <17 years female 0.59–0.86 52–76
17 to <19 years male 0.69–1.10 61–97
17 to <19 years female 0.60–0.88 53–78

Use of Endogenous Substrates age group. At birth, the plasma creatinine value
in Clinical Medicine for the Estimation reflects maternal concentrations, giving rise to
of GFR higher creatinine values in the first days of extra-
uterine life. The GFR itself at term birth is only
Creatinine approximately 30 mL/min/1.73 m2 and lower in
Creatinine, a waste product, is the nitrogen- premature neonates but increases rapidly in the
containing cyclic anhydride of phosphocreatine, first 18 months of life to stabilize at normal adult
an intracellular component of the skeletal muscle, values of approximately 104 mL/min/1.73 m2
involved in the temporary storage and transfer of from where it remains relatively constant
high-energy phosphate moieties required for mus- (Fig. 1). Best available normative data for GFR
cle contraction (Table 1). Creatine itself is synthe- in premature neonates and children < 15 years of
sized in the liver, pancreas, and kidneys prior to age are presented in Tables 2 and 3 respectively.
transport to the muscle tissue where it is phos- As can be appreciated from the presented data,
phorylated and utilized in a cyclical manner. Syn- both plasma creatinine and GFR are subject to
thesis is however self-limiting to a certain extent, wide interindividual variation, and patients with
and production decreases at very high plasma relatively low muscle mass and significant renal
creatinine levels. Approximately 1–2 % per day dysfunction may still have a plasma creatinine
of total muscle creatine spontaneously converts to level within reference intervals, making it difficult
creatinine, giving rise to a stable production rate. to identify early stages of renal dysfunction with-
The stable plasma concentration of creatinine is out the hindsight of serial measurements or clear-
therefore a function of muscle mass relative to the ance studies. With the loss of nephrons, the GFR
body surface area and the GFR. Creatinine clear- falls, and plasma creatinine tends to increasingly
ance tends to overestimate the true GFR because overestimate the GFR [14, 15]. This is due, in
although primarily filtered at the glomerulus, cre- part, to the fact that the tubular secretion of creat-
atinine is also secreted into urine by tubular epi- inine contributes an increasing fraction of total
thelial cells and is also reabsorbed to a certain creatinine excreted, leading to a clearance that is
extent [10, 11]; these effects are more pronounced increasingly greater than actual GFR.
at high levels of plasma creatinine. Because of the Creatinine is routinely measured in plasma and
combination of effects of relative muscle mass urine using either the chemical Jaffe method or
and evolving GFR, the plasma concentration of enzymatically by means of either creatininase or
creatinine in childhood changes over time; fur- creatininase-/creatinase-based assays. A major
thermore, with the onset of puberty, creatinine limitation of the Jaffe reaction is the fact that the
levels are higher in males compared females, and reagent (picric acid) cross-reacts with protein,
sex-specific reference intervals are required in this glucose, ascorbic acid, ketones, and
616 G. Watt et al.

Fig. 1 GFR values derived 160


from 51Cr-EDTA clearance,
corrected for body surface
area in children under 140
2 years. The 10th, 25th,
50th, 75th, and 90th 120
percentiles are represented

GFR ml/min / 1.73 m2


[13]
100

80

60

40

20

0
0 0.5 1 1.5 2
age (years)

Table 2 Reference intervals for GFR (mL/min per 1.73 m2) in very preterm infants during the first month of life based on
creatinine clearance (3rd to 97th percentiles) [12]
Day 27 weeks GA 28 weeks GA 29 weeks GA 30 weeks GA 31 weeks GA
7 7.9–18.9 10.7–21.7 13.6–24.6 16.4–27.4 19.3–30.3
14 10.7–21.7 13.5–24.6 16.4–27.4 19.3–30.3 22.1–33.1
21 12.5–23.5 15.3–26.3 18.2–29.2 21–32 23.9–34.9
28 15.5–26.5 18.3–29.4 21.2–32.2 24–35 26.9–37.9

Table 3 Reference intervals for GFR (mL/min per aligned to those originally used to derive creati-
1.73 m2) in children <0.1 to >2 years of age based on nine clearance, GFR, and estimated GFR (eGFR)
51
Cr-EDTA clearance (mean 2 SD) [13]
51
reference intervals. To correct this discrepancy,
Age in years Cr-EDTA clearance (mL/min/1.73 m2) the Creatinine Standardization Program was initi-
0.1 34–70
ated by the National Kidney Disease Education
0.1–0.3 33.1–90.3
Program’s laboratory working group in collabora-
0.3–0.66 43.9–99.5
tion with the International Federation of Clinical
0.66–1 48–117.2
Chemistry and the European Federation of Clini-
1–1.5 55.9–127.1
1.5–2 58.3–130.7
cal Chemistry and Laboratory Medicine to reduce
2 63.6–145.2 interlaboratory variation in creatinine assay cali-
bration and to enable more accurate estimates of
glomerular filtration rate [16]. As of 2011, all
cephalosporins, among others, and although these major Jaffe and enzymatic creatinine assays
limitations are corrected for in modern assays by worldwide were standardized to a reference mate-
employing kinetic assays that measure the rate of rial, NIST SRM 967, traceable to an accurate
chromogen formation during a creatinine-specific isotope dilution mass spectrometric (IDMS)
window, assay manufacturers until recently still method. This standardization has also resulted in
calibrated their assays to overestimate creatinine the need for reformulation of reference intervals
by a factor of 20–30 % so as to yield results for GFR and creatinine, and eGFR equations
21 Laboratory Investigation of the Child with Suspected Renal Disease 617

Table 4 Recommended equations for estimating GFR (mL/min/1.73 m2) based on plasma creatinine values and
demographic characteristics
Age Recommended equation Mass units – mg/dL, height in cm SI units – creat in μmol/L, height in cm
<18 Bedside IDMS-traceable GFR = (0.413  height)/creat GFR = (36.20  height)/creat
years Schwartz eGFR for children
<18 years old [19]
18 IDMS-traceable MDRD GFR = 175  (creatinine)–1.154 GFR = 175  (creatinine /88.4)1.154
years Study Equation eGFR for  (age)0.203  (0.742 if  (age)0.203  (0.742 if female) 
Adults [17] female)  (1.212 if African (1.212 if African American)
American)
CKD-EPI (2009) (where GFR = 141  min(creat/κ, 1)α GFR = 141  min((creat/88.4)/κ, 1)α
κ = 0.7 for females and 0.9  max (creat/κ, 1)1.209  0.993  max (creat/κ, 1)1.209  0.993 Age
for males, α is 0.329 for Age
[1.018 if female] [1.159 if [1.018 if female] [1.159 if black]
females and 0.411 for black]
males, min indicates the
minimum of creat/κ or 1, and
max indicates the maximum
of creat/κ or 1) [18]

based on plasma creatinine. Currently, the best with extremes of relative muscle mass or creatine
performing eGFR equations based on plasma cre- intake (amputees, paraplegics, obese patients;
atinine values in adults are the IDMS-traceable muscle-wasting disease, neuromuscular disor-
Modification of Diet in Renal Disease (MDRD) ders; malnutrition or patients on a vegetarian or
Study equation and the Chronic Kidney Disease low-meat diet). In these patients, formal creatinine
Epidemiology Collaboration (CKD-EPI) equa- clearance with timed urine collection is still
tions [17, 18] and the Bedside IDMS-traceable recommended. Estimated GFR equations are sub-
Schwartz equation for children under the age of ject to higher variability at low plasma creatinine
18 years [19]; the latter equation was derived levels, limiting their reliability in identifying early
using an enzymatic creatinine method. It is impor- stages of renal dysfunction. Most laboratories
tant to realize that all eGFR equations derived only report numerical eGFR results below a
prior to the IDMS standardization of creatinine threshold of 60 mL/min/1.73 m2. Estimated GFR
such as the Cockroft-Gault and original Schwartz equations are however very useful in monitoring
formula are no longer valid and should not be used progression of renal dysfunction over time in an
under any circumstances with standardized creat- individual patient. The recommended creatinine-
inine methods as they will overestimate the GFR. based equations are presented in Table 4.
Despite standardization of creatinine measure- To effectively calculate creatine clearance,
ments internationally, evaluations have demon- urine collections must be accurately timed to
strated a small positive bias, most prominent in avoid erroneous results. Ideally, a 24-h (1,440
pediatric samples at low levels of creatinine min) urine collection is required to calculate
between Jaffe and enzymatic methods, and as urine flow rate in mL/min which may be difficult
such, it is recommended that only enzymatic cre- to achieve in small children. Under-collection will
atinine methods be used when estimating GFR in report a falsely decreased GFR and over-
children with the Bedside IDMS-traceable collection the opposite. To complete the clearance
Schwartz equation [20]. As a general rule, eGFR calculation, plasma creatinine should ideally be
equations are superior to creatinine clearance- measured simultaneously with the urine collection
derived GFRs in the majority of patients with using the same assay methodology, and as with
normal relative muscle mass but should not be eGFR calculations, clearance studies should not
used in patients with unstable creatinine levels be performed in patients with unstable creatinine
(acutely ill patients, acute renal failure) or those levels.
618 G. Watt et al.

Table 5 Equations for estimating GFR (mL/min/1.73 m2) based on plasma creatinine  urea and cystatin C values and
demographic characteristics
Mass units – creat in mg/dL, cys C
in mg/L, urea in mg/dL, height SI units – creat in μmol/L, cys C in mg/L,
Age Recommended equation in m urea in mmol/L, height in m
<18 IDMS-traceable Schwartz GFR = 39.1  (height/ GFR = 39.1  (height/(creat/
years eGFR-creat-cys for creat)0.516  (1.8/cystatin C) 0.294 88.4))0.516  (1.8/cys C) 0.294 
children <18 years old  (30/urea)0.169  (1.099)male  (30/(urea/0.357)0.169  (1.099)male 
[19] (height/1.4)0.188 (height/1.4)0.188
Adults CKD-EPI creatinine- GFR = 135  min(creat/κ, 1)α GFR = 135  min((creat/88.4)/κ, 1)α 
cystatin C equation (2012)  max (creat/κ, 1)0.601  min max ((creat/88.4)/κ, 1)0.601  min (cys
(where κ = 0.7 for (cys C/0.8, 1)0.375  max(cys C/0.8, 1)0.375  max(cys C/0.8, 1)0.711
females and 0.9 for males, C/0.8, 1)0.711  0.995 Age  0.995 age [0.969 if female] [1.08 if
α is 0.248 for females [0.969 if female] [1.08 if black]
and 0.207 for males, min black]
indicates the minimum of
creat/κ or 1, and max
indicates the maximum of
creat/κ or 1) [23]

Cystatin C reference material, ERM-DA471/IFCC across


Cystatin C can also be used as an endogenous assay platforms. As with creatinine, standardiza-
marker of renal function and to estimate GFR. tion has led to the need for reevaluation or
Cystatin C is a nonglycosylated low molecular reformulation of eGFR-creat-cys equations, and
weight protein of 13.3 kd produced in all cells current recommendations are that equations
with relative constancy. Plasma levels are less should only be implemented in those populations
significantly influenced by age, gender, body hab- from which they were developed [27].
itus, or composition compared to creatinine
[21]. Renal handling of cystatin C is by complete
filtration with near total reabsorption and catabo- Use of Exogenous Substrates
lism in the proximal tubule and little or no urinary for Measurement of GFR
excretion [22]. Reference intervals for plasma
cystatin C are presented in Table 5. Contrary to Inulin
earlier meta-analyses, the best available current This is the classic marker for measuring GFR. The
data indicates that plasma cystatin C-based test requires a constant infusion of inulin and
eGFR equations (eGFR-cys) do not more accu- urinary catheter placement, and it is not routinely
rately predict GFR than the creatinine-based equa- used in clinical practice [28]. Reference values for
tions (eGFR-creat) and that equations derived inulin clearance are demonstrated in Fig. 2.
from both analytes (eGFR-creat-cys) such as the
51
CKD-EPI creatinine-cystatin C 2012 are more Cr-EDTA
precise with less bias at the critical threshold of Availability of this radiopharmaceutical is not
60 mL/min/m2 [23–26]. Equations based on cre- universal, but 51Cr-EDTA is regarded as the best
atinine and cystatin C for both adult and pediatric radiopharmaceutical tracer for the measurement
populations are presented in Table 5. Presently, a of kidney function because it is eliminated exclu-
widespread implementation of eGFR-creat-cys sively by glomerular filtration ([19, 99]). The ref-
equations in favor of eGFR-creat is limited by erence single-injection plasma disappearance
the fact that cystatin C is far more expensive curve method consists of an injection of 51Cr-
than creatinine to assay, utilizing nephelometric EDTA and blood samples drawn at 2 and
or immunoturbidimetric methodologies, and has 4 h. There is no collection of urine. Measuring
only recently been standardized to an acceptable the decrease in activity in the blood allows one to
21 Laboratory Investigation of the Child with Suspected Renal Disease 619

Fig. 2 Inulin clearance as a 250

Inulin clearance, ml/min per 1.73 m2


function of age X  SD

200

150
120
100

50

0
Neonate 2–8 9–22 1–5.9 6–11.9 12–19 2–12 Adult Adult
days days months months months years

calculate the GFR [99]. It is recommended that the absolute differential renal function [33]. It should
two-sample method is used because improving however be noted that when combining imaging
the accuracy over the two-sample method would and GFR determination a larger dose of radiophar-
require at least 13 samples drawn between 2 and maceutical is administered. In addition, 99mTc-
4 h [29]. Correction factors are employed to cor- DTPA image quality is poor in patients with
rect for the initial fast clearance of activity from poor renal function and immature kidneys
the blood. The reproducibility of 51Cr EDTA GFR [33]. If there is reason to suspect a decrease in
measurement differs in patients with normal and renal function imaging with 99mTc MAG3 in com-
poor renal function. The day-to-day variation in bination with a separate GFR test, using a
measurement of GFR using this method is 4.1 % low-dose 99mTc-DTPA or 51Cr-EDTA is advised.
in patients with a GFR of greater than 30 mL/min
and 11.5 % in patients with a GFR less than The Two-Sample Method
30 mL/min [30]). This method is valid in patients The early exponential is neglected in this simpli-
down to a GFR of 15 mL/min/1.73 m2 [13, 19]. fied method.
The effective radiation dose that the patient Clearance is expressed as
receives from this investigation is approximately
D
0.011 mSv per examination [99]. This compares Cl ¼
favorably with the average radiation dose of A
where
0.1 mSv received by an anterior/posterior and
lateral chest X-ray [31].
Cl = clearance
D = dose
99m
Tc-DTPA A = the estimated area under the plasma curve
This radiopharmaceutical is readily available.
Variations in protein binding have been described The dose (D) is calculated from the following
as a problem in the past. This problem is rarely measurements:
encountered with the newer DTPA preparations
available commercially [32]. ðDm  RÞ  ADS  DS

When using the slope intercept method to per- AS
form this test, the same method is used as the one
described for 51Cr-EDTA [99]. This agent is rou-
tinely used as an imaging agent. This gives the Dm = dose measured before intravenous injec-
advantage of combining a renogram with the GFR tion: if the weighing technique is used, this is
determination and allows the clinician to calculate the weight of the dose.
620 G. Watt et al.

R = residual activity in the syringe after injec- blood leads to an overestimation of GFR, and a
tion: for the weighing technique, this is the correction factor should be employed. Currently,
weight of the empty syringe after injection. the correction factors by Chantler and Brøchner-
ADS = activity in 1 mL of the dilution of the Mortensen are routinely employed [99]. The
standard. Brøchner-Mortensen correction factor results in
DS = the dilution factor of the standard, for underestimation of renal clearance if the clearance
example, the drop is diluted in a 100 or values are higher than 130 mL/min [34]. The
500 mL flask of water. Chantler correction factor underestimates low
AS = the activity of the standard: if the weighing clearance values [99].
technique is used, this is the weight of the The Chantler method:
aliquot used to make the standard.
GFR ¼ 0:87  Cl1 in ml=min=1:73m2
The estimated area under the curve (A), is
obtained from the following equation: The Brøchner-Mortensen method:

y0 GFR ¼ 1:01  Cl1  0:0017



b
 Cl21 in ml=min=1:73m2
where
Newer correction factors by Fleming and
Y0 is the intercept of the exponential of the late Brøchner-Mortensen and Jødal employ correction
plasma sample (2 and 4 h) at time 0 and b is the factors for body surface areas [35].
slope of the exponential. Fleming’s method:

Using only the plasma sample activities and GFR ¼ ððCl1 Þ=ð1 þ FÞÞ  ðCl1 Þ in ml=min=1:73m2
plasma times, the equation can then be expressed Where F ¼ 0:00170 min=ml
as
Brøchner-Mortensen and Jødal:
 
p
D  In 1 GFR ¼ ððCl1 Þ=ð1 þ FBSA ÞÞ  ðCl1 Þ in ml=min=1:73m2
p2
Cl ¼ exp ððt1  lnp2 Þ  ðt2  lnp1 ÞÞ=ðt2  t1 Þ Where FBSA ¼ 0:00185:BSA0:3
ðt 2  t 1 Þ

where These newer correction factors do not have the


problems of overestimations seen with the
D = administered activity in counts per minute Chantler method and underestimation of the
P1 = plasma activity in counts per minute/mL at Brøchner-Mortensen method [34, 35].
time 1
P2 = plasma activity in counts per minute/mL at The Distribution Volume Method or
time 2 One-Sample Method
This method has been validated for both
51
The clearance calculated from this equation is Cr-EDTA and 99mTc-DTPA. In this method, a
then corrected for body surface area. single blood sample is drawn at 110 to 130 min
Clearance corrected for body surface area after the injection of the tracer. The volume
(BSA) = Cl1 of distribution in this method is calculated and
Cl1 = Cl  1.73/the patient’s body surface not measured [36]. It is important to note
area in mL/min/1.73 m2 that this method is not accurate in patients with
The omission of early blood samples used to clearance values below 30–35 mL/min/1.73 m2
measure the initial fast clearance of activity from [19, 37].
21 Laboratory Investigation of the Child with Suspected Renal Disease 621

When using the one-sample method, the GFR S1 = mean iothalamate concentration of the
is calculated as follows: three serum samples also in mg/mL
Infusate = the mL/min of the infusion
Cl ¼ ð2:602  V120Þ  0:273 This result is then corrected for surface
V120 is the “virtual distribution of volume,” area [38].
where the dose injected is divided by the
120-min plasma concentration. It is rare that a
plasma sample is taken at exactly 120 min after Iohexol
injection and a correction factor for the blood This nonionic low osmolar contrast agent is rou-
sample time is employed. This correction factor tinely used in high doses for intravenous-
is not valid if the blood sample is taken outside the contrasted radiological examinations. Low doses
allowed time window of 110–130 min: of iohexol are used for the measurement of GFR.
Iohexol is excreted completely unmetabolized in
P120 ¼ Pt  e0:008ðt120Þ the urine; is not reabsorbed, metabolized, or
secreted by the kidney; and has a low
The final result is then corrected for body surface toxicity [19].
area [36, 99]. The HPLC method is used to assay the sera for
the iohexol concentration. Different methods have
125
I Iothalamate been described with a four-sample method being
At the time of this publication, no standardized used in routine clinical practice with success. The
guideline for the determination of GFR using 125I dose is given over 1 min. Blood samples are then
iothalamate in children is available. Protocols vary taken at approximately 5, 120, 180, and 240 min
from single injection and subcutaneous injection to after dosing with the times of blood samples accu-
constant infusion techniques, with or without the rately recorded. The 5-min sample is taken to
addition of urinary clearance measurements [28]. check that the dose has been administered intra-
venously and not, inadvertently, subcutaneously.
Nonradioactive Iothalamate The GFR is calculated using a single compart-
Nonradioactive iothalamate can be measured using mental model based on the iohexol clearance
high-performance liquid chromatography. A prim- rate between 120 and 240 min. Results are then
ing dose of iothalamate is given to achieve a corrected using the formula proposed by
plasma serum level of 1 mg/dL. The prime is Brøchner-Mortensen. The GFR is then corrected
followed by a constant infusion to maintain a con- for body surface area.
sistent plasma level. Due to differences in volumes A two-sample method was also recently
of distribution, children less than three years of age described by Ng et al.; this method employs a
are infused over 8 h, and children older than three correction factor similar to the newer correction
years are infused over 6 h. Blood samples are then factors by Fleming and Brøchner-Mortensen and
drawn at 120 min before, 60 min before, and at the Jødal [39]. In terms of clinical application, the
end of the infusion. If the final three samples varied two-sample method of iohexol with low toxicity
by less than 10 %, the patients have achieved and only two blood samples required is gaining
steady state, and the calculation can then be popularity.
performed using the following equation:

I1
Iocl ¼  infusate
S1
Proteinuria
where
Iocl = iothalamate clearance in mL/min Normal values for urinary total protein excretion
I1 = concentration of the iothalamate infusion are <240 mg/m2/day in children <6 months of
in mg/mL age and <150 mg/m2/day in older children,
622 G. Watt et al.

whereas excretion >3 g/1.73 m2/day is classified immunoglobulin light-chain production in the
as nephrotic range proteinuria in all age groups plasma cell dyscrasias (multiple myeloma).
[40, 41]. Approximately half of normal urine pro-
tein is secreted by the tubular epithelium in the
form of Tamm-Horsfall protein or uromodulin, a Measurement of Urinary Protein
glycoprotein. The balance is made up of albumin,
accounting for approximately 40 % of the total Two screening tests are initially used to quantita-
and other low molecular weight plasma proteins, tively assess urine protein excretion, namely, uri-
such as beta-2-microglobulin. The normal glo- nary dipstick analysis and the urine total protein
merulus retards the passage of proteins into the assay using a colorimetric automated assay such
Bowman space by charge-repelling negatively as sulfosalicylic acid. It is important to realize that
charged proteins such as albumin and by size extremes of urine dilution and concentration can
exclusion of larger proteins. Freely filtered low significantly affect protein concentration in urine
molecular weight (LMW) proteins less than and a dilute urine may underestimate the degree of
25,000 Da are normally reabsorbed by the proxi- pathological proteinuria, whereas highly concen-
mal tubule through endocytosis at the luminal trated but normal urine may have a protein con-
membrane. Excessive proteinuria can therefore centration of up to 1 g/L. For accurate assessment,
be classified as: protein should be measured quantitatively and
expressed as a protein/creatinine ratio or based
• Glomerular proteinuria – due to increased fil- on a 24-h urine collection and expressed as
tration of protein across the glomerular mem- mg/m2/day. Normal values as a function of age
brane. This is the most common cause of are presented in Table 6.
proteinuria in children and is seen with glomer- Urinary dipstick estimates albumin concentra-
ular disease and other non-pathologic condi- tion by a specific colorimetric reaction with
tions such as orthostatic proteinuria, fever, and tetrabromophenol blue, yielding a green color.
excessive exercise. Albumin is the major urine For this reason, it is unsuitable as a screening
protein present, but as disease severity pro- test for detecting tubular or overflow proteinuria.
gresses and glomerular selectivity decreases, False-positive results can occur with highly alka-
larger proteins such as immunoglobulins line urine or in urine contaminated with antiseptic
appear in urine. Orthostatic proteinuria is agents such as benzalkonium chloride and chlor-
quite common during adolescence, affecting hexidine and/or containing iodinated contrast
2–5 % of the adolescent population [42]. media [43]. Typical dipstick results are expressed
• Tubular proteinuria – due to reduced semiquantitatively as follows:
reabsorption of freely filtered low molecular
weight proteins such as alpha-1- • Trace: – <0.3 g/L
microglobulin, beta-2-microglobulin, and • 1+:0.3 to <1 g/L
retinol-binding protein. This form of protein-
uria is seen in a variety of tubulointerstitial
diseases and is often associated with other Table 6 Normal values of protein excretion as a function
of age
defects of proximal tubular function such as
glycosuria, aminoaciduria, phosphaturia, and Age g/g creatinine g/mol creatinine
proximal renal tubular acidosis, known as the 0–6 months 0.70 80
Fanconi syndrome. 6–12 months 0.55 60
• Overflow proteinuria – due to increased levels 1–2 years 0.40 45
of low molecular weight proteins in the plasma 2–3 years 0.30 30
3–5 years 0.20 20
that overwhelm tubular reabsorptive capacity.
5–7 years 0.15 19
This form of proteinuria is rarely seen in chil-
7–17 years 0.15 18
dren and usually associated with
21 Laboratory Investigation of the Child with Suspected Renal Disease 623

• 2+:1 to 3 g/L protein loss becomes nonselective. Highly


• 3+: 3 to <20 g/L selective proteinuria will have an IgG/albumin
• 4+: >20 g/L ratio of <0.1.
Tubular proteinuria can be differentiated from
Routine laboratory colorimetric total protein glomerular proteinuria by the selective
assays however detect all proteins present in immunonephelometric measurement of specific
urine and can be used to assess all forms of low molecular weight proteins such as beta-2-
proteinuria. microglobulin, alpha-1-microglobulin, lysozyme,
Albumin is accurately quantified in urine using and retinol-binding protein. In tubular proteinuria,
albumin-specific immunoassays with high sensi- these levels are typically increased 10- to 100-fold
tivities that enable the recognition of subtle above normal. A relatively simple and
glomerular injury such as that associated with underutilized method to asses and differentiate
early diabetic nephropathy and hypertensive variable degrees of glomerular from tubular pro-
renal disease. Although variable and influenced teinuria is by analysis of urinary protein by gel or
by age, albuminuria at a level of <30 mg/g capillary electrophoresis, where the ratios of high
Cr (<3 mg/mmol) is considered normal, whereas to low molecular weight plasma proteins present
levels between 30 and 300 mg/g Cr (3–30 in urine are visualized. This test is performed by
mg/mmol) are considered moderately increased most laboratories, usually for the diagnosis and
and levels >300 mg/g Cr (>30 mg/mmol) are monitoring of myeloma.
considered severely increased. Albuminuria can While normal urine contains very little protein,
be used in this way along with GFR to screen for a tubular proteinuria pattern consists primarily of
and classify renal disease severity, although LMW proteins, the hallmark being an alpha-2-
current data suggest that total protein should globulin doublet consisting of alpha-2-
preferentially used in children despite the fact microglobulins, with some albumin. In more
that acceptable guidelines and cutoffs have not severe tubular proteinuria, beta-2-microglobulin
been formulated yet [44]. In addition to the is also present, just cathodal to the normal location
above, albumin is also measured quantitatively of transferrin. In contrast, urine with selective and
together with a larger protein such as IgG as a nonselective glomerular proteinuria demonstrates
measure of glomerular selectivity in glomerular larger amounts of albumin along with varying
disease. Whereas loss of anionic charge alone degrees of higher molecular weight proteins,
results in increased albumin excretion (selective resembling the protein pattern present in serum;
proteinuria), as glomerular injury worsens, see Fig. 3 [98].

Fig. 3 Urine protein URINE AGAROSE


electrophoresis on agarose PROTEIN ELECTROPHORESIS +
gel: lanes 1 and 2 – tubular
proteinuria, showing Albumin
albumin band and alpha-2-
globulin doublet; lane 3 –
selective glomerular
proteinuria with albumin
and transferrin and some α-2 doublet
gamma globulins; lanes Transferrin
4 and 5 – moderate and
severe nonselective Immunoglobulins
glomerular proteinuria; lane
6 - normal serum sample

1 2 3 4 5 6 -
624 G. Watt et al.

Renal Biomarkers patients at risk of AKI. In all likelihood, a panel


of biomarkers will be developed and used in a
Recent efforts have been made over the last few similar manner to a urine dipstick [48].
years to identify novel urinary biomarkers to iden- Patients with chronic kidney disease (CKD)
tify early acute kidney injury (AKI) allowing early have traditional risk factors such as hypertension,
identification and management before AKI is hypercholesterolemia, and obesity as well as
established. A need for precise assessment is nontraditional risks such as inflammation, ane-
important as there is an independent association mia, and bone and mineral abnormalities. Fibro-
between AKI and mortality rate of critically ill blast growth factor-23 (FGF-23) has recently
patients of up to 60 %. emerged as powerful predictor of adverse out-
Until now, reliance has been placed on reduced comes in patients with CKD. Elevated FGF-23
urine output as well as rising serum creatinine to exerts its negative impact through mechanisms
make the diagnosis of AKI [45]. This is a concern, of action independent of role as regulator of phos-
as serum creatinine changes can occur without phorus hemostasis [49]. These FGF-23 levels can
kidney damage and similarly, functional changes be measured in the laboratory setting.
only occur after significant damage has occurred.
An ideal biomarker should be accurate, reliable,
and easy to measure, which makes urine the ideal
fluid. Biomarkers for AKI that have been Urinalysis
suggested include: neutrophil gelatinase-
associated lipocalin (NGAL), interleukin 18, kid- Likely the most frequently requested but least under-
ney injury molecule-1, and liver fatty acid- stood laboratory test for examining renal function or
binding protein [46]. The most promising of injury to the kidney or urinary tract, the urinalysis, is
these markers is NGAL, a protein that mediates easy to perform and is used as a screening test, a
innate immunity by inhibiting bacterial growth diagnostic test, and as a follow-up examination to
through iron sequestration. It is expressed princi- test the efficacy of therapy. Its utility is at times
pally in neutrophils but also in renal tubular cells overestimated, and the risk of either false positives
and increases in urine and plasma within hours or false negatives should be understood [50].
after AKI. Due to its low molecular mass and Typical urinalysis usually characterizes three
protease resistance, it is stable and easily mea- aspects of urine:
sured in urine [47]. Plasma and urinary NGAL
elevations are able to detect AKI far sooner than • Physical character: color, clarity, specific grav-
plasma creatinine, with higher sensitivity and ity, and/or osmolality
specificity and have also been shown to have • Chemical character: pH, glucose, protein,
good prognostic value in predicting outcome in blood, ketones, urobilinogen, conjugated bili-
many clinical scenarios associated with AKI. Fur- rubin, leukocyte esterases, and nitrites
ther clinical trials on outcomes and cost/benefits • Microscopic character: red cells (RBC), white
comparing the use of NGAL and other novel bio- cells (WBC), tubular casts, organisms, and
markers to standard clinical practices are however crystals
still required to demonstrate the clinical utility of
these biomarkers. Development of specific refer- Normal urine should be clear. Urine may be
ence intervals, action cutoffs, and intervention turbid or cloudy due to the presence of crystals
protocols are also required before NGAL can be (e.g., calcium phosphate in alkaline urine) or cel-
implemented into widespread clinical lular components – WBC, RBC, or both. Urine
practice [46]. may also have unusual color or odors; examples
Ultimately, there is a search for the renal equiv- include a grey or black discoloration in urine that
alent of troponin T or I that would allow early has been alkalinized or exposed to air in patients
recognition and therapeutic intervention in with alkaptonuria, development of a wine red
21 Laboratory Investigation of the Child with Suspected Renal Disease 625

color in urine exposed to air and light in Hematuria may be due to renal trauma, glo-
patients with some porphyrias, a distinct pleasant merular injury, interstitial or tubular injury, stone
caramelized sugar odor with maple syrup urine disease, cystic disease, malignancy, or trauma to
disease, and a noxious cheesy odor in patients the urinary tract. Red cells can appear in urine
with glutaric aciduria type 2 or isovaleric even though they derive from outside the urinary
acidemia. tract. Two examples include menses or excoria-
The most commonly used method for some tion of the epidermis at the urethral meatus. Nor-
physical, chemical, and even cellular elements mal urine has few, if any, intact red cells, generally
present in urine is a dipstick test which employs <2 red blood cell/high-powered field on micros-
a colorimetric reaction to identify abnormalities. copy on a urine specimen, which has been
These impregnated sticks should be used appro- centrifuged. This small number will not result in a
priately. Certain rules pertain: positive result on dipstick. At the other extreme,
urine will appear red with red cells, hemoglobin,
• Urine tested should be around body or even or myoglobin in urine. Because hematuria is a
room temperature. Refrigeration will affect the nonspecific finding, the finding usually indicates
test results. some injury but usually does not provide more
• The observations of change should be made information. There are two exceptions. First, dys-
after 15 s and up to 1 min after dipping the morphic red cells seen using phase contrast micros-
reagent stick in urine. Too rapid or too late, an copy on microscopic examination of the urine may
observation of color change can give a false help determine the site of injury along the urinary
reading. tract. If 75 % or greater of red cells seen are dys-
• Moisture, sunlight, cold, heat, or time (strips morphic, the kidney, and most likely the glomeru-
have expiration dates) will result in false lus, is the site of injury. If <25 % of RBC are
readings. dysmorphic, the injury site is the urinary tract –
• A number of substances present in urine may renal pelvis or below [53]. Second, if a dipstick is
yield false-positive or false-negative results on positive for RBCs but no RBCs are noted on
the dipstick test; examples include: microscopy, then hemoglobinuria or myoglobinuria
– False-positive protein readings with high should be suspected, and direct measurement of
levels of ascorbic acid, iodinated contrast hemoglobin or myoglobin should be undertaken.
agents, some antiseptic agents, and highly
alkaline urine.
– False-negative glucose readings with high Tubular Function
levels of ascorbic acid.
– False-positive urobilinogen readings in The renal tubules are responsible for modification
patients with acute porphyrias and elevated of the glomerular ultrafiltrate by reabsorption and,
urinary porphobilinogen. to a lesser extent, secretion of solutes and water in
– False-positive hemoglobin reading can be order to maintain an optimal internal milieu, in the
obtained with myoglobinuria. process, producing a concentrated urine. When
assessing tubular function, therefore, the
When leukocytes, especially neutrophils, are reabsorptive and/or secretory capacity, as well as
present, leukocyte esterase may be positive. tubular ability to concentrate urine, is assessed.
Nitrite is found in urine as a result of metabolic Commonly measured urinary solutes for the
release by bacteria. Unfortunately, neither is sen- assessment of reabsorptive (particularly proximal
sitive or specific enough to be able to definitively tubular) function are sodium, phosphate, glucose,
diagnose urinary tract infections even when used bicarbonate, calcium, and amino acids. Further
together [51]. Routine urinalysis as a screening testing involves assessing distal tubular capacity
tool in the general population remains for secreting potassium and hydrogen ions and for
controversial [52]. water reabsorption or excretion [54, 55].
626 G. Watt et al.

Fractional Excretion (FE) diet and extracellular volume status. With extra-
cellular volume contraction, enhanced sodium
Reabsorptive capacity can be assessed by calcu- reabsorption results in enhanced chloride
lating the FE of the solute. This refers to the reabsorption. During metabolic alkalosis, the uri-
fraction of filtered solute that is eventually nary chloride concentration can be useful diag-
excreted in urine, normalized to the relative excre- nostically. A urinary chloride concentration >10
tion of creatinine. The FE can be calculated as mmol/L is consistent with diuretic-induced renal
follows, where U represents concentration in loss, whereas urine chloride concentration under
urine, P concentration in plasma, and X the solute 10 mmol/L is consistent with gastric loss and
of interest,  100 to obtain the result as a volume contraction.
percentage:

UX  PCreatinine
FEX ¼  100 Calcium
PX  UCreatinine
Urine calcium measurement is used in the inves-
This calculation is most accurate when performed
tigation of hyper- and hypocalcemia,
on a 24-h urine collection, with a blood sample
normocalcemic hypercalciuria, parathyroid func-
taken during this collection.
tion, and renal calcium-sensing receptor function.
In children, hypercalciuria is defined as calciuria
>4 mg/kg/day (0.1mmol/kg/day) based on a 24-h
Sodium
urine sample collection [58]. The urine calcium/
creatinine ratio is commonly used to screen for
The FE of sodium (FE-Na) is often used to differ-
hypercalciuria defined as >0.2 mg/mg (0.6 mmol/
entiate prerenal azotemia, due to decreased renal
mmol) [59]. A fasting random calcium/creatinine
perfusion, from intrinsic renal failure. As sodium is
ratio on the second morning voided urine sample
almost completely reabsorbed in the functional but
correlates best with a 24-h urine calcium measure-
underperfused kidney, an FE-Na <1 % implies
ment [60]. In infants, values vary with age: 0–6
appropriate tubular reabsorption and therefore
months <0.8 mg/mg (2.3 mmol/mmol), 6–12
prerenal disease. It is not valid in patients receiving
months <0.6 mg/mg (1.7 mmol/mmol), and 1–2
diuretics where it can be falsely elevated. It may
years <0.5 mg/mg (1.4 mmol/mmol) [61]. In chil-
also be falsely depressed and therefore inaccurate
dren with decreased muscle mass, and therefore
with certain intrinsic causes of kidney injury such
decreased creatinine excretion, the calcium/osmo-
as acute glomerulonephritis, acute interstitial
lality ratio is a comparable alternative [62]. The
nephritis, radiocontrast-induced nephropathy, and
FE of calcium, based on a 24-h urine collection
rhabdomyolysis with myoglobinuria and in
and measurement of calcium and creatinine con-
patients with chronic low-flow states such as con-
centrations in plasma and urine, is another alter-
gestive cardiac failure. The FE of urea may be a
native and is particularly useful in assessing
better discriminator in these patients, with an
calcium-sensing receptor function or parathyroid
FE-urea <35 % supporting the diagnosis of
hormone function.
prerenal failure and >50 % suggestive of an intrin-
sic renal problem although there are differing opin-
ions on these cutoffs [55–57].
Magnesium

Chloride Magnesium in the ultrafiltrate is reabsorbed pri-


marily in the loop of Henle. The FE of magnesium
Urinary chloride excretion is tied to urinary (FE-Mg) is typically performed to determine ade-
sodium excretion. Urinary chloride depends on quacy of renal handling of magnesium. Normally,
21 Laboratory Investigation of the Child with Suspected Renal Disease 627

Fig. 4 Determining the


Tmp/GFR: a straight line
drawn through the plasma
phosphate concentration
and the TRP will pass
through the TmP/GFR [65]

approximately 5 % of filtered magnesium is reabsorption threshold of phosphate per glomeru-


excreted. With hypomagnesemia, the FE-Mg can lar filtration rate (TmP/GFR). The TmP/GFR rep-
fall to 1 %. In the presence of hypomagnesemia, resents the serum phosphate concentration at
an FE-Mg 5 % suggests renal wasting of mag- which phosphate begins to appear in urine [65]
nesium [63, 64]. and can either be calculated or determined from a
nomogram (Fig. 4), using the TRP and the serum
phosphate concentration. This calculation
Phosphate requires fasting urine and plasma phosphate and
creatinine measurements. Results should be com-
Renal phosphate handling may be abnormal even pared with age-appropriate reference intervals. A
in the presence of a normal serum phosphate level. typical reference interval in children aged 2–15
The tubular phosphate reabsorptive capacity is years is 3.6–7.6 mg/dL (1.15–2.44 mmol/L) [66].
therefore more effectively assessed by calculating Decreased TmP/GFR is associated with vitamin D
the tubular reabsorption of phosphate (TRP) deficiency and hyperparathyroidism, while
which can be considered the converse of the FE increased TmP/GFR is seen with hypo- and
and is calculated as follows: pseudohypoparathyroidism:

TRP ¼ 1  FEphosphate TmP=GFR ¼ TRPserumphosphate ðmg=dL or mmol=LÞ

The best indicator of renal tubular handling of It can be calculated directly using the following
phosphate, however, is the tubular maximum equation:
628 G. Watt et al.

TmP=GFR ðin mg=dL or mmol=LÞ defect, with increased glutamate and aspartate
excretion), Hartnup’s disease (neutral amino acid
Pphosphate  Uphosphate  Pcreatinine
¼ transporter defect leading to increased excretion
Ucreatinine
of the neutral amino acids), lysinuric protein intol-
erance (cationic amino acid transporter leading to
increased excretion of lysine, ornithine, and argi-
Glucose
nine), and iminoglycinuria (increased proline,
hydroxyproline, and glycine excretion) [68, 70].
Glucose is reabsorbed by the proximal tubule. The
Urine collection should preferably be accom-
renal threshold for reabsorption is a plasma con-
panied by plasma analysis for comparison. Urine
centration of 9.9 (180 mg/dL) mmol/L, implying
and plasma samples must be frozen immediately
that at normal plasma glucose concentrations,
after collection to maintain analyte integrity.
glycosuria is absent. When the plasma glucose
Urine amino acid concentrations are reported as
concentration exceeds this threshold as in diabetes
a ratio to creatinine concentration to accommo-
mellitus, the proximal tubule’s capacity to reab-
date for varying degrees of dilution.
sorb glucose is exceeded and glycosuria ensues.
Familial renal glycosuria is a rare benign condi-
tion due to a defective renal glucose transport and
Potassium and Transtubular Gradient
is associated with isolated glycosuria and normal
of Potassium
plasma glucose concentration [67]. Here, the renal
threshold is lowered such that glycosuria occurs at
In contrast to the previous analytes discussed,
a normal plasma glucose concentration. Glycos-
urine potassium concentration is determined by
uria may also occur with other tubular disorders
renal secretion in the distal tubule, which, in
such as in the Fanconi syndrome, cystinosis, and
turn, is associated with sodium uptake, via miner-
hereditary tyrosinemia. In these cases, it is usually
alocorticoid action. Urine potassium however is
associated with other tubular losses of phosphate,
not an accurate indication of adequate potassium
uric acid, amino acids, and bicarbonate [68, 69].
handling as it does not take into account the effect
of water movement across the distal tubule. Renal
handling of potassium is therefore assessed by
Urine Amino Acid Analysis calculating the transtubular potassium gradient
(TTKG) which uses the serum/urine osmolality
Amino acids are almost entirely reabsorbed in the ratio to adjust for the concentrating effects occur-
proximal tubule. As with glucose and phosphate, ring in the medullary collecting duct, thereby esti-
there is a maximum capacity of reabsorption for mating the potassium concentration at the point in
each amino acid. Any condition leading to the collecting duct where tubular fluid is isotonic
increased plasma concentrations, such that this with that of plasma [71, 72]:
maximum capacity is exceeded, will lead to
increased excretion in the urine. Generalized ami- UKþ  Posmolality
TTKG ¼
noaciduria, in the presence of normal plasma con- PKþ  U osmolality
centrations, indicates proximal tubular damage or
dysfunction (e.g., the Fanconi syndrome, A high TTKG is suggestive of renal potassium
cystinosis, and Lowe syndrome), while defects wasting, which would be appropriate in the pres-
of individual amino acid transporters are associ- ence of hyperkalemia but inappropriate in hypo-
ated with excretion of specific amino acids or kalemia. Causes of a high TTKG in the setting of
groups of amino acids. These include cystinuria hypokalemia include increased mineralocorticoid
(dibasic transporter defect with elevated cystine, activity due to hyperaldosteronism or pseudohy-
arginine, ornithine, and lysine excretion), dicar- peraldosteronism (Cushing syndrome or Liddle
boxylic aciduria (dicarboxylic acid transporter syndrome). A low TTKG suggests impaired distal
21 Laboratory Investigation of the Child with Suspected Renal Disease 629

secretion of potassium which may occur in min- Urine pH should be measured immediately after
eralocorticoid deficiency such as type IV renal collection or kept refrigerated to prevent bacterial
tubular acidosis. In adults, a TTKG >5 means action which may alter the pH (typically
aldosterone is active, while <3 suggests limited increases, but may decrease in the presence of
or no mineralocorticoid activity. In children with glucose) [74].
hyperkalemia, a TTKG <4.1 (or 4.9 in infants)
suggests decreased mineralocorticoid activity. Fractional Excretion of Bicarbonate
With hypokalemia, a TTKG >2 is indicative of This test requires the patient’s serum bicarbonate
ongoing aldosterone secretion. It is important to (HCO3) concentration to be restored to normal
note that the TTKG can only be used in the levels via intravenous administration prior to mea-
presence of sufficient distal delivery of sodium suring HCO3 in urine and plasma along with
(urine sodium >25 mmol/L) so that sodium creatinine to calculate the FE-HCO3. An
delivery is not rate limiting for potassium secre- FE-HCO3 >15 % confirms the diagnosis of
tion and a urine osmolality equal to or greater than proximal RTA [73]. Again urine and plasma sam-
that of serum, implying adequate vasopressin ples must be collected anaerobically and prefera-
levels – required for optimal distal tubular secre- bly measured without delay.
tion of potassium [71].

Estimating Urine Ammonium Production


Assessment of Renal Acid/Base The normal renal response to acidosis is to
Homeostasis increase the excretion of ammonium (NH4+) in
order to efficiently buffer excreted protons,
The following tests are useful in ascertaining the together with an equal increase in Cl ions to
site of tubular dysfunction in the presence of a maintain electroneutrality. With distal RTA, there
suspected tubular cause for metabolic acidosis – is decreased excretion of ammonium ions, and
renal tubular acidosis (RTA). Proximal and distal therefore concomitantly, Cl excretion is not
RTA differ in the mechanism by which the acido- appropriately increased. The urine ammonium
sis occurs. In the former, there is a lowered thresh- concentration is therefore an indicator of appro-
old for the loss of bicarbonate, while in the latter, priate hydrogen excretion by the distal tubule. As
the problem is an inability to excrete protons. The this analyte is not routinely measured, two surro-
following tests are useful to distinguish the types gate methods for estimating ammonium excretion
of RTA. can be used:

Urine pH (a) Urine anion gap:


In the normal kidney, the introduction of an acid
load leads to acidification of the urine, with Urine anion gap ¼ ðNaþ þ Kþ Þ
decreased excretion of bicarbonate and increased  Cl ðin mmol=LÞ
buffering of acid in the form of ammonium chlo-
ride and sodium or potassium phosphates, yield- Bicarbonate levels are usually insignificant in
ing a urine pH of <5.5. With both forms of RTA, a acidic urine (pH < 6) and can therefore be
paradoxically, relatively alkaline urine is often ignored in the calculation. The normal renal
found in the setting of a normal anion gap meta- response to acidosis results in a negative gap
bolic acidosis. In proximal RTA, however, the due to the increase in Cl, while in distal RTA,
distal tubule is still able to excrete protons, and this gap remains positive, implying defective
therefore the urine pH can still achieve a pH of ammonium production [61]. The urinary
<5.5 provided the plasma bicarbonate concentra- anion gap has also been used in neonates,
tion is below the renal threshold [73]. In contrast, but is more difficult to interpret especially in
with distal RTA, the urine pH is always >6.0 [61]. the premature [75].
630 G. Watt et al.

(b) Estimated ammonium concentration using alternative methods are recommended. The urine
the urine osmolar gap maximum PCO2 is measured after collection
This method makes the same assumptions under oil and in glass to prevent air contamina-
as the urine anion gap that the gap is generally tion, following intravenous sodium bicarbonate
caused by an increase in ammonium chloride. administration with a urine pH >7. This test can
It has been shown to correlate well with net be performed using either sodium bicarbonate or
acid excretion and ammonium levels and is acetazolamide or both [78–81]. Bicarbonate
the best indirect test for assessment of ammo- administration results in a large delivery of
nium excretion [76, 77]: HCO 3 to the distal tubule, and HCO3 now


becomes almost the only acceptor of excreted


protons in the lumen, resulting in the production
Estimated urine ammoniumðmmol=LÞ
of carbonic acid which, in turn, dissociates into
¼ urine
 osmolar gap
2 
carbon dioxide and water. Therefore, with normal
¼ U osmolality  U osmolarity
2
distal hydrogen ion secretion, the urine PCO2 is
expected to rise. With defective distal hydrogen
Urine osmolarity (mOsm/L) can be calcu-
ion secretion, the PCO2 will be similar to that in
lated using the following formula (all analytes
blood. Acetazolamide, a carbonic anhydrase
in mmol/L):
inhibitor, enhances this effect. These tests need
not be performed if urine pH is <5.5 which
Urineosmolarity ¼ 2½Naþ þ Kþ  þ Urea þ glucose implies adequate distal acidification.

To convert from urea nitrogen in mg/dL to


urea in mmol/L, divide by 2.8. For glucose, Urate, Oxalate, Citrate
convert from mg/dL to mmol/L by dividing
by 18. Urate, derived from purine catabolism, is excreted
predominantly by the proximal tubules but also
In the event that distal tubular dysfunction is filtered and reabsorbed. Abnormal serum urate
still not demonstrable, other assessments of uri- levels in children should always be investigated
nary acidification may be required. Three methods as a number of rare disorders may otherwise be
are employed to stimulate and therefore assess missed; in addition, elevated levels carry the risk
distal hydrogen ion secretion: of nephrolithiasis and AKI. Increased serum
levels are ascribed to either systemic
(a) Acid loading by administration of ammonium overproduction or undersecretion in the kidney.
chloride (NH4Cl) and measuring the change Examples of overproduction in children include
in urine pH: pH <5.5 is normal, whereas pH the tumor lysis syndrome and rare metabolic dis-
remains above 6.5 in distal RTA. orders such as Lesch-Nyhan syndrome and glyco-
(b) Increasing sodium delivery to the distal tubule gen storage disease type 1. Idiopathic gout is rare
by administration of furosemide in combina- in children. Undersecretion in the kidney may be
tion with mineralocorticoid stimulation due to metabolic abnormalities or competition
(fludrocortisone) and measuring the urine pH with other chronically elevated anions such as
response. lactate, salicylate, and ketones. Decreased serum
(c) Increasing the delivery of HCO3 to the distal urate levels may be associated with hepatocellular
tubule and measuring the increase in urine disease, increased renal losses in the Fanconi syn-
PCO2. drome, deficiency of xanthine oxidase activity
(molybdenum cofactor deficiency), and purine
While the urine acidification response to nucleoside phosphorylase deficiency [82]. Frac-
NH4Cl is commonly described, this test is not tional excretion of uric acid (Fe-UA) is useful for
the preferred method in children, and the differentiating the causes of both hypouricemia
21 Laboratory Investigation of the Child with Suspected Renal Disease 631

and hyperuricemia in children; Fe-UA is very 24-h urine collection, and other causes excluded;
high in young infants at approximately 28 %, these include hyperglycemia, hypokalemia, and
dropping rapidly in the first months of life to an hypercalcemia. Polyuria is defined as >4 mL/kg/
average of 8 % in childhood, with normal adult h in infants and children and >6 mL/kg/h in the
values of approximately 5 % attained in adoles- newborn [88]. An early morning urine osmolality
cence [83]. In children over 2 years of age, uric >800 mOsm/kg H2O implies normal concentrat-
acid excretion is approximately (520  145 ing ability with no further testing required.
mg/1.73 m2/24 h) 3,093  875 μmol/1.73
m2/24 h [84]. Water Deprivation Test
Oxalate is usually only measured in urine to A water deprivation test is performed to establish
diagnose hyperoxaluria as it forms highly insolu- renal ability to concentrate urine. The water dep-
ble crystals in urine and, like urate, may precipi- rivation test should only be performed if the base-
tate nephrolithiasis and/or AKI. Normal oxalate line serum sodium and osmolality are within
excretion is in the range of 20–50 mg/1.73 m2/ reference intervals and urine osmolality is below
24 h (0.3–0.55 mmol/1.73 m2/24 h). In infants, the 300 mosm/kg [88]. The test involves fluid restric-
value may be slightly higher [85]. Above this tion for a number of hours until significant dehy-
range is suspicious for hyperoxaluria. The pri- dration has occurred, accompanied by hourly
mary hyperoxalurias constitute a number of met- measurements of osmolality and sodium in
abolic disorders of oxalate metabolism, whereas serum and urine, as well as weight. The test is
secondary hyperoxaluria may be idiopathic or stopped when either 2–5 % body weight loss has
associated with ethylene glycol poisoning and been reached, when the upper limit of the refer-
fat malabsorption syndromes where oxalate is ence interval has been exceeded for serum osmo-
preferentially absorbed from the gut. lality or sodium, or when a normal urine response
Citrate excretion in urine is important in reduc- to dehydration has occurred, typically urine osmo-
ing the risk of stone formation especially calcium- lality >800 mosm/kg. The test is usually limited
containing stones. Hypocitraturia contributes to to 4 h in infants and 7 h in children. If the urine
stone formation. Citrate excretion below osmolality exceeds >800 mosm/kg, both central
300 mg/g creatinine (0.176 mol/mol) in girls and and nephrogenic diabetes can be excluded, and a
125 mg/g/ creatinine (0.074 mol/mol) in boys tentative diagnosis of primary polydipsia made
suggests hypocitraturia [86]. [88, 89]. In children where the urine osmolality
has not risen above 300 mosm/kg, vasopressin
(1-desamino-8-argininovasopressin) is adminis-
Renal Tubular Concentrating Ability tered intranasally. Partial fluid restriction is
recommended by some (50–75 % of usual fluid
The concentrating ability of the kidney relies on intake in infants), to avoid water overload. Intake
the action of vasopressin on vasopressin receptors and output are strictly monitored. Urine is col-
in the renal collecting ducts, resulting in insertion lected after 4 h (2 h in infants) for osmolality
of aquaporin-2 channels into the apical membrane measurement. If urine osmolality then increases
facilitating the reabsorption of water. Defective by >50 %, a diagnosis of central diabetes
concentration of urine with associated polyuria insipidus can be inferred, whereas an increase of
and polydipsia can therefore be ascribed to either <50 % implies a nephrogenic cause for diabetes
vasopressin deficiency (central diabetes insipidus.
insipidus), loss of vasopressin action In those cases where the urine osmolality fol-
(nephrogenic diabetes insipidus), or primary poly- lowing water deprivation falls between 300 and
dipsia, defined as water intake of more than 800 mosm/kg, further investigation is required to
2 L/m2/day [87]. differentiate between primary polydipsia, partial
When a diagnosis of diabetes insipidus is central, and partial nephrogenic diabetes
suspected, polyuria must first be confirmed by a insipidus [89].
632 G. Watt et al.

It is important to realize that current methods used in conjunction with post-osmotic stimulus
for serum osmolality measurement may not sodium concentration [92].
perform at the level of accuracy required for the
water deprivation test and therefore serum
sodium is a better measure of dehydration. Parathyroid Hormone
A basal sodium concentration above the reference
interval in the presence of a urine osmolality of Circulating parathyroid hormone (PTH) consists
<300 mosm/kg makes the water deprivation of a range of immunoreactive fragments. Biolog-
test unnecessary and potentially dangerous. ically active PTH 1–84 is the principle hormone
Vasopressin can be administered immediately. making up approximately 15–20 % of circulating
The water deprivation test must not be performed levels. It exerts its effects via the first 34 amino
in patients with renal insufficiency, hypovolemia, acids on the amino terminal of the molecule,
uncorrected hypothyroidism, and uncontrolled interacting with the type 1 PTH receptor, and has
diabetes insipidus. Dehydration may develop a very short half-life of approximately 3 min. Fur-
rapidly; therefore, careful monitoring of ther degradation in the circulation and in the para-
water balance must occur via hourly body thyroid gland results in additional PTH fragments
weight and serum sodium and osmolality containing carboxyl (C-terminal) or amino
measurements. (N-terminal) terminal portions of the peptide.
These fragments have much longer half-lives
Direct Vasopressin Measurement and so make up the majority of PTH type peptides
Urine osmolality measurement following the in plasma. In addition, some of the N-terminally
water deprivation test is an indirect measurement truncated fragment PTH(7–84) interact with other
of vasopressin activity. In locations where vaso- PTH receptors such as the C-PTH receptor and
pressin measurement is readily available, proto- may play important roles in the regulation of bone
cols include direct vasopressin measurement as resorption and serum calcium concentration [93].
part of the hourly biochemical measurements. Automated routine laboratory PTH immunoas-
Vasopressin and urinary osmolality increase in says rely on antibodies directed at different epitopes
normal subjects and with primary polydipsia, on the PTH molecule. The first widely used immu-
while neither increases with central diabetes noassay called the intact PTH assay employs a cap-
insipidus. In nephrogenic diabetes insipidus, ture antibody directed toward C-terminal epitopes
vasopressin levels increase as for normal subjects, 39–84 and a second detection antibody directed
while urine osmolality does not increase toward N-terminal epitopes 13–34. This assay mea-
[90]. Vasopressin measurement is useful in differ- sures biologically active PTH 1–84 together with
entiating partial diabetes insipidus. Unfortunately, large C-terminal fragments lacking portions of the
vasopressin assays are not readily available, and N-terminus and can therefore overestimate PTH
vasopressin is a highly unstable analyte with a levels in uremic patients [94]. To address this issue,
short half-life. a more specific bioactive PTH 1–84 assay was devel-
oped, utilizing a similar capture antibody to the intact
Co-peptin (C-Terminal ProAVP) Direct PTH assays but with a detection antibody
Assay directed against the extreme N-terminal epitopes
Co-peptin, the C-terminal part of the vasopressin 1–4 [95]. Both assays are adequate for making a
precursor, is co-secreted with vasopressin from diagnosis of primary hyperparathyroidism, but evi-
the neurohypophysis. It is superior to vasopressin dence suggests that bioactive PTH 1–84 assays have
in that is more stable ex vivo and is easier to higher diagnostic accuracy in patients with renal
measure [91]. It has been shown to be a good failure and primary hypoparathyroidism and in
differentiator in partial diabetes insipidus when patients with PTH-producing carcinoma [96, 100].
21 Laboratory Investigation of the Child with Suspected Renal Disease 633

References 15. Fong J, Johnston S, Valentino T, Notterman D.


Length/serum creatinine ratio does not predict mea-
1. Haycock GB, Schwartz GJ, Wisotsky DH. Geometric sured creatinine clearance in critically ill children.
method for measuring body surface area: a height- Clin Pharmacol Ther. 1995;58:192–1977.
weight formula validated in infants, children and 16. Myers GL, Miller WG, Coresh J, Fleming J,
adults. J Pediatr. 1978;93:62–6. Greenberg N, Greene T, Hostetter T, Levey AS,
2. Guignard JP, Santos F. Laboratory investigations. In: Panteghini M, Welch M, Eckfeldt JH, National Kid-
Avner ED, Harmon WE, Niaudet P, editors. Pediatric ney Disease Education Program Laboratory Working
nephrology. 5th ed. Philadelphia: Lippincott Williams Group. Recommendations for improving serum cre-
Wilkins; 2004. p. 349–424. atinine measurement: a report from the Laboratory
3. Durand E, Prigent A. The basics of renal imaging and Working Group of the National Kidney Disease Edu-
function studies. J Nucl Med. 2002;46:249–67. cation Program. Clin Chem. 2006;52(1):5–18.
4. Odlund B, Hällgren R, Sohtell M, Lindström B. Is 17. Levey AS, Coresh J, Greene T, Marsh J, Stevens LA,
125I-iothalamate an ideal marker for glomerular Kusek JW, Van Lente F, Chronic Kidney Disease
filtration? Kidney Int. 1985;27:9–16. Epidemiology Collaboration. Expressing the Modifi-
5. Bhatt MK, Bartlett ML, Mallitt KA, McTaggart S, cation of Diet in Renal Disease Study equation for
Ravi Kumar AS. Correlation of various published estimating glomerular filtration rate with standardized
radionuclide glomerular filtration rate estimation serum creatinine values. Clin Chem. 2007;53(4):
techniques and proposed paediatric normative data. 766–72.
Nucl Med Commun. 2011;32(11):1088–94. 18. Levey AS, Stevens LA, Schmid CH, Zhang YL,
6. Nilsson-Ehle P, Grubb A. New markers for determi- Castro 3rd AF, Feldman HI, Kusek JW, Eggers P,
nation of GFR: iohexol clearance and cystatin C Van Lente F, Greene T, Coresh J, CKD-EPI (Chronic
serum concentration. Kidney Int. 1994;46:S17–19. Kidney Disease Epidemiology Collaboration). A new
7. Krustzen E, Back SE, Nilsson-Ehle P. Determination equation to estimate glomerular filtration rate. Ann
of glomerular filtration rate using iohexol clearance Intern Med. 2009;150(9):604–12.
and capillary sampling. Scand J Clin Lab Invest. 19. Schwartz GJ, Muñoz A, Schneider MF, Mak RH,
1990;50:279–83. Kaskel F, Warady BA, Furth SL. New equations to
8. Gaspari F, Perico N, Ruggenenti P, Mosconi L, estimate GFR in children with CKD. J Am Soc
Amuchastegui CS, Guerini E, Daina E, Remuzzi Nephrol. 2009;20(3):629–37.
G. Plasma clearance of nonradioactive iohexol as a 20. Greenberg N, Roberts WL, Bachmann LM, Wright
measure of glomerular filtration rate. J Am Soc EC, Dalton RN, Zakowski JJ, Miller WG. Specificity
Nephrol. 1995;6:257–63. characteristics of seven commercial creatinine mea-
9. Colantonio DA, Kyriakopoulou L, Chan MK, Daly surement procedures using enzymatic and Jaffe
CH, Brinc D, Venner AA, Pasic MD, Armbruster D, method principles. Clin Chem. 2012;58:391–401.
Adeli K. Closing the gaps in pediatric laboratory 21. Finney H, Newman DJ, Thakkar H, Fell JM, Price
reference intervals: a CALIPER database of 40 CP. Reference ranges for plasma cystatin C and cre-
biochemical markers in a healthy and multiethnic atinine measurements in premature infants, neonates
population of children. Clin Chem. 2012;58(5): and older children. Arch Dis Child. 2000;82:71–5.
854–68. 22. Tenstad O, Roald AB, Grubb A, Aukland K. Renal
10. Namnum P, Insogna K, Baggish D, Hayslett handling of radiolabelled human cystatin C in the rat.
JP. Evidence for bidirectional net movement of creat- Scand J Clin Lab Invest. 1996;56:409–14; Keevil BG,
inine in the rat kidney. Am J Physiol. 1983;244: Kilpatrick ES, Nichols SP, Maylor PW. Biological
F719–23. variation of cystatin C: implications for the assess-
11. Sjöstrom PA, Odlind BG, Wolgast M. Extensive tubu- ment of glomerular filtration rate. Clin Chem
lar secretion and reabsorption of creatinine in humans. 1998;44:1535–1539.
Scand J Urol Nephrol. 1988;22:129–31. 23. Inker LA, Schmid CH, Tighiouart H, Eckfeldt JH,
12. Vieux R, Hascoet JM, Merdariu D, Fresson J, Guille- Feldman HI, Greene T, Kusek JW, Manzi J, Van
min F. Glomerular filtration rate reference values in Lente F, Zhang YL, Coresh J, Levey AS, CKD-EPI
very preterm infants. Pediatrics. 2010;125(5): Investigators. Estimating glomerular filtration rate
1186–92. from serum creatinine and cystatin C. N Engl J Med.
13. Piepsz A, Tondeur M, Ham H. Revisiting normal (51) 2012;367:20–9.
Cr-ethylenediaminetetraacetic acid clearance values 24. Inker LA, Eckfeldt J, Levey AS, Leiendecker-Foster-
in children. Eur J Nucl Med Mol Imaging. 2006;33 C, Rynders G, Manzi J, Waheed S, Coresh
(12):1477–82. J. Expressing the CKD-EPI (Chronic Kidney Disease
14. Doolan PD, Alpen EL, Theil GB. A clinical appraisal Epidemiology Collaboration) cystatin C equations for
of the plasma concentration and endogenous clear- estimating GFR with standardized serum cystatin C
ance of creatinine. Am J Med. 1962;32:65–79. values. Am J Kidney Dis. 2011;58(4):682–4.
634 G. Watt et al.

25. Roos JF, Doust J, Tett SE, Kirkpatrick CM. Diagnos- plasma sample method. J Nucl Med. 1991;32(6):
tic accuracy of cystatin C compared to serum creati- 1294–7.
nine for the estimation of renal dysfunction in 37. Ham HR, De Sadeleer C, Hall M, Piepsz A. Which
adults and children-a meta-analysis. Clin Biochem. single blood sample method should be used to esti-
2007;40(5–6):383–91. mate 51Cr-EDTA clearance in adolescents? Nucl Med
26. Dharnidharka VR, Kwon C, Stevens G. Serum Commun. 2004;25(2):155–7.
cystatin C is superior to serum creatinine as a marker 38. Staples A, LeBlond R, Watkins S, Wong C, Brandt J.
of kidney function: a meta-analysis. Am J Kidney Validation of the revised Schwartz estimating equa-
Dis. 2002;40(2):221–6. tion in a predominantly non-CKD population. Pediatr
27. Grubb A, Horio M, Hansson LO, Björk J, Nyman U, Nephrol. 2010;25(11):2321–6.
Flodin M, Larsson A, Bökenkamp A, Yasuda Y, 39. Ng DK, Schwartz GJ, Jacobson LP, Palella FJ,
Blufpand H, Lindström V, Zegers I, Althaus H, Margolick JB, Warady BA, Furth SL, Muñoz
Blirup-Jensen S, Itoh Y, Sjöström P, Nordin G, A. Universal GFR determination based on two time
Christensson A, Klima H, Sunde K, Hjort- points during plasma iohexol disappearance. Kidney
Christensen P, Armbruster D, Ferrero C. Generation Int. 2011;80(4):423–30.
of a new cystatin C-based estimating equation for 40. Hogg RJ, Furth S, Lemley KV, Portman R, Schwartz
glomerular filtration rate by use of 7 assays standard- GJ, Coresh J, Balk E, Lau J, Levin A, Kausz AT,
ized to the international calibrator. Clin Chem. Eknoyan G, Levey AS. National Kidney Founda-
2014;60(7):974–86. tion’s Kidney Disease Outcomes Quality Initiative
28. Levey AS, Greene T, Schluchter MD, et al. clinical practice guidelines for chronic kidney
Glomerular filtration rate measurements in clinical disease in children and adolescents: evaluation, clas-
trials. modification of diet in renal disease study sification, and stratification. Pediatrics. 2003;111:
group and the diabetes control and complications 1416–21.
trial research group. J Am Soc Nephrol. 1993;4(5): 41. Hogg RJ, Portman RJ, Milliner D, Lemley KV,
1159–71. Eddy A, Ingelfinger J. Evaluation and management
29. De Sadeleer C, Van Laere K, Georges B, Piepsz A, of proteinuria and nephrotic syndrome in children:
Ham HR. Influence of time interval and number of recommendations from a pediatric nephrology panel
blood samples on the error in renal clearance deter- established at the National Kidney Foundation con-
mination using a mono-exponential model: a monte ference on proteinuria, albuminuria, risk, assessment,
carlo simulation. Nucl Med Commun. 2000;21(8): detection, and elimination (PARADE). Pediatrics.
741–5. 2000;105:1242–9.
30. Brochner-Mortensen J, Rodbro P. Selection of routine 42. Houser MT, Jahn MF, Kobayashi A, Walburn J.
method for determination of glomerular filtration rate Assessment of urinary protein excretion in the ado-
in adult patients. Scand J Clin Lab Invest. 1976;36(1): lescent: effect of body position and exercise. J Pediatr.
35–43. 1986;109:556–61.
31. Fahey FH, Treves ST, Adelstein SJ. Minimizing 43. Morcos SK, el-Nahas AM, Brown P, Haylor J. Effect
and communicating radiation risk in pediatric of iodinated water soluble contrast media on urinary
nuclear medicine. J Nucl Med. 2011;52(8):1240–51. protein assays. BMJ. 1992;305:29.1.
32. Rehling M, Nielsen LE, Marqversen J. Protein 44. Trachtenberg F, Barregard L. The effect of age, sex,
binding of 99Tcm-DTPA compared with other and race on urinary markers of kidney damage in
GFR tracers. Nucl Med Commun. 2001;22(6): children. Am J Kidney Dis. 2007;50:938–45.
617–23. 45. Goldstein SL. Acute kidney injury biomarkers: renal
33. Gordon I, Piepsz A, Sixt R, Auspices of Paediatric angina and the need for a renal troponin I. BMC Med.
Committee of European Association of Nuclear Med- 2011;9:135.
icine. Guidelines for standard and diuretic renogram 46. Ronco C, Legrand M, Goldstein SL, Hur M, Tran N,
in children. Eur J Nucl Med Mol Imaging. 2011; Howell EC, Cantaluppi V, Cruz DN, Damman K,
38(6):1175–88. Bagshaw SM, Di Somma S, Lewington A. Neutrophil
34. Fleming JS. An improved equation for correcting gelatinase-associated lipocalin: ready for routine clin-
slope-intercept measurements of glomerular filtration ical use? An international perspective. Blood Purif.
rate for the single exponential approximation. Nucl 2014;37(4):271–85.
Med Commun. 2007;28(4):315–20. 47. Bennett M, Dent CL, Ma Q, Dastrala S, Grenier F,
35. Jodal L, Brochner-Mortensen J. Reassessment of a Workman R, Syed H, Ali S, Barasch J,
classical single injection 51Cr-EDTA clearance Devarajan P. Urine NGAL predicts severity of
method for determination of renal function in children acute kidney injury after cardiac surgery: a prospec-
and adults. Part I: analytically correct relationship tive study. Clin J Am Soc Nephrol. 2008;3(3):
between total and one-pool clearance. Scand J Clin 665–73.
Lab Invest. 2009;69(3):305–13. 48. Krawczeski CD, Goldstein SL, Woo JG, Wang Y,
36. Ham HR, Piepsz A. Estimation of glomerular filtra- Piyaphanee N, Ma Q, Bennett M, Devarajan P.
tion rate in infants and in children using a single- Temporal relationship and predictive value of urinary
21 Laboratory Investigation of the Child with Suspected Renal Disease 635

acute kidney injury biomarkers after pediatric cardio- 65. Singh J, Moghal N, Pearce SH, Cheetham T. The
pulmonary bypass. J Am Coll Cardiol. 2011;58(22): investigation of hypocalcaemia and rickets. Arch
2301–9. Dis Child. 2003;88(5):403–7.
49. Kovesdy CP, Quarles LD. Fibroblast growth factor- 66. Shaw N, Wheeldon J, Brocklebank J. Indices of intact
23: what we know, what we don’t know, and what we serum parathyroid hormone and renal excretion of
need to know. Nephrol Dial Transplant. 2013;28(9): calcium, phosphate and magnesium. Arch Dis Child.
2228–36. 1990;65:1208–11.
50. Friedman AL. Urinalysis: oft obtained, oft ignored. 67. Calado J, Santer R, Rueff J. Effect of kidney disease
Contemp Pediatr. 1991;8:31–51. on glucose handling (including genetic defects).
51. Huicho L, Campos-Sanchez M, Alano C. Meta- Kidney Int Suppl. 2011;120:S7–13.
analysis of urine screening tests for determining the 68. Bökenkamp A, Ludwig M. Disorders of the renal
risk of urinary tract infection in children. Pediatr proximal tubule. Nephron Physiol. 2011;118(1):1–6.
Infect Dis J. 2002;21:1–7. 69. Sirac C, Bridoux F, Essig M, Devuyst O, Touchard G,
52. Linshaw MA, Gruskin AB. The routine urinalysis: to Cogné M. Toward understanding renal Fanconi syn-
keep or not to keep: that is the question. Pediatrics. drome: step by step advances through experimental
1997;100:1031–2. models. Contrib Nephrol. 2011;169:247–61.
53. Fassett RG, Horgan B, Mathew TH. The detection of 70. Duran N. Amino acids. In: Blau N, Duran M, Gibson
glomerular bleeding by phase contrast microscopy. KM, editors. Laboratory guide to the methods in
Lancet. 1982;1:1432–4. biochemical genetics. Berlin: Springer; 2008.
54. Arvind B, Anurag B, Shina M. Approach to p. 53–89.
renal tubular disorders. Indian J Pediatr. 2005;72(9): 71. Choi MJ, Ziyadeh FN. The utility of the transtubular
771–6. potassium gradient in the evaluation of hyperkalemia.
55. Delaney MP, Price CP, Lamb EJ. Kidney disease. In: J Am Soc Nephrol. 2008;19(3):424–6.
Burtis CA, Ashwood ER, Bruns DE, editors. Tietz 72. West ML, Marsden PA, Richardson RM, Zettle RM,
textbook of clinical chemistry and molecular diagnos- Halperin ML. New clinical approach to evaluate dis-
tics. 5th ed. London: Elsevier/Saunders; 2012. orders of potassium excretion. Miner Electrolyte
p. 1523–607. Metab. 1986;12(4):234–8.
56. Fahimi D, Mohajeri S, Hajizadeh N, et al. Compari- 73. Haque SK, Ariceta G, Batlle D. Proximal renal tubu-
son between fractional excretions of urea and sodium lar acidosis: a not so rare disorder of multiple etiolo-
in children with acute kidney injury. Pediatr Nephrol. gies. Nephrol Dial Transplant. 2012;27:4273–87.
2009;24(12):2409–12. 74. Meerkin M, et al. Section II general clinical tests. In:
57. Gotfried J, Wiesen J, Raina R, Nally Jr JV. Finding the Wu AHB, editor. Tietz clinical guide to laboratory
cause of acute kidney injury: which index of frac- tests. 4th ed. St. Louis: Saunders/Elsevier; 2006.
tional excretion is better? Cleve Clin J Med. p. 32–1202.
2012;79(2):121–6. 75. Sulyok E, Guignard JP. Relationship of urinary anion
58. Srivastava T, Schwaderer A. Diagnosis and manage- gap to urinary ammonium excretion in the neonate.
ment of hypercalciuria in children. Curr Opin Pediatr. Biol Neonate. 1990;57:98–106.
2009;21(2):214–19. 76. Kim GH, Han JS, Kim YS, Joo KW, Kim S, Lee
59. Alconcher LF, Castro C, Quintana D, et al. Urinary JS. Evaluation of urine acidification by urine anion
calcium excretion in healthy school children. Pediatr gap and urine osmolal gap in chronic metabolic aci-
Nephrol. 1997;11:186–8. dosis. Am J Kidney Dis. 1996;27(1):42–7.
60. Foley KF, Boccuzzi L. Urine calcium: laboratory 77. Halperin ML, Kamel KS, Goldstein MB. Tools to use
measurement and clinical utility. Lab Med. to diagnose acid-base disorders. In: Halperin ML,
2010;41(11):683–6. Kamel KS, Goldstein MB, editors. Fluid, electrolyte,
61. Escobar L, Mejía N, Gil H, Santos F. Distal renal and acid-base physiology-a problem-based approach.
tubular acidosis: a hereditary disease with an inade- 4th ed. Philadelphia: Saunders; 2010. p. 39–59.
quate urinary H+ excretion. Nefrologia. 2013;33(3): 78. García Nieto V, Monge M, Hernández Hernández L,
289–96. Callejón A, Yanes MI, García Rodríguez VE. Study
62. Sevgi M, Erkin S. Quantification of hypercalciuria of the renal acidification capacity in children diag-
with the urine calcium osmolality ratio in children. nosed of idiopathic hypercalciuria. Nefrologia.
Pediatr Nephrol. 2005;20(11):1562–5. 2003;23:219–24.
63. Cole DEC, Quamme GA. Inherited disorders of renal 79. Batlle DC. Segmental characterization of defects in
magnesium handling. J Am Soc Nephrol. 2000;11: collecting tubule acidification. Kidney Int. 1986;30:
1937–47. 546–54.
64. Bangert SK, Lapsley M. Renal tubular disorders and 80. Viljoen A, Norden AGW, Karet FE. Replacing the
renal stone disease. In: Marshall W, Bangert SK, short ammonium chloride test. Kidney Int. 2007;72:
editors. Clinical biochemistry-metabolic and clinical 1163.
aspects. 2nd ed. London: Churchill/Livingston; 2008. 81. Walsh SB, Shirley DG, Wrong OM, Unwin RJ.
p. 174–85. Urinary acidification assessed by simultaneous
636 G. Watt et al.

furosemide and fludrocortisone treatment: an alterna- and indirect water deprivation tests. J Clin Endocrinol
tive to ammonium chloride. Kidney Int. 2007;71: Metab. 2011;96:1506–15.
1310–16. 93. Murray TM, Rao LG, Divieti P, Bringhurst
82. Lamb E, Newman DJ, Price CP. Kidney function FR. Parathyroid hormone secretion and action: evi-
tests. In: Burtis A, Ashwood ER, Bruns D, editors. dence for discrete receptors for the carboxyl-terminal
Tietz textbook of clinical chemistry and molecular region and related biological actions of carboxyl-
diagnostics. 4th ed. St. Louis: Elsevier/Saunders; terminal ligands. Endocr Rev. 2005;26:78–113.
2006. p. 805–7. 94. Lepage R, Roy L, Brossard JH, Rousseau L, Dorais C,
83. Stiburkova B, Bleyer AJ. Changes in serum urate and Lazure C, D’Amour P. A non-(1-84) circulating para-
urate excretion with age. Adv Chronic Kidney Dis. thyroid hormone (PTH) fragment interferes signifi-
2012;19(6):372–6. cantly with intact PTH commercial assay
84. Stapleton FB, Linshaw MA, Hassanein K, et al. Uric measurements in uremic samples. Clin Chem. 1998;
acid excretion in normal children. J Pediatr. 44:805–9.
1978;92:911–14. 95. Savoca R, Bock A, Kraenzlin ME, Schmid HR, Huber
85. DeSanto NG, Di Iorio B, Capasso G, et al. Population AR. An automated ‘bio-intact’ PTH assay: a step
based data on urinary excretion of calcium, magne- towards standardisation and improved correlation
sium, oxalate, phosphate and uric acid in children with parathyroid function in renal disease. Clin
from Cimitile (Southern Italy). Pediatr Nephrol. Chim Acta. 2004;343:167–71.
1992;6:149–57. 96. Gao P, Scheibel S, D’Amour P, John MR, Rao SD,
86. Norman ME, Feldman NI, Cohn RM, et al. Urinary Schmidt-Gayk H, Cantor TL. Development of a novel
citrate excretion in the diagnosis of distal tubular immunoradiometric assay exclusively for biologi-
acidosis. J Pediatr. 1978;82:394–400. cally active whole parathyroid hormone 1-84: impli-
87. Saborio P, Tipton GA, Chan JC. Diabetes insipidus. cations for improvement of accurate assessment of
Pediatr Rev. 2000;21:122–9. parathyroid function. J Bone Miner Res. 2001;16:
88. Mishra G, Chandrashekhar SR. Management of dia- 605–14.
betes insipidus in children. Indian J Endocrinol 97. Piepsz A, Ham HR. Pediatric applications of renal
Metab. 2011;15(7):180–7. nuclear medicine. Semin Nucl Med. 2006;36(1):
89. Fenske W, Allolio B. Current state and future per- 16–35.
spectives in the diagnosis of diabetes insipidus: a 98. Winter EW. Urine protein electrophoresis. In: Harris
clinical review. J Clin Endocrinol Metab. 2012; NS, Winter EW, editors. Multiple myeloma and relate
97(10):3426–37. serum protein disorders – an electrophoretic guide. 1st
90. Shimura N. Urinary arginine vasopressin (AVP) mea- ed. New York: DemosMedical; 2012. p. 83–116.
surement in children: water deprivation test incorpo- 99. Piepsz A, Colarinha P, Gordon I, Hahn K, Olivier P,
rating urinary AVP. Acta Paediatr Jpn. 1993;35(4): Sixt R, van Velzen J; Paediatric Committee of the
320–4. European Association of Nuclear Medicine. Guide-
91. Morgenthaler NG, Struck J, Alonso C, Bergmann A. lines for glomerular filtration rate determination in
Assay for the measurement of copeptin, a stable pep- children. Eur J Nucl Med. 2001;28(3):BP31–6.
tide derived from the precursor of vasopressin. Clin 100. Cavalier E, Daly AF, Betea D, Pruteanu-Apetrii PN,
Chem. 2006;52:112–19. Delanaye P, Stubbs P, Bradwell AR, Chapelle JP,
92. Fenske W, Quinkler M, Lorenz D, Zopf K, Haagen U, Beckers A. The ratio of parathyroid hormone as mea-
Papassotiriou J, Pfeiffer AF, Fassnacht M, Störk S, sured by third- and second-generation assays as a
Allolio B. Copeptin in the differential diagnosis of the marker for parathyroid carcinoma. J Clin Endocrinol
polydipsia-polyuria syndrome – revisiting the direct Metab. 2010; 95(8):3745–9.
Growth and Development of the Child
with Renal Disease 22
Bethany Foster

Contents Introduction
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 637
Normal growth and development is a central goal
Normal Growth and Development . . . . . . . . . . . . . . . . 637
Bone Age . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 646
in the management of children with chronic kid-
ney disease (CKD). This chapter will review the
Growth and Development in Chronic Kidney
growth patterns observed in children with CKD as
Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 647
Protein Energy Wasting in CKD . . . . . . . . . . . . . . . . . . . . 649 well as the causes of growth impairment and the
signs and causes of protein energy wasting (PEW)
Evaluation of Growth and Development
in CKD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 652 in pediatric CKD. The methods for assessing
growth and nutritional status, along with the lim-
Optimizing Growth in Children with CKD . . . . . . 655
Nutrition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 656
itations of these measures in CKD, will also be
Fluid and Sodium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 658 reviewed. Finally, management strategies to opti-
Acidosis Control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 658 mize growth among children with CKD will be
Control of Hyperparathyroidism and Metabolic outlined. However, before considering the growth
Bone Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 658
Recombinant Human Growth Hormone
abnormalities associated with childhood CKD,
Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 659 normal growth and development and the tools
Enhanced Dialytic Clearance . . . . . . . . . . . . . . . . . . . . . . . . 660 available to assess the adequacy of the growth of
Minimized Exposure to Corticosteroids . . . . . . . . . . . . . 660 an individual child will be reviewed.
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 660
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 660
Normal Growth and Development

Normal phases of growth: Normal growth can be


divided into three phases – infancy, childhood,
and puberty. Each phase is primarily influenced
by a different factor [1–3]. The infancy phase of
growth is essentially a continuation of fetal
growth and, like fetal growth, is driven primarily
by nutrition. Nutritional intake stimulates insulin
and insulin-like growth factors, the major regula-
B. Foster (*)
tors of growth in this phase. Growth is most rapid
The Research Institute of the McGill University Health
Centre, Montreal, QC, Canada in the first 2–3 months of life, with weight gains
e-mail: bethany.foster@mcgill.ca of ~30 g per day in the first 2 months and
# Springer-Verlag Berlin Heidelberg 2016 637
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_20
638 B. Foster

20–25 g/day in the third month. Linear growth is completion of genital development indicated
similarly brisk in the first 3 months of life, but delayed sexual maturation [7].
shows rapid deceleration thereafter [2]. At 6–12 Growth measurements: Linear growth in
months of age, the early, nutrition-dominated children is assessed by measuring recumbent
infancy phase of growth is replaced by the child- length (in children 24 months of age or younger)
hood phase of growth. Growth hormone is the or standing height (children >24 months old) and
major driver of growth in the childhood phase; weight. These measures are then compared with
during this phase, growth velocity is greater than the distributions among healthy children of the
it is at the end of the infancy phase and decelerates same age to determine the adequacy of growth.
slowly over time [3]. In the childhood phase, Similarly, weight is compared with the distribu-
growth rates of ~7–8 cm/year are typical at tion of weights among healthy children of the
3 years of age and of ~6–7 cm/year at 10 years same age. For children under 3 years of age,
of age. Finally, the onset of puberty marks the head circumference is also assessed.
beginning of the pubertal phase of growth, Body mass index (BMI): BMI is calculated by
influenced mainly by sex hormones [1–4]. Puber- dividing weight (kg) by height (m) squared and
tal growth patterns differ for girls and boys. Girls provides a means of evaluating the appropriate-
experience a major increase in growth velocity ness of a child’s weight relative to height, at a
during the first 2 years of puberty, followed by specific age. Pediatric BMI reference values
an additional 2–3 years of slower growth. In con- have been available for children over 2 years of
trast, boys show a smaller increase in growth age since 2000 (US Centers for Disease Control
velocity in early puberty. In fact some boys expe- (CDC) [8]) and from birth since 2006 (World
rience a decline in growth velocity for 12–18 Health Organization (WHO) [9]). These reference
months in early puberty before the growth spurt curves are necessary because, unlike in adults,
in mid-puberty. Like in girls, the growth spurt raw BMI is not meaningful in children. Normal
lasts about 2 years and is followed by 2–3 addi- BMI changes with age-related changes in body
tional years of gradual growth. proportions and body composition. Therefore,
Normal puberty: There is substantial varia- BMI is expressed as a percentile or standard devi-
tion in the age of onset of puberty in healthy ation score relative to age in healthy children [10],
children. Puberty typically begins earlier in girls allowing meaningful and consistent interpretation
than in boys, and sexual development is also of BMI regardless of age. The BMI percentile is
completed at a younger age in girls than boys. used to determine whether a child’s weight-for-
Sexual development is assessed using Tanner height can be classified into a specific “at risk”
staging (Fig. 1) [5]. For girls, breast and pubic category, including underweight, overweight, or
hair development are staged separately. For boys, obese. However, the cutoffs defining these cate-
genital (testicular volume and penile size) and gories vary somewhat depending on the source of
pubic hair development are staged separately. the reference data. A BMI-for-age less than the
Pubic hair and breast (girls)/genital (boys) staging fifth percentile represents underweight according
may be discordant. In boys, testicular volume to the US CDC [11, 12]. The WHO defines under-
should be measured using a Prader orchiometer; weight or “thinness” as a BMI-for-age less than
a testicular volume of 4 ml signals the onset of the third percentile (standard deviation score less
puberty [6]. than 2.0). It is important to recognize that these
Sexual development is considered to be are both distributional cutoffs; no evidence-based
delayed in a girl if there is no breast development definitions exist linking underweight or thinness
by 14 years, or if there is more than a 5-year to poor outcomes in the general population. The
interval between breast development and menar- CDC and the WHO agree on the definition of
che, or no menarche by 16 years. In boys, the overweight (BMI-for-age greater than the 85th
absence of testicular enlargement by 14 years or percentile or more than about 1 SD above the
an interval of more than 5 years between onset and average), but definitions of obesity differ [13].
22 Growth and Development of the Child with Renal Disease 639

Fig. 1 Stages of pubertal development for boys and girls enlargement of testes and scrotum, increasing darkening
(a–c). Stages of genital development for boys (a). (1) Pre- of scrotal skin. Stages of breast development for girls (b).
adolescent – testes, scrotum, and penis are of about the (1) Preadolescent – elevation of papilla only. (2) Breast bud
same size and proportions as in early childhood. stage – elevation of papilla as a small mound, enlargement
(2) Enlargement of scrotum and of testes – the skin of the of areolar diameter. (3) Further enlargement and elevation
scrotum reddens and changes in texture; little or no of breast and areola, with no separation of their contours.
enlargement of penis at this stage. (3) Enlargement of (4) Projection of areola and papilla to form a secondary
penis, which occurs at first mainly in length – further mound; the development of the areolar mound does not
growth of testes and scrotum. Increased size of penis with occur in all girls. (5) Mature stage – projection of papilla
growth in breadth and development of glans, further only, caused by recession of the areola to the general
640 B. Foster

The CDC defines obesity as a BMI-for-age greater distributions of height, weight, and BMI relative
than the 95th percentile (>1.64 SD above the to age, were created by merging data from the
average), whereas the WHO defines obesity as a 1977 American National Center for Health Statis-
BMI-for-age >2 SD above the average (> ~ 97th tics (NCHS)/WHO growth reference (1–24 years)
percentile). with data from the WHO <5-year old growth
Growth reference values: In 2006, the WHO standards’ cross-sectional sample (18–71 months)
released growth reference standards indicating the to smooth out the transition between the two
typical distribution of length/height, weight, and samples.
BMI relative to age for children from birth to The American CDC published revised Ameri-
5 years of age [9]; growth velocity standards are can growth reference charts for infants and chil-
also available for infants up to 24 months [14]. All dren from birth to 20 years of age in 2000 [8]. The
WHO growth charts can be found at http://www. CDC reference curves and the WHO Growth
who.int/childgrowth/en/. These growth charts are Standards differ very little after 2 years of age.
referred to as growth standards because the chil- Because the WHO Growth Standards represent
dren contributing measurements were specifically ideal growth, the WHO Growth Standards should
selected to represent children growing under ideal be used as the reference for children from birth to
conditions: they had nonsmoking mothers, were 2 years. The recommendations as to which growth
from areas of high socioeconomic status, and reference should be used after 2 years of age differ
received regular pediatric healthcare, including by country, with some countries recommending
immunizations. Longitudinal growth data, from country-specific reference curves and other coun-
birth to 24 months, were collected in a subset of tries recommending the WHO Growth Standards
882 infants, all of whom were breastfed for at least to age 5 years and the WHO Growth Reference
4 months. These longitudinal data were used to 2007 after 5 years.
generate the growth velocity standards. The refer- Growth velocity: Growth velocity, measured
ence population used to generate the WHO in cm/year, may be calculated to determine if the
Growth Standards was of broad ethnic diversity, growth rate is appropriate for age. The WHO
including children from Brazil, Ghana, India, growth velocity standards represent the best avail-
Norway, Oman, and the USA. Although there able reference values for children younger than
may be some theoretical advantages to country- 2 years; reference values are available for growth
specific growth charts, it is notable that ethnicity in 2-, 3-, 4-, and 6-month increments. Reference
had very little impact on length in the WHO values are also available for older children
population; only 3 % of the variability in length [16, 17]. These were determined by taking serial
within the population could be attributed to the measurements of healthy, growing children to
country of origin [9]. This suggests that the determine growth rates. It is important to remem-
growth standards reflect a reasonable expectation ber that the Tanner growth velocity reference
for growth regardless of ethnicity. values (Fig. 2) are based on measurements taken
The growth of children 5–19 years old is at 1-year intervals. The Baumgartner tables
represented in the WHO Growth Reference 2007 (Table 1) provide growth data at 6-month intervals
[15]. These curves, showing the typical to allow determination of growth velocity based

Fig. 1 (continued) contour of the breast. Stages of pubic resembles adult in type, but the area covered is still con-
hair development for boys and girls (a–c). (1) Preadoles- siderably smaller than in the adult; no spread to the medial
cent – the vellus over the pubes is not developed further surface of the thighs. (5) Adult in quantity and type with
than over the abdominal wall. (2) Sparse growth of long, distribution of the horizontal (classically feminine) pattern;
slightly pigmented downy hair, or only slightly curled, spread to the medial surface of thighs but not up to linea
appearing chiefly at the penis or along the labia. (3) Con- alba or elsewhere above the base of the inverse triangle
siderably darker, coarser, and more curled; the hair spreads (From (39a) Tanner J. Growth at adolescence. Oxford:
sparsely over the junction of the pubes. (4) Hair now Blackwell, 1962, with permission)
22 Growth and Development of the Child with Renal Disease 641

Fig. 2 (continued)
642 B. Foster

Fig. 2 (a) Growth velocity for age in boys 2–19 years (From [17]). (b) Growth velocity for age in girls 2–19 years (From
Ref. [17])
Table 1 Growth velocity tables in 6-month increments (Reproduced from Baumgartner et al. 1986 [16])
22

Percentiles of 6-mo increments


Age at end of interval 3 (2 SD) 5 10 (1 SD) 25 50 (mean) 75 90 (+ 1SD) 95 97 (+ 2SD) n
mo cm/6 mo
Recumbent length of boys: birth to 36 mo of age in 6-mo increment (Fels Longitudinal Study)
6 12.22 (12.00) 12.96 13.71 (14.49) 15.33 17.09 (16.98) 18.57 19.91 (19.47) 20.79 21.70 (21.96) 271
9 8.39 (819) 8.90 9.39 (9.72) 10.22 11.08 (11.25) 12.14 13.26 (12.77) 13.93 14.51 (14.30) 265
12 5.89 (5.64) 6.20 6.69 (7.06) 7.55 8.51 (8.48) 9.29 10.14 (9.90) 10.71 10.91 (11.32) 286
18 4.24 (4.03) 4.57 4.99 (5.29) 5.82 6.51 (6.56) 7.31 8.05 (7.82) 8.67 9.02 (9.08) 286
24 3.26 (2.94) 3.58 3.89 (4.17) 4.57 5.40 (5.41) 6.13 6.85 (6.64) 7.39 7.57 (7.88) 280
30 2.84 (2.73) 3.00 3.49 (3.70) 4.00 4.63 (4.67) 5.29 5.77 (5.63) 6.10 6.63 (6.60) 270
36 2.33 (2.23) 2.47 3.02 (3.18) 3.58 4.05 (4.12) 4.66 5.38 (5.07) 5.75 6.01 (6.01) 266
Recumbent length of girls: birth to 36 mo of age in 6-mo increment (Fels Longitudinal Study)
6 12.00 (11.55) 12.36 13.26 (13.84) 14.51 16.06 (16.13) 17.66 18.85 (18.42) 19.76 20.34 (20.71) 254
9 8.18 (7.92) 8.40 9.10 (9.38) 9.78 10.82 (10.83) 11.77 12.58 (12.29) 13.22 13.53 (13.74) 248
12 5.94 (5.43) 6.20 6.76 (6.99) 7.41 8.49 (8.56) 9.43 10.34 (10.12) 10.87 11.32 (11.69) 263
18 3.94 (4.17) 4.61 5.19 (5.46) 6.00 6.79 (6.76) 7.50 8.15 (8.05) 8.81 9.17 (9.35) 262
24 3.72 (3.41) 4.00 4.40 (4.57) 4.95 5.59 (5.73) 6.45 7.09 (6.89) 7.60 7.87 (8.04) 259
Growth and Development of the Child with Renal Disease

30 2.90 (2.70) 3.19 3.54 (3.71) 4.02 4.66 (4.72) 5.23 6.00 (5.74) 6.39 6.77 (6.75) 245
36 2.54 (2.29) 2.72 2.99 (3.22) 3.62 4.13 (4.16) 4.67 5.25 (5.10) 5.78 6.16 (6.04) 246
Stature of boys: 3 to 18 yr of age in 6-mo increment (Fels Longitudinal Study)
3.5 2.54 (2.22) 2.64 2.84 (3.03) 3.36 3.81 (3.84) 4.33 4.72 (4.65) 5.00 5.32 (3.46) 208
4.0 2.14 (2.09) 2.49 2.69 (2.84) 3.11 3.59 (3.60) 4.02 4.41 (4.35) 4.77 4.86 (5.11) 233
4.5 2.27 (2.21) 2.52 2.73 (2.88) 3.05 3.53 (3.55) 4.03 4.40 (4.23) 4.59 4.77 (4.89) 244
5.0 2.08 (2.03) 2.34 2.67 (2.77) 3.05 3.53 (3.50) 3.92 4.41 (4.24) 4.66 4.93 (4.97) 262
5.5 2.25 (2.15) 2.33 2.67 (2.79) 3.02 3.45 (3.43) 3.79 4.20 (4.07) 4.51 4.62 (4.71) 241
6.0 1.99 (2.00) 2.07 2.43 (2.63) 2.93 3.35 (3.30) 3.67 4.10 (3.90) 4.23 4.48 (4.53) 240
6.5 2.00 (1.92) 2.07 2.31 (2.55) 2.76 3.21 (3.19) 3.60 3.96 (3.82) 4.22 4.39 (4.45) 233
7.0 1.99 (1.99) 2.24 2.45 (2.58) 2.70 3.19 (3.17) 3.53 3.93 (3.75) 4.11 4.26 (4.34) 235
7.5 1.83 (1.72) 2.01 2.20 (2.38) 2.62 2.99 (3.03) 3.45 3.88 (3.69) 4.11 4.29 (4.34) 229
8.0 1.69 (1.69) 1.86 2.18 (2.34) 2.51 3.02 (2.98) 3.45 3.75 (3.62) 3.90 4.18 (4.27) 226
8.5 1.80 (1.82) 2.01 2.23 (2.36) 2.59 2.96 (2.90) 3.21 3.56 (3.44) 3.79 3.86 (3.98) 214
9.0 1.77 (1.73) 1.89 2.11 (2.28) 2.45 2.84 (2.83) 3.21 3.52 (3.37) 3.67 3.70 (3.92) 212
9.5 1.46 (1.43) 1.61 1.83 (2.02) 2.28 2.67 (2.62) 2.96 3.36 (3.22) 3.64 3.75 (3.82) 204
643

(continued)
644

Table 1 (continued)
Percentiles of 6-mo increments
Age at end of interval 3 (2 SD) 5 10 (1 SD) 25 50 (mean) 75 90 (+ 1SD) 95 97 (+ 2SD) n
mo cm/6 mo
10.0 1.80 (1.56) 1.89 2.06 (2.15) 2.35 2.70 (2.73) 3.06 3.44 (3.31) 3.69 3.91 (3.89) 202
10.5 1.44 (1.30) 1.56 1.83 (1.94) 2.14 2.56 (2.57) 2.93 3.38 (3.20) 3.67 3.94 (3.84) 208
11.0 1.49 (1.31) 1.68 1.87 (1.96) 2.22 2.52 (2.61) 2.96 3.43 (3.26) 3.74 4.11 (3.91) 204
11.5 1.58 (1.22) 1.69 1.86 (1.96) 2.20 2.57 (2.70) 3.06 3.70 (3.44) 4.00 4.18 (4.18) 198
12.0 1.49 (0.99) 1.60 1.86 (1.93) 2.24 2.73 (2.86) 3.27 4.07 (3.79) 4.68 5.01 (4.72) 196
12.5 1.31 (0.67) 1.49 1.83 (1.85) 2.20 2.77 (3.03) 3.53 4.93 (4.20) 5.44 5.83 (5.38) 195
13.0 1.61 (1.05) 1.87 2.10 (2.28) 2.55 3.29 (3.52) 4.45 5.22 (4.75) 5.75 6.06 (5.98) 191
13.5 1.27 (1.26) 1.55 2.19 (2.42) 2.75 3.49 (3.58) 4.41 5.03 (4.74) 5.31 5.55 (5.89) 188
14.0 1.53 (1.49) 1.84 2.15 (2.65) 2.93 4.01 (3.81) 4.64 5.21 (4.97) 5.49 5.71 (6.13) 190
14.5 1.40 (1.00) 1.51 1.75 (2.28) 2.60 3.59 (3.57) 4.54 5.26 (4.85) 5.48 5.63 (6.13) 186
15.0 0.65 (0.67) 0.90 1.42 (1.90) 2.26 3.19 (3.13) 4.01 4.66 (4.36) 5.11 5.22 (5.59) 179
15.5 0.04 (0.54) 0.32 0.66 (0.87) 1.24 2.05 (2.29) 3.26 4.28 (3.70) 4.66 4.95 (5.12) 178
16.0 0.28 (0.62) 0.12 0.60 (0.58) 0.94 1.56 (1.78) 2.48 3.57 (2.98) 3.91 4.21 (4.18) 176
16 5 0.67 (1.07) 0.50 0.03 (0.01) 0.44 0.94 (1.08) 1.50 2.44 (2.16) 2.98 3.81 (3.23) 155
17.0 0.78 (0.98) 0.55 0.16 (0.04) 0.25 0.83 (0.89) 1.33 1.94 (1.83) 2.85 3.24 (2.76) 153
17.5 0.84 (1.03) 0.66 0.37 (0.23) 0.12 0.41 (0.57) 1.00 1.43 (1.37) 1.73 2.26 (2.17) 137
18.0 0.87 (0.83) 0.55 0.33 (0.24) 0.03 0.32 (0.36) 0.72 1.01 (0.96) 1.45 1.61 (1.56) 137
Stature of girls: 3 to 18 yr of age in 6-mo increment (Fels Longitudinal Study)
3.5 2.55 (2.31) 2.84 2.98 (3.09) 3.34 3.79 (3.87) 4.33 4.84 (4.65) 5.24 5.52 (5.43) 195
4.0 2.50 (2.37) 2.72 2.91 (3.04) 3.25 3.65 (3.71) 4.09 4.49 (4.38) 4.97 5.21 (5.05) 211
4.5 2.09 (2.07) 2.23 2.71 (2.82) 3.06 3.54 (3.57) 4.05 4.46 (4.31) 4.81 4.99 (5.06) 230
5.0 2.20 (2.06) 2.33 2.52 (2.80) 3.02 3.52 (3.54) 4.03 4.36 (4.27) 4.84 4.89 (5.01) 240
5.5 2.24 (2.08) 2.42 2.57 (2.74) 2.98 3.39 (3.39) 3.79 4.16 (4.05) 4.48 4.55 (4.71) 226
6.0 2.07 (1.99) 2.23 2.46 (2.64) 2.84 3.30 (3.29) 3.75 4.10 (3.95) 4.49 4.62 (4.60) 227
B. Foster
22

6.5 1.85 (1.76) 1.93 2.20 (2.42) 2.60 3.11 (3.08) 3.49 3.94 (3.74) 4.29 4.35 (4.41) 223
7.0 1.96 (1.86) 2.11 2.36 (2.48) 2.66 3.08 (3.09) 3.50 3.81 (3.71) 3.98 4.17 (4.33) 223
7.5 1.86 (1.81) 1.98 2.23 (2.42) 2.64 3.07 (3.02) 3.39 3.67 (3.63) 4.03 4.14 (4.23) 221
8.0 1.62 (1.64) 1.82 2.17 (2.31) 2.56 3.01 (2.98) 3.35 3.85 (3.65) 4.07 4.20 (4.32) 222
8.5 1.77 (1.58) 1.86 1.99 (2.22) 2.41 2.88 (2.86) 3.27 3.68 (3.50) 3.90 4.09 (4.14) 222
9.0 1.81 (1.70) 1.93 2.13 (2.28) 2.47 2.84 (2.85) 3.22 3.53 (3.43) 3.77 4.08 (4.01) 216
9.5 1.64 (1.50) 1.78 1.95 (2.16) 2.36 2.80 (2.83) 3.29 3.58 (3.49) 3.92 4.09 (4.16) 219
10.0 1.63 (1.31) 1.70 1.99 (2.11) 2.38 2.91 (2.90) 3.28 3.83 (3.70) 4.38 4.61 (4.49) 220
10.5 1.57 (1.13) 1.64 1.99 (2.07) 2.37 2.82 (3.01) 3.56 4.21 (3.95) 4.70 5.27 (4.90) 212
11.0 1.67 (1.38) 1.87 2.08 (2.27) 2.53 3.06 (3.16) 3.74 4.33 (4.05) 4.76 4.92 (4.94) 203
11.5 1.61 (1.41) 1.84 2.09 (2.38) 2.54 3.34 (3.35) 4.02 4.63 (4.32) 4.84 5.17 (5.29) 202
12.0 1.67 (1.22) 1.60 1.98 (2.25) 2.65 3.32 (3.28) 3.98 4.52 (4.31) 4.86 4.98 (5.34) 196
12.5 0.79 (1.04) 1.11 1.61 (2.07) 2.41 3.19 (3.10) 3.87 4.38 (4.13) 4.58 4.72 (5.16) 186
13.0 0.36 (0.37) 0.52 1.05 (1.52) 1.99 2.76 (2.67) 3.46 4.09 (3.82) 4.21 4.43 (4.97) 185
13.5 0.25 (0.21) 0.35 0.57 (0.92) 1.20 1.98 (2.05) 2.90 3.56 (3.19) 3.92 4.24 (4.32) 184
14.0 0.28 (0.71) 0.09 0.28 (0.43) 0.78 1.33 (1.58) 2.35 3.11 (2.72) 3.66 4.09 (3.86) 179
14.5 0.32 (0.79) 0.21 0.04 (0.16) 0.47 0.88 (1.11) 1.65 2.77 (2.06) 2.97 3.32 (3.01) 154
15.0 0.62 (0.87) 0.47 0.16 (0.04) 0.24 0.68 (0.78) 1.24 1.75 (1.60) 2.33 2.51 (2.43) 150
Growth and Development of the Child with Renal Disease

15.5 0.88 (0.88) 0.67 0.43 (0.20) 0.07 0.49 (0.49) 0.83 1.27 (1.17) 1.62 1.91 (1.85) 143
16.0 0.53 (0.68) 0.48 0.33 (0.13) 0.04 0.46 (0.42) 0.75 1.08 (0.97) 1.42 1.54 (1.52) 139
16.5 0.66 (0.70) 0.61 0.37 (0.21) 0.05 0.23 (0.28) 0.63 0.91 (0.77) 1.02 1.19 (1.25) 132
I7.0 1.14 (0.94) 0.84 0.65 (0.38) 0.13 0.19 (0.17) 0.56 0.81 (0.73) 0.94 1.08 (1.29) 133
17.5 0.81 (0.97) 0.74 0.51 (0.41) 0.21 0.04 (0.14) 0.38 0.97 (0.70) 1.25 1.42 (1.26) 126
18.0 0.80 (0.91) 0.78 0.64 (0.39) 0.15 0.09 (0.13) 0.46 0.83 (0.65) 1.05 1.08 (1.17) 123
645
646 B. Foster

on shorter intervals. Although it is common prac- to the mother’s height and averaged with the
tice to measure height at even shorter intervals and father’s height. This average of parental heights
extrapolate, this approach should be used cau- is then plotted at age 20 years on the growth chart
tiously in children over 2 years old. Growth rates relevant for the sex of the child; the heights 8.5 cm
are not constant even over a 6-month interval; above and below the calculated midparental
briefer intervals may capture periods of slower height represent two standard deviations around
or more rapid growth than may be observed over this estimate [22].
a longer period. Nevertheless, height may be mea-
sured more frequently and plotted on the growth
curves to give a general impression of growth Bone Age
adequacy.
Special populations: Special growth curves An assessment of skeletal development will give
are available for a variety of special populations, an idea as to the remaining growth potential of the
including Trisomy 21, Turner syndrome, and pre- long bones. By convention, bone age is evaluated
mature infants [18, 19]. Diagnosis-specific curves by obtaining a left hand and wrist radiograph. This
were constructed from fairly small reference is compared with the images of “standard” hand/
populations, but provide useful guidance on wrist radiographs from children of different ages
expected growth in these unique groups. Prema- in the Greulich and Pyle skeletal age atlas
ture infants should have growth assessed relative [23]. The age of the child in the atlas’ standard
to their corrected age rather than chronological image that is closest in appearance to the child’s
age. Corrected age is determined by subtracting X-ray represents the bone age. This provides a
the number of weeks early that the child was born fairly rough estimate of bone age, since bone age
from the chronological age. Separate curves are images are available at 1-year intervals. In addi-
available for low birth weight (1,500–2,500 g) tion, discrepancies between the maturity of bones
and very low birth weight (<1,500 g) infants in the hand and the wrist may be difficult to
[20, 21]. reconcile, and interobserver variability is com-
Frequency of growth assessment: Among mon. Furthermore, the Greulich and Pyle atlas
healthy children, growth should be assessed at was generated with images from white children
each well-child health visit. In the first year of of high socioeconomic status living in the USA in
life, these are suggested to occur at birth, 1–2 the 1930s; the applicability of these standards to
weeks of age, and at 1, 2, 4, 6, 9, and 12 months. an ethnically diverse, contemporary population is
Between 1 and 3 years old, assessments are debatable [24–26].
suggested every 6 months, at 18, 24, 30, and Tanner and Whitehouse developed another
36 months. After 3 years, annual assessments are method of estimating skeletal maturation, in
sufficient. which 20 bones of the hand are scored into
Estimating height potential: When assessing one of eight categories; the scores are translated
the adequacy of growth, it is important to consider into a bone age [27]. This method has superior
the child’s height not only relative to the appro- reproducibility to the Greulich and Pyle method
priate reference group but also relative to his or and allows assessment of bone age at 0.1-year
her genetic potential. Although many factors increments. However, it is significantly more
influence growth in addition to genetic factors, a time consuming and does not show major
child’s adult height potential may be estimated advantages over the Greulich and Pyle atlas in
from the sex-adjusted midparental height. most populations. Bone age should be recognized
Midparental height is calculated from the heights as an estimate of skeletal maturity, rather than
of the biological parents. For girls, 13 cm is a precise measure, and should be interpreted in
subtracted from the father’s height and averaged the context of the sexual maturity and overall
with the mother’s height. For boys, 13 cm is added health.
22 Growth and Development of the Child with Renal Disease 647

Growth and Development in Chronic


Kidney Disease

Growth disturbance is one of the hallmarks of


CKD in childhood [28–32]. Short stature is com-
mon among children with CKD, with younger age
of onset, and more advanced CKD associated
with larger height deficits. Short stature has long
been recognized as a complication of CKD, with
30–50 % of individuals with childhood onset
CKD being reported as having height deficits as
adults [33, 34]. CKD affects each of the phases of
growth in a slightly different way.
Disruption of normal phases of growth in
CKD: During the infancy phase of growth, poor
nutritional intake may result in growth restriction.
In addition, the childhood phase of growth may be
delayed in onset until 2–3 years of age in about
40 % of boys and 27 % of girls with CKD; 60 % of
both boys and girls may show a transient resump-
tion of the infancy pattern of growth after starting
the childhood phase [4]. Because linear growth is
quite slow at the end of the infancy phase and Fig. 3 Typical growth pattern in patients with congenital
chronic kidney failure is shown in red. Loss of statural
faster at the onset of the childhood phase, delayed height predominates in infancy and adolescence, with
onset of the childhood phase or return to the growth parallel to the normal curves in mid-childhood.
infancy phase will result in overall slower growth The shaded area represents the normal range (3–97 %)
than normal. (Reproduced with permission from Mehls and Schaefer
[150])
Children with CKD growing in the childhood
phase may have normal growth velocity. How-
ever, children with lower glomerular filtration pituitary gland. In CKD, GnRH secretion may be
rate (GFR) have been shown to have significantly blunted, resulting in a decreased frequency of LH
lower growth velocity than children with higher pulses [37]. Although LH levels may be increased
GFR [29, 35]. In addition, the onset of pubertal in CKD due to decreased renal clearance, pubertal
growth is delayed among children with CKD, and patients receiving maintenance dialysis produce
the pubertal growth spurt is shorter [36]. The typ- substantially less LH overnight than healthy peers
ical growth pattern for a child with CKD is illus- [38, 39]. As a result of these abnormalities, the
trated in Fig. 3. onset of puberty may be delayed in children with
Together, the disturbances in the normal pat- CKD. However, both boys and girls with CKD do
tern of growth may lead to severe short stature. eventually progress through the normal stages of
Children with CKD from a very young age are puberty and attain full sexual maturation. Because
particularly severely affected; failure to grow at a growth and pubertal development are so tightly
normal rate during critical periods when growth is linked, it is important that pubertal development
normally rapid is very difficult to catch up later. be carefully monitored in conjunction with
Pubertal development in CKD: In healthy growth assessment.
children, gonadotropin-releasing hormone Etiology of growth impairment in CKD:
(GnRH) stimulates the release of luteinizing hor- There are a variety of reasons that children with
mone (LH) in pulsatile nocturnal bursts from the CKD experience impaired growth. As noted
648 B. Foster

above, poor caloric intake – common among The interplay between CKD-related metabolic
infants with CKD – has the most important effect bone disease and growth is complex; the effects of
on growth in the first 1–3 years of life. Anorexia calcium and phosphate metabolism and parathy-
appears to be the primary reason for growth failure roid hormone (PTH) levels on growth have not yet
in infants and very young children with CKD been elucidated. Whereas some small studies
[40–42]. Anorexia has been observed in infants showed a positive association between PTH levels
with glomerular filtration rates as high as and growth, others found that high PTH levels
70 ml/min/1.73 m2 [43]. A variety of factors may were associated with poor growth [55]. Normal
cause anorexia in CKD, including disturbances in growth has been demonstrated in children with
the appetite-regulating hormones leptin and CKD and PTH at ~1.5 times the upper limit of
ghrelin [44, 45], altered taste [46], gastroesopha- normal [56], children with PTH <2 times the
geal reflux, delayed gastric emptying, and ele- upper limit of normal [57], and even among chil-
vated levels of several cytokines such as IL-1, dren with adynamic bone disease [58]. Contro-
IL-6, and tumor necrosis factor (TNF)-α versy remains regarding the optimal target PTH
[47]. Although some older children may have level to best support growth and healthy bones.
poor energy intake, this is not common; most The very therapies given to control PTH levels –
older children have normal energy intake relative cholecalciferol and active vitamin D – have direct
to body size [31]. Developmental abnormalities inhibitory effects on the chondrocytes of the
and/or psychological factors may also contribute growth plate; both dose and dosing schedule of
to poor intake. Children with a history of anorexia these medications may influence growth [55].
may also have poor oral-motor skills, resulting in Disturbances in the growth hormone (GH) axis
very slow eating and low intake. The struggles also play an important role in the growth retarda-
around eating between parents and anorectic chil- tion associated with CKD [59]. The nature of the
dren, and the use of nasogastric tubes, may also disturbance depends somewhat on age. In puber-
result in negative psychological reactions to food tal children GH release is reduced, possibly due to
and further food refusal [48, 49]. Caloric intake diminished stimulation by sex hormones, whereas
may also be limited by frequent vomiting in CKD. in prepubertal children GH release is normal or
A large proportion of children with CKD have even increased [38, 60, 61]. Poor growth in the
poor urinary concentrating ability and therefore face of normal GH levels suggests a resistance to
high urine volumes. Such children often feel the actions of GH. GH resistance may be due to
thirsty and prefer to drink water over milk or either reduced GH receptor density or a defect in
other fluids. The volume of water taken may be signal transduction following GH binding to the
large enough to diminish their appetite for milk or receptor; either of these would result in decreased
food. Children with CKD may also experience insulin-like growth factor-1 (IGF-1) production –
salt wasting, resulting in chronic volume deple- which is normally stimulated by GH. Signal trans-
tion; this will also limit growth. Chronic meta- duction following GH binding to its receptor
bolic acidosis may also contribute to poor involves the activation of Janus kinase 2 (JAK2),
growth [50]. The mechanisms by which acidosis which then activates signal transducer and activa-
impairs growth include decreased thyroid hor- tor of transcription (STAT) proteins [62]. Acti-
mone levels, disturbances in the growth hormone vated STAT proteins upregulate transcription of
IGF axis and response to IGF-1, and promoting GH-regulated genes. The activation of JAK2 and
catabolism [22, 51, 52]. STAT proteins appears to be impaired in uremia
The role of CKD-related anemia in growth [62, 63]. Another family of proteins known as
impairment is not clear. Although some evidence SOCS (suppressor of cytokine signaling) also
suggests an association between hemoglobin level plays a role in control of GH signaling. SOCS
and growth [53], this relation may be confounded by are induced by GH and provide negative feedback
other factors. No studies have demonstrated a clear by inhibiting activation of JAK2 and STAT pro-
growth benefit with erythropoietin therapy [54]. teins [62, 64]. SOCS are also upregulated in
22 Growth and Development of the Child with Renal Disease 649

response to inflammation and may in this way the setting of CKD, PEW is the more widely used
play a role in PEW [65]. term. PEW is distinct from simple malnutrition.
Individuals with CKD also show low levels of Malnutrition is caused by inadequate intake and
circulating growth hormone-binding protein accompanied by adaptive responses, including
(GHBP), a product of proteolytic cleavage of the hunger, a protective decrease in energy expendi-
extracellular domain of the GH receptor. ture, and preferential use of fat stores for energy
Although GHBP levels are associated with with preservation of lean body mass. In contrast,
growth rate and with response to recombinant maladaptive responses prevail in PEW. PEW is
human GH therapy in children with CKD characterized by anorexia and increased meta-
[66, 67], it is not clear whether GHBP levels bolic rate; fat mass is preserved, or even increased,
are a reliable marker of GH receptor while muscle mass is reduced [72]. Although poor
expression [68]. nutritional intake may contribute, other factors
As noted above, one of the major effects of GH may be more important in the pathogenesis of
is to stimulate IGF-1 production. However, PEW in CKD; systemic inflammation and meta-
despite GH resistance, IGF-1 levels are normal bolic acidosis have been shown to play a role
in children with CKD and only slightly decreased [73–78]. Figure 4 summarizes the purported
in ESRD [67]. There is evidence of IGF-1 resis- causes and consequences of PEW in CKD. Ure-
tance in CKD. This appears to be mediated in two mic toxins are suspected as the root cause of
separate ways. First, increased levels of endocrine disturbances such as growth hormone
IGF-binding proteins in CKD will lead to a reduc- resistance and abnormal neuropeptide signaling –
tion in free (active) IGF-1. Second, signal trans- both of which are also believed to contribute to
duction following binding of IGF-1 to its receptor PEW [72, 78]. Interestingly, current growth hor-
may be impaired [69, 70]. mone use was found to be protective against skel-
etal muscle wasting in a study of children with
CKD [79]. Importantly, unlike malnutrition, in
Protein Energy Wasting in CKD which increased food intake or altered dietary
composition usually leads to rapid weight gain
The term “protein energy wasting” (PEW) is used and restoration of normal body composition, cal-
to describe the wasting syndrome seen in some orie supplements or other dietary manipulations
patients with CKD [71]. The term “cachexia” is are less successful in treating patients with PEW
also used to describe this syndrome; however, in [75, 76, 80–82].

Fig. 4 Schematic Causes of PEW Consequences of PEW


illustrating the causes and
consequences of PEW in ↓ Dietary intake
pediatric CKD. Double- Loss of kidney function
headed arrows indicate that Uremic toxins
the factor may be both a
cause and a consequence of Infection
PEW (Adapted, with
permission, from Ikizler Comorbid conditions
et al. KI 2013 [78]) Protein
Cardiovascular disease
energy
wasting
Dialysis-associated
catabolism

Metabolic derangements Poor growth (?)


(hyperparathyroidism,
metabolic acidosis,
hypogonadism, GH resistance) Inflammation
650 B. Foster

In adults with CKD, at least one of the criteria Table 2 Proposed criteria for protein energy wasting in
in each of the following categories must be met to adults and children with CKD
make a diagnosis PEW: (1) biochemical distur- Criteria for PEW in adults Modified criteria for PEW
bances, including serum albumin <38 g/L, with CKD in children with CKD
prealbumin <30 g/L (dialysis patients only), and Serum chemistry
total cholesterol <100 mg/dl; (2) low body Serum albumin <3.8 g per Serum albumin <3.8 g per
100 ml 100 ml
weight, reduced body fat, or weight loss,
Serum prealbumin Serum transferrin <140
defined by body mass index <23, unintentional (transthyretin) <30 mg per mg/dl
weight loss of 5 % over 3 months or 10 % over 100 ml (for maintenance
6 months, or total body fat percentage <10 %; dialysis patients only;
levels may vary according
(3) decreased muscle mass, defined as loss of
to GFR level for patients
muscle mass of 5 % over 3 months or 10 % with CKD stages 2–5)
over 6 months or mid-arm muscle circumference Serum cholesterol Total cholesterol <100
>10 % below the reference average, or low uri- <100 mg per 100 ml mg/dl
nary creatinine appearance [83]; and (4) low pro- C-reactive protein >3
tein or energy intake, defined as protein intake mg/L
Body mass
<0.6 g/day for at least 2 months for dialysis
BMI <23 BMI-for-height-age and
patients and <0.8 g/day for at least 2 months for
sex <5th percentile
patients with CKD stage 2–5. Unintentional weight loss Unintentional decrease in
In pediatric CKD, there is no consensus on the over time: 5 % over BMI-for-height-age and
criteria that should be used to define PEW. There 3 months or 10 % over sex percentile
are significant challenges in adapting the adult 6 months
criteria to growing children. For example, weight Total body fat percentage –
<10 %
loss is insensitive as a criterion in children who are
Muscle mass
expected to be gaining weight with growth.
Muscle wasting: reduced Mid-upper arm
Efforts have been made to adapt the adult criteria muscle mass 5 % over circumference for height-
to children, with limited success [72, 84]. The 3 months or 10 % over age and sex <5th
criteria for PEW in adults with CKD and proposed 6 months percentile
modified criteria for children are summarized in Reduced mid-arm muscle Decrease in mid-upper
circumference area arm circumference for
Table 2. (reduction >10 % in height-age and sex
There should be a sound rationale for each of relation to 50th percentile percentile
the criteria accepted as important in defining of reference population)
PEW. Ideally, the abnormalities making up the Creatinine appearance –
syndrome of PEW should predict poor clinical Dietary intake
outcomes. A clear outcome-based definition has Unintentional low DPI Self-reported poor appetite
<0.80 g kg1 day1 for at
yet to be determined for either adults or children least 2 months for dialysis
with CKD. The evidence that the proposed criteria patients or <0.6 g kg1
for PEW predict adverse outcomes is reviewed day1 for patients with
below. CKD stages 2–5
Biochemical disturbances: Hypoalbuminemia Unintentional low DEI
<25 kcal kg1 day1 for
has been consistently demonstrated to be a pre- at least 2 months
dictor of mortality in both adults [85] and children Growth
[86] treated with dialysis. However, N/A Height-for-age and sex
hypoalbuminemia is a nonspecific predictor of <3rd percentile
PEW in CKD [87–94]. Low serum albumin may Low height velocity or a
be a consequence of both volume overload and decrease in height-for-age
and sex percentile
systemic inflammation [90, 91] – both of which
are independently associated with adverse
22 Growth and Development of the Child with Renal Disease 651

outcomes. In the absence of inflammatory use different cutoffs to define underweight, and
markers, hypoalbuminemia is not predictive of neither definition has been shown to indepen-
increased mortality [94]. Although it is agreed dently predict adverse outcomes in children
that hypoalbuminemia is neither necessary nor with CKD.
sufficient for a diagnosis of PEW, it is considered Unintentional weight loss, independent of
an important biochemical indicator of PEW given absolute BMI, is a reasonable criterion for PEW
its strong association with mortality. in adults; weight loss is associated with adverse
The limitations of serum prealbumin (also outcomes [71]. However, while weight loss would
known as transthyretin) are similar to those of certainly be cause for concern, weight loss would
albumin. However, adult hemodialysis patients be an insensitive indication of PEW in growing
with prealbumin concentrations <20 mg/dl were children; weight must always be assessed relative
demonstrated to have an elevated death risk even to height in children. A crossing down of BMI
when albumin levels were normal [4]. Low serum percentiles is a more appropriate diagnostic crite-
transferrin level [95] and low serum cholesterol rion for pediatric PEW [72, 84].
have also been suggested as possible indicators of Linear growth failure has also been suggested
PEW; however, their ability to predict poor out- as an indicator or PEW in children [84, 102] and
comes is not established. There are other circulat- has been associated with a greater mortality risk in
ing inflammatory markers that are commonly CKD [103, 104]. However, poor growth may have
elevated in PEW and may predict mortality, such many causes distinct from PEW in the setting of
as C-reactive protein, and proinflammatory cyto- CKD, including inadequate caloric intake,
kines such as IL-6 [96, 97]; however, these were delayed sexual maturation, and growth hormone
not included as part of the adult diagnostic criteria insensitivity. Importantly, some of the causes of
for PEW. PEW may independently cause growth
Low body weight, weight loss, or reduced failure, even if PEW is absent. Therefore, growth
body fat: In adults, stable body weight, within a failure may have poor specificity as a criterion
certain range relative to height, is expected in for PEW.
healthy individuals. Therefore, low body weight Decreased muscle mass: Given that muscle
or weight loss may be important indicators of wasting is a central feature of PEW, reduced mus-
PEW in adults. Despite the fact that it provides cle mass is probably the single most valid criterion
little information about body composition, BMI for the presence of PEW in CKD [71]. However,
may be a particularly useful method of evaluation although muscle mass can be estimated
for PEW because BMI is strongly correlated with using techniques such as dual-energy X-ray
lean body mass at the low end of the BMI spec- absorptiometry (DXA), there is currently no prac-
trum [98]. PEW is characterized by loss of lean tical method of measuring muscle mass in the
mass; therefore low BMI may be a reasonable way clinical setting. Mid-arm muscle circumference
to identify lean mass deficits. In addition, low has been suggested as a surrogate for
BMI is associated with an elevated mortality risk muscle mass, but has not been validated for this
in both adults [99, 100] and children [101] on purpose. While indirect measures, such as
maintenance dialysis. creatinine appearance (estimated by the
BMI is expected to be fairly stable in adults; quantification of creatinine in a 24-h urine collec-
therefore the PEW criterion of BMI < 23 is log- tion and in the collected spent dialysate), may
ical in adults. In children, however, BMI must be have some value in adults with CKD [71], refer-
expressed as a percentile or Z-score relative to age ence values for creatinine appearance are not
and sex due to the expected changes in BMI with available for children, making interpretation
growth and development. The BMI percentile impossible.
cutoff below which a diagnosis of PEW should Low protein or energy intake: Poor appetite is
be considered in children with CKD is not clear. associated with an increased mortality risk in adult
As noted above, the WHO and the American CDC dialysis patients [105]. While anorexia leading to
652 B. Foster

poor energy intake is common among infants with Evaluation of Growth


CKD, both calorie and protein intake are usually and Development in CKD
appropriate for body size in older children with
CKD [31]. Nevertheless, the presence of anorexia Because of the high risk for impaired growth, and
may be a sign of PEW in some children. the unquantified (but at least theoretical) risk for
A study of children with CKD stages 2–3 in the protein energy wasting in children with CKD, it is
Chronic Kidney Disease in Children (CKiD) recommended that growth and nutritional status
cohort tested a variety of different pediatric PEW be evaluated at regular intervals. The National
definitions in predicting hospitalization rates Kidney Foundation Kidney Disease Outcomes
[84]. The criteria for PEW in that study included Quality Initiative 2008 update to the Clinical
the following: (1) biochemical, serum albumin Practice Guideline for Nutrition in Children with
<38 g/L, transferrin <140 mg/dl, total cholesterol CKD provided guidance on which measures to
<100 mg/dl, and C-reactive protein >3 mg/L; follow and at what intervals [22]. These guide-
(2) low body mass, BMI-for-height-age and sex lines were developed by a working group with
<5th percentile, or a decrease in BMI-for-height- international representation. Table 3 summarizes
age and sex percentile of 10 % over 12 months the recommendations. Recommendations differ
from an initial BMI <80th percentile; slightly for children under 3 years old versus
(3) decreased muscle mass, mid-arm muscle cir- older children, based on the different factors
cumference for height-age and sex <5th percen- influencing growth among infants compared
tile or a decrease in mid-arm muscle with older children with CKD. Methods of
circumference for height-age and sex percentile assessing each suggested parameter, and the ratio-
of 10 % over 12 months; (4) decreased appe- nale for assessing it, are detailed below.
tite, as a surrogate for low protein or energy Dietary intake: A skilled registered dietician is
intake; and (5) poor growth, height-for-age and an important member of the care team for children
sex <3rd percentile or a decrease in height-for- with CKD. Dieticians are able to assess dietary
age and sex percentile of 10 % over 12 months. intake using a variety of methods. The guidelines
PEW was alternately defined as having at least suggest that dietary intake be assessed regularly
one criterion in two of categories 1–4, in three of using a 3-day diet diary, but acknowledge that
categories 1–4, or in three of categories 1–4 plus a three 24-h recalls may be better suited to adoles-
growth criterion. Although there was a trend cents. Dietary records allow the dietician to esti-
toward a higher risk of hospitalization in children mate daily intake of calories, macronutrients
with PEW regardless of the definition used, the (carbohydrates, protein, and fat), vitamins, and
increased risk was only statistically significant minerals. Prospective 3-day dietary diaries have
when poor growth was included in the PEW def- been shown to give unbiased estimates of energy
inition. Because PEW is not yet clearly defined in intake in normal weight children under 10 years
children with CKD, it is not possible to estimate old; however, underreporting is common among
the prevalence. However, the study mentioned adolescents [106, 107], making 24-h recalls a
above noted a prevalence of 7–20 % depending potentially more suitable option in adolescents.
on the definition used; there was no clear relation- The 24-h recall method is limited by its inability
ship between the stage of CKD and prevalence to capture the day-to-day variability in dietary
of PEW. intake. A more complete evaluation of intake
Further research is needed to better understand can be made by examining three 24-h recalls,
the pathogenesis and manifestations of PEW in including 1 weekend day and 2 weekdays.
pediatric CKD. However, the concept is a useful Information on dietary intake allows the
one because it has highlighted the distinctions treating team to evaluate the adequacy of a
between simple intake-related malnutrition and patient’s intake before significant adverse changes
more complex wasting syndromes, with multiple in body composition result. This is particularly
causes. important in infants, in whom poor intake is
22 Growth and Development of the Child with Renal Disease 653

Table 3 Recommended parameters and frequency of assessment for children with CKD stages 2–5 (including dialysis).
a
Applies only to adolescents receiving hemodialysis (Reproduced from K/DOQI [22])
Minimum interval
Age 0– < 1 year Age 1–3 years Age >3 years
CKD CKD CKD CKD CKD CKD CKD CKD CKD
Measure 2–3 4–5 5D 2–3 4–5 5D CKD2 3 4–5 5D
Dietary intake 0.5–3 0.5–3 0.5–2 1–3 1–3 1–3 6–12 6 3–4 3–4
Height- or 0.5–1.5 0.5–1.5 0.5–1 1–3 1–2 1 3–6 3–6 1–3 1–3
length-for-age
percentile or SD
score
Height- or 0.5–2 0.5–2 0.5–1 1–6 1–3 1–2 6 6 6 6
length velocity-
for-age
percentile or SD
score
Estimated dry 0.5–1.5 0.5–1.5 0.25–1 1–3 1–2 0.5–1 3–6 3–6 1–3 1–3
weight and
weight-for-age
percentile or SD
score
BMI-for-height- 0.5–1.5 0.5–1.5 0.5–1 1–3 1–2 1 3–6 3–6 1–3 1–3
age percentile or
SD score
Head 0.5–1.5 0.5–1.5 0.5–1 1–3 1–2 1–2 N/A N/A N/A N/A
circumference-
for-age
percentile or SD
score
nPCR N/A N/A N/A N/A N/A N/A N/A N/A N/A 1a

expected. Nutritional supplements should be ini- used to calculate the standard deviation score (cal-
tiated without delay in infants with poor intake if culated by subtracting the child’s height from the
there is any evidence of inadequate weight gain or mean height for children of the same age and sex
poor growth. Dietary intake information is also and dividing by the standard deviation for chil-
useful to determine intake of calcium, phospho- dren of the same age and sex). A standard devia-
rus, sodium, and potassium. These minerals and tion score of zero represents an average height; a
electrolytes must be carefully controlled in indi- positive standard deviation score indicates above
viduals with CKD. average height, and a negative standard deviation
Height- or Length-for-Age Percentile or score indicates below average height.
Standard Deviation Score: Length should be Height- or Length Velocity-for-Age Percentile
measured in infants younger than 2 years using a or Standard Deviation Score: The growth veloc-
length board, and height should be measured in ity (change in height per unit time) can be deter-
older children using a wall-mounted stadiometer. mined by recording serial height measurements.
Ideally the same well-trained person should take As noted above, it is also possible to calculate
the measurement at each assessment. Stature mea- growth velocity standard deviation scores using
surements should then be plotted on the growth WHO Reference Standards in infants under
curves recommended for use in the country where 2 years or using reference data from the Fels
the child is being treated. Alternatively, or in Longitudinal Study [16] in children over 2 years
addition to the visual assessment provided by the old. A negative height velocity standard deviation
curves, reference data for healthy children can be score indicates that the child is growing at a lower
654 B. Foster

than average rate, resulting in a crossing down of isolation because it will be low in children who
percentiles. A positive height velocity standard are short for age. Therefore, weight-for-age
deviation score may indicate catch-up growth. should be interpreted in the context of the height-
The determination of height velocity standard for-age percentile or standard deviation score.
deviation score or percentile provides a quantifi- Appropriateness of weight relative to height can
able measure of relative growth velocity and can be assessed using weight-for-length percentiles in
be a useful adjunct to plotting heights on the younger children or BMI in children of all ages.
growth curves. There are some caveats to the assessment of
Estimated dry weight and weight-for-age and BMI in children with CKD due to the high prev-
body mass index-for-age percentiles or standard alence of growth retardation and delayed matura-
deviation scores: In children with predialysis tion. If BMI is expressed relative to chronological
CKD without edema, measuring weight and plot- age in a child with growth and/or maturational
ting it on weight-for-age curves are fairly simple. delay, this will result in inappropriate underesti-
In children being treated with dialysis, or children mation of his/her BMI compared to peers of sim-
with edema related to nephrotic syndrome, weight ilar height and developmental age. It has been
assessment is more challenging. The first step is to shown that this problem can be avoided by
estimate the euvolemic weight. This is generally expressing BMI relative to height-age in children
referred to as the “target dry weight,” since vol- with CKD [109]. Height-age is the age at which
ume overload is the more common abnormality in the child’s actual height would be on the 50th
CKD. However, some children with CKD may percentile. Although this approach works well
also be at risk for volume depletion due to high for most children with CKD, it may not be appro-
urine volumes (e.g., renal dysplasia, cystinosis). priate for all children. For example, when sexual
Physical findings such as edema and jugular maturation is complete, it may be more appropri-
venous distention indicate volume overload. ate to express BMI relative to chronological age.
Hypertension is also commonly a sign of fluid As in healthy children, the BMI percentile can
overload in patients on maintenance dialysis be used to classify children with CKD as under-
[108]. Hypoalbuminemia may also be a marker weight, normal weight, overweight, or obese.
for fluid overload. In contrast, thirst, tachycardia, However, it must be recognized that the risk asso-
decreased skin turgor, and sunken eyes and/or ciated with any of these categories in a CKD
fontanelle signify volume depletion. In mainte- population remains controversial. Although obe-
nance dialysis patients, response to dialytic fluid sity and overweight have been associated with
removal (hypotension, cramping) may also give adverse outcomes in otherwise healthy adults,
clues as to the volume status of the patient. Esti- the ability of these categories to predict poor out-
mation of target dry weight is difficult, particu- comes in the CKD population is unknown. Large
larly in growing children, in whom weight is studies of adult hemodialysis patients showed an
expected to increase over time. Errors in the esti- inverse relationship between BMI and mortality
mation of target dry weight may have an impor- risk. These studies found no clear BMI threshold
tant impact on the assessment of the above which the mortality risk stabilized or
appropriateness of body weight relative to height. began to increase, even as BMI increased above
For example, an overestimation of target dry 30 [100, 110]. A smaller study, also of adult
weight representing 10 % of the body weight hemodialysis patients, found a U-shaped relation-
will result in a BMI standard deviation score ship between BMI and mortality risk, with higher
about 0.5–1.0 SD units above what it would be mortality risk among those with BMI <17 or >23
at true target dry weight. compared to those with BMI between 17.0 and
Once the target dry weight has been estimated, 18.9 [111]. A similar U-shaped association has
weight should be plotted on the weight-for-age been seen in children with stage 5 CKD: mortality
curves and the standard deviation score calcu- risk was lowest among children with a BMI SD
lated. Weight-for-age cannot be interpreted in score around +0.5. A 1.0 SD unit difference in
22 Growth and Development of the Child with Renal Disease 655

BMI SD score in either direction was associated in kg), V2 = predialysis total body water in deci-
with a 6 % higher risk of mortality [101] – which liters (5.8 predialysis weight in kg), and t = time
represents a fairly small effect. All of these studies (min) from the end of the dialysis treatment to the
can only conclude an association between BMI beginning of the following treatment. Adult stud-
and outcome; a causal relationship has not been ies suggest that pre- and postdialysis BUN levels
established. Nonetheless, BMI, interpreted in the from the same treatment can be used to calculate
overall clinical context, can provide useful infor- nPCR [118].
mation to clinicians caring for children with CKD. The value of nPCR in children with CKD –
Head circumference-for-age percentile or SD beyond as a means of estimating protein intake
score: Like for healthy children 3 years of age, – is not clear. Higher nPCR was shown to be
head circumference should be measured regularly associated with subsequent weight gain and
and plotted on the head circumference-for-age lower nPCR with future weight loss among ado-
curves in children 3 years old with CKD. The lescents treated with dialysis [119, 120]. It is
WHO Growth Standards are the most appropriate suggested that nPCR level be targeted to the
reference curves [112]. Infants with CKD have age-specific recommended protein intake.
been shown to be at risk for poor head growth – Frequency of growth and nutritional assess-
which reflects poor brain growth [113, 114]. Head ment: Given the high risk among children with
circumference assessment should be coupled with CKD for disturbances in growth and nutritional
intermittent developmental assessments in this status, it is suggested that these parameters be
population. assessed at least twice as frequently as in healthy
Normalized protein catabolic rate (nPCR): children and more often with severe CKD and/or
The protein catabolic rate (PCR) provides an when there is an ongoing concern.
objective measure of dietary protein intake in
patients receiving maintenance hemodialysis.
When PCR is expressed normalized for the Optimizing Growth in Children
patient’s weight, it is referred to as the normalized with CKD
PCR (nPCR). Although nPCR was originally cal-
culated using formal urea kinetic modeling [115], Why is growth important? Growth may be one
simpler algebraic formulas appear to provide of the most important outcomes in pediatric CKD.
almost identical results to more complex model- There are two main reasons that growth is so
ing [116]. The nPCR can be calculated using the important. First, growth is an excellent index of
modified Borah equation, where G represents the overall health. Second, stature has important
urea generation rate in mg/min and V1 represents effects on self-esteem and perceived quality
the total body water at the end of dialysis in liters of life.
(0.58 weight in kg) [117]: Large retrospective cohort studies of children
with ESRD showed a strong association between
nPCR ¼ 5:43 G=V1 þ 0:17 stature and risk for morbidity and mortality [103,
104]. A study of children recorded in the United
The urea generation rate is determined from the States Renal Data Systems (USRDS) Pediatric
rise in blood urea nitrogen (BUN) from the end of Growth and Development Study compared hospi-
one HD treatment to the beginning of the next talization rates and mortality rates over 5 years of
treatment and is calculated as follow-up among children with moderate or
severe growth failure, defined as a height velocity
Gðmg=minÞ ¼ ½ðC2 V2 Þ  ðC1 V1 Þ=t SD score calculated over a 1-year interval of 2.0
to 3.0 (moderate) or < 3.0 (severe)
where C1 = postdialysis BUN (mg/dl), C2 = [103]. Compared with children with height veloc-
predialysis BUN (mg/dl), V1 = postdialysis total ity SD scores of > 2.0, those with moderate
body water in deciliters (5.8 postdialysis weight growth failure had a relative risk (RR) for
656 B. Foster

hospitalization over the next 5 years of 1.24 (95 % associations between better physical and social
confidence interval (CI) 1.2, 1.3) and those with functioning and both of catch-up growth and treat-
severe growth failure a RR of 1.14 (95 % CI 1.1, ment with growth hormone [124].
1.2), after adjustment for age, gender, race, cause Because of the important associations between
and duration of ESRD, and treatment modality growth and morbidity, mortality, and quality of
(dialysis or transplant). Similarly, moderate and life, efforts should be made to optimize growth in
severe growth failure were associated with a sig- children with CKD. Optimal growth may also be
nificantly higher risk of death (RR 2.01 (95 % CI especially important in infants with CKD who
1.1, 3.6) for moderate and 2.9 (95 % CI 1.6, 5.3) must reach a specific body size before being eli-
for severe) [77]. A study from the North American gible for a kidney transplant. Growth can be opti-
Pediatric Renal Trials and Collaborative Studies mized by systematically addressing each of the
registry had similar findings [104]. Children with potential contributors to growth failure in the
a height-for-age SD scores of < 2.5 at the time pediatric CKD population.
of dialysis initiation had significantly more hospi-
tal days per month of dialysis than patients with
height-for-age SD score > 2.5 [78]. Height-for- Nutrition
age SD score < 2.5 was also associated with a
two times higher risk of death, after adjustment for Adequate energy intake is necessary for normal
age, sex, race, primary renal disease, dialysis growth. Assessment of dietary intake combined
modality, and whether the patient was listed for with examination of weight and height trajectories
kidney transplant. These studies should not be will help determine whether poor energy intake
interpreted as showing that short stature causes may be contributing to poor growth. Children
morbidity or death. Rather, they show that growth with CKD should take in 100 % of the estimated
failure is a marker for illness severity. Efforts energy requirement based on sex, age, weight,
should be directed toward the root causes of height, and physical activity level [22]. The equa-
growth failure in order to improve outcomes. tions used to calculate estimated energy require-
Short stature is highly visible and may be one ment are provided in Table 4 and physical activity
of the most obvious features distinguishing indi- coefficients in Table 5. Children experiencing
viduals with childhood onset CKD from their inadequate growth who have insufficient energy
healthy peers. Although existing studies of adult intake will need dietary supplements. The vast
survivors of pediatric CKD cannot draw causal majority of infants <2 years old with moderate
links between stature and quality of life, associa- to severe CKD will be unable to take sufficient
tions were quite strong [121–123]. Among calories by mouth and will need tube feeding. It is
244 adults who had received a kidney transplant important to intervene preemptively, before
in childhood, greater final adult height was asso- stunting is evident; height potential lost in infancy
ciated with a significantly higher likelihood of is very difficult to catch up later. Infants in whom
being married or living outside of the parents tube feedings are started before major height def-
home [121]. Short stature, defined as a final icits are noted have superior final adult height
adult height >2.5 standard deviations below the [125]. Supplemental tube feedings should be con-
mean, was also associated with a lower likelihood sidered for any infant failing to meet the energy or
of full-time employment [121]. In a small study of fluid requirements for normal growth [22].
39 adults with childhood onset ESRD, 36 % Tube feedings can be delivered either via a
reported dissatisfaction with their height com- nasogastric tube or via a gastrostomy tube.
pared with 4 % of healthy age-matched controls; Gastrostomy feeding has cosmetic advantages
greater satisfaction with adult height was signifi- (hidden under clothing) and avoids negative oro-
cantly positively correlated with a better per- pharyngeal stimulation associated with chronic
ceived quality of life [123]. A study of presence of and frequent replacement of an NG
433 children with CKD found significant tube [126]. The unpleasant stimulation of the
22 Growth and Development of the Child with Renal Disease 657

oropharynx associated with nasogastric tube use In infants weighing less than 4 kg, nasogastric
may exacerbate oral feeding aversion and contrib- feeding is the route of choice [126].
ute to poor development of oral-motor skills In contrast to infants and very young children,
[126]. In addition, a gastrostomy avoids the poten- in whom the growth benefits of tube feeding are
tial complications associated with nasogastric well established [42, 127, 128], there is little evi-
tubes such as otitis, sinusitis, pulmonary aspira- dence supporting the use of supplemental tube
tion, exacerbation of gastroesophageal reflux, and feeding to promote growth in older children with
nasoseptal erosion [126]. Despite the advantages CKD. In fact, there is some evidence that poor
of gastrostomy feeding, nasogastric tube feeding intake in older children with CKD may be a con-
still has a place in the nutritional care of children sequence of poor growth, rather than the cause:
with CKD, especially when tube feedings are spontaneous calorie intake increased by almost
expected to be needed for only a short time. 12 % during treatment with recombinant human
growth hormone in a study of 33 children with
Table 4 Equations to estimate energy requirements for CKD [129]. Nevertheless, selected older children
children (Reproduced from K/DOQI [22]). PA represents may need, and benefit from, tube feeding. Older
the physical activity coefficient children treated with nutritional supplements
Estimated energy requirement (EER) should be observed closely for evidence of a
(Kcal/day) = total energy expenditure + response in the form of improved growth. Thera-
Age energy deposition
pies of proven benefit, such as growth hormone,
0–3 EER = [89 X weight (kg) 100] + 175
months should not be unduly delayed by prolonged trials
4–6 EER = [89 X weight (kg) 100] + 56 of calorie supplementation [59].
months In addition to energy intake, the protein intake
7–12 EER = [89 X weight (kg) 100] + 22 of children with CKD should also be monitored
months and controlled. While children with CKD tend to
13–35 EER = [89 X weight (kg) 100] + 20 spontaneously consume less protein than their
months
healthy counterparts, it is rare that spontaneous
3–8 years Boys: EER = 88.5–61.9 X age (y) + PA X
[26.7 X weight (kg) +903 X height (m)] + protein intake does not meet requirements. Rather,
20 protein intake among children with CKD is often
Girls: EER = 135.3–30.8 X age (y) + PA X in the range of 1.5–2 times the recommended
[10 X weight (kg) +934 X height (m)] + 20 dietary allowance [130–132]. In view of the
9–19 Boys: EER = 88.5–61.9 X age (y) + PA X
important correlations between protein intake
years [26.7 X weight (kg) + 903 X height (m)] +
25 and phosphorus intake, and the negative effects
Girls: EER = 135.3–30.8 X age (y) + PA X of phosphorus overload on cardiovascular health,
[10 X weight (kg) + 934 X height (m)] + 25 the current recommendations are to maintain

Table 5 Physical activity coefficients for the determination of energy requirements in children 3–18 years of age (From
Health Canada: http://www.hc-sc.gc.ca/fn-an/alt_formats/hpfb-dgpsa/pdf/nutrition/dri_tables-eng.pdf)
Level of physical activity
Sedentary Low active Active Very active
ADL +
Typical ADL +30–60 min of 60 min of ADL + 60 min of daily moderate
activities of daily moderate activity daily activity + an additional 60 min of
daily living (e.g., walking at 5–7 moderate vigorous activity or 120 min of moderate
Gender (ADL) only km/h) activity activity
Boys 1.0 1.13 1.26 1.42
Girls 1.0 1.16 1.31 1.56
658 B. Foster

Table 6 Recommended dietary protein intake in children with CKD stages 3–5 and 5D (dialysis) (Reproduced from
K/DOQI [22])
Dietary reference intake (DRI)
DRI Recommended for CKD Recommended for CKD Recommended Recommended for
(g/kg/ stage 3 (g/kg/day) stages 4–5 (g/kg/day) for hemodialysis peritoneal dialysis
Age day) (100–140 % DRI) (100–120 % DRI) (g/kg/day)a (g/kg/day)b
0–6 1.5 1.5–2.1 1.5–1.8 1.6 1.8
months
7–12 1.2 1.2–1.7 1.2–1.5 1.3 1.5
months
1–3 1.05 1.05–1.5 1.05–1.25 1.15 1.3
years
4–13 0.95 0.95–1.35 0.95–1.15 1.05 1.1
years
14–19 0.85 0.85–1.2 0.85–1.05 0.95 1.0
years
a
DRI + 0.1 g/kg/day to compensate for dialytic losses
b
DRI + 0.15–0.3 g/kg/day depending on patient age to compensate for peritoneal losses

protein intake at 100–140 % of the dietary refer- sodium intakes of 2–4 mEq per 100 ml of fluid are
ence intake for children with CKD stage 3 and at suggested for infants with polyuric CKD [125].
100–120 % for those with CKD stages 4–5
(including 5D – dialysis) [22]. Low protein
diets are not recommended. The recommended Acidosis Control
dietary protein intake by age is summarized in
Table 6. Current recommendations suggest that the bicar-
bonate level be corrected to at least 22 mmol/L
[22]. This can be achieved using oral supplements
Fluid and Sodium of sodium bicarbonate or sodium citrate. Citrate
should be avoided in the rare situation where
Many children with CKD, including those with aluminum-based phosphate binders are being
renal dysplasia, obstructive uropathy, used due to enhanced aluminum absorption in
nephronophthisis, and cystinosis, are polyuric. the presence of citrate. For children being treated
When urine volumes are high, there may also be with hemodialysis, acidosis may also be corrected
substantial sodium losses. Infants on peritoneal by a higher bicarbonate concentration in the bath
dialysis may also experience large sodium losses or by an increased dose of dialysis.
through the dialysate [133]. Chronic sodium and
water losses may result in significant volume
depletion, which will impair linear growth [134] Control of Hyperparathyroidism
and may even result in neurologic injury and Metabolic Bone Disease
[22]. When sodium losses are significant, sodium
supplements are required to support optimal The metabolic bone disease associated with CKD
growth [40, 41, 125, 135]. In infants with polyuric (formerly referred to as renal osteodystrophy) has
CKD, the volume of fluid and doses of sodium a negative impact on growth. Management of
needed to promote adequate growth can rarely be CKD-related metabolic bone disease is dealt
taken orally. Tube feeding facilitates delivery of with in ▶ Chap. 66, “Management of Chronic
adequate sodium and fluid, as well as calories Kidney Disease in Children” of this book. The
[125]. Fluid volumes of 180–240 ml/kg/day and main components of therapy include dietary
22 Growth and Development of the Child with Renal Disease 659

phosphate restriction, the use of intestinal phos- SDS; the most pronounced effects are seen in the
phate binders to limit phosphate absorption, and first year of treatment [139]. Importantly, the fre-
active vitamin D therapy. quency of adverse effects did not differ between
rhGH-treated subjects and the control group
[139]. Although prepubertal children with CKD
Recombinant Human Growth Hormone treated with rhGH have been shown to achieve
Therapy normal final adult height [59], the impact of rhGH
treatment on final height in pubertal patients is
Recombinant human growth hormone (rhGH) less clear. While some studies showed a good
therapy has become the standard of care for chil- growth response to rhGH in pubertal patients,
dren with CKD with evidence of growth failure. without advancing bone maturation beyond that
Numerous studies have demonstrated its safety expected [136], most studies suggested a more
and effectiveness [136–138]. A Cochrane review vigorous response in prepubertal than pubertal
including 15 randomized control trials of rhGH children [59, 139–141]. Most agree that existing
treatment in children with predialysis CKD, those evidence supports early initiation of rhGH to opti-
on dialysis, and with kidney transplants con- mize final height. Unfortunately rhGH remains an
cluded that rhGH treatment significantly improves underutilized therapy [142]. A consensus state-
height SDS, height velocity, and height velocity ment, published in 2006, provided guidance on

Consider GH therapy in patients with CKD stage 2-5 and 5D AND height SDS <-
1.88 or height-for-age <3rd percentile

Assess and treat complicating factors for poor growth including: acidosis, inadequate calorie intake, salt-
wasting, metabolic bone disease, and hypothyroidism

No Is growth velocity improved? Yes

Perform baseline assessments for GH therapy:


Continue current therapy
(a) Recalculate height SDS, height velocity SDS, and height velocity
(b) Assess pubertal stage, bone age, hip and knee X-rays, fundoscopic
exam, chemistries, PTH, and thyroid studies

Start GH therapy:
0.05 mg/kg/day SC (0.35 mg/kg/week)

Monitor during GH therapy:


(a) Every 3-4 months: height, weight, head
circumference (until 3 years old), pubertal stage,
nutritional evaluation, fundoscopic exam,
chemistrics, PTH, and toxicity
(b) Every year: Bone age
(c) For symptoms: hip and knee X-rays

Is growth adequate? (i.e. growth Yes


No
velocity 2cm/yr> baseline)

Assess and correct: Is growth Continue GH therapy and adjust dose


adequate? every 3-4 months based on weight
(a) Dose for weight Discontinue GH therapy in the following circumstances:
(b) Metabolic status (a) Achieved height goal based on mid-parental
(c) Nutrition height or 50th percentile for age

(d) Adherence (b) Closed epiphyses


(c) Active neoplasia
(d) Slipped femoral epiphyses
(e) Intracranial hypertension
If height velocity remains <2 cm/yr
Consider pediatric (f) Nonadherence over baseline and reason for
No
endocrine consult discontinuation resolved, consider
(g) Severe hyperparathyroidism per CKD stage:
reinitiation of GH therapy
Stage 2-4: PTH>400 pg/ml
Stage 5: PTH>900 pg/ml

Fig. 5 Proposed algorithm for evaluation and treatment of growth failure in children with CKD (Modified with
permission from Mahan et al. 2006 [137])
660 B. Foster

the evaluation and treatment of growth failure in difference in growth outcomes at 3 years
children with CKD in the form of a practical posttransplant [149]. Therefore, other reasons for
algorithm [137] (Fig. 5). growth failure besides steroid use should be
sought, and treated, before considering reduction
or discontinuation of steroids.
Enhanced Dialytic Clearance

The first evidence that enhanced dialytic clearance


Summary
may promote better growth in children was a 1999
study of 12 children 7 months to 14 years of age
Kidney disease can have a profound effect on
being treated with hemodialysis [143]. By provid-
growth and development. In the past, a normal
ing enhanced clearance (average Kt/V of 2.0 per
final adult height was out of reach for the majority
treatment; average of 14.8 h per week of dialysis),
of children with CKD. However, much has been
these children were able to achieve catch-up
learned about the causes of growth failure in pedi-
growth, without growth hormone therapy. Fol-
atric CKD, and effective treatments are readily
lowing that study, others have also demonstrated
available. It is now a realistic goal to have children
improved growth with intensified dialysis therapy
with CKD reach (or at least come close to) their
[144]. A study of 15 children (median age 8 years;
full height potential. This requires close monitor-
interquartile range 6–10 years) treated with
ing by a multidisciplinary team, a strong commit-
hemodiafiltration 3 h per day, 6 days per week,
ment by the family, and early intervention to
achieving an average single treatment Kt/Vof 1.4,
prevent growth deficits.
showed an increase in height velocity to 14.3 
3.8 cm/year during intensified therapy compared
with 3.8  1.1 cm/year with conventional dialysis
doses [145]. There are no randomized trials of the References
impact of enhanced clearance on growth. How-
ever, the existing evidence is quite compelling. 1. Karlberg J. On the construction of the infancy-
Intensified dialysis should be considered if growth childhood-puberty growth standard. Acta Paediatr
cannot be optimized using other means of improv- Scand Suppl. 1989;356:26–37.
2. Karlberg J, Engstrom I, Karlberg P, Fryer
ing growth. JG. Analysis of linear growth using a mathematical
model. I. From birth to three years. Acta Paediatr
Scand. 1987;76(3):478–88.
Minimized Exposure to Corticosteroids 3. Karlberg J, Fryer JG, Engstrom I, Karlberg
P. Analysis of linear growth using a mathematical
model. II. From 3 to 21 years of age. Acta Paediatr
Many children with CKD are treated with cortico- Scand Suppl. 1987;337:12–29.
steroids, either for an underlying condition or fol- 4. Karlberg J, Schaefer F, Hennicke M, Wingen AM,
lowing a kidney transplant. The growth Rigden S, Mehls O. Early age-dependent growth
impairment in chronic renal failure. European Study
suppressing effects of corticosteroids are well Group for Nutritional Treatment of Chronic Renal
known [146, 147]. When growth is inadequate in Failure in Childhood. Pediatr Nephrol. 1996;10
a child with CKD who is being treated with corti- (3):283–7.
costeroids, efforts should be made to reduce the 5. Tanner JM. Growth at adolescence. Oxford: Black-
well Scientific; 1962.
dose or discontinue corticosteroid therapy if possi- 6. Prader A. Testicular size: assessment and clinical
ble. An observational study of steroid minimization importance. Triangle. 1966;7(6):240–3.
after renal transplantation, by early withdrawal, 7. Blondell RD, Foster MB, Dave KC. Disorders of
appeared to result in improved growth outcomes puberty. Am Fam Physician. 1999;60(1):209–18.
223–4.
in pediatric transplant recipients [148]. However, a 8. Kuczmarski RJ, Ogden CL, Grummer-Strawn LM,
randomized trial of steroid-based immunosuppres- Flegal KM, Guo SS, Wei R, et al. CDC growth charts:
sion versus complete steroid avoidance showed no United States. Adv Data. 2000;314:1–17.
22 Growth and Development of the Child with Renal Disease 661

9. World Health Organization. WHO child growth stan- Greulich and Pyle skeletal age standards to black
dards: length/height-for-age, weight-for-age, weight- and white children of today. Am J Dis Child.
for-length, weight-for-height, and body mass index- 1993;147(12):1329–33.
for-age. Methods and development. Geneva: World 25. Mora S, Boechat MI, Pietka E, Huang HK, Gilsanz
Health Organization; 2006. V. Skeletal age determinations in children of European
10. Kuczmarski RJ, Ogden CL, Guo SS. 2000 CDC and African descent: applicability of the Greulich and
growth charts for the United States: methods and Pyle standards. Pediatr Res. 2001;50(5):624–8.
development. Vital Health Stat. 2002;11(246): 26. Koc A, Karaoglanoglu M, Erdogan M, Kosecik M,
127–36. Cesur Y. Assessment of bone ages: is the Greulich-
11. Centers for Disease Control. Recommended BMI-for- Pyle method sufficient for Turkish boys? Pediatr Int.
age cut-offs. http://www.cdc.gov/nccdphp/dnpa/ 2001;43(6):662–5.
growthcharts/training/modules/module1/text/page5a. 27. Tanner J, Whitehouse R, Cameron N. Assessment of
htm Accessed 30 June 2014. skeletal maturity and prediction of adult height (TW2
12. World Health Organization. Physical status: the use method). 2nd ed. London: Academic; 1983.
and interpretation of anthropometry. Geneva: World 28. Abitbol CL, Warady BA, Massie MD, Baluarte HJ,
Health Organization; 1995. Report No. 854 Fleischman LE, Geary DF, et al. Linear growth and
13. Barlow SE. Expert committee recommendations anthropometric and nutritional measurements in chil-
regarding the prevention, assessment, and treatment dren with mild to moderate renal insufficiency: a
of child and adolescent overweight and obesity: sum- report of the growth failure in children with renal
mary report. Pediatrics. 2007;120 Suppl 4:S164–92. diseases study. J Pediatr. 1990;116(2):S46–54.
14. World Health Organization Multicentre Growth Ref- 29. Betts PR, Magrath G. Growth pattern and dietary
erence Study Group. WHO child growth standards: intake of children with chronic renal insufficiency.
growth velocity based on weight, length and head Br Med J. 1974;2(912):189–93.
circumference: methods and development. Geneva: 30. Arnold WC, Danford D, Holliday MA. Effects of
World Health Organization; 2009. caloric supplementation on growth in children with
15. de Onis M, Onyango A, Borghi E, Siyam A, Nishida uremia. Kidney Int. 1983;24(2):205–9.
CJS. Development of a WHO growth reference for 31. Norman LJ, Coleman JE, Macdonald IA, Tomsett
school-aged children and adolescents. Bull World AM, Watson AR. Nutrition and growth in relation to
Health Organ. 2007;85:660–7. severity of renal disease in children. Pediatr Nephrol.
16. Baumgartner RN, Roche AF, Himes JH. Incremental 2000;15(3–4):259–65.
growth tables: supplementary to previously published 32. Salusky IB, Fine RN, Nelson P, Blumenkrantz MJ,
charts. Am J Clin Nutr. 1986;43(5):711–22. Kopple JD. Nutritional status of children undergoing
17. Tanner JM, Davies PS. Clinical longitudinal stan- continuous ambulatory peritoneal dialysis. Am J Clin
dards for height and height velocity for North Amer- Nutr. 1983;38:599–611.
ican children. J Pediatr. 1985;107(3):317–29. 33. Offner G, Latta K, Hoyer PF, Baum HJ, Ehrich JH,
18. Cronk C, Crocker AC, Pueschel SM, Shea AM, Pichlmayr R, et al. Kidney transplanted children come
Zackai E, Pickens G, et al. Growth charts for children of age. Kidney Int. 1999;55(4):1509–17.
with down syndrome: 1 month to 18 years of age. 34. Hokken-Koelega AC, van Zaal MA, van Bergen W,
Pediatrics. 1988;81(1):102–10. de Ridder MA, Stijnen T, Wolff ED, et al. Final height
19. Frias JL, Davenport ML. Health supervision for chil- and its predictive factors after renal transplantation in
dren with turner syndrome. Pediatrics. 2003;111 childhood. Pediatr Res. 1994;36(3):323–8.
(3):692–702. 35. Schaefer F, Wingen AM, Hennicke M, Rigden S,
20. Guo SS, Wholihan K, Roche AF, Chumlea WC, Mehls O. Growth charts for prepubertal children
Casey PH. Weight-for-length reference data for pre- with chronic renal failure due to congenital renal
term, low-birth-weight infants. Arch Pediatr Adolesc disorders. European Study Group for Nutritional
Med. 1996;150(9):964–70. Treatment of Chronic Renal Failure in Childhood.
21. Guo SS, Roche AF, Chumlea WC, Casey PH, Moore Pediatr Nephrol. 1996;10(3):288–93.
WM. Growth in weight, recumbent length, and head 36. Schaefer F, Seidel C, Binding A, Gasser T, Largo RH,
circumference for preterm low-birthweight infants Prader A, et al. Pubertal growth in chronic renal
during the first three years of life using gestation- failure. Pediatr Res. 1990;28(1):5–10.
adjusted ages. Early Hum Dev. 1997;47(3):305–25. 37. Daschner M, Philippin B, Nguyen T, Wiesner RJ,
22. K/DOQI. KDOQI clinical practice guideline for nutrition Walz C, Oh J, et al. Circulating inhibitor of gonado-
in children with CKD: 2008 update. Executive summary. tropin releasing hormone secretion by hypothalamic
Am J Kidney Dis. 2009;53(3 Suppl 2):S11–104. neurons in uremia. Kidney Int. 2002;62(5):1582–90.
23. Greulich W, Pyle S. Radiographic atlas of skeletal 38. Feneberg R, Schaefer F, Veldhuis JD. Neuroendocrine
development of the hand and wrist. Stanford: adaptations in renal disease. Pediatr Nephrol. 2003;18
Stanford University Press; 1959. (6):492–7.
24. Loder RT, Estle DT, Morrison K, Eggleston D, Fish 39. Schaefer F, Veldhuis JD, Robertson WR, Dunger D,
DN, Greenfield ML, et al. Applicability of the Scharer K. Immunoreactive and bioactive luteinizing
662 B. Foster

hormone in pubertal patients with chronic renal fail- 55. Waller S. Parathyroid hormone and growth in chronic
ure. Cooperative Study Group on Pubertal Develop- kidney disease. Pediatr Nephrol. 2011;26(2):
ment in Chronic Renal Failure. Kidney Int. 1994;45 195–204.
(5):1465–76. 56. Cansick J, Waller S, Ridout D, Rees L. Growth and
40. Claris-Appiani A, Ardissino GL, Dacco V, Funari C, PTH in prepubertal children on long-term dialysis.
Terzi F. Catch-up growth in children with Pediatr Nephrol. 2007;22(9):1349–54.
chronic renal failure treated with long-term enteral 57. Waller S, Ledermann S, Trompeter R, van’t Hoff W,
nutrition. JPEN J Parenter Enteral Nutr. 1995;19 Ridout D, Rees L. Catch-up growth with normal
(3):175–8. parathyroid hormone levels in chronic renal failure.
41. Kari JA, Gonzalez C, Ledermann SE, Shaw V, Rees Pediatr Nephrol. 2003;18(12):1236–41.
L. Outcome and growth of infants with severe chronic 58. Waller S, Shroff R, Freemont AJ, Rees L. Bone
renal failure. Kidney Int. 2000;57(4):1681–7. histomorphometry in children prior to commencing
42. Ledermann SE, Shaw V, Trompeter RS. Long-term renal replacement therapy. Pediatr Nephrol. 2008;
enteral nutrition in infants and young children with 23(9):1523–9.
chronic renal failure. Pediatr Nephrol. 1999;13(9): 59. Haffner D, Schaefer F, Nissel R, Wuhl E, Tonshoff B,
870–5. Mehls O. Effect of growth hormone treatment on the
43. Abitbol CL, Zilleruelo G, Montane B, Strauss adult height of children with chronic renal failure.
J. Growth of uremic infants on forced feeding regi- German Study Group for Growth Hormone Treat-
mens. Pediatr Nephrol. 1993;7(2):173–7. ment in Chronic Renal Failure. N Engl J Med.
44. Mak RH, Cheung W, Cone RD, Marks DL. Leptin 2000;343(13):923–30.
and inflammation-associated cachexia in chronic kid- 60. Schaefer F, Baumann G, Haffner D, Faunt LM, John-
ney disease. Kidney Int. 2006;69(5):794–7. son ML, Mercado M, et al. Multifactorial control of
45. Mak RH, Cheung W, Purnell J. Ghrelin in chronic the elimination kinetics of unbound (free) growth
kidney disease: too much or too little? Perit Dial Int. hormone (GH) in the human: regulation by age, adi-
2007;27(1):51–5. posity, renal function, and steady state concentrations
46. Bellisle F, Dartois AM, Kleinknecht C, Broyer of GH in plasma. J Clin Endocrinol Metab.
M. Alteration of the taste for sugar in renal insuffi- 1996;81(1):22–31.
ciency: study in the child. Nephrologie. 1995; 61. Tonshoff B, Veldhuis JD, Heinrich U, Mehls
16(2):203–8. O. Deconvolution analysis of spontaneous nocturnal
47. Cheung WW, Paik KH, Mak RH. Inflammation and growth hormone secretion in prepubertal children
cachexia in chronic kidney disease. Pediatr Nephrol. with preterminal chronic renal failure and with
2010;25(4):711–24. end-stage renal disease. Pediatr Res. 1995;37(1):
48. Coleman JE, Warady BA. Supplemental tube feeding. 86–93.
In: Warady BA, Schaefer F, Fine RN, Alexander SR, 62. Rabkin R, Sun DF, Chen Y, Tan J, Schaefer F. Growth
editors. Pediatric dialysis. Dordrecht: Kleuwer; 2004. hormone resistance in uremia, a role for impaired
p. 243–57. JAK/STAT signaling. Pediatr Nephrol. 2005;20(3):
49. Dello Strologo L, Principato F, Sinibaldi D, Appiani 313–8.
AC, Terzi F, Dartois AM, et al. Feeding dysfunction 63. Schaefer F, Chen Y, Tsao T, Nouri P, Rabkin
in infants with severe chronic renal failure after long- R. Impaired JAK-STAT signal transduction contrib-
term nasogastric tube feeding. Pediatr Nephrol. utes to growth hormone resistance in chronic uremia.
1997;11(1):84–6. J Clin Invest. 2001;108(3):467–75.
50. Donckerwolcke R, Yang WN, Chan JC. Growth fail- 64. Wormald S, Hilton DJ. Inhibitors of cytokine signal
ure in children with renal tubular acidosis. Semin transduction. J Biol Chem. 2004;279(2):821–4.
Nephrol. 1989;9(1):72–4. 65. Kaysen GA. The microinflammatory state in uremia:
51. Kuizon BD, Salusky IB. Growth retardation in chil- causes and potential consequences. J Am Soc
dren with chronic renal failure. J Bone Miner Res. Nephrol. 2001;12(7):1549–57.
1999;14(10):1680–90. 66. Postel-Vinay MC, Tar A, Crosnier H, Broyer M,
52. Boirie Y, Broyer M, Gagnadoux MF, Niaudet P, Rappaport R, Tonshoff B, et al. Plasma growth
Bresson JL. Alterations of protein metabolism by hormone-binding activity is low in uraemic children.
metabolic acidosis in children with chronic renal fail- Pediatr Nephrol. 1991;5(4):545–7.
ure. Kidney Int. 2000;58(1):236–41. 67. Tonshoff B, Cronin MJ, Reichert M, Haffner D,
53. Boehm M, Riesenhuber A, Winkelmayer WC, Wingen AM, Blum WF, et al. Reduced concentration
Arbeiter K, Mueller T, Aufricht C. Early erythropoi- of serum growth hormone (GH)-binding protein in
etin therapy is associated with improved growth in children with chronic renal failure: correlation with
children with chronic kidney disease. Pediatr GH insensitivity. The European Study Group for
Nephrol. 2007;22(8):1189–93. nutritional treatment of chronic renal failure in child-
54. Jabs K. The effects of recombinant human erythro- hood the German Study Group for growth hormone
poietin on growth and nutritional status. Pediatr treatment in chronic renal failure. J Clin Endocrinol
Nephrol. 1996;10(3):324–7. Metab. 1997;82(4):1007–13.
22 Growth and Development of the Child with Renal Disease 663

68. Greenstein J, Guest S, Tan JC, Tummala P, Busque S, 82. Mitch WE. Proteolytic mechanisms, not malnutrition,
Rabkin R. Circulating growth hormone binding pro- cause loss of muscle mass in kidney failure. J Ren
tein levels and mononuclear cell growth hormone Nutr. 2006;16(3):208–11.
receptor expression in uremia. J Ren Nutr. 2006;16 83. Forbes GB, Bruining GJ. Urinary creatinine excretion
(2):141–9. and lean body mass. Am J Clin Nutr. 1976;29
69. Haffner D, Nissel RI. Growth and puberty in chronic (12):1359–66.
kidney disease. In: Geary D, Schaefer F, editors. 84. Abraham AG, Mak RH, Mitsnefes M, White C,
Comprehensive pediatric nephrology. Philadelphia: Moxey-Mims M, Warady B, et al. Protein energy
Mosby Elsevier; 2008. p. 709–32. wasting in children with chronic kidney disease.
70. Ding H, Gao XL, Hirschberg R, Vadgama JV, Kopple Pediatr Nephrol. 2014;29(7):1231–8.
JD. Impaired actions of insulin-like growth factor 85. Lowrie EG, Lew NL. Death risk in hemodialysis
1 on protein synthesis and degradation in skeletal patients: the predictive value of commonly measured
muscle of rats with chronic renal failure. Evidence variables and an evaluation of death rate differences
for a postreceptor defect. J Clin Invest. 1996;97 between facilities. Am J Kidney Dis. 1990;15:
(4):1064–75. 458–82.
71. Fouque D, Kalantar-Zadeh K, Kopple J, Cano N, 86. Wong CS, Hingorani S, Gillen DL, Sherrard DJ,
Chauveau P, Cuppari L, et al. A proposed nomencla- Watkins SL, Brandt JR, et al. Hypoalbuminemia and
ture and diagnostic criteria for protein-energy wasting risk of death in pediatric patients with end-stage renal
in acute and chronic kidney disease. Kidney Int. disease. Kidney Int. 2002;61(2):630–7.
2008;73(4):391–8. 87. Bergstrom J, Lindholm B. Malnutrition, cardiac dis-
72. Mak R, Cheung WW, Zhan JY, Shen Q, Foster ease, and mortality: an integrated point of view. Am J
BJ. Cachexia and protein energy wasting in children Kidney Dis. 1998;32(5):834–41.
with chronic kidney disease. Pediatr Nephrol. 88. Foster BJ, Leonard MB. Measuring nutritional status
2012;27:173–181. in children with chronic kidney disease. Am J Clin
73. Kalantar-Zadeh K, Mehrotra R, Fouque D, Kopple Nutr. 2004;80(4):801–14.
JD. Metabolic acidosis and malnutrition- 89. Graf L, Candelaria S, Doyle M, Kaskel F. Nutrition
inflammation complex syndrome in chronic renal assessment and hormonal influences on body compo-
failure. Semin Dial. 2004;17(6):455–65. sition in children with chronic kidney disease. Adv
74. Mak RH, Cheung W. Energy homeostasis and Chronic Kidney Dis. 2007;14(2):215–23.
cachexia in chronic kidney disease. Pediatr Nephrol. 90. Jones CH, Wells L, Stoves J, Farquhar F, Woodrow
2006;21(12):1807–14. G. Can a reduction in extracellular fluid volume result
75. Mak RH, Cheung W, Cone RD, Marks in increased serum albumin in peritoneal dialysis
DL. Mechanisms of disease: cytokine and adipokine patients? Am J Kidney Dis. 2002;39(4):872–5.
signaling in uremic cachexia. Nat Clin Pract Nephrol. 91. Kalantar-Zadeh K, Kopple JD. Relative contributions
2006;2(9):527–34. of nutrition and inflammation to clinical outcome in
76. Mitch WE. Insights into the abnormalities of chronic dialysis patients. Am J Kidney Dis. 2001;38
renal disease attributed to malnutrition. J Am Soc (6):1343–50.
Nephrol. 2002;13 Suppl 1:S22–7. 92. Kalantar-Zadeh K, Kopple JD, Block G, Humphreys
77. Mitch WE, Maroni BJ. Factors causing malnutrition MH. A malnutrition-inflammation score is correlated
in patients with chronic uremia. Am J Kidney Dis. with morbidity and mortality in maintenance hemo-
1999;33(1):176–9. dialysis patients. [Comment]. Am J Kidney Dis.
78. Ikizler TA, Cano NJ, Franch H, Fouque D, 2001;38(6):1251–63.
Himmelfarb J, Kalantar-Zadeh K, et al. Prevention 93. Kaysen GA. Malnutrition and the acute-phase reac-
and treatment of protein energy wasting in tion in dialysis patients-how to measure and how to
chronic kidney disease patients: a consensus distinguish. Nephrol Dial Transplant. 2000;15(10):
statement by the International Society of Renal Nutri- 1521–4.
tion and Metabolism. Kidney Int. 2013;84(6): 94. Yeun JY, Levine RA, Mantadilok V, Kaysen
1096–107. GA. C-Reactive protein predicts all-cause and cardio-
79. Foster BJ, Kalkwarf HJ, Shults J, Zemel BS, vascular mortality in hemodialysis patients. Am J
Wetzsteon RJ, Thayu M, et al. Association of chronic Kidney Dis. 2000;35(3):469–76.
kidney disease with muscle deficits in children. J Am 95. Kalantar-Zadeh K, Kleiner M, Dunne E, Ahern K,
Soc Nephrol. 2010;22(2):377–86. Nelson M, Koslowe R, et al. Total iron-binding
80. Mak RH, Cheung W. Cachexia in chronic kidney capacity-estimated transferrin correlates with
disease: role of inflammation and neuropeptide sig- the nutritional subjective global assessment in hemo-
naling. Curr Opin Nephrol Hypertens. 2007;16 dialysis patients. Am J Kidney Dis. 1998;31
(1):27–31. (2):263–72.
81. Mitch WE. Malnutrition: a frequent misdiagnosis for 96. Stenvinkel P. Malnutrition and chronic inflammation
hemodialysis patients. J Clin Invest. 2002;110(4): as risk factors for cardiovascular disease in chronic
437–9. renal failure. Blood Purif. 2001;19(2):143–51.
664 B. Foster

97. Bologa RM, Levine DM, Parker TS, Cheigh JS, hemodialysis patients. Clin Nephrol. 1998;50(1):
Serur D, Stenzel KH, et al. Interleukin-6 predicts 44–50.
hypoalbuminemia, hypocholesterolemia, and mortal- 112. Group WMGRS. WHO child growth standards: head
ity in hemodialysis patients. Am J Kidney Dis. circumference-for-age, arm circumference-for-age,
1998;32(1):107–14. triceps skinfold-for-age and subscapular skinfold-
98. Freedman DS, Ogden CL, Berenson GS, Horlick for-age, methods and development. Geneva: WHO
M. Body mass index and body fatness in childhood. Multicentre Growth Reference Study Group; 2007.
Curr Opin Clin Nutr Metab Care. 2005;8(6):618–23. 113. So SK, Chang PN, Najarian JS, Mauer SM, Simmons
99. Kalantar-Zadeh K, Abbott KC, Salahudeen AK, RL, Nevins TE. Growth and development in infants
Kilpatrick RD, Horwich TB. Survival advantages of after renal transplantation. J Pediatr. 1987;110(3):
obesity in dialysis patients. Am J Clin Nutr. 343–50.
2005;81(3):543–54. 114. Warady BA, Belden B, Kohaut E. Neurodeve-
100. Leavey SF, Strawderman RL, Jones CA, Port FK, lopmental outcome of children initiating peritoneal
Held PJ. Simple nutritional indicators as independent dialysis in early infancy. Pediatr Nephrol. 1999;
predictors of mortality in hemodialysis patients. Am J 13(9):759–65.
Kidney Dis. 1998;31(6):997–1006. 115. Gotch FA, Sargent JA. A mechanistic analysis of the
101. Wong CS, Gipson DS, Gillen DL, Emerson S, National Cooperative Dialysis Study (NCDS). Kid-
Koepsell T, Sherrard DJ, et al. Anthropometric mea- ney Int. 1985;28(3):526–34.
sures and risk of death in children with end-stage renal 116. Goldstein SL. Hemodialysis in the pediatric patient:
disease. Am J Kidney Dis. 2000;36(4):811–9. state of the art. Adv Ren Replace Ther. 2001;8(3):
102. Evans WJ, Morley JE, Argiles J, Bales C, Baracos V, 173–9.
Guttridge D, et al. Cachexia: a new definition. Clin 117. Borah MF, Schoenfeld PY, Gotch FA, Sargent JA,
Nutr. 2008;27(6):793–9. Wolfsen M, Humphreys MH. Nitrogen balance dur-
103. Furth SL, Hwang W, Yang C, Neu AM, Fivush BA, ing intermittent dialysis therapy of uremia. Kidney
Powe NR. Growth failure, risk of hospitalization and Int. 1978;14(5):491–500.
death for children with end-stage renal disease. 118. Depner TA, Cheer A. Modeling urea kinetics with two
Pediatr Nephrol. 2002;17(6):450–5. vs three BUN measurements A critical comparison.
104. Furth SL, Stablein D, Fine RN, Powe NR, Fivush BA. ASAIO Trans. 1989;35(3):499–502.
Adverse clinical outcomes associated with short stat- 119. Goldstein SL, Baronette S, Gambrell TV, Currier H,
ure at dialysis initiation: a report of the North Amer- Brewer ED. nPCR assessment and IDPN treatment of
ican Pediatric Renal Transplant Cooperative Study. malnutrition in pediatric hemodialysis patients.
Pediatrics. 2002;109(5):909–13. Pediatr Nephrol. 2002;17(7):531–4.
105. Kalantar-Zadeh K, Block G, McAllister CJ, 120. Orellana P, Juarez-Congelosi M, Goldstein SL.
Humphreys MH, Kopple JD. Appetite and inflamma- Intradialytic parenteral nutrition treatment and bio-
tion, nutrition, anemia, and clinical outcome in hemo- chemical marker assessment for malnutrition in ado-
dialysis patients. Am J Clin Nutr. 2004;80(2): lescent maintenance hemodialysis patients. J Ren
299–307. Nutr. 2005;15(3):312–7.
106. Bandini LG, Cyr H, Must A, Dietz WH. Validity of 121. Broyer M, Le Bihan C, Charbit M, Guest G, Tete MJ,
reported energy intake in preadolescent girls. Am J Gagnadoux MF, et al. Long-term social outcome of
Clin Nutr. 1997;65(4 Suppl):1138S–41. children after kidney transplantation. Transplantation.
107. Champagne CM, Baker NB, DeLany JP, Harsha DW, 2004;77(7):1033–7.
Bray GA. Assessment of energy intake 122. Busschbach JJ, Rikken B, Grobbee DE, De Charro
underreporting by doubly labeled water and observa- FT, Wit JM. Quality of life in short adults. Horm Res.
tions on reported nutrient intakes in children. J Am 1998;49(1):32–8.
Diet Assoc. 1998;98(4):426–33. 123. Rosenkranz J, Reichwald-Klugger E, Oh J, Turzer M,
108. Charra B, Calemard E, Cuche M, Laurent G. Control Mehls O, Schaefer F. Psychosocial rehabilitation and
of hypertension and prolonged survival on mainte- satisfaction with life in adults with childhood-onset of
nance hemodialysis. Nephron. 1983;33(2):96–9. end-stage renal disease. Pediatr Nephrol. 2005;20(9):
109. Gao T, Leonard MB, Zemel B, Kalkwarf HJ, Foster 1288–94.
BJ. Interpretation of Body Mass Index in Children 124. Al-Uzri A, Matheson M, Gipson DS, Mendley SR,
with CKD. Clin J Am Soc Nephrol. 2012;7(4): Hooper SR, Yadin O, et al. The impact of short stature
558–64. on health-related quality of life in children with
110. Pifer TB, McCullough KP, Port FK, Goodkin DA, chronic kidney disease. J Pediatr. 2013;163(3):
Maroni BJ, Held PJ, et al. Mortality risk in hemodi- 736–41e1.
alysis patients and changes in nutritional indicators: 125. Parekh RS, Flynn JT, Smoyer WE, Milne JL,
DOPPS. Kidney Int. 2002;62(6):2238–45. Kershaw DB, Bunchman TE, et al. Improved growth
111. Kaizu Y, Tsunega Y, Yoneyama T, Sakao T, Hibi I, in young children with severe chronic renal insuffi-
Miyaji K, et al. Overweight as another nutritional risk ciency who use specified nutritional therapy. J Am
factor for the long-term survival of non-diabetic Soc Nephrol. 2001;12(11):2418–26.
22 Growth and Development of the Child with Renal Disease 665

126. Rees L, Brandt ML. Tube feeding in children with controlled study. Genentech Cooperative Study
chronic kidney disease: technical and practical issues. Group. J Pediatr. 1994;124(3):374–82.
Pediatr Nephrol. 2010;25:699–704. 139. Vimalachandra D, Hodson EM, Willis NS, Craig JC,
127. Coleman JE, Watson AR, Rance CH, Moore E. Cowell C, Knight JF. Growth hormone for children
Gastrostomy buttons for nutritional support on with chronic kidney disease. Cochrane Database Syst
chronic dialysis. Nephrol Dial Transplant. Rev. 2006;3, CD003264.
1998;13(8):2041–6. 140. Nissel R, Lindberg A, Mehls O, Haffner D. Factors
128. Ramage IJ, Geary DF, Harvey E, Secker DJ, Balfe JA, predicting the near-final height in growth hormone-
Balfe JW. Efficacy of gastrostomy feeding in infants treated children and adolescents with chronic kidney
and older children receiving chronic peritoneal dialy- disease. J Clin Endocrinol Metab. 2008;93(4):
sis. Perit Dial Int. 1999;19(3):231–6. 1359–65.
129. Zadik Z, Frishberg Y, Drukker A, Blachar Y, Lotan D, 141. Rees L, Ward G, Rigden SP. Growth over 10 years
Levi S, et al. Excessive dietary protein and suboptimal following a 1-year trial of growth hormone therapy.
caloric intake have a negative effect on the growth Pediatr Nephrol. 2000;14(4):309–14.
of children with chronic renal disease before and 142. Gorman G, Fivush B, Frankenfield D, Warady B,
during growth hormone therapy. Metabolism. 1998; Watkins S, Brem A, et al. Short stature and growth
47(3):264–8. hormone use in pediatric hemodialysis patients.
130. Foreman JW, Abitbol CL, Trachtman H, Garin EH, Pediatr Nephrol. 2005;20(12):1794–800.
Feld LG, Strife CF, et al. Nutritional intake in children 143. Tom A, McCauley L, Bell L, Rodd C, Espinosa P,
with renal insufficiency: a report of the growth failure Yu G, et al. Growth during maintenance hemodialy-
in children with renal diseases study. J Am Coll Nutr. sis: impact of enhanced nutrition and clearance.
1996;15(6):579–85. J Pediatr. 1999;134(4):464–71.
131. Ratsch IM, Catassi C, Verrina E, Gusmano R, 144. Fischbach M, Terzic J, Menouer S, Dheu C, Soskin S,
Appiani A, Bettinelli A, et al. Energy and nutrient Helmstetter A, et al. Intensified and daily hemodialy-
intake of patients with mild-to-moderate chronic renal sis in children might improve statural growth. Pediatr
failure compared with healthy children: an Italian Nephrol. 2006;21(11):1746–52.
multicentre study. Eur J Pediatr. 1992;151(9):701–5. 145. Fischbach M, Terzic J, Menouer S, Dheu C, Seuge L,
132. Wingen AM, Fabian-Bach C, Schaefer F, Mehls Zalosczic A. Daily on line haemodiafiltration
O. Randomised multicentre study of a low-protein promotes catch-up growth in children on chronic
diet on the progression of chronic renal failure in dialysis. Nephrol Dial Transplant. 2010;25(3):
children. European Study Group of Nutritional Treat- 867–73.
ment of Chronic Renal Failure in Childhood. Lancet. 146. Mehls O, Wuhl E, Tonshoff B, Schaefer F, Nissel R,
1997;349(9059):1117–23. Haffner D. Growth hormone treatment in short chil-
133. Rees L, Shaw V. Nutrition in children with CRF and dren with chronic kidney disease. Acta Paediatr.
on dialysis. Pediatr Nephrol. 2007;22(10):1689–702. 2008;97(9):1159–64.
134. Wassner SJ, Kulin HE. Diminished linear growth 147. Tonshoff B, Mehls O. Interaction between glucocor-
associated with chronic salt depletion. Clin Pediatr ticoids and the somatotrophic axis. Acta Paediatr
(Phila). 1990;29(12):719–21. Suppl. 1996;417:72–5.
135. Foster BJ, Borzych D. Tube feeding in children with 148. Grenda R, Watson A, Trompeter R, Tonshoff B,
chronic renal insufficiency. In: Warady BA, Jaray J, Fitzpatrick M, et al. A randomized trial to
Schaefer F, Alexander S, editors. Peritoneal dialysis. assess the impact of early steroid withdrawal on
New York: Springer; 2011. growth in pediatric renal transplantation: the TWIST
136. Hokken-Koelega A, Mulder P, De Jong R, Lilien M, study. Am J Transplant. 2010;10(4):828–36.
Donckerwolcke R, Groothof J. Long-term effects of 149. Sarwal MM, Ettenger RB, Dharnidharka V,
growth hormone treatment on growth and puberty in Benfield M, Mathias R, Portale A, et al. Complete
patients with chronic renal insufficiency. Pediatr steroid avoidance is effective and safe in children with
Nephrol. 2000;14(7):701–6. renal transplants: a multicenter randomized trial with
137. Mahan JD, Warady BA. Assessment and treatment of three-year follow-up. Am J Transplant. 2012;12(10):
short stature in pediatric patients with chronic kidney 2719–29.
disease: a consensus statement. Pediatr Nephrol. 150. Mehls O, Schaefer F. Endocrine, metabolic and
2006;21(7):917–30. growth disorders - patterns of growth and maturation
138. Fine RN, Kohaut EC, Brown D, Perlman AJ. Growth in chronic renal failure - impact of developmental
after recombinant human growth hormone treatment stage. In: Holliday MA, Barratt TM, Avner ED,
in children with chronic renal failure: report of a (eds.). Pediatric Nephrology. Baltimore, Williams &
multicenter randomized double-blind placebo- Wilkins; 1994 pp. 1260–1263.
Diagnostic Imaging of the Child with
Suspected Renal Disease 23
Jonathan Loewen and Larry A. Greenbaum

Contents Diagnostic Procedures


Diagnostic Procedures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 667
Ultrasound . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 667 Imaging is essential for the diagnosis and man-
Voiding Cystourethrography . . . . . . . . . . . . . . . . . . . . . . . . . 670 agement of many disorders of the urinary tract
Intravenous Pyelography . . . . . . . . . . . . . . . . . . . . . . . . . . . . 671 (UT). Clinicians should be familiar with the avail-
Computed Tomography . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 672
Nuclear Medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 673 able techniques, including their indications, limi-
Magnetic Resonance Imaging . . . . . . . . . . . . . . . . . . . . . . . 677 tations, and potential complications.
Congenital Kidney and Urological
Ultrasound (US) is the most widely used
Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 678 modality because of the information provided,
Fetal Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 678 safety, and low cost. Intravenous pyelography
Hereditary Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 685 (IVP) is currently seldom utilized, but voiding
Infections of the Urinary Tract . . . . . . . . . . . . . . . . . . . . 686 cystourethrography (VCUG) and nuclear medi-
Renal Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 690 cine studies continue to have important roles.
Renal Transplant . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 690
Computed tomography (CT) is readily available
Urolithiasis and Nephrocalcinosis . . . . . . . . . . . . . . . . . 691 and can acquire images rapidly, generally obviat-
Urolithiasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 691 ing the need for sedation. Magnetic resonance
Nephrocalcinosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 691
imaging (MRI) generates anatomically precise
Trauma to the Urinary Tract . . . . . . . . . . . . . . . . . . . . . . 691 images without radiation exposure, but at high
Renal and Urinary Tract Tumors . . . . . . . . . . . . . . . . . 692 direct financial cost and with the need for sedation
Role of Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 692 in young children. Imaging studies should be
Imaging of Specific Renal Tumors . . . . . . . . . . . . . . . . . . 692 carefully selected, with consideration of diagnos-
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 696 tic utility, cost, and potential complications such
as contrast nephropathy and radiation exposure.

Ultrasound

US is the principal imaging modality for visuali-


zation of the UT. US provides a rapid assessment
of kidney shape and size, and it is especially good
J. Loewen (*) • L.A. Greenbaum
at identifying hydronephrosis. Studies are com-
Department of Pediatric Radiology, Emory University
School of Medicine, Atlanta, GA, USA pleted without discomfort or need for sedation,
e-mail: jonathan.loewen@choa.org; lgreen6@emory.edu and there are no known complications or side
# Springer-Verlag Berlin Heidelberg 2016 667
E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_21
668 J. Loewen and L.A. Greenbaum

effects. In addition, US is relatively inexpensive and evaluation of the renal pelvis, ureters, and
and readily available. The skill of the examiner bladder. The ureters are not usually visible, but
can significantly influence the information may be seen when dilated due to underlying
obtained via US. Lack of patient cooperation and pathology.
large body habitus may also limit image quality. There are published standards for US determi-
Because of its low cost and the absence of side nation of renal length for fetuses, preterm infants,
effects, US is the optimal modality for clinical full-term neonates, infants, and older children
situations requiring repeated evaluations. For ([2–7]; Table 1). Length measurements should
example, US can assess hydronephrosis before be made with the patient supine or in the contra-
and after an intervention such as a catheter, stent, lateral decubitus position; length may be
or nephrostomy tube placement. US is also ideal underestimated with the patient prone [8, 9].
as a screening examination for children who need Gestational age and birth weight correlate with
surveillance for kidney stones or malignancy. kidney length in newborns [2, 5]. In older chil-
Advances in sonographic technology including dren, age is a convenient parameter, but length or
compound imaging, harmonic imaging, pano- height is a better predictor of normal kidney
ramic imaging, and 3D imaging have improved length [4, 10, 11]. A multivariable approach
image quality [1]. allows for improved prediction equations for
renal length [12], but such equations are not
Technique widely used in clinical practice.
Images are generally obtained with the patient In adults and children, the average left kidney
supine, which optimizes visualization of the is slightly longer than the average right kidney
upper poles, using the spleen and liver as acoustic [4, 5, 10], although this is only observed in
windows. The contralateral decubitus position
may permit better visualization in some patients.
In certain situations, placing the patient in the Table 1 Renal length
prone position may allow for better visualization Range (2 SD
of the lower pole if subtle pathology is Age Mean length (cm) in cm)
present. Ideally, the bladder should be visualized Term newborn 4.48 3.86–5.10
while filled with urine. A cooperative child 2 months 5.28 3.96–6.60
should be asked not to void for a few hours prior 6 months 6.15 4.81–7.49
to the exam. In a patient who is not toilet 1.5 years 6.65 5.57–7.73
trained, the bladder should be visualized first 2.5 years 7.36 6.28–8.44
to allow imaging of the bladder before the 3.5 years 7.36 6.18–8.54
4.5 years 7.87 6.87–8.87
patient voids.
5.5 years 8.09 7.01–9.17
US transducers produce sound waves of
6.5 years 7.83 6.39–9.27
varying frequencies. High-frequency sound
7.5 years 8.33 7.31–9.35
waves do not penetrate tissue as deeply, but pro-
8.5 years 8.90 7.14–10.66
vide greater resolution. In most situations, the 9.5 years 9.20 7.40–11.00
sonographer should use the highest frequency 10.5 years 9.17 7.53–10.81
that penetrates to the desired depth. Hence, high- 11.5 years 9.60 8.32–10.88
frequency US sound permits excellent resolution 12.5 years 10.42 8.68–12.16
in infants and small children. Conversely, resolu- 13.5 years 9.79 8.29–11.29
tion is decreased in very large or obese 14.5 years 10.05 8.81–11.29
individuals. 15.5 years 10.93 9.41–12.45
US evaluation of the UT should include mea- 16.5 years 10.04 8.32–11.76
surement of renal lengths and assessment of renal 17.5 years 10.53 9.95–11.11
shape, thickness and echogenicity of the cortex, 18.5 years 10.81 8.55–13.07
the presence of corticomedullary differentiation, Adapted from Rosenbaum et al. [7]
23 Diagnostic Imaging of the Child with Suspected Renal Disease 669

Fig. 1 (a) Normal ultrasound of a neonatal kidney. Med- kidney. Note decreasing cortical echogenicity, decreased
ullary pyramids are hypoechoic with a relatively thin cor- conspicuity of medullary pyramids, and increased renal
tex. (b) Normal 2-year-old kidney. (c) Normal 16-year-old sinus fat echoes with increasing age

slightly more than 50 % of measurements of indi- The appearance of the kidneys changes with
vidual patients (the remainder have kidneys of age. The relative volume of medullary tissue com-
equal length or a longer right kidney) [11]. In pared to cortical tissue is increased in neonates
infants and adults, the median left renal length is and infants (Fig. 1a; [16]). Cortical echogenicity
about 3 mm longer than the median right renal is increased in the term newborn kidney, with the
length [4, 13]. In adults and older teenagers, males majority having the same echogenicity as the liver
have longer kidneys, which is explained by or spleen [6]. Renal cortical echogenicity is fur-
their increased height compared to females. ther increased in premature infants, with some
Black children have shorter kidneys than white kidneys having cortical echogenicity greater than
children [12]. the echogenicity of the liver or spleen [17]. Corti-
Measurement of renal length in children has a cal echogenicity decreases after birth, and only a
fairly high rate of intraobserver and interobserver small percentage of normal kidneys of term
variation [14]; hence, US measurement of renal infants have cortical echogenicity equivalent to
length lacks the accuracy to allow assessment of the liver or spleen by 6 months. Cortical
renal growth over limited time periods. Measure- echogenicity is compared to the liver or spleen,
ment of the left kidney length is less accurate, but this comparison may not be valid if there is
probably because the spleen provides a smaller underlying liver disease. Transient increased cor-
acoustic window than the liver. Nevertheless, tical echogenicity of the kidneys may occur in
renal length discrepancy in children can be children with a variety of nonrenal illnesses [18].
used to predict abnormalities on technetium The kidneys of term newborns have prominent
99m-dimercaptosuccinic acid (99mTc-DMSA) medullary pyramids that are relatively hypoechoic
renal scintigraphy [15]. [16]. This results in marked corticomedullary
670 J. Loewen and L.A. Greenbaum

Table 2 Society of fetal urology grading system for in adults and children who are 6 years of age or
hydronephrosis older, but higher values are observed in children
Grade Ultrasound less than 6 years [28].
0 No hydronephrosis Power Doppler mode allows increased sensi-
1 Only renal pelvis is seen tivity for blood flow detection, but does not pro-
2 Renal pelvis and a few calices are seen vide information on flow direction or velocity.
3 Virtually all calices are seen Power Doppler US is superior to color Doppler
4 Similar to grade 3, but parenchymal thinning US in detecting intrarenal blood flow [29] and in
present
identifying areas of decreased perfusion within
the kidney [30]. Power Doppler increases the sen-
differentiation. The echogenicity of the medullary sitivity of US for detecting acute pyelonephritis
pyramids decreases after birth, and prominent [31]. Power Doppler is very sensitive to motion
medullary pyramids are not normally present by artifact and hence requires a very cooperative
1 year. In contrast, the central sinus echo is min- patient, limiting its utility in young children. 3D
imal at birth and gradually increases during the US of the kidneys can also be performed, with
first 10 years of childhood [6] (Fig. 1b, c). subtraction of a dilated collected system in order
Hydronephrosis may indicate obstruction or to calculate renal parenchymal volumes [32].
vesicoureteral reflux (VUR), or it may be a normal Voiding urosonography (VUS) is an alternative
variant, especially when mild. The Society for technique for detecting VUR that uses intravesical
Fetal Urology grading system for hydronephrosis instillation of a US contrast agent [33, 34]. The
is predictive of the presence or absence of obstruc- principal advantage is the elimination of radiation
tion (Table 2; [19, 20]). Mild hydronephrosis is exposure, but bladder catheterization is still nec-
common in newborns and occurs in a higher per- essary. VUS does not provide adequate imaging
centage of males than females [21]. of the bladder and urethra and thus should only be
An increase in bladder wall thickness may be used for the assessment of VUR. The results with
an indication of pathology, especially chronic VUS correlate well with conventional VCUG [33,
increased bladder pressure. Measurement of blad- 35–38]. It is not yet clear if operator skill or other
der wall thickness is highly dependent on the barriers such as availability of US contrast agents
degree of bladder filling, but does not appear to may limit the widespread use of this
be dependent on patient age or gender [22], technique [32].
although male neonates may have slightly greater
bladder wall thickness than females [21]. The Application
upper limit of normal is 5 mm for an empty US is the initial imaging modality in a variety of
bladder and 3 mm for a well-distended bladder clinical situations, including evaluation of renal
[22]. US can be used to accurately estimate blad- failure, kidney transplant dysfunction, hematuria,
der volume [23, 24]. US measurement of bladder screening for renal malignancy, abdominal
volume after voiding is a useful parameter for mass, and follow-up of abnormal prenatal US.
assessing bladder function. US may detect Procedural applications of US in pediatric
ureteroceles or bladder masses. nephrology include pre-biopsy or real-time local-
Doppler US permits evaluation of blood flow ization of native and transplant kidneys as well as
and has been utilized to identify renal artery dis- venous localization prior to hemodialysis catheter
ease, renal vein thrombosis, tumor thrombosis in placement [39].
the renal vein and inferior vena cava, and arterio-
venous fistulas [25–27]. Resistance to blood flow
can be quantified by measuring the resistive index Voiding Cystourethrography
(RI), which is calculated from the formula: RI =
(peak systolic velocity-end diastolic velocity)/ VCUG is the most frequently performed fluoro-
peak systolic velocity. A normal RI is below 0.7 scopic examination and the gold standard for the
23 Diagnostic Imaging of the Child with Suspected Renal Disease 671

diagnosis of VUR [1]. Most clinical studies of


VUR have utilized VCUG. Moreover, VCUG is
the only modality that detects VUR and provides
detailed information about the bladder and
urethra.
Radiation exposure is the most important side
effect of VCUG. The radiation dose has been
reduced through technological innovations, and
a variety of additional approaches are available
to further reduce radiation exposure [40, 41].
A known allergy to contrast material is a contra-
indication to the study [42]. Because of the need
for catheterization, VCUG creates a small risk of
iatrogenic infection and trauma to the urethra,
although these risks are low [43]. In addition,
many children and families find catheterization
frightening and emotionally traumatic. Sedation
with midazolam appears safe and effective
[44–46].

Technique
VCUG is done using fluoroscopy. An initial film Fig. 2 Voiding cystourethrogram. Frontal voiding image
prior to bladder catheterization detects calcifica- of normal 5-year-old girl
tions or bony abnormalities; an abnormal bowel
gas pattern may suggest a mass. The bladder is advances including digital fluoroscopy with
sterilely catheterized using a feeding tube (5 F in pulsed fluoroscopy and last-image-hold options
young infants; 8 F in older children), and urine can have made it possible to diminish radiation
be obtained for culture. After emptying the blad- dosage [1].
der of urine, contrast material is infused to fill the
bladder based on estimates of bladder capacity,
but using gravity to avoid overfilling. The bladder Intravenous Pyelography
must be filled to capacity to insure an adequate
study. The availability of US and other imaging modal-
An early film during filling of the bladder is ities such as MR urography has dramatically
useful for identifying filling defects, such as decreased the use of intravenous pyelography
caused by a ureterocele or bladder tumor. Films (IVP) [50]. The risks of IVP include radiation
are taken before and after voiding, with VUR and contrast exposure; intravenous access is
graded based on the International Reflux Study necessary.
in Children [47]. In young children, who may not
allow their bladder to fill to capacity, the sensitiv- Technique
ity of VCUG for detecting VUR improves with An initial supine film is performed prior to con-
multiple cycles of bladder filling and voiding trast injection. Approximately 5–7 min after con-
[48, 49], but this increases the length and radiation trast, a film centered on the kidneys allows
exposure of the procedure. A film during voiding visualization of the nephrogram and the calices.
permits visualization of the urethra and is essential Subsequent films are full length and taken 15 and
in male children (Figs. 2 and 3). At the end of 30 min after injection. Delayed films are useful in
voiding, a final film identifies residual urine and cases of urinary obstruction. Bowel preparation
previously unidentified VUR. Technological does not improve image quality [51, 52], although
672 J. Loewen and L.A. Greenbaum

Fig. 3 Voiding
cystourethrogram. Oblique
voiding image of a normal
1-year-old boy

a short fasting period is usually recommended. most situations. Multiple strategies and innova-
The radiation exposure of IVP can be reduced by tions have reduced the radiation dose with CT
adjusting the number of films based on the clinical [55, 57], but it remains an important concern
question and the results of initial films. Nonionic [58]. Sedation is currently rarely necessary due
contrast medium improves image quality and to rapid image acquisition [59, 60], which also
decreases the risk of contrast nephropathy. decreases motion artifact [57].

Application Technique
Poor renal function limits the utility of IVP due to Thin-section helical imaging, now performed on
inadequate contrast accumulation in the kidneys. all CT scanners, allows for multiplanar
Moreover, poor renal function increases the risk reformatted images to be obtained quickly and
of contrast nephropathy. IVP is currently rarely displays the kidneys, ureters, and bladder in
used in the evaluation of obstructive disease; US three planes. Oral contrast, while often unneces-
and nuclear medicine studies are the preferred sary for evaluation of the UT, is useful for exam-
imaging modalities. 99mTc-DMSA has replaced inations where differentiating bowel from
IVP for detection of renal scarring, but IVP pro- pathology is necessary. Time constraints (acute
vides excellent detail and is certainly an appropri- trauma) or risk of aspiration may prevent the use
ate substitute [53, 54]. IVP is useful for evaluation of oral contrast. For younger children, excessive
of ectopic ureters, ureteroceles, and fusion anom- movement can be prevented by using blankets or
alies, although alternative imaging (US, MRI, CT) adhesive tape.
is generally utilized when available. Intravenous contrast is usually indicated, with
the notable exception of nephrolithiasis evalua-
tion. Low osmolality nonionic contrast is
Computed Tomography preferred; contrast is dosed based on weight
[57, 61]. Neonates and infants require a longer
CT provides excellent anatomic resolution of the delay from contrast injection to imaging.
UT (Fig. 4), and CT has many applications. Radi-
ation exposure is an important risk of CT and Application
should limit its use to cases where the detail pro- CT is the preferred modality for initial evaluation
vided outweighs the risk of the study [55, 56]. of possible symptomatic nephrolithiasis, although
IV contrast is necessary for optimal studies in US is generally utilized for surveillance to
23 Diagnostic Imaging of the Child with Suspected Renal Disease 673

Fig. 4 CT of a normal 11-year-old boy. (a) Axial image through the mid-pole of both kidneys. (b) Coronal image. (c)
Sagittal image

minimize radiation exposure. CT is used for renal typically utilized to answer a specific question.
tumor staging and is the optimal modality when Nuclear medicine is often a complementary tech-
trauma of the UT is suspected. CT provides excel- nique. While there is radiation exposure, it is less
lent resolution of renal scarring, but 99mTc-DMSA than other modalities such as VCUG, IVP, or CT.
is more commonly used. CT angiography is useful
in assessing for renal artery stenosis [62, 63]. Technique
Diminishing radiation exposure by using age-/
weight-based exposure parameters and by limit- Dynamic Renal Scintigraphy
ing the field of view is suggested. Efforts should Technetium 99m-diethylene triamine pentaacetic
be made to achieve the lowest radiation dose acid (99mTc-DTPA) and technetium
needed in order to generate a diagnostic exam 99m-mercaptoacetyltriglycine (99mTc-MAG3)
[64, 65]. are the radiotracers currently utilized for dynamic
renal scintigraphy. 99mTc-DTPA is primarily
excreted via glomerular filtration; it is not
Nuclear Medicine reabsorbed or secreted by the tubules; its clear-
ance is slightly lower than inulin. 99mTc-MAG3 is
A variety of radiotracers are utilized for imaging predominantly excreted via tubular secretion. For
of the UT [66, 67]. The radiotracers emit photons, most indications, 99mTc-MAG3 is superior to
99m
which are detected by a scintillation detector. Tc-DTPA because 99mTc-MAG3 is more rap-
Nuclear medicine imaging does not provide ana- idly excreted and thus provides a superior renal/
tomic detail; it provides functional data that is background ratio. The nuclear radiotracers have a
674 J. Loewen and L.A. Greenbaum

Fig. 5 Posterior image of a normal 99mTc-MAG3 renal scan in an 8-year-old with hydronephrosis by ultrasound

lower allergic reaction profile than radiographic Following administration of a diuretic, radio-
contrast media [68]. tracer drains rapidly from a non-obstructed renal
Patients are encouraged to consume liquids pelvis (Figs. 5 and 6); there is limited washout of
immediately prior to the procedure as this facili- radiotracer if obstruction is present. A half-life of
tates exam interpretation, but also decreases the radiotracer disappearance greater than 20 min
radiation dose to the urinary bladder [68]. Patients indicates obstruction [71]. There is a good corre-
should also ideally void before injection of iso- lation between the test results and histological
tope. The uncooperative child should be changes [72], and the test is sensitive and specific
immobilized, but sedation is rarely necessary. [71, 73]. Diuresis scintigraphy is accurate in new-
Images are obtained with the patient supine; the borns, despite their lower glomerular filtration rate
scintillation detector is below the patient for [73, 74], although follow-up imaging of normal
native kidneys and above the patient for renal results is necessary to ensure that obstruction does
transplants. After bolus injection of radiotracer, not subsequently develop [75]. Potential causes of
dynamic images are recorded for 30 min. The false-positive results are a massively dilated renal
initial images assess renal perfusion, and later pelvis or a kidney with very poor function [76],
images evaluate excretion. Differential renal func- but technical modifications may allow for more
tion is calculated using the early images, typically accurate assessment of a poorly functioning kid-
during the second minute, prior to radiotracer ney [77–79]. False-positive results can also be
appearance in the collecting system. decreased by obtaining images after the patient
Diuresis scintigraphy is performed to evaluate voids to avoid bladder pressure effects and after
a patient for renal obstruction [69, 70]; use of a placing the child in the erect position to maximize
diuretic is only necessary if radiotracer does not gravitational drainage [80–82].
rapidly drain from the renal pelvis during initial
imaging. A dilated renal pelvis that is not Cortical Scintigraphy
99m
obstructed may have slow drainage of radiotracer Tc-DMSA is the preferred radiotracer for
due to the increased pelvic volume. Administra- imaging of the renal cortex [83, 84]. 99mTc-
tion of diuretic allows differentiation of an DMSA is taken up by the cells of the proximal
obstructed kidney from a non-obstructed dilated tubule, where it binds to sulfhydryl groups
renal pelvis. Furosemide is given 15–20 min after (Fig. 7). The accumulation of 99mTc-DMSA in
administration of radiotracer. the kidney is relatively slow; hence, imaging is
23 Diagnostic Imaging of the Child with Suspected Renal Disease 675

Fig. 6 Time-activity curve


from 99mTc-MAG3 renal
scan showing symmetric
prompt excretion of
radiotracer, indicating no
obstruction

Fig. 7 (a) Posterior image and coned oblique images of normal kidneys on DSMA scan in a 3-year-old with a urinary
tract infection. (b) Regions of interest drawn for split-function calculation

typically performed 1.5–3 h after injection, impression explains flattening of the superolateral
although diagnostic accuracy may improve with surface of the left kidney [84]. The poles of nor-
an increased delay in imaging if renal function is mal kidneys may have relatively decreased uptake
significantly impaired. Posterior and left and right of radiotracer. Nevertheless, there is good
posterior oblique views should be obtained interobserver agreement in the interpretation of
99m
[83, 84], but there is minimal loss in diagnostic Tc-DMSA scans for scarring and acute pyelo-
accuracy if only a posterior view is possible in a nephritis [86, 87].
99m
restless child [85]. Anterior imaging is preferred Tc-DMSA is not specific for scars or acute
for pelvic or horseshoe kidneys. Additional pin- pyelonephritis. Alternative explanations for
hole images are useful for improving resolution defects such as cysts, hydronephrosis, infarcts,
when imaging infants. masses, or a dysplastic half of a duplex kidney
A variety of factors may complicate interpreta- can only be identified using alternative imaging
tion of 99mTc-DMSA imaging. Fetal lobulations strategies (e.g., US). Children with proximal
may give the false impression that there are areas tubulopathies have decreased uptake of 99mTc-
of decreased radiotracer uptake. The splenic DMSA and increased urinary excretion [88].
676 J. Loewen and L.A. Greenbaum

Single-photon emission computed tomogra- Table 3 Drugs that interfere with uptake of MIBG
phy (SPECT) is an alternative to planar scintigra- Propranolol
phy. SPECT appears to offer superior sensitivity Labetalol
for detecting renal scars [89, 90], although with a Calcium channel blockers
higher rate of false-positive results [91, 92]. The Phenylephrine
clinical relevance of the increased detection of Pseudoephedrine
small scars via SPECT is not clear. An Tricyclic antidepressants
intrarenicular septum, a common normal variant, Phenothiazines
may lead to a photopenic area with SPECT, but Monoamine oxidase inhibitors
can be differentiated from a scar because of pre-
served cortical uptake [93]. The increased imag- interfere with MIBG uptake into tissue and should
ing time required with SPECT may necessitate be discontinued prior to MIBG imaging (Table 3).
sedation in young children. MIBG radiotracer is injected, with images
Split function is determined by outlining the obtained at various times depending on the iso-
appropriate regions of the kidneys and subtracting tope. MIBG accumulates in a variety of organs,
the activity of an appropriate background area in including the liver, heart, and salivary glands;
the posterior image. Normal split function is 50 % isotope is detected in the kidney and bladder due
+/ 6 %. Estimates of split function must be to renal excretion. Tumors can be identified
corrected for attenuation if there is an anteriorly because of the stronger radiotracer uptake, which
placed ectopic kidney. Such correction is not is typically focal. 123I has a much lower radiation
accurate with a pelvic kidney due to the effect of burden than 131I and results in superior
the pelvic bone. Imaging may need to be delayed images [98].
for up to 24 h in an obstructed kidney because
isotope accumulation in the renal pelvis may Radionuclide Cystography
cause an error in estimating split function, with Radionuclide cystography (RNC) is an alternative
an overestimation of the function of the obstructed to VCUG for the detection of VUR (Fig. 8;
kidney if the image is obtained prematurely. A [99–101]). In RNC, a catheter is placed into the
similar approach may be necessary for a bladder, and a solution containing a radioisotope
non-obstructed, severely dilated renal pelvis, is infused, with complete filling of the bladder.
although injection of furosemide, with the subse- RNC, because it allows for continuous monitor-
quent diuresis clearing radiotracer from the ing, is more sensitive at detecting VUR than
dilated pelvis, is an alternative strategy. Estimates VCUG, which cannot utilize continuous monitor-
of split function are reliable, with good ing due to radiation concerns. Furthermore, the
interobserver agreement, except in cases where radiation burden is considerably less than VCUG.
renal function is significantly impaired [94]. However, radionuclide cystography does not per-
mit evaluation of the bladder or the urethra.
Adrenal Gland Scintigraphy
Metaiodobenzylguanidine (MIBG), when labeled Applications
with either 131I or 123I, is used for the detection of 99m
Tc-DMSA is the preferred radiopharmaceuti-
catecholamine-secreting tumors, including pheo- cal for evaluation of the renal cortex and the gold
chromocytomas and neuroblastomas [95–97]. standard for the identification of pyelonephritis
MIBG, a guanethidine analog that structurally and renal scars [102]. 99mTc-DMSA can be uti-
resembles norepinephrine, is taken up by sympa- lized to determine split renal function and to iden-
thetic nerve cells. The patient should receive oral tify and characterize renal infarcts or anatomically
nonradioactive iodine before and after administra- abnormal kidneys such as horseshoe kidneys, pel-
tion of radiolabeled MIBG to compete with free vic kidneys, and crossed fused ectopia [103, 104].
radioactive iodine, minimizing radioactive expo- Dynamic renal imaging with 99mTc-MAG3 or
99m
sure to the thyroid gland. A variety of medications Tc-DTPA permits evaluation of renal blood
23 Diagnostic Imaging of the Child with Suspected Renal Disease 677

flow, split renal function, kidney location, and etiology such as VUR, high urine flow, or relieved
urinary drainage. Dynamic renal imaging allows obstruction [105]. Dynamic renal imaging can be
determination of whether hydronephrosis is sec- utilized to determine split renal function and to
ondary to renal obstruction or an alternative identify renal artery stenosis when used with
captopril [106].

Magnetic Resonance Imaging

MRI, with its ability to image the abdomen in


multiple planes and with sequences showing dif-
ferent signal characteristics, provides excellent
resolution of the UT without radiation exposure
(Figs. 9 and 10) [107]. Faster MR sequences
allowing for rapid image acquisition and the con-
tinued trend in reducing radiation exposure in the
pediatric population have also increased imaging
of the urinary tract by MRI [1].
Gadolinium-based contrast at standard doses is
significantly less nephrotoxic than iodinated con-
trast [108, 109], but gadolinium-based contrast is
nephrotoxic when used at radiographic doses for
angiographic procedures [110–112]. Gadolinium-
based contrast is the dominant risk factor for the
development of nephrogenic systemic fibrosis
(NSF) [113], an entity that occurs almost exclu-
sively in patients with end-stage renal disease or
severe renal impairment. Most cases of NSF have
been associated with gadodiamide, although other
Fig. 8 Radionuclide cystogram. Posterior image from a
nuclear cystogram in a 4-year-old demonstrating left gadolinium-based contrast agents may lead to
vesicoureteral reflux NSF [114]. Gadolinium-based contrast should be

Fig. 9 Renal MRI of a normal 5-year-old. (a) Axial T2-weighted image through the mid-pole of the kidneys. (b) Coronal
T2-weighted image
678 J. Loewen and L.A. Greenbaum

superior contrast and temporal and spatial resolu-


tion when compared to renal scintigraphy [1].

Application
Conventional MRI is useful in characterization of
solid and cystic renal masses, renal vascular dis-
ease, and congenital abnormalities including
lower urogenital tract malformations and complex
post-traumatic conditions [64, 117, 118]. The
most common indication for MRU is
hydronephrosis. This technique allows for simul-
taneous anatomic and physiologic assessment of
the parenchyma, collecting system, and surround-
ing tissues [1, 119]. The goal of the exam is to
determine whether obstruction of a
hydronephrotic system is present based on its
response to a fluid/diuretic challenge [119, 120].

Congenital Kidney and Urological


Disorders

Fetal Imaging
Fig. 10 Renal MRI. 3D image of the kidneys and ureters The widespread use of fetal US has led to a dra-
in a normal 12-year-old
matic increase in postnatal imaging of the
UT. Most congenital abnormalities of the UT are
now detected prenatally, and this has led to a
used cautiously in patients with moderate to decrease in the diagnosis of obstructive uropathies
severe renal impairment [115]. as a consequence of urinary tract infection (UTI),
Sedation is typically necessary for infants and potentially diminishing the role of renal US in
young children. MRI is the most expensive imag- children with UTI [121, 122].
ing modality, but it may replace multiple other Fetal kidney length relates to gestational age,
studies and provide superior resolution in certain and normal values are available (Table 4; [3]).
clinical situations. Fetal US is excellent at identifying
hydronephrosis, and this may identify infants
Technique with UT obstruction or VUR. Oligohydramnios,
Magnetic resonance urography (MRU) provides which may indicate poor urine output due to renal
superb anatomic detail combined with functional insufficiency, is easily quantified and followed.
information [116]. In this technique, a gadolinium Persistent oligohydramnios is associated with pul-
chelate (Gd-DTPA) is injected, and the kidneys monary hypoplasia. The fetal bladder, which may
are imaged over time. In addition to anatomic be distended with impaired bladder emptying, and
assessment of renal parenchyma and collecting the echogenicity of the renal parenchyma are also
system anatomy, post-processing of MRU data visualized via fetal US.
derived from the study allows for determination Hydronephrosis is the most common renal
of split differential renal function, transit time of abnormality on fetal US, and ureteropelvic junc-
the contrast agent through the kidney, and an tion (UPJ) obstruction is the most common path-
estimate of glomerular filtration rate. MRU has ologic condition, although a high percentage of
23 Diagnostic Imaging of the Child with Suspected Renal Disease 679

Table 4 Mean renal lengths for various gestational ages


Gestational age Mean 95 %
(Weeks) length (cm) SD CI n
18 2.2 0.3 1.6–2.8 14
19 2.3 0.4 1.5–3.1 23
20 2.6 0.4 1.8–3.4 22
21 2.7 0.3 2.1–3.2 20
22 2.7 0.3 2.0–3.4 18
23 3.0 0.4 2.2–3.7 13
24 3.1 0.6 1.9–4.4 13
25 3.3 0.4 2.5–4.2 9
26 3.4 0.4 2.4–4.4 9
27 3.5 0.4 2.7–4.4 15
28 3.4 0.4 2.6–4.2 19
29 3.6 0.7 2.3–4.8 12
30 3.8 0.4 2.9–4.6 24
31 3.7 0.5 2.8–4.6 23
32 4.1 0.5 3.1–5.1 23 Fig. 11 Sagittal T2-weighted fetal MR image of a
33 4.0 0.3 3.3–4.7 28 28-week fetus with dilated bladder and posterior urethra
34 4.2 0.4 3.3–5.0 36 (arrow) in the setting of posterior urethral valves
35 4.2 0.5 3.2–5.2 17
36 4.2 0.4 3.3–5.0 36 evaluation [125]. The severity of hydronephrosis
37 4.2 0.4 3.3–5.1 40 is determined by ultrasonography after 48 h of life
38 4.4 0.6 3.2–5.6 32 by the measurement of the anterior posterior
39 4.2 0.3 3.5–4.8 17 diameter of the pelvis. A diameter less than
40 4.3 0.5 3.2–5.3 10 7 mm is considered as normal, 7–8 mm is a mild
41 4.5 0.3 3.9–5.1 4 hydronephrosis, 9–16 mm is a moderate
Gestational age is an average of the gestational ages in hydronephrosis, and more than 16 mm is a severe
weeks determined on the basis of biparietal diameter, fem-
oral length, and abdominal circumference. SD standard
hydronephrosis which requires a voiding
deviation; 95 % confidence interval; n number of fetuses. cystourethrogram and a functional renal scan
A t distribution was used when n < 30 (From Cohen diuretic renography [126].
et al. [3]) Prenatal US can detect children with a variety
of other pathologic conditions, including
posterior urethral valves, multicystic dysplastic
kidneys are ultimately diagnosed as kidney, VUR, megaureter, duplex anomalies
normal [123]. There is a high rate of false- with upper pole hydronephrosis, single kidney,
negative results when a postnatal US to follow- polycystic kidney disease, and Eagle-Barrett
up on prenatal hydronephrosis is done in the first syndrome [127].
few days of life, presumably because of low urine While fetal US remains the primary means of
output due to physiologic dehydration in utero imaging, fetal MRI has a role in further
[124]. Hence, the initial postnatal US should characterization of complex fetal genitourinary
either be done at least 4 weeks after birth or the abnormalities. The pregnant patient is most fre-
child should have a second US. Deferral of the quently scanned in the supine position during the
initial US is inappropriate if the prenatal US second or third trimester. Intravenous contrast
suggests the presence of disease that would material is not used for the exam. The most com-
require immediate intervention (e.g., bilateral monly encountered GU abnormalities on fetal
hydronephrosis or oligohydramnios). The degree MRI include both upper urinary tract obstruction
of prenatal hydronephrosis is highly predictive of (UPJ stenosis) and lower urinary tract obstruction
the likelihood of pathology on postnatal such as posterior urethral valves (Fig. 11).
680 J. Loewen and L.A. Greenbaum

Fig. 12 Ectopic ureterocele with vesicoureteral reflux. (a) left duplex system with eversion of the ureterocele
Voiding cystourethrogram shows rounded filling defect in (asteric). Opacification of the kidney indicates intrarenal
bladder base from ureterocele. (b) Voiding cystoure- reflux
throgram shows reflux into the lower pole moiety of the

More complex abnormalities such as cloacal information and superior anatomic detail without
malformations and bladder exstrophy can also be exposure to radiation, albeit with the need for
imaged [128]. sedation (Fig. 14; [131, 132]).
A second alternative technique for diagnosing
Megaureter obstruction is the measurement of the resistive
Megaureter may be primary or secondary to VUR index (RI) by Doppler US, with an elevated RI
or obstruction. VCUG permits identification of suggesting obstruction. The sensitivity and spec-
VUR, and diuretic scintigraphy establishes the ificity of this approach is improved by adminis-
presence of obstruction. MRU provides excellent tering normal saline and furosemide prior to US
anatomic detail, often allowing identification of [133]. The principal advantage is the elimination
the site of obstruction [129]. of radiation exposure, but this approach is not yet
widely utilized.
Ureterocele
Ureteroceles may be demonstrated by US, Abnormal Kidney Number or Location
VCUG, IVP, or MRU (Fig. 12). An everting Unilateral renal agenesis must be distinguished
ureterocele may be confused with a bladder diver- from an ectopic kidney, which can be located
ticulum on VCUG [130]. Ureteroceles may anywhere along the course of renal ascent. Such
obstruct the ureter during voiding. kidneys are often smaller in size. Detection by US
is possible, but MRI, IVP, or DMSA is often
Ureteropelvic Junction Obstruction necessary (Fig. 15). IVP may have limited sensi-
UPJ obstruction is one of the most common con- tivity if the ectopic kidney has poor renal function.
genital kidney disorders, and most cases are now
diagnosed by fetal US. Dynamic renal scintigra- Horseshoe Kidney and Crossed Fused
phy remains the gold standard imaging technique Ectopia
for diagnosing UPJ obstruction (Fig. 13). MRI is Horseshoe kidney, with an incidence of 1 of
an alternative approach that provides functional 400 births, is the most common fusion anomaly.
23 Diagnostic Imaging of the Child with Suspected Renal Disease 681

99m
Fig. 13 Left ureteropelvic junction obstruction. (a) US Tc-MAG3 scan shows a normal right kidney. On the
image of the left kidney shows dilatation of the renal pelvis left, little drainage occurs in response to furosemide. T ½
and calices. (b) 99mTc-MAG3 renal scan shows retention of exceeds 20 min
radiotracer in the left kidney. (c) Time-activity curve from
682 J. Loewen and L.A. Greenbaum

Crossed ectopia occurs in an estimated 1 of


1,000–2,000 births. While US can be diagnostic,
additional imaging, including IVP, MRI, CT, or
DMSA, is often necessary to establish the diag-
nosis (Fig. 16).

Renal Dysplasia
Renal dysplasia is a histological diagnosis char-
acterized by primitive ducts and cartilage. US
evaluation demonstrates increased echogenicity;
most dysplastic kidneys are decreased in size. US
may detect cysts, which may vary in size and
number.
The incidence of multicystic dysplastic kidney
(MCDK) is estimated at 1 in 2,500 newborn
[134]; it is one of the common prenatally detected
fetal anomalies. Postnatal US shows cysts of vary-
ing size that do not appear to communicate with
each other or the collecting system and a small
amount of abnormal-appearing renal parenchyma.
It may be difficult to differentiate MCDK from
severe hydronephrosis [135]. In MCDK, there is
Fig. 14 Left ureteropelvic junction obstruction. MR typically no communication between cysts, and
urogram in patient with UPJ obstruction shows dilatation the larger cysts are not medial (Fig. 17). In
of the left renal pelvis and calices hydronephrosis, the calices extend outward from
the dilated renal pelvis, and there is functional
renal parenchyma surrounding the central cystic
structure. If the diagnosis is unclear, a DMSA
shows uptake of tracer if hydronephrosis is pre-
sent, but usually no uptake with MCDK (Fig. 18).
Alternatively, MRI can distinguish these entities.

Duplex Collecting System


A duplex collecting system is a common inciden-
tal finding and usually of no clinical significance
unless hydronephrosis is also present. Associated
anomalies include ureterocele, VUR, ectopic ure-
ters, renal dysplasia, and UPJ obstruction
(Figs. 19 and 20; [136]). The upper pole is more
likely to be associated with obstruction at the
distal ureter, while the lower pole is associated
with VUR or UPJ obstruction. US can usually
demonstrate division of the kidney, but is not
reliable in distinguishing between bifid ureter
and complete ureteral duplication. IVP is being
supplanted by MRU for delineating the anatomy
Fig. 15 Crossed fused ectopia. DMSA renal scan anterior of the ureteral duplication when such information
image demonstrates a single fused renal moiety on the right is necessary for planning surgery [129]. MRU is
23 Diagnostic Imaging of the Child with Suspected Renal Disease 683

Fig. 16 Horseshoe kidney.


CT scan of abdomen shows
a horseshoe kidney with a
bridge of renal tissue fused
across the midline
(arrowhead)

Fig. 17 Left multicystic


dysplastic kidney. Renal US
in an infant shows multiple
noncommunication cysts in
the left kidney

especially useful for characterization of the distal unit is poorly functioning. MRU may allow detec-
ureter [137]. VCUG may demonstrate VUR or tion of the ectopic ureter if other studies are unsuc-
ureterocele. cessful [137] and has largely replaced IVP in the
evaluation of ectopic ureteral insertion. VCUG is
Ectopic Ureters useful for showing reflux and may indicate the
Ectopic ureters are three times more common in insertion point of the ureter if it is proximal to
girls, and approximately 80 % are associated with the external sphincter. DMSA is useful for identi-
a duplicated collecting system and ectopic inser- fying an ectopic kidney that is the source of an
tion of the upper pole ureter. Greater distance from ectopic ureter in a patient with unexplained incon-
the normal ureter insertion site is associated with tinence [103, 104]. By indicating relative renal
more severe ipsilateral renal abnormalities function, DMSA may also help with surgical
[138]. US can demonstrate duplication, planning.
hydronephrosis, megaureter, and abnormal renal
parenchyma. IVP is useful for delineating the Bladder Abnormalities
course of the ectopic ureter, although lack of con- US and VCUG are the standard imaging modalities
trast may limit the study if the associated renal for patients with a neurogenic bladder (Fig. 21).
684 J. Loewen and L.A. Greenbaum

VCUG readily identifies bladder diverticula, which Urethral Abnormalities


are easily missed by US. Bladder wall thickening, VCUG is the definitive test for diagnosing
consistent with high bladder pressures, can be posterior urethral valves (Fig. 22). The prostatic
quantified by US. urethra is dilated, the membranous urethra is
stenotic, and the distal urethra appears
normal. Secondary VUR, hydronephrosis,
and bladder abnormalities are often
present (Fig. 23). Other urethral abnormalities
(anterior urethral valves, urethral duplication,
Cowper’s duct cyst, urethral diverticulum,
megalourethra) are also best visualized via
VCUG.

Vesicoureteral Reflux
VCUG is the gold standard for diagnosing VUR,
with VUR graded based on the International
Reflux Study in Children (Fig. 24; [47]). RNC
and VUS, because of decreased or no
radiation burden, respectively, are appropriate
substitutes for VCUG in clinical situations
where the purpose of the study is solely to assess
the patient for VUR. Examples include follow-up
studies in children with previously diagnosed
VUR, screening siblings of children with VUR,
and potentially for the initial evaluation of
girls with urinary tract infections. Renal US
lacks adequate sensitivity and specificity for diag-
Fig. 18 Left multicystic dysplastic kidney. DMSA renal
nosing VUR [139–142], although high-grade
scan (posterior image) shows no uptake in the left VUR is more likely if hydronephrosis is
multicystic dysplastic kidney present [143].

Fig. 19 Duplex system


with ectopic ureterocele.
Renal US shows mild
hydronephrosis
(arrowheads) of the upper
and lower poles. A band of
renal tissue extends across
the mid-pole, indicating a
duplex system
23 Diagnostic Imaging of the Child with Suspected Renal Disease 685

Fig. 20 Duplex system with ectopic ureterocele. MRU


shows hydronephrosis of the right upper pole
Fig. 21 Neurogenic bladder in a 16-year-old with a spinal
cord injury. Voiding cystourethrogram shows pear-shaped
bladder with trabeculation
Hereditary Disorders

Cystic Kidney Diseases


In autosomal recessive polycystic kidney disease detecting macroscopic cysts and associated com-
(ARPKD), intravenous pyelogram (IVP) shows plications (Figs. 26 and 27). The number and size
enlarged kidneys with a delayed nephrogram of cysts in children increases with age, and those
[144]. Radial streaks due to contrast in the dilated children with more cysts have increased kidney
collecting ducts are usually present in infancy, but size [148]. ADPKD rarely causes renal failure in
may not be visible in older children. Because of childhood [32, 149]. In adults, kidney size mea-
concerns regarding intravenous contrast, a renal sured by MRI predicts decline in renal function
US is now the usual initial diagnostic test [150]. Increased renal echogenicity may be seen
(Fig. 25). US shows enlarged kidneys with in some children [147]. Children occasionally
increased echogenicity and poor corticomedullary have marked disease asymmetry, which can create
differentiation; a hypoechoic rim is often visible diagnostic confusion [151]. Both ARPKD and
[145]. Older children have increased medullary ADPKD can have associated liver abnormalities
echogenicity, which may resemble [152]. Extrarenal complications of ADPKD
nephrocalcinosis [146]. Multiple small cysts may include liver cyst infection/hemorrhage, intracra-
be visible by US [146, 147], but this is quite nial aneurysms, and valvular heart disease [153].
variable. Macrocysts are sometimes visible in US in nephronophthisis shows hyperechoic
older children [147]. kidneys with loss of corticomedullary differentia-
In autosomal dominant polycystic kidney dis- tion; they are of normal or slightly decreased size.
ease (ADPKD), US, CT, and MRI are useful for Medullary cysts are a hallmark of the disease, but
686 J. Loewen and L.A. Greenbaum

they are not always detected by US. CT is a more Tuberous Sclerosis


sensitive method for identifying medullary cysts Angiomyolipomas are the most common renal
and thin-section CT is recommended [154]. lesion in tuberous sclerosis (TS) and are readily
The diagnosis of medullary sponge kidney is seen by CT or ultrasound (Figs. 28 and 29). Chil-
based on characteristic IVP changes: stagnation of dren with disruption of the contiguous PKD1 and
contrast in one or more renal papillae due to TSC2 genes usually have severe cystic disease and
dilation of the collecting ducts. The resultant are at high risk for hypertension and early kidney
image has been described as a “pyramidal failure [156]. Renal malignancies, including renal
blush.” In medullary cystic disease, the kidneys cell carcinoma and malignant angiomyolipomas,
are usually normal or small, and US or CT may are a serious concern in TS, and both are seen
show a few medullary cysts [155]. in children, sometimes in early childhood
[157, 158]. All children with TS should have a
renal ultrasound at diagnosis and follow-up renal
ultrasounds every 1–3 years, with frequency dic-
tated by the specific clinical situation
[159]. Patients with extensive or rapidly changing
lesions require more frequent follow-up. Those
with more severe kidney disease may require CT
or MRI to screen for malignant changes. Differ-
entiating angiomyolipomas from malignancy
requires careful comparison of sequential images.

Infections of the Urinary Tract

Clinical and laboratory criteria do not reliably


differentiate pyelonephritis from cystitis
[160–162]. DMSA is the gold standard for diag-
nosing acute pyelonephritis, although MRI and
CT provide similar sensitivity [163]. US may
Fig. 22 Voiding cystourethrogram shows dilatation of the detect loss of corticomedullary differentiation
posterior urethra (asteric) in a boy with posterior urethral
and renal enlargement, but lacks adequate
valves

Fig. 23 Renal US shows


dilatation of the right renal
pelvis, calices, and ureter in
a boy with posterior urethral
valves
23 Diagnostic Imaging of the Child with Suspected Renal Disease 687

Fig. 24 Voiding
cystourethrogram shows
bilateral grade
4 vesicoureteral reflux

Fig. 25 Autosomal
recessive polycystic kidney
disease in an infant. (a)
Longitudinal view shows
diffuse enlargement of the
right kidney, which is in
increased in echogenicity.
(b) Transverse image
through the mid-pole of
both kidneys

sensitivity and specificity when compared to from renal scarring, which is associated with
DMSA [164–167]. sharper borders and loss of cortical volume.
The decreased uptake using DMSA with DMSA may allow diagnosis of pyelonephritis in
pyelonephritis may be focal, multifocal, or dif- children who have received antibiotics prior to
fuse. Acute pyelonephritis must be differentiated urine culture or who have a negative culture
688 J. Loewen and L.A. Greenbaum

Fig. 26 Autosomal
dominant polycystic kidney
disease. Renal US,
longitudinal image, and
bilateral renal cysts of
varying sizes in a 15-year-
old

Fig. 27 CT in a 10-year-
old with autosomal
dominant polycystic kidney
disease and bilateral cysts of
varying sizes. Left kidney
complicated by cyst
hemorrhage and perinephric
hematoma (arrows)

Fig. 28 Angiomyolipomas
in tuberous sclerosis. Renal
US, longitudinal image,
demonstrates multiple small
echogenic lesions
throughout the kidney
23 Diagnostic Imaging of the Child with Suspected Renal Disease 689

Fig. 29 Angiomyolipomas
in tuberous sclerosis. Renal
CT, axial image, with
multiple fat-containing
lesions in the kidneys

Fig. 30 Renal scarring in a


10-year-old with
vesicoureteral reflux.
DMSA renal scan shows a
small right kidney with
diffuse renal scarring.
Uptake in the right kidney is
20 %

despite clinical and laboratory evidence 6 months after an acute infection to


suggesting pyelonephritis [168]. differentiate a transient lesion due to infection
DMSA is the current standard modality for from a scar [101, 170, 171]. MRI has the potential
diagnosing renal scarring (Fig. 30; [169]), with to differentiate acute pyelonephritis from renal
increased sensitivity when compared to US or IVP scarring, perhaps avoiding the need for radiation
[170]. Scars appear as photopenic areas with vol- exposure and obviating the need for two diagnos-
ume loss. DMSA should be deferred at least tic tests [172].
690 J. Loewen and L.A. Greenbaum

Fig. 31 Acute
glomerulonephritis in a
3-year-old. Renal US shows
the large echogenic right
kidney

All boys and young girls with a first UTI are Renal parenchymal disease is often associated
conventionally evaluated with a renal US and with increased echogenicity (Fig. 31;
VCUG [173]. The necessity of this approach has [178–180]), although a normal US examination
been questioned, especially given the lack of evi- does not exclude this possibility. The kidneys
dence that prophylactic antibiotics are effective in generally have a normal appearance in prerenal
preventing UTIs or renal damage [174]. Moreover, azotemia. The kidneys may be normal or have
because of the widespread use of prenatal US in increased echogenicity in acute tubular necrosis.
many countries, the diagnostic yield of US exam- Echogenic kidneys, which may be moderately
ination in children with UTI is low [175]. In girls, enlarged, are often seen in glomerular disease
the radiation burden of the initial imaging can be [179, 181]. The kidneys are enlarged and very
reduced by using RNC instead of VCUG. echogenic in HIV nephropathy [182]. The US
An alternative strategy is to perform DMSA scin- findings in renal parenchymal disease are rarely
tigraphy as an initial test in children with UTI and specific [180, 181]. US is commonly used for
only perform VCUG or RNC in children with an pre-biopsy localization [32].
abnormal DMSA [176].

Renal Transplant
Renal Failure
US is the primary imaging modality for evaluation
US is the initial imaging modality when a patient of the renal transplant [183, 184]. US can identify
presents with unexplained renal failure. Imaging posttransplant complications, including abnormal
must include assessment of renal size, fluid collections (e.g., lymphoceles, urinomas)
echogenicity, morphology, cystic disease, and (Fig. 32), obstruction, and mass lesions
hydronephrosis [32, 177]. US generally allows [32, 185]. Doppler US can detect vascular disease
differentiation of chronic renal failure (small, after kidney transplant (renal artery thrombosis,
echogenic kidneys with a thin cortex) from acute renal artery stenosis, renal vein thrombosis, arte-
renal failure. The kidneys may be large if chronic riovenous fistula) [27, 184, 186, 187]. There are a
renal failure is secondary to polycystic kidney number of potential US abnormalities seen in
disease. US readily detects hydronephrosis, patients with acute graft rejection (increased
which suggests that renal failure is due to an renal size and echogenicity, decreased or absent
obstructive etiology. Hydronephrosis may also central sinus echoes) [188], but US does not have
be secondary to VUR, papillary necrosis, or high adequate sensitivity or specificity; kidney biopsy
urine output; such hydronephrosis is rarely severe. remains necessary for the diagnosis of rejection.
23 Diagnostic Imaging of the Child with Suspected Renal Disease 691

Fig. 32 Lymphocele in a
15-year-old with renal
transplant. Renal US shows
a complex, loculated fluid
mass adjacent (L ) to the
transplanted kidney (T )

Acute rejection, vascular thrombosis, and acute likely secondary to the US beam interacting with
tubular necrosis (ATN) are the most common stone, can also assist in calculus detection
complications in the immediate postoperative [32]. US is less sensitive than CT in detecting
period. Long-term complications include arterial calculi in symptomatic children (Fig. 33; [193]).
stenosis, ureteral stenosis, chronic rejection, and CT is also superior to US for identification of
posttransplant lymphoproliferative disorder bladder stones, especially if the bladder is
[32, 189]. augmented.
Radionuclide imaging (99mTc-DTPA or 99mTc-
MAG3) is useful when US does not provide an
explanation for graft dysfunction. Absence of Nephrocalcinosis
flow to the kidney occurs with arterial or venous
obstruction or with hyperacute rejection. In con- Nephrocalcinosis is typically detected by US
trast, a graft with ATN has normal renal perfusion, (Fig. 34), although it may be seen via CT. Plain
but delayed or no excretion. Radionuclide imag- x-ray only detects nephrocalcinosis in severe
ing is useful for demonstrating urinary obstruction cases [194]. Causes of cortical nephrocalcinosis
or that a perinephric fluid collection is due to a include primary hyperoxaluria [195] and cortical
urine leak [190–192]. necrosis, while there are many entities that may
cause medullary nephrocalcinosis [194, 196].

Urolithiasis and Nephrocalcinosis


Trauma to the Urinary Tract
Urolithiasis
CT with contrast is the primary modality for eval-
US is a sensitive test for detecting calculi in the uating a patient with suspected trauma to the kid-
kidney and the proximal and distal ureter. US does ney (Fig. 35; [197, 198]). CT angiography permits
not easily detect stones in the middle portion of detailed evaluation of the renal artery and vein.
the ureter, although hydronephrosis due to US is an alternative imaging modality that may
obstruction may suggest the diagnosis. Acoustic identify large lacerations, hematomas, and
shadowing is a useful finding, but may not be urinomas; US with Doppler readily identifies
present with smaller calculi. Twinkle artifact, significant injuries of the renal artery or vein.
692 J. Loewen and L.A. Greenbaum

Fig. 33 Renal calculus in the mid-pole of the left kidney. (a) Coronal image. (b) Axial image

Fig. 34 Medullary
nephrocalcinosis in a
12-year-old with renal
tubular acidosis. Renal US,
longitudinal image, shows
echogenic renal pyramids

US suggests bladder laceration when there is a contrast allows for identification of tumor calcifi-
large amount of fluid in the cul-de-sac. When cations; contrast allows for better differentiation
indicated, VCUG can potentially identify the site of tumor from normal tissue. US is sensitive for
of bladder leak and detect urethral injury. the detection of bladder tumors in children [199].

Renal and Urinary Tract Tumors Imaging of Specific Renal Tumors

Role of Imaging Wilms Tumor


US and CT are complementary in the evaluation
US is especially suited for initial evaluation, while and follow-up of Wilms tumor (Figs. 36 and 37).
CT or MRI is necessary for defining tumor extent US with Doppler may provide superior imaging of
and the presence of metastases. CT without tumor in the inferior vena cava in some patients
23 Diagnostic Imaging of the Child with Suspected Renal Disease 693

Fig. 35 Motor vehicle collision resulting in extensive left renal laceration with a large perinephric hematoma. (a) Axial
CT. (b) Coronal CT

[200]. Radiologic staging of Wilms tumor echogenicity [205]. CT may demonstrate “con-
includes chest x-ray, US of the abdomen and trast inversion” of the lesions in renal lymphoma:
pelvis, and CT of the chest, abdomen, and pelvis. increased density compared to normal paren-
Chest x-ray appears to be an appropriate substitute chyma without contrast and decreased density
for chest CT to identify pulmonary metastases with contrast [205].
[201]. MRI is also an effective modality for eval-
uation of Wilms tumor [202]. US is a sensitive Vascular Disease and Hypertension
approach for screening patients for tumor recur-
rence, although CT or MRI is necessary to identify Renal Artery Stenosis
nephrogenic rests [203]. Doppler US lacks adequate sensitivity for diag-
US is the preferred modality for Wilms tumor nosing renal artery stenosis in children [206–208],
screening, which is necessary in children with although it may be a useful screening test in adults
syndromes that significantly increase the risk of [209]. US may demonstrate a small affected
Wilms tumor (e.g., hemihypertrophy, Denys- kidney if the stenosis is severe and long-standing;
Drash syndrome, Beckwith-Wiedemann syn- the contralateral kidney may have increased
drome) [204]. Screening frequency and duration echogenicity [210]. Renal scintigraphy before
varies depending on the syndrome. and after captopril has good sensitivity in identi-
fying renovascular disease in children [106]. CT
Lymphoma and MRI angiography provide excellent visuali-
Children with lymphoma may have renal involve- zation of the extraparenchymal renal artery
ment. By US, renal lymphoma has decreased (Fig. 38), the most common site of disease, but
694 J. Loewen and L.A. Greenbaum

Fig. 36 Bilateral Wilms tumor in a 5-year-old with a palpable abdominal mass. (a) Axial CT. (b) Coronal CT

do not visualize distal branch vessel lesions from a normal-appearing kidney to a small,
[65]. When clinical suspicion is high, conven- echogenic kidney with little or no function. In
tional angiography remains necessary to evaluate perinatal renal vein thrombosis, renal length is
children with suspected hypertension due to renal negatively correlated with outcome [214].
artery disease. US may directly demonstrate clots in the renal
vein and inferior vena cava. Doppler US may
Renal Vein Thrombosis show absent renal venous flow and pulsatility.
US is utilized to diagnose renal vein thrombosis. The arterial RI is frequently elevated. Clot retrac-
The appearance on US varies depending on the tion and formation of collaterals after a few days
timing of imaging. Within the first few days of clot may create diagnostic confusion due to a return of
formation, imaging may demonstrate pathogno- some venous flow and a decrease in arterial RI. In
monic echogenic streaks due to clot in interlobular neonates, adrenal hemorrhage, which is readily
and interlobar veins [211, 212]. Renal enlarge- seen by US, frequently accompanies renal vein
ment occurs during the first week and is associated thrombosis and assists in confirming the diagnosis
with an echogenic cortex and less echogenic med- [215, 216]. This association is more common on
ullary pyramids. Beyond the first week, there is a the left due to the common drainage of the renal
loss of corticomedullary differentiation, and vein and adrenal vein.
echogenic streaks are no longer visible [211]. An
additional later finding is calcification of thrombi Pheochromocytoma
within renal veins, including large or small Pheochromocytoma may be strongly suspected
veins [213]. Long-term imaging outcomes vary based on clinical and laboratory evaluation, but
23 Diagnostic Imaging of the Child with Suspected Renal Disease 695

Fig. 37 Wilms tumor in a 2-year-old. (a) Coronal CT Longitudinal US image with tumor filling the dilated IVC
image shows a large right renal mass, pulmonary nodules, extending to the diaphragm (arrows) and entering the right
and tumor extension into the IVC to the diaphragm. (b) atrium (a)

Fig. 38 Renal artery


stenosis in a 13-year-old
with neurofibromatosis and
hypertension. Axial
maximal intensity CT
image shows narrowing of
both renal arteries near their
origins from the aorta

imaging is necessary to confirm the diagnosis and lesions or tumors in other locations (Fig. 39).
allow for staging and resection, which is usually Hypertensive crisis may occur if ionic contrast is
curative. US may identify a pheochromocytoma utilized, but nonionic contrast appears safe at the
in or near the adrenal gland, but US lacks the doses used for CT [217]. Chest CT or MRI is
sensitivity of CT or MRI, especially for small indicated if an abdominal lesion is not detected
696 J. Loewen and L.A. Greenbaum

Fig. 39 Pheochromocytoma in a 14-year-old. (a) Axial CT image shows a right pelvic mass (P). (b) Sagittal T2-weighted
MR image shows a right pelvic mass extending into the adjacent neural foramen (arrow)

MIBG scintigraphy, a complementary imaging


modality, may be appropriate as an initial test, to
confirm the diagnosis in equivocal cases or to
search for disease when the CT or MRI is negative
(Fig. 40; [218, 219]). Positron emission tomogra-
phy (PET) scan is a potential alternative if all
other studies are negative and clinical suspicion
remains high [220].

References
1. Darge K, Grattan-Smith JD, Riccabona M. Pediatric
uroradiology: state of the art. Pediatr Radiol.
2011;41(1):82–91.
2. Chiara A, Chirico G, Barbarini M, De Vecchi E,
Rondini G. Ultrasonic evaluation of kidney length in
term and preterm infants. Eur J Pediatr. 1989;149(2):
94–5.
3. Cohen HL, Cooper J, Eisenberg P, Mandel FS, Gross
BR, Goldman MA, et al. Normal length of fetal kid-
neys: sonographic study in 397 obstetric patients.
AJR Am J Roentgenol. 1991;157(3):545–8.
4. Blane CE, Bookstein FL, DiPietro MA, Kelsch RC.
Sonographic standards for normal infant kidney
length. AJR Am J Roentgenol. 1985;145(6):1289–91.
Fig. 40 Pheochromocytoma. Posterior 123I-MIBG image 5. Fitzsimons RB. Kidney length in the newborn mea-
shows tumor uptake in the pelvis sured by ultrasound. Acta Paediatr Scand. 1983;
72(6):885–7.
6. Han BK, Babcock DS. Sonographic measurements
or for staging of malignant disease. Pheochromo- and appearance of normal kidneys in children. AJR
Am J Roentgenol. 1985;145(3):611–6.
cytomas enhance on contrast CT, although 7. Rosenbaum DM, Korngold E, Teele RL. Sonographic
nonenhancing foci of necrosis and hemorrhage assessment of renal length in normal children. AJR
may be present. Am J Roentgenol. 1984;142(3):467–9.
23 Diagnostic Imaging of the Child with Suspected Renal Disease 697

8. Carrico CW, Zerin JM. Sonographic measurement of 25. Middleton WD, Kellman GM, Melson GL, Madrazo
renal length in children: does the position of the BL. Postbiopsy renal transplant arteriovenous fistu-
patient matter? Pediatr Radiol. 1996;26(8):553–5. las: color Doppler US characteristics. Radiology.
9. De Sanctis JT, Connolly SA, Bramson RT. Effect of 1989;171(1):253–7.
patient position on sonographically measured renal 26. Riccabona M, Schwinger W, Ring E. Arteriovenous
length in neonates, infants, and children. AJR Am J fistula after renal biopsy in children. J Ultrasound
Roentgenol. 1998;170(5):1381–3. Med. 1998;17(8):505–8.
10. Haugstvedt S, Lundberg J. Kidney size in normal 27. Mutze S, Turk I, Schonberger B, Filimonow SI,
children measured by sonography. Scand J Urol Bollow M, Petersein J, et al. Colour-coded duplex
Nephrol. 1980;14(3):251–5. sonography in the diagnostic assessment of vascular
11. Dinkel E, Ertel M, Dittrich M, Peters H, Berres M, complications after kidney transplantation in chil-
Schulte-Wissermann H. Kidney size in childhood. dren. Pediatr Radiol. 1997;27(12):898–902.
Sonographical growth charts for kidney length and 28. Kuzmic AC, Brkljacic B, Ivankovic D, Galesic K.
volume. Pediatr Radiol. 1985;15(1):38–43. Doppler sonographic renal resistance index in healthy
12. Chen JJ, Pugach J, Patel M, Luisiri A, Steinhardt GF. children. Eur Radiol. 2000;10(10):1644–8.
The renal length nomogram: multivariable approach. 29. Bude RO, Rubin JM, Adler RS. Power versus con-
J Urol. 2002;168(5):2149–52. ventional color Doppler sonography: comparison in
13. Emamian SA, Nielsen MB, Pedersen JF, Ytte L. the depiction of normal intrarenal vasculature. Radi-
Kidney dimensions at sonography: correlation with ology. 1994;192(3):777–80.
age, sex, and habitus in 665 adult volunteers. AJR Am 30. Riccabona M, Ring E, Schwinger W, Aigner R.
J Roentgenol. 1993;160(1):83–6. Amplitude coded-colour Doppler sonography in
14. Schlesinger AE, Hernandez RJ, Zerin JM, Marks TI, paediatric renal disease. Eur Radiol. 2001;11(5):
Kelsch RC. Interobserver and intraobserver variations 861–6.
in sonographic renal length measurements in children. 31. Dacher JN, Pfister C, Monroc M, Eurin D, LeDosseur
AJR Am J Roentgenol. 1991;156(5):1029–32. P. Power Doppler sonographic pattern of acute pyelo-
15. Khazaei MR, Mackie F, Rosenberg AR, Kainer G. nephritis in children: comparison with CT. AJR Am J
Renal length discrepancy by ultrasound is a reliable Roentgenol. 1996;166(6):1451–5.
predictor of an abnormal DMSA scan in children. 32. Carmichael J, Easty M. Imaging chronic renal disease
Pediatr Nephrol. 2008;23(1):99–105. and renal transplant in children. Pediatr Radiol.
16. Hricak H, Slovis TL, Callen CW, Callen PW, 2010;40(6):963–74.
Romanski RN. Neonatal kidneys: sonographic ana- 33. Darge K. Diagnosis of vesicoureteral reflux with
tomic correlation. Radiology. 1983;147(3):699–702. ultrasonography. Pediatr Nephrol. 2002;17(1):52–60.
17. Erwin BC, Carroll BA, Muller H. A sonographic 34. Darge K. Voiding urosonography with US contrast
assessment of neonatal renal parameters. J Ultrasound agents for the diagnosis of vesicoureteric reflux in
Med. 1985;4(5):217–20. children. II. Comparison with radiological examina-
18. Wiersma F, Toorenvliet BR, Ruige M, Holscher HC. tions. Pediatr Radiol. 2008;38(1):54–63.
Increased echogenicity of renal cortex: a transient 35. Berrocal T, Gaya F, Arjonilla A, Lonergan GJ.
feature in acutely ill children. AJR Am J Roentgenol. Vesicoureteral reflux: diagnosis and grading
2008;190(1):240–3. with echo-enhanced cystosonography versus
19. Fernbach SK, Maizels M, Conway JJ. Ultrasound voiding cystourethrography. Radiology. 2001;
grading of hydronephrosis: introduction to the system 221(2):359–65.
used by the Society for Fetal Urology. Pediatr Radiol. 36. Darge K, Troeger J, Duetting T, Zieger B,
1993;23(6):478–80. Rohrschneider W, Moehring K, et al. Reflux in
20. Maizels M, Reisman ME, Flom LS, Nelson J, young patients: comparison of voiding US of the
Fernbach S, Firlit CF, et al. Grading nephroureteral bladder and retrovesical space with echo enhance-
dilatation detected in the first year of life: correlation ment versus voiding cystourethrography for diagno-
with obstruction. J Urol. 1992;148(2 Pt 2):609–14. sis. Radiology. 1999;210(1):201–7.
21. Yamazaki Y, Yago R, Toma H. Sonographic charac- 37. Bosio M. Cystosonography with echocontrast: a new
teristics of the urinary tract in healthy neonates. imaging modality to detect vesicoureteric reflux in
J Urol. 2001;166(3):1054–7. children. Pediatr Radiol. 1998;28(4):250–5.
22. Jequier S, Rousseau O. Sonographic measurements 38. Valentini AL, Salvaggio E, Manzoni C, Rendeli C,
of the normal bladder wall in children. AJR Am Destito C, Summaria V, et al. Contrast-enhanced
J Roentgenol. 1987;149(3):563–6. gray-scale and color Doppler voiding urosonography
23. Hiraoka M, Hori C, Tsuchida S, Sudo M. versus voiding cystourethrography in the diagnosis
Ultrasonographic evaluation of bladder volume in and grading of vesicoureteral reflux. J Clin Ultra-
young children. Pediatr Nephrol. 1995;9(3):351–3. sound. 2001;29(2):65–71.
24. Hiraoka M, Tsukahara H, Tsuchida S, Hori C, Sudo 39. O'Neill WC. Renal relevant radiology: use of ultra-
M. Ultrasonographic evaluation of bladder volume in sound in kidney disease and nephrology procedures.
children. Pediatr Nephrol. 1993;7(5):533–5. Clin J Am Soc Nephrol. 2014;9(2):373–81.
698 J. Loewen and L.A. Greenbaum

40. Kleinman PK, Diamond DA, Karellas A, Spevak MR, 55. Kalra MK, Singh S, Blake MA. CT of the urinary
Nimkin K, Belanger P. Tailored low-dose fluoro- tract: turning attention to radiation dose. Radiol Clin
scopic voiding cystourethrography for the North Am. 2008;46(1):1–9.
reevaluation of vesicoureteral reflux in girls. AJR 56. Goske MJ, Applegate KE, Boylan J, Butler PF,
Am J Roentgenol. 1994;162(5):1151–4. Callahan MJ, Coley BD, et al. The image gently
41. Hernandez RJ, Goodsitt MM. Reduction of radiation campaign: working together to change practice. AJR
dose in pediatric patients using pulsed fluoroscopy. Am J Roentgenol. 2008;190(2):273–4.
AJR Am J Roentgenol. 1996;167(5):1247–53. 57. Siegel MJ, Luker GD. Pediatric applications of helical
42. Weese DL, Greenberg HM, Zimmern PE. Contrast (spiral) CT. Radiol Clin North Am. 1995;33(5):
media reactions during voiding cystourethrography 997–1022.
or retrograde pyelography. Urology. 1993;41(1): 58. Brenner DJ, Hall EJ. Computed tomography–an
81–4. increasing source of radiation exposure. N Engl J
43. Vates TS, Shull MJ, Underberg-Davis SJ, Fleisher Med. 2007;357(22):2277–84.
MH. Complications of voiding cystourethrography 59. White KS. Invited article: helical/spiral CT scanning:
in the evaluation of infants with prenatally detected a pediatric radiology perspective. Pediatr Radiol.
hydronephrosis. J Urol. 1999;162(3 Pt 2):1221–3. 1996;26(1):5–14.
44. Schmit P, Sfez M. Management of anxious and pain- 60. Pappas JN, Donnelly LF, Frush DP. Reduced fre-
ful manifestations in pediatric uroradiology. J Radiol. quency of sedation of young children with
1997;78(5):367–72. multisection helical CT. Radiology. 2000;215(3):
45. Ljungman G, Kreuger A, Andreasson S, Gordh T, 897–9.
Sorensen S. Midazolam nasal spray reduces 61. Frush DP, Donnelly LF. Helical CT in children: tech-
procedural anxiety in children. Pediatrics. 2000; nical considerations and body applications. Radiol-
105(1 Pt 1):73–8. ogy. 1998;209(1):37–48.
46. Elder JS, Longenecker R. Premedication with oral 62. Glockner JF, Vrtiska TJ. Renal MR and CT angiogra-
midazolam for voiding cystourethrography in chil- phy: current concepts. Abdom Imaging. 2007;32(3):
dren: safety and efficacy. AJR Am J Roentgenol. 407–20.
1995;164(5):1229–32. 63. Rountas C, Vlychou M, Vassiou K, Liakopoulos V,
47. Lebowitz RL, Olbing H, Parkkulainen KV, Smellie Kapsalaki E, Koukoulis G, et al. Imaging modalities
JM, Tamminen-Mobius TE. International system of for renal artery stenosis in suspected renovascular
radiographic grading of vesicoureteric reflux. Interna- hypertension: prospective intraindividual comparison
tional Reflux Study in Children. Pediatr Radiol. of color Doppler US, CT angiography, GD-enhanced
1985;15(2):105–9. MR angiography, and digital subtraction angiogra-
48. Jequier S, Jequier JC. Reliability of voiding cystoure- phy. Ren Fail. 2007;29(3):295–302.
thrography to detect reflux. AJR Am J Roentgenol. 64. Riccabona M. Urinary tract imaging in infancy.
1989;153(4):807–10. Pediatr Radiol. 2009;39 Suppl 3:436–45.
49. Paltiel HJ, Rupich RC, Kiruluta HG. Enhanced detec- 65. Kurian J, Epelman M, Darge K, Meyers K, Nijs E,
tion of vesicoureteral reflux in infants and children Hellinger JC. The role of CT angiography in the
with use of cyclic voiding cystourethrography. Radi- evaluation of pediatric renovascular hypertension.
ology. 1992;184(3):753–5. Pediatr Radiol. 2013;43(4):490–501. quiz 487–9.
50. Andresen R, Wegner HE. Intravenous urography 66. Linebarger JS, Roy ML. Focus on diagnosis: common
revisited in the age of ultrasound and computerized nuclear medicine studies in pediatrics. Pediatr Rev.
tomography: diagnostic yield in cases of renal colic, 2007;28(11):415–7.
suspected pelvic and abdominal malignancies, 67. He W, Fischman AJ. Nuclear imaging in the genito-
suspected renal mass, and acute pyelonephritis. Urol urinary tract: recent advances and future directions.
Int. 1997;58(4):221–6. Radiol Clin North Am. 2008;46(1):25–43. v.
51. Bailey SR, Tyrrell PN, Hale M. A trial to assess 68. Taylor AT. Radionuclides in nephrourology, part 1:
the effectiveness of bowel preparation prior to Radiopharmaceuticals, quality control, and quantita-
intravenous urography. Clin Radiol. 1991;44(5): tive indices. J Nucl Med. 2014;55(4):608–15.
335–7. 69. Conway JJ, Maizels M. The “well tempered” diuretic
52. Bradley AJ, Taylor PM. Does bowel preparation renogram: a standard method to examine the asymp-
improve the quality of intravenous urography? Br J tomatic neonate with hydronephrosis or hydroureter-
Radiol. 1996;69(826):906–9. onephrosis. A report from combined meetings of The
53. Stokland E, Hellstrom M, Hansson S, Jodal U, Society for Fetal Urology and members of The Pedi-
Oden A, Jacobsson B. Reliability of ultrasonography atric Nuclear Medicine Council–The Society of
in identification of reflux nephropathy in children. Nuclear Medicin. J Nucl Med. 1992;33(11):2047–51.
BMJ. 1994;309(6949):235–9. 70. O'Reilly P, Aurell M, Britton K, Kletter K,
54. Smellie JM. The intravenous urogram in the detection Rosenthal L, Testa T. Consensus on diuresis renogra-
and evaluation of renal damage following urinary phy for investigating the dilated upper urinary tract.
tract infection. Pediatr Nephrol. 1995;9(2):213–9. Radionuclides in Nephrourology Group Consensus
23 Diagnostic Imaging of the Child with Suspected Renal Disease 699

Committee on Diuresis Renography. J Nucl Med. 86. Ladron De Guevara D, Franken P, De Sadeleer C,
1996;37(11):1872–6. Ham H, Piepsz A. Interobserver reproducibility in
71. Kass EJ, Majd M, Belman AB. Comparison of the reporting on 99mTc-DMSA scintigraphy for detec-
diuretic renogram and the pressure perfusion study in tion of late renal sequelae. J Nucl Med. 2001;42(4):
children. J Urol. 1985;134(1):92–6. 564–6.
72. English PJ, Testa HJ, Gosling JA, Cohen SJ. Idio- 87. De Sadeleer C, Tondeur M, Melis K, Van Espen MB,
pathic hydronephrosis in childhood–a comparison Verelst J, Ham H, et al. A multicenter trial on
between diuresis renography and upper urinary tract interobserver reproducibility in reporting on 99mTc-
morphology. Br J Urol. 1982;54(6):603–7. DMSA planar scintigraphy: a Belgian survey. J Nucl
73. Wong DC, Rossleigh MA, Farnsworth RH. Med. 2000;41(1):23–6.
F + 0 diuresis renography in infants and children. J 88. van Luyk WH, Ensing GJ, Piers DA. Low renal
Nucl Med. 1999;40(11):1805–11. uptake of 99mTc-DMSA in patients with proximal
74. Eising EG, Bonzel KE, Zander C, Farahati J, Reiners tubular dysfunction. Eur J Nucl Med. 1983;8(9):
C. Value of diuresis renography in the post-natal 404–5.
period of assumed physiological renal immaturity. 89. Takeda M, Katayama Y, Tsutsui T, Takahashi H,
Nucl Med Commun. 1997;18(11):1008–16. Komeyama T, Mizusawa T, et al. Value of dimercap-
75. Wong JC, Rossleigh MA, Farnsworth RH. Utility of tosuccinic acid single photon emission computed
technetium-99m-MAG3 diuretic renography in the tomography and magnetic resonance imaging in
neonatal period. J Nucl Med. 1995;36(12):2214–9. detecting renal injury in pediatric patients with
76. Gordon I, Colarinha P, Fettich J, Fischer S, Frokier J, vesicoureteral reflux. Comparison with dimercapto-
Hahn K, et al. Guidelines for standard and diuretic succinic acid planar scintigraphy and
renography in children. Eur J Nucl Med. 2001;28(3): intravenous pyelography. Eur Urol. 1994;25(4):
BP21–30. 320–5.
77. Anderson PJ, Rangarajan V, Gordon I. Assessment of 90. Itoh K, Yamashita T, Tsukamoto E, Nonomura K,
drainage in PUJ dilatation: pelvic excretion efficiency Furudate M, Koyanagi T. Qualitative and quantitative
as an index of renal function. Nucl Med Commun. evaluation of renal parenchymal damage by 99mTc-
1997;18(9):823–6. DMSA planar and SPECT scintigraphy. Ann Nucl
78. Saunders CA, Choong KK, Larcos G, Farlow D, Med. 1995;9(1):23–8.
Gruenewald SM. Assessment of pediatric 91. De Sadeleer C, Bossuyt A, Goes E, Piepsz A. Renal
hydronephrosis using output efficiency. J Nucl Med. technetium-99m-DMSA SPECT in normal volun-
1997;38(9):1483–6. teers. J Nucl Med. 1996;37(8):1346–9.
79. Chaiwatanarat T, Padhy AK, Bomanji JB, Nimmon 92. Craig JC, Wheeler DM, Irwig L, Howman-Giles
CC, Sonmezoglu K, Britton KE. Validation of renal RB. How accurate is dimercaptosuccinic acid scintig-
output efficiency as an objective quantitative param- raphy for the diagnosis of acute pyelonephritis? A
eter in the evaluation of upper urinary tract obstruc- meta-analysis of experimental studies. J Nucl Med.
tion. J Nucl Med. 1993;34(5):845–8. 2000;41(6):986–93.
80. Rossleigh MA, Leighton DM, Farnsworth RH. Diure- 93. Rossleigh MA. The interrenicular septum. A normal
sis renography. The need for an additional view after anatomical variant seen on DMSA SPECT. Clin Nucl
gravity-assisted drainage. Clin Nucl Med. 1993; Med. 1994;19(11):953–5.
18(3):210–3. 94. Tondeur M, Melis K, De Sadeleer C, Verelst J, Van
81. Gordon I, Mialdea-Fernandez RM, Peters Espen MB, Ham H, et al. Inter-observer reproducibil-
AM. Pelviureteric junction obstruction. The value of ity of relative 99Tcm-DMSA uptake. Nucl Med
a post-micturition view in 99mTc DTPA diuretic Commun. 2000;21(5):449–53.
renography. Br J Urol. 1988;61(5):409–12. 95. Manger WM, Eisenhofer G. Pheochromocytoma:
82. Jones DA, Lupton EW, George NJ. Effect of bladder diagnosis and management update. Curr Hypertens
filling on upper tract urodynamics in man. Br J Urol. Rep. 2004;6(6):477–84.
1990;65(5):492–6. 96. Lenders JW, Eisenhofer G, Mannelli M, Pacak K.
83. Piepsz A, Colarinha P, Gordon I, Hahn K, Olivier P, Phaeochromocytoma. Lancet. 2005;366(9486):
Roca I, et al. Guidelines for 99mTc-DMSA scintigra- 665–75.
phy in children. Eur J Nucl Med. 2001;28(3): 97. Avram AM, Fig LM, Gross MD. Adrenal gland scin-
BP37–41. tigraphy. Semin Nucl Med. 2006;36(3):212–27.
84. Mandell GA, Eggli DF, Gilday DL, Heyman S, Leon- 98. Stabin MG, Gelfand MJ. Dosimetry of pediatric
ard JC, Miller JH, et al. Procedure guideline for renal nuclear medicine procedures. Q J Nucl Med.
cortical scintigraphy in children. Society of Nuclear 1998;42(2):93–112.
Medicine. J Nucl Med. 1997;38(10):1644–6. 99. D'Errico G. The role of nuclear medicine in evalua-
85. Mannes F, Bultynck E, Van Roijen N, Van Der tion of vesicoureteral reflux and/or reflux nephropa-
Mauten L, Piepsz A, Ham HR. Utility of posterior thy. Rays. 2002;27(2):149–54.
oblique views in (99m)Tc-DMSA renal scintigraphy 100. Unver T, Alpay H, Biyikli NK, Ones T. Comparison
in children. J Nucl Med Technol. 2003;31(2):72–3. of direct radionuclide cystography and voiding
700 J. Loewen and L.A. Greenbaum

cystourethrography in detecting vesicoureteral reflux. 115. Ersoy H, Rybicki FJ. Biochemical safety profiles of
Pediatr Int. 2006;48(3):287–91. gadolinium-based extracellular contrast agents and
101. Piepsz A, Ham HR. Pediatric applications of renal nephrogenic systemic fibrosis. J Magn Reson Imag-
nuclear medicine. Semin Nucl Med. 2006;36(1): ing. 2007;26(5):1190–7.
16–35. 116. Grattan-Smith JD, Little SB, Jones RA. MR
102. Taylor AT. Radionuclides in nephrourology, Part 2: urography in children: how we do it. Pediatr Radiol.
pitfalls and diagnostic applications. J Nucl Med. 2008;38 Suppl 1:S3–17.
2014;55(5):786–98. 117. Ehammer T, Riccabona M, Maier E. High resolution
103. Pattaras JG, Rushton HG, Majd M. The role of 99m MR for evaluation of lower urogenital tract
technetium dimercapto-succinic acid renal scans in malformations in infants and children: feasibility and
the evaluation of occult ectopic ureters in girls with preliminary experiences. Eur J Radiol. 2011;78(3):
paradoxical incontinence. J Urol. 1999;162(3 Pt 1): 388–93.
821–5. 118. Figueroa VH, Chavhan GB, Oudjhane K, Farhat
104. Gharagozloo AM, Lebowitz RL. Detection of a W. Utility of MR urography in children suspected of
poorly functioning malpositioned kidney with single having ectopic ureter. Pediatr Radiol. 2014;44(8):
ectopic ureter in girls with urinary dribbling: imaging 956–62.
evaluation in five patients. AJR Am J Roentgenol. 119. Grattan-Smith JD. MR urography: anatomy and phys-
1995;164(4):957–61. iology. Pediatr Radiol. 2008;38(2):S275–80.
105. Itoh K. 99mTc-MAG3: review of pharmacokinetics, 120. Zhang JL, Morrell G, Rusinek H, Sigmund EE,
clinical application to renal diseases and quantifica- Chandarana H, Lerman LO, et al. New magnetic
tion of renal function. Ann Nucl Med. 2001;15(3): resonance imaging methods in nephrology. Kidney
179–90. Int. 2014;85(4):768–78.
106. Lagomarsino E, Orellana P, Munoz J, Velasquez C, 121. Alon US, Ganapathy S. Should renal ultrasonography
Cavagnaro F, Valdes F. Captopril scintigraphy in the be done routinely in children with first urinary tract
study of arterial hypertension in pediatrics. Pediatr infection? Clin Pediatr. 1999;38(1):21–5.
Nephrol. 2004;19(1):66–70. 122. Miron D, Daas A, Sakran W, Lumelsky D, Koren A,
107. Prasad SR, Dalrymple NC, Surabhi VR. Cross- Horovitz Y. Is omitting post urinary-tract-infection
sectional imaging evaluation of renal masses. Radiol renal ultrasound safe after normal antenatal ultra-
Clin North Am. 2008;46(1):95–111. vi-vii. sound? An observational study. Arch Dis Child.
108. Townsend RR, Cohen DL, Katholi R, Swan SK, 2007;92(6):502–4.
Davies BE, Bensel K, et al. Safety of intravenous 123. Homsy YL, Saad F, Laberge I, Williot P, Pison
gadolinium (Gd-BOPTA) infusion in patients with C. Transitional hydronephrosis of the newborn and
renal insufficiency. Am J Kidney Dis. 2000;36(6): infant. J Urol. 1990;144(2 Pt 2):579–83.
1207–12. 124. Dejter Jr SW, Gibbons MD. The fate of infant kidneys
109. Prince MR, Arnoldus C, Frisoli JK. Nephrotoxicity of with fetal hydronephrosis but initially normal postna-
high-dose gadolinium compared with iodinated con- tal sonography. J Urol. 1989;142(2 Pt 2):661–2.
trast. J Magn Reson Imaging. 1996;6(1):162–6. 125. Lee RS, Cendron M, Kinnamon DD, Nguyen
110. Briguori C, Colombo A, Airoldi F, Melzi G, Michev I, HT. Antenatal hydronephrosis as a predictor of post-
Carlino M, et al. Gadolinium-based contrast agents natal outcome: a meta-analysis. Pediatrics. 2006;
and nephrotoxicity in patients undergoing coronary 118(2):586–93.
artery procedures. Catheter Cardiovasc Interv. 2006; 126. Yoshioka K, Takemura T, Hattori S. Tubulointerstitial
67(2):175–80. nephritis antigen: primary structure, expression
111. Sam 2nd AD, Morasch MD, Collins J, Song G, and role in health and disease. Nephron. 2002;90(1):
Chen R, Pereles FS. Safety of gadolinium contrast 1–7.
angiography in patients with chronic renal insuffi- 127. van Eerde AM, Meutgeert MH, de Jong TP, Giltay
ciency. J Vasc Surg. 2003;38(2):313–8. JC. Vesico-ureteral reflux in children with prenatally
112. Thomsen HS, Almen T, Morcos SK. Gadolinium- detected hydronephrosis: a systematic review. Ultra-
containing contrast media for radiographic examina- sound Obstet Gynecol. 2007;29(4):463–9.
tions: a position paper. Eur Radiol. 2002;12(10): 128. Chauvin NA, Epelman M, Victoria T, Johnson
2600–5. AM. Complex genitourinary abnormalities on fetal
113. Marckmann P, Skov L, Rossen K, Dupont A, Damholt MRI: imaging findings and approach to diagnosis.
MB, Heaf JG, et al. Nephrogenic systemic fibrosis: AJR Am J Roentgenol. 2012;199(2):W222–31.
suspected causative role of gadodiamide used for 129. Wille S, von Knobloch R, Klose KJ, Heidenreich A,
contrast-enhanced magnetic resonance imaging. J Hofmann R. Magnetic resonance urography in pedi-
Am Soc Nephrol. 2006;17(9):2359–62. atric urology. Scand J Urol Nephrol. 2003;37(1):
114. Sadowski EA, Bennett LK, Chan MR, Wentland AL, 16–21.
Garrett AL, Garrett RW, et al. Nephrogenic systemic 130. Zerin JM, Baker DR, Casale JA. Single-system
fibrosis: risk factors and incidence estimation. Radi- ureteroceles in infants and children: imaging features.
ology. 2007;243(1):148–57. Pediatr Radiol. 2000;30(3):139–46.
23 Diagnostic Imaging of the Child with Suspected Renal Disease 701

131. Jones RA, Perez-Brayfield MR, Kirsch AJ, Grattan- 146. Gagnadoux MF, Habib R, Levy M, Brunelle F,
Smith JD. Renal transit time with MR urography in Broyer M. Cystic renal diseases in children. Adv
children. Radiology. 2004;233(1):41–50. Nephrol Necker Hosp. 1989;18:33–57.
132. Grattan-Smith JD, Perez-Bayfield MR, Jones RA, 147. Avni FE, Guissard G, Hall M, Janssen F,
Little S, Broecker B, Smith EA, et al. MR imaging DeMaertelaer V, Rypens F. Hereditary polycystic kid-
of kidneys: functional evaluation using F-15 perfu- ney diseases in children: changing sonographic pat-
sion imaging. Pediatr Radiol. 2003;33(5):293–304. terns through childhood. Pediatr Radiol. 2002;32(3):
133. Shokeir AA, Provoost AP, el-Azab M, Dawaba M, 169–74.
Nijman RJ RJ. Renal Doppler ultrasound in children 148. Fick GM, Duley IT, Johnson AM, Strain JD, Manco-
with obstructive uropathy: effect of intravenous nor- Johnson ML, Gabow PA. The spectrum of autosomal
mal saline fluid load and furosemide. J Urol. dominant polycystic kidney disease in children. J Am
1996;156(4):1455–8. Soc Nephrol. 1994;4(9):1654–60.
134. Dillon E, Ryall A. A 10 year audit of antenatal ultra- 149. Epelman M, Victoria T, Meyers KE, Chauvin N,
sound detection of renal disease. Br J Radiol. 1998; Servaes S, Darge K. Postnatal imaging of neonates
71(845):497–500. with prenatally diagnosed genitourinary abnormali-
135. Singh I, Sharma D, Singh N, Jain BK, Minocha ties: a practical approach. Pediatr Radiol. 2012;
VR. Hydronephrotic obstructed kidney mimicking 42(1):S124–41.
a congenital multicystic kidney: case report with 150. Grantham JJ, Torres VE, Chapman AB, Guay-
review of literature. Int Urol Nephrol. 2002;34(2): Woodford LM, Bae KT, King Jr BF, et al. Volume
179–82. progression in polycystic kidney disease. N Engl J
136. Fernbach SK, Feinstein KA, Spencer K, Lindstrom Med. 2006;354(20):2122–30.
CA. Ureteral duplication and its complications. 151. Fick-Brosnahan G, Johnson AM, Strain JD,
Radiographics. 1997;17(1):109–27. Gabow PA. Renal asymmetry in children with auto-
137. Avni FE, Nicaise N, Hall M, Janssens F, Collier F, somal dominant polycystic kidney disease. Am J Kid-
Matos C, et al. The role of MR imaging for the ney Dis. 1999;34(4):639–45.
assessment of complicated duplex kidneys in chil- 152. Chung EM, Conran RM, Schroeder JW, Rohena-
dren: preliminary report. Pediatr Radiol. 2001;31(4): Quinquilla IR, Rooks VJ. From the radiologic pathol-
215–23. ogy archives: pediatric polycystic kidney disease and
138. Schulman CC. The single ectopic ureter. Eur Urol. other ciliopathies: radiologic-pathologic correlation.
1976;2(2):64–9. Radiographics. 2014;34(1):155–78.
139. Berrocal T, Pinilla I, Gutierrez J, Prieto C, de Pablo L, 153. Rahbari-Oskoui F, Mittal A, Mittal P, Chapman A.
Del Hoyo ML. Mild hydronephrosis in newborns and Renal relevant radiology: radiologic imaging in auto-
infants: can ultrasound predict the presence of somal dominant polycystic kidney disease. Clin J Am
vesicoureteral reflux. Pediatr Nephrol. 2007;22(1): Soc Nephrol. 2014;9(2):406–15.
91–6. 154. Elzouki AY, al-Suhaibani H, Mirza K, al-Sowailem
140. Foresman WH, Hulbert Jr WC, Rabinowitz R. Does AM. Thin-section computed tomography scans detect
urinary tract ultrasonography at hospitalization for medullary cysts in patients believed to have juvenile
acute pyelonephritis predict vesicoureteral reflux? nephronophthisis. Am J Kidney Dis. 1996;27(2):
J Urol. 2001;165(6 Pt 2):2232–4. 216–9.
141. Evans ED, Meyer JS, Harty MP, Bellah RD. 155. Dahan K, Fuchshuber A, Adamis S, Smaers M,
Assessment of increase in renal pelvic size on post- Kroiss S, Loute G, et al. Familial juvenile
void sonography as a predictor of vesicoureteral hyperuricemic nephropathy and autosomal dominant
reflux. Pediatr Radiol. 1999;29(4):291–4. medullary cystic kidney disease type 2: two facets of
142. Blane CE, DiPietro MA, Zerin JM, Sedman AB, the same disease? J Am Soc Nephrol. 2001;12(11):
Bloom DA. Renal sonography is not a reliable screen- 2348–57.
ing examination for vesicoureteral reflux. J Urol. 156. Sampson JR, Maheshwar MM, Aspinwall R,
1993;150(2 Pt 2):752–5. Thompson P, Cheadle JP, Ravine D, et al. Renal cystic
143. Aboutaleb H, Bolduc S, Bagli DJ, Khoury AE. disease in tuberous sclerosis: role of the polycystic
Correlation of vesicoureteral reflux with degree of kidney disease 1 gene. Am J Hum Genet. 1997;61(4):
hydronephrosis and the impact of antireflux surgery. 843–51.
J Urol. 2003;170(4 Pt 2):1560–2. 157. Ewalt DH, Sheffield E, Sparagana SP, Delgado MR,
144. Kääriäinen H, Jääskeläinen J, Kivisaari L, Roach ES. Renal lesion growth in children with
Koskimies O, Norio R. Dominant and recessive poly- tuberous sclerosis complex. J Urol. 1998;160(1):
cystic kidney disease in children: classification by 141–5.
intravenous pyelography, ultrasound, and computed 158. Al-Saleem T, Wessner LL, Scheithauer BW,
tomography. Pediatr Radiol. 1988;18(1):45–50. Patterson K, Roach ES, Dreyer SJ, et al. Malignant
145. Mercado-Deane MG, Beeson JE, John SD. US of tumors of the kidney, brain, and soft tissues in chil-
renal insufficiency in neonates. Radiographics. dren and young adults with the tuberous sclerosis
2002;22(6):1429–38. complex. Cancer. 1998;83(10):2208–16.
702 J. Loewen and L.A. Greenbaum

159. Roach ES, DiMario FJ, Kandt RS, Northrup resonance imaging for children with suspected acute
H. Tuberous sclerosis consensus conference: recom- pyelonephritis. J Urol. 2003;169(6):2308–11.
mendations for diagnostic evaluation. National 173. American Academy of Pediatrics. Practice parameter:
Tuberous Sclerosis Association. J Child Neurol. the diagnosis, treatment, and evaluation of the initial
1999;14(6):401–7. urinary tract infection in febrile infants and young
160. Jakobsson B, Soderlundh S, Berg U. Diagnostic sig- children. Pediatrics. 1999;103(4):843–52.
nificance of 99mTc-dimercaptosuccinic acid (DMSA) 174. Williams GJ, Wei L, Lee A, Craig JC. Long-term
scintigraphy in urinary tract infection. Arch Dis antibiotics for preventing recurrent urinary tract infec-
Child. 1992;67(11):1338–42. tion in children. Cochrane Database Syst Rev. 2006;3,
161. Benador D, Benador N, Slosman D, Mermillod B, CD001534.
Girardin E. Are younger children at highest risk of 175. Hoberman A, Charron M, Hickey RW, Baskin M,
renal sequelae after pyelonephritis? Lancet. 1997; Kearney DH, Wald ER. Imaging studies after a first
349(9044):17–9. febrile urinary tract infection in young children. N
162. Majd M, Rushton HG, Jantausch B, Wiedermann Engl J Med. 2003;348(3):195–202.
BL. Relationship among vesicoureteral reflux, 176. Hansson S, Dhamey M, Sigstrom O, Sixt R,
P-fimbriated Escherichia coli, and acute pyelonephri- Stokland E, Wennerstrom M, et al. Dimercapto-
tis in children with febrile urinary tract infection. succinic acid scintigraphy instead of voiding
J Pediatr. 1991;119(4):578–85. cystourethrography for infants with urinary tract
163. Majd M, Nussbaum Blask AR, Markle BM, Shalaby- infection. J Urol. 2004;172(3):1071–3.
Rana E, Pohl HG, Park JS, et al. Acute pyelonephritis: 177. Faubel S, Patel NU, Lockhart ME, Cadnapaphornchai
comparison of diagnosis with 99mTc-DMSA, MA. Renal relevant radiology: use of ultrasonogra-
SPECT, spiral CT, MR imaging, and power Doppler phy in patients with AKI. Clin J Am Soc Nephrol.
US in an experimental pig model. Radiology. 2014;9(2):382–94.
2001;218(1):101–8. 178. Hiraoka M, Hori C, Tsuchida S, Tsukahara H, Sudo
164. Mastin ST, Drane WE, Iravani A. Tc-99m DMSA M. Ultrasonographic findings of acute tubuloin-
SPECT imaging in patients with acute symptoms or terstitial nephritis. Am J Nephrol. 1996;16(2):154–8.
history of UTI. Comparison with ultrasonography. 179. Kenney PJ, Brinsko RE, Patel DV, Spitzer RE,
Clin Nucl Med. 1995;20(5):407–12. Farrar FM. Sonography of the kidneys in hemolytic
165. Hitzel A, Liard A, Vera P, Manrique A, Menard JF, uremic syndrome. Invest Radiol. 1986;21(7):547–50.
Dacher JN. Color and power Doppler sonography 180. Rosenfield AT, Siegel NJ. Renal parenchymal dis-
versus DMSA scintigraphy in acute pyelonephritis ease: histopathologic-sonographic correlation. AJR
and in prediction of renal scarring. J Nucl Med. Am J Roentgenol. 1981;137(4):793–8.
2002;43(1):27–32. 181. Stanley JH, Cornella R, Loevinger E, Schabel SI,
166. Ataei N, Madani A, Habibi R, Khorasani Curry NS. Sonography of systemic lupus nephritis.
M. Evaluation of acute pyelonephritis with DMSA AJR Am J Roentgenol. 1984;142(6):1165–8.
scans in children presenting after the age of 5 years. 182. Di Fiori JL, Rodrigue D, Kaptein EM, Ralls
Pediatr Nephrol. 2005;20(10):1439–44. PW. Diagnostic sonography of HIV-associated
167. Wang YT, Chiu NT, Chen MJ, Huang JJ, Chou HH, nephropathy: new observations and clinical correla-
Chiou YY. Correlation of renal ultrasonographic tion. AJR Am J Roentgenol. 1998;171(3):713–6.
findings with inflammatory volume from dimercapto- 183. Pozniak MA, Kelcz F, Dodd 3rd GD. Renal transplant
succinic acid renal scans in children with acute pyelo- ultrasound: imaging and Doppler. Semin Ultrasound
nephritis. J Urol. 2005;173(1):190–4. CT MR. 1991;12(4):319–34.
168. Levtchenko EN, Lahy C, Levy J, Ham HR, Piepsz A. 184. Surratt JT, Siegel MJ, Middleton WD. Sonography of
Role of Tc-99m DMSA scintigraphy in the diagnosis complications in pediatric renal allografts. Radio-
of culture negative pyelonephritis. Pediatr Nephrol. graphics. 1990;10(4):687–99.
2001;16(6):503–6. 185. Nixon JN, Biyyam DR, Stanescu L, Phillips GS, Finn
169. Arnold AJ, Brownless SM, Carty HM, Rickwood LS, Parisi MT. Imaging of pediatric renal transplants
AM. Detection of renal scarring by DMSA and their complications: a pictorial review. Radio-
scanning–an experimental study. J Pediatr Surg. graphics. 2013;33(5):1227–51.
1990;25(4):391–3. 186. Herz DB, McLorie GA, Hafez AT, Rodgers-Herz C,
170. Stokland E, Hellstrom M, Jakobsson B, Sixt El-Ghoneimi A, Shuckett B, et al. High resolution
R. Imaging of renal scarring. Acta Paediatr Suppl. ultrasound characterization of early allograft hemo-
1999;88(431):13–21. dynamics in pediatric living related renal transplanta-
171. Goldraich NP, Goldraich IH. Update on tion. J Urol. 2001;166(5):1853–8.
dimercaptosuccinic acid renal scanning in children 187. Gainza FJ, Minguela I, Lopez-Vidaur I, Ruiz LM,
with urinary tract infection. Pediatr Nephrol. 1995; Lampreabe I. Evaluation of complications due to per-
9(2):221–6. cutaneous renal biopsy in allografts and native kid-
172. Weiser AC, Amukele SA, Leonidas JC, Palmer neys with color-coded Doppler sonography. Clin
LS. The role of gadolinium enhanced magnetic Nephrol. 1995;43(5):303–8.
23 Diagnostic Imaging of the Child with Suspected Renal Disease 703

188. Slovis TL, Babcock DS, Hricak H, Han BK, Rose G, sonography in Wilms tumor. Pediatr Hematol Oncol.
McEnery P, et al. Renal transplant rejection: sono- 2002;19(2):107–15.
graphic evaluation in children. Radiology. 1984; 204. Choyke PL, Siegel MJ, Craft AW, Green DM,
153(3):659–65. DeBaun MR. Screening for Wilms tumor in children
189. Sharfuddin A. Renal relevant radiology: imaging in with Beckwith-Wiedemann syndrome or idiopathic
kidney transplantation. Clin J Am Soc Nephrol. hemihypertrophy. Med Pediatr Oncol. 1999;32(3):
2014;9(2):416–29. 196–200.
190. Nankivell BJ, Cohn DA, Spicer ST, Evans SG, Chap- 205. Ng YY, Healy JC, Vincent JM, Kingston JE,
man JR, Gruenewald SM. Diagnosis of kidney trans- Armstrong P, Reznek RH. The radiology of
plant obstruction using Mag3 diuretic renography. non-Hodgkin's lymphoma in childhood: a review of
Clin Transplant. 2001;15(1):11–8. 80 cases. Clin Radiol. 1994;49(9):594–600.
191. Goodear M, Barratt L, Wycherley A. Intraperitoneal 206. Brun P, Kchouk H, Mouchet B, Baudouin V,
urine leak in a patient with a renal transplant on Raynaud A, Loirat C, et al. Value of Doppler ultra-
Tc-99m MAG3 imaging. Clin Nucl Med. 1998; sound for the diagnosis of renal artery stenosis in
23(11):789–90. children. Pediatr Nephrol. 1997;11(1):27–30.
192. Tulchinsky M, Malpani AR, Eggli DF. Diagnosis of 207. Broekhuizen-de Gast HS, Tiel-van Buul MM, Van
urinoma by MAG3 scintigraphy in a renal transplant Beek EJ. Severe hypertension in children with
patient. Clin Nucl Med. 1995;20(1):80–1. renovascular disease. Clin Nucl Med. 2001;26(7):
193. Palmer JS, Donaher ER, O'Riordan MA, Dell 606–9.
KM. Diagnosis of pediatric urolithiasis: role of ultra- 208. Garel L, Dubois J, Robitaille P, Russo P, Filiatrault D,
sound and computerized tomography. J Urol. Grignon A, et al. Renovascular hypertension in chil-
2005;174(4 Pt 1):1413–6. dren: curability predicted with negative intrarenal
194. Nayir A, Kadioglu A, Sirin A, Emre S, Tonguc E, Doppler US results. Radiology. 1995;195(2):401–5.
Bilge I. Causes of increased renal medullary 209. Williams GJ, Macaskill P, Chan SF, Karplus TE,
echogenicity in Turkish children. Pediatr Nephrol. Yung W, Hodson EM, et al. Comparative accuracy
1995;9(6):729–33. of renal duplex sonographic parameters in the diag-
195. Akhan O, Ozmen MN, Coskun M, Ozen S, Akata D, nosis of renal artery stenosis: paired and unpaired
Saatci U. Systemic oxalosis: pathognomonic renal analysis. AJR Am J Roentgenol. 2007;188(3):
and specific extrarenal findings on US and CT. Pediatr 798–811.
Radiol. 1995;25(1):15–6. 210. Enriquez G, Castello F, Sousa P, Aso C, Lucaya
196. Shultz PK, Strife JL, Strife CF, McDaniel J. Increased cortical echogenicity of the normal kid-
JD. Hyperechoic renal medullary pyramids in infants ney in infants with unilateral renal artery stenosis:
and children. Radiology. 1991;181(1):163–7. report of two cases. J Ultrasound Med. 1997;16(1):
197. Morey AF, Bruce JE, McAninch JW. Efficacy of 59–63.
radiographic imaging in pediatric blunt renal trauma. 211. Hibbert J, Howlett DC, Greenwood KL, MacDonald
J Urol. 1996;156(6):2014–8. LM, Saunders AJ. The ultrasound appearances of
198. Stein JP, Kaji DM, Eastham J, Freeman JA, Esrig D, neonatal renal vein thrombosis. Br J Radiol.
Hardy BE. Blunt renal trauma in the pediatric popu- 1997;70(839):1191–4.
lation: indications for radiographic evaluation. Urol- 212. Lalmand B, Avni EF, Nasr A, Ketelbant P,
ogy. 1994;44(3):406–10. Struyven J. Perinatal renal vein thrombosis. Sono-
199. Dennery MP, Rushton HG, Belman AB. Sonography graphic demonstration. J Ultrasound Med. 1990;
for the detection and follow-up of primary 9(8):437–42.
nonsarcomatous bladder tumors in children. Urology. 213. Brill PW, Mitty HA, Strauss L. Renal vein thrombo-
2002;59(1):119–21. sis: a cause of intrarenal calcification in the newborn.
200. Patel K, Mahboubi S, Sherman NH, Rosenberg Pediatr Radiol. 1977;6(3):172–5.
HK. Diagnosis and follow-up of children with 214. Winyard PJ, Bharucha T, De Bruyn R, Dillon MJ,
Wilms’ tumor: correlative study of ultrasound and van't Hoff W, Trompeter RS, et al. Perinatal renal
computed tomography. Report of eighteen cases. venous thrombosis: presenting renal length predicts
Ann Radiol (Paris). 1991;34(6–7):376–82. outcome. Arch Dis Child Fetal Neonatal Ed. 2006;
201. Wootton-Gorges SL, Albano EA, Riggs JM, Ihrke H, 91(4):F273–8.
Rumack CM, Strain JD. Chest radiography versus 215. Lebowitz JM, Belman AB. Simultaneous idiopathic
chest CT in the evaluation for pulmonary metastases adrenal hemorrhage and renal vein thrombosis in the
in patients with Wilms' tumor: a retrospective review. newborn. J Urol. 1983;129(3):574–6.
Pediatr Radiol. 2000;30(8):533–7. 216. Bennett WG, Wood BP. Radiological case of the
202. Hoffer FA. Magnetic resonance imaging of abdomi- month. Left renal vein thrombosis and left adrenal
nal masses in the pediatric patient. Semin Ultrasound hemorrhage. Am J Dis Child. 1991;145(11):
CT MR. 2005;26(4):212–23. 1299–300.
203. Daw NC, Kauffman WM, Bodner SM, Pratt CB, 217. Bessell-Browne R, O'Malley ME. CT of pheochro-
Hoffer FA. Patterns of abdominal relapse and role of mocytoma and paraganglioma: risk of adverse events
704 J. Loewen and L.A. Greenbaum

with i.v. administration of nonionic contrast material. phaeochromocytomas–experience and meta-analysis.


AJR Am J Roentgenol. 2007;188(4):970–4. Anticancer Res. 2006;26(2B):1599–604.
218. Greenblatt DY, Shenker Y, Chen H. The utility of 220. Mackenzie IS, Gurnell M, Balan KK, Simpson H,
metaiodobenzylguanidine (MIBG) scintigraphy in Chatterjee K, Brown MJ. The use of 18-fluoro-
patients with pheochromocytoma. Ann Surg Oncol. dihydroxyphenylalanine and 18-fluorodeoxyglucose
2008;15(3):900–5. positron emission tomography scanning in the
219. Van Der Horst-Schrivers AN, Jager PL, Boezen HM, assessment of metaiodobenzylguanidine-negative
Schouten JP, Kema IP, Links TP. Iodine-123 phaeochromocytoma. Eur J Endocrinol. 2007;
metaiodobenzylguanidine scintigraphy in localising 157(4):533–7.
Pediatric Renal Pathology
24
Agnes B. Fogo

Contents Light Microscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 713


Immunofluorescence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 714
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 706 Electron Microscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 715
Renal Biopsy Indications . . . . . . . . . . . . . . . . . . . . . . . . . . . 706 Basic Renal Lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 715
Hematuria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 706 Normal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 715
Proteinuria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 706 Overall Pattern . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 717
Nephrotic Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 706 Specific Glomerular Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . 718
Acute Nephritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 707 Glomerular Basement Membrane . . . . . . . . . . . . . . . . . . . 720
Acute Kidney Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 707 Tubules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 720
Rapidly Progressive Glomerulonephritis . . . . . . . . . . . . 707 Interstitium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
Chronic Kidney Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 707 Vessels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 721
Systemic Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 707
Follow-Up of Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 708 Clinical Pathological Correlations . . . . . . . . . . . . . . . . 722
Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 708 Diagnostic Findings in Selected Renal Diseases . . . . 722
Anti-glomerular Basement Membrane Antibody
Obtaining Tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 708 Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 733
General Considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 708
Contraindications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 709 Prognostic Implications of Biopsy Findings . . . . . . 737
Biopsy Technique . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 709 Renal Transplant Biopsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 738
Aspiration Biopsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 712
New Methods for the Future . . . . . . . . . . . . . . . . . . . . . . . 741
Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 712
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 742
Assessment of the Renal Biopsy . . . . . . . . . . . . . . . . . . . 712
Adequacy of Sample . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 712
Allotment of Tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 713

A.B. Fogo (*)


Department of Pathology, Microbiology and Immunology,
Vanderbilt University Medical Center, Nashville, TN, USA
e-mail: agnes.fogo@vanderbilt.edu

# Springer-Verlag Berlin Heidelberg 2016 705


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_22
706 A.B. Fogo

Introduction information can be of importance in avoiding


further repeated invasive evaluation in the patient.
This chapter reviews the usual circumstances in
which biopsies are obtained, methods of obtaining
the biopsy material and analyzing the tissue, and Proteinuria
considerations in allocation of biopsy tissue.
Lesions in the kidney are described and specific Isolated sub-nephrotic proteinuria may be pos-
terminology defined, and the distinct characteristic tural or due to tubulointerstitial disease. These
morphologic findings in various diseases are briefly possibilities should be evaluated completely
described and illustrated. Last, experimental tech- before renal biopsy is considered. Any glomerular
niques that may provide important pathogenic, disease may cause mild to moderate proteinuria as
prognostic, or diagnostic information are discussed. the only manifestation, and biopsy may yield the
diagnosis even at an early stage.

Renal Biopsy Indications


Nephrotic Syndrome
The indications for renal biopsy vary according to
the ethnic and age characteristics of the popula- Numerous children with nephrotic syndrome
tion studied and the geographic location because (NS) were studied when renal biopsy first became
these factors influence the incidence of various available. The biopsies showed so-called minimal
renal diseases. The indications discussed below change disease (MCD) in the vast majority of
present the most common settings in children for cases. The efficacy of corticosteroids in this setting
which renal biopsy is undertaken. has obviated the need for renal biopsies in most of
these cases. Therefore, young children (i.e., age
1–7 years) with NS will typically undergo a thera-
Hematuria peutic trial of corticosteroids without a biopsy.
However, in infants with NS, in older children, or
Isolated hematuria (i.e., without proteinuria and in those with evidence of nephritis (hypertension,
with normal function of the kidney) may be due to hematuria, low C3, or decreased renal function) or
hypercalciuria or familial or urologic disease failing corticosteroid therapy, renal biopsy is often
[1–3]. Once these disorders are ruled out, a glo- performed. In these patients, disease other than
merular origin of persistent isolated hematuria MCD (e.g., focal segmental glomerulosclerosis
should be considered. Red blood cell casts or [FSGS], membranoproliferative glomerulonephri-
dysmorphic red blood cells indicate likely glomer- tis [MPGN], IgA nephropathy, membranous
ular origin of hematuria. Renal biopsy may define glomerulopathy, C3 glomerulopathy, or more
the underlying abnormality in these patients. The rarely, in infants less than 1 year, Finnish-type
most common findings are mesangial proliferative congenital nephrotic syndrome or diffuse
disease or IgA nephropathy. Less common disor- mesangial sclerosis) is often present [4–9]. Children
ders include hereditary nephritis (Alport syn- with steroid-resistant NS and FSGS on biopsy may
drome) and thin basement membrane lesion. The have a podocyte gene mutation, such as podocin, as
latter may be familial (benign familial hematuria) cause of their disease. Genetic testing is important
or sporadic. One-quarter to nearly one-half of the to identify these children, as 10–30 % of children
patients with isolated hematuria have normal with sporadic steroid-resistant NS and FSGS have
biopsies [1–4]. Renal biopsy may, therefore, such mutations, and most do not respond to con-
define the pathology and provide assurance of a tinued immunosuppression and will not have recur-
benign prognosis in some patients or diagnose a rence of the genetic disease in a kidney transplant
possible hereditary disease, which may initiate [10–12]. These genetically induced lesions do not
screening of other family members. Last, the show specific renal biopsy morphologic findings.
24 Pediatric Renal Pathology 707

Acute Nephritis may be distinguished only by specific serologic


studies (see below) or renal biopsy must be treated
The child with acute glomerulonephritis may need a urgently to avoid severe chronic kidney damage.
biopsy when the course is not typical of acute Although serum anti-glomerular basement mem-
postinfectious disease or if urinary abnormalities per- brane (GBM) antibody or anti-neutrophil cytoplas-
sist. Although the primary disease process may be mic antibody (ANCA) titers may provide useful
evident in systemic conditions, such as Henoch- information, the ANCA test in particular is not
Schönlein purpura or systemic lupus erythematosus diagnostic of a specific condition; rather, it is a
(SLE), renal biopsy often is indicated to assess sever- screening test for necrotizing vasculitides
ity of injury, to guide therapy and prognosis. Differ- [18, 19]. ANCA positivity may be detected by
entiation of specific types of proliferative lesions indirect immunofluorescence to detect binding of
such as MPGN, the spectrum of C3 glomerulopathies serum antibodies to test neutrophils in a perinuclear
(C3GP), including dense deposit disease (DDD) and (p-ANCA) or cytoplasmic (c-ANCA) pattern.
C3-dominant glomerulonephritis is made by renal ANCA are now routinely tested more sensitively
biopsy. This distinction has important implications and specifically by ELISA to detect antibodies to
for treatment. C3GP is due to complement myeloperoxidase (MPO) or proteinase-3 (PR3).
dysregulation, due to genetic deficiency of key com- ANCA positivity can also be present in severe
plement regulatory molecules and/or antibodies to crescentic immune complex disease and was
C3 convertase (C3 nephritic factor), directing a dif- detected, in 19 % of such patients in a large series
ferent approach to treatment [13]. Further, morpho- [20, 21]. ANCA, particularly p-ANCA (MPO), are
logic recurrence of DDD in the transplant is also present in about 20 % of anti-GBM antibody
invariable and causes worse graft outcome [14–16]. disease [21]. Thus, kidney biopsy will allow spe-
cific diagnosis not only of the cause of RPGN but
also its activity and chronicity, aiding in treatment
Acute Kidney Injury strategies.

The cause of acute kidney injury may be clinically


obvious, or there may be multiple potential cul- Chronic Kidney Disease
prits. When prerenal and obstructive causes are
not apparent, renal parenchymal disease should Patients with chronic kidney disease of uncertain
be considered. When acute kidney injury is associ- etiology are candidates for renal biopsy. Although
ated with nephritis, NS, or evidence of vasculitis or renal biopsy of the small, shrunken kidney is more
systemic diseases, biopsy is usually performed. risky because of the greater incidence of bleeding
Other common causes include acute tubular necro- complications, the diagnosis of primary disease can
sis or injury (often caused by drug or ischemic be important. This information allows assessment
injury), vascular disease, and interstitial nephritis. of existing severity of morphologic lesions, deter-
These conditions can often be diagnosed without mination of risk of recurrence in eventual renal
renal biopsy. However, when the cause remains transplant, and suitability of deceased versus
uncertain after complete evaluation, renal biopsy living-related donor transplantation. If the disease
may be necessary for diagnosis [17]. has a familial basis or recurs frequently with resul-
tant graft loss, deceased donor transplantation may
be preferable to living-related donor transplant [16].
Rapidly Progressive
Glomerulonephritis
Systemic Diseases
Renal biopsy is considered an urgent procedure in
the patient with rapidly progressive glomerulone- The severity of renal involvement in systemic
phritis (RPGN). Various systemic vasculitides that disease, such as chronic or recurrent hemolytic-
708 A.B. Fogo

uremic syndrome (HUS), Henoch-Schönlein pur- in creatinine supports calcineurin inhibitor toxic-
pura, diabetes mellitus, or SLE, may not be appar- ity because this drug commonly causes a decline
ent without renal biopsy. Patients with diabetes in the glomerular filtration rate (GFR) by vaso-
and renal abnormalities that are atypical for the constriction and not overt structural lesions. The
expected clinical course of diabetic nephropathy absence of findings of acute rejection in renal
are often biopsied to determine whether condi- biopsy thus can assist in avoiding unnecessary
tions other than, or in addition to, diabetes are immunosuppressive therapy with its potential for
affecting the kidney. Severity of lesions and increased morbidity and mortality. Even a diag-
stage of chronicity and activity impart prognostic nosis of chronic allograft nephropathy (CAN, also
information and may affect therapeutic decisions called IFTA; interstitial fibrosis/tubular atrophy),
(see below). The most extensively studied disease which is not amenable to immunosuppressive
in this regard is SLE. Differentiation of specific therapy, has important therapeutic implications
class of lupus nephritis by the International Soci- for the patient.
ety of Nephrology/Renal Pathology Society Diseases that recur in the transplant with high
(ISN/RPS) classification (see below) may be dif- frequency include IgA nephropathy, MPGN,
ficult without renal biopsy [22–24]. Overall, evi- dense deposit disease (DDD, also previously
dence indicates that renal biopsy findings may be known as MPGN type II), FSGS, atypical,
more sensitive than clinical assessment alone in diarrhea-negative HUS (caused by complement
evaluating the severity of renal involvement in dysregulation defect), and membranous
SLE [25–27]. glomerulopathy. The latter two also occur de
novo in the transplant, likely related to antibody-
mediated rejection. Metabolic diseases such as
Follow-Up of Disease oxalosis and diabetic nephropathy can also cause
recurrent disease in the renal transplant, if liver or
With improved therapeutic modalities available pancreas transplantation does not cure the primary
for intervention in chronic progressive kidney abnormality [16]. Alport syndrome is caused by
disease, sequential or follow-up biopsies are mutation in one of the type IV collagen genes,
becoming increasingly necessary to evaluate ther- resulting in abnormal GBM assembly and struc-
apeutic efficacy. Additional therapy may be indi- ture. Patients with Alport may develop anti-GBM
cated, or alternatively, cytotoxic therapy with its antibody disease in the transplant because of anti-
side effects may be withheld, if the biopsy shows bodies against its normal type IV basement mem-
end-stage histology. Intervention with, for exam- brane collagen [29, 30].
ple, angiotensin-converting enzyme (ACE) inhib-
itors or angiotensin type 1 receptor blockers
(ARBs) can alter the course of chronic progres- Obtaining Tissue
sive renal disease [28].
General Considerations

Transplantation Percutaneous renal biopsy is the most common


method for obtaining tissue for the kidney. In large
Kidney transplant biopsies are useful in assessing series, major complications are rare. The tech-
episodes of clinically suspected rejection, investi- nique, first done in 1951 by Iverson and Brun,
gating the cause of decreased renal function or allows tissue yield in 93–95 % of biopsies, with
urine output, and detecting the development of more than 87 % of these being adequate
de novo or recurrent disease. Occasionally, infec- [31–33]. The biopsy findings altered diagnoses
tion may be diagnosed by renal biopsy. Drug in half of the cases in one series, indicating differ-
toxicity may be diagnosed by morphologic find- ent therapeutic approaches in approximately
ings. The absence of lesions in a patient with a rise one-third of those cases [31]. Although some
24 Pediatric Renal Pathology 709

renal diseases show diagnostic features by light is essential. Adequate control of hypertension
microscopy (LM) (Table 1), special studies add to before the procedure is important as hypertension
the sensitivity of the study. For the renal biopsy to is a risk for post biopsy bleeding [37]. Before
be most useful, it must be evaluated appropriately biopsy is done, ultrasound examination must con-
by an experienced renal pathologist. Biopsies firm that there are two kidneys in normal position.
must be examined by special LM, IF, and electron The biopsy optimally is timed so that an experi-
microscopy (EM) for the most accurate diagnosis enced technician or pathologist can attend to
[32, 34]. If the nephrologist’s hospital does not ensure prompt processing of the biopsy tissue.
provide these services, arrangements must be Food and drink should have been withheld for
made to send tissue in appropriate fixatives (see at least 6 h before biopsy, and the child should be
below) to a reference laboratory with these capa- lightly sedated. The child lies in the prone position
bilities. If such services cannot be provided, it is with a sandbag or rolled sheet under the abdomen,
doubtful whether the institution should be under- and the skin of the flank is “prepped” and draped
taking renal biopsies. in sterile fashion. Although the left kidney is
usually preferred, either side can be chosen for
biopsy. The lower pole of the kidney is marked on
Contraindications the skin with a pen after localization by any of
several imaging techniques, such as computed
Contraindications (relative or absolute) to percu- tomography (CT) or ultrasound, the last being
taneous biopsy are solitary, ectopic, or horseshoe the most commonly used method. Local anes-
kidney; bleeding diathesis; abnormal renal vascu- thetic is infiltrated first in the skin and then in
lar supply; and uncontrolled hypertension deeper tissues, taking care not to enter the kidney.
[31–33, 35]. In the era of ultrasound guidance Conventional or spring-loaded needles are used
and automated biopsy instruments, solitary kid- for renal biopsies. The conventional biopsy nee-
ney may be biopsied safely in selected patients dle is inserted to the desired position as the patient
[36]. Relative contraindications include obesity, holds his or her breath. In younger children who
uncooperative patients, hydronephrosis, ascites, cannot reliably cooperate in this manner, biopsy is
and small shrunken kidneys, all associated with done under anesthesia. The cannula is advanced
greater risk for complications. Open or modified over the obturator once the needle is in correct
open biopsy is preferable if the biopsy informa- position, the needle is advanced, and the entire
tion is crucial in these conditions. Percutaneous needle with the core of tissue is then removed.
biopsy is contraindicated if the kidney has tumors, The use of an automatic spring-loaded biopsy
large cysts, abscesses, or pyelonephritis because system is now widespread because of the simplic-
the needle track may facilitate spread of malignant ity and ease of the technique [38, 39]. The kidney
cells or infection. Open biopsy allows selection of is localized with ultrasound guidance and the
specific areas for biopsy in these situations. depth of the kidney as judged by ultrasound. The
biopsy needle is advanced to the kidney capsule
under ultrasound observation and guidance. The
Biopsy Technique patient may hold his or her breath for only a few
seconds while the spring-loaded needle is acti-
The patient may be brought to the hospital on the vated, causing the obturator to automatically
day of the biopsy. Laboratory evaluation must advance into the kidney, and the entire needle is
include a complete blood cell count with normal then removed with the tissue core. The speed of
platelet count, partial thromboplastin, and pro- automated biopsy needles, however, minimizes
thrombin times. On rare occasions, infusions of the need for the patient to hold respirations,
platelets or fresh frozen plasma or DDAVP may required with conventional needles. It is important
be necessary to allow renal biopsy in critical clin- to note that the caliber of the needle used with any
ical situations in which histopathologic diagnosis of these techniques directly impacts the adequacy
710 A.B. Fogo

Table 1 Characteristic abnormalities of glomerular diseases


Disease and IF staining
typical clinical
presentation LM pattern Mesangial Subepithelial Subendothelial EM, other findings
Hematuria/nephritis
Alport’s syndrome Early: normal    Thin and thick,
Late: sclerosis basket-weaving
GBM
Mesangial lupus Mesangial +   Immune deposits
nephritis ISN/RPS proliferation All Igs, C3, by EM, reticular
II C1q aggregates in
endothelial cells
Focal lupus Proliferative and/or + + (few) + (scattered) Immune deposits
nephritis ISN/RPS sclerosing, <50% All Igs, C3, C1q by EM, reticular
III of glomeruli aggregates in
endothelial cells
Diffuse lupus Proliferative and/or + + + (wire loop) Immune deposits
nephritis ISN/RPS sclerosing, 50% All Igs, C3, C1q by EM, reticular
IV of glomeruli, wire aggregates in
loops endothelial cells
IgA nephropathy Mesangial +   Immune deposits
proliferation Predominant by EM
or
codominant
IgA
Henoch-Schönlein Mesangial  + +/ +/ Immune deposits
purpura endocapillary  Predominant  by EM
hypercellularity,  or
crescents codominant
IgA
Postinfectious GN Endocapillary + +  Irregular, hump-
hypercellularity, Coarsely Coarsely like subepithelial
PMNs granular granular deposits on top of
IgG, C3 IgG, C3 GBM by EM
Hemolytic-uremic Glomerular/    Increased lamina
syndrome arteriolar rara interna by EM,
thrombosis swollen
endothelial cells,
fibrin tactoids, no
deposits
MPGN Endocapillary +  + Subendothelial
hypercellularity, IgG, C3 IgG, C3 immune deposits
lobular, double by EM, cellular
contour GBMs interposition
C3 GP: Mesangial,    Discontinuous
endocapillary Dominant
proliferation C3, scanty or
no Ig
C3-dominant GN Double contours in C3GN: usual C3GN: chunky, irregular C3GN:
C3GN mesangial capillary wall subendothelial/
mesangial,
occasional
subepithelial
deposits by EM
(continued)
24 Pediatric Renal Pathology 711

Table 1 (continued)
Disease and IF staining
typical clinical
presentation LM pattern Mesangial Subepithelial Subendothelial EM, other findings
DDD Ribbonlike DDD: DDD: ribbonlike, C3 DDD:
capillary wall in globular C3 intramembranous,
DDD mesangial dense
deposits by EM
Nephrotic syndrome
Minimal change Normal    Effacement of
disease podocyte foot
processes
No deposits
Focal segmental Segmental +/   Effacement of
glomerulosclerosis glomerulosclerosis, IgM, C3 podocyte foot
glomerular processes, no
hypertrophy deposits
Diabetic Increased mesangial    Thick GBM
nephropathy matrix,  nodular, without deposits
thick GBM,
hyalinized arterioles
Membranous Thick GBM, spikes Scattered +  Subepithelial,
lupus nephritis on Jones’ stain mesangial immune
ISN/RPS V deposits
+ All Igs, C3,  Reticular
C1q aggregates in
endothelial cells
Primary Thick GBM, spikes  +  Subepithelial
membranous GP on Jones’ stain  IgG, C3  immune deposits
PLA2R
RPGN
Anti-GBM Necrosis of  Linear  No deposits by EM
antibody disease glomeruli, staining of
crescents; all lesions GBM
of similar activity/ IgG linear
chronicity (C3 chunky/
granular)
Granulomatosis Focal segmental    No deposits by EM
with polyangiitis necrosis of
glomeruli,
crescents; variable
activity
Microscopic Focal segmental    No deposits by EM
polyangiitis necrosis of
glomeruli,
crescents; variable
activity
Abbreviations: GP glomerulopathy, PLA2R phospholipase A2 receptor

of the specimen [40]. When 18-gauge needles are distortion and with fewer passes necessary to
used with this method, the resulting cores are very obtain adequate tissue.
small and there can be artifact along the edges. Usually, two cores of kidney cortex are neces-
The use of a 16-gauge needle thus is more likely to sary for optimum evaluation or three cores if
provide an adequate tissue sample without 18-gauge needles are used. If tissue cannot be
712 A.B. Fogo

obtained after several passes, the biopsy should be a series of 5,120 biopsies. In a series totaling
attempted on another day. After biopsy, a dressing 1,820 biopsies in children, 1 nephrectomy
is applied, and the child is kept supine in bed for resulted [46–51]. Transient microscopic hematu-
6 h and monitored with frequent checks of vital ria is universal after biopsy, although macroscopic
signs and urine for hematuria. hematuria is seen in only 5 % and requires trans-
Increasingly, percutaneous native and trans- fusion in up to 2.3 % of patients. Perirenal hema-
plant renal biopsies are performed as same day toma is most often asymptomatic and can be seen
procedures in pediatric patients so that hospital by CT in up to 85 % of biopsies. Symptomatic
admission is not required. Those with post biopsy hematoma is rare, occurring in less than 2 %.
perinephric hematomas, post biopsy gross hema- Arteriovenous fistulae are usually symptomatic
turia, or very young children can be observed with hematuria, hypertension, or cardiac failure
overnight so that hemostasis is assured in only 0.5 % of biopsies, although bruits may be
[41–43]. Open biopsy can be performed under detected in as many as 75 % of patients. Most
general anesthesia. The kidney can be directly fistulae heal within a few months. Other reported
visualized even through a small incision. complications include inadvertent puncture of
Although a larger sample may be obtained with other viscera or major renal vessels, sepsis, renal
a wedge biopsy, it is preferable to also perform a infection, and seeding of cancer. Death is rare, less
needle biopsy to sample deeper cortex and than 0.1 % in reviews of large series. Complica-
medulla for assessment of diseases that preferen- tion rates appear to be slightly higher in less
tially involve juxtamedullary glomeruli (see developed countries [46, 48]. Complications for
below). the spring-loaded needle biopsy system appear to
be similar to conventional needles if the same
gauge is used [55]. Complications relate in part
Aspiration Biopsy to the number of passes made to obtain tissue.
Therefore, the potential lower rate of complication
Fine-needle aspiration (FNA) biopsy technique is with an 18-gauge spring-loaded needle in some
used most often to diagnose mass lesion of the studies is offset largely by the need for more
kidney. FNA can also obtain material for culture. passes for adequate samples.
A modified FNA technique has been described for
collection of glomeruli from either native or trans-
plant kidneys to analyze glomerular lesions. This Assessment of the Renal Biopsy
FNA technique has limitations in sample size and
is not suitable for study of vascular or fibrotic Adequacy of Sample
processes or diagnosis of antibody-mediated
rejection. The less-invasive nature of the proce- Cores of fat and connective tissue will float when
dure makes it amenable to serial monitoring of placed in saline, but a core of renal parenchyma
interstitial cellular infiltrates in transplanted kid- will sink. The biopsy sample should also be visu-
neys, but its use has declined due to less diagnos- ally inspected with a dissecting microscope or
tic yield versus the gold standard core biopsy hand lens. Glomeruli are visualized as small red
approach [44, 45]. dots in the biopsy core. Scarred glomeruli may be
difficult to identify since they are not perfused. In
diffuse disease, such as membranous
Complications glomerulopathy, one glomerulus may be adequate
for diagnosis. However, other diseases, such as
Important complications occur in 5–10 % of crescentic glomerulonephritis, FSGS, or lupus
patients [31, 46–54]. Major complications, usu- nephritis, may be focal. The greater the number
ally bleeding, leading to nephrectomy occurred in of glomeruli sampled, the lower the probability of
5 patients in a review of 8,081 [54] and 1 patient in missing a focally distributed lesion [56]. If only
24 Pediatric Renal Pathology 713

10 % of glomeruli in the kidney are involved by clean smooth surface, such as a wax board, for
the focal process, a biopsy sample of only 10 glo- cutting.
meruli has a 35 % probability of missing the It may be difficult to identify the cortical part of
lesion, decreasing to 12 % if the biopsy contains the specimen even after inspection with a hand
20 glomeruli. When one-fourth of glomeruli are lens or dissecting microscope when scarring or
involved in the kidney, there is only a 5 % chance severe injury is present. Therefore, we recom-
of missing the abnormal glomeruli in a biopsy of mend cutting two 1-mm pieces with a sharp
10 glomeruli. A biopsy sample of 20–25 glomer- blade from each end of each core for electron
uli for light microscopic assessment is sufficient to microscopic studies. The remaining core is then
distinguish between mild disease (less than 20 % divided into specimens for IF and LM. When
of glomeruli involved), moderate disease (20–50 multiple cores are obtained, this allocation should
% of glomeruli involved), or severe disease (more be applied to each core, rather than allocating one
than 50 % of glomeruli involved). The sample site complete core to one study. This approach will
must also be considered in evaluating the ade- maximize chances of adequacy of tissue with
quacy of tissue. Even a large biopsy, consisting glomeruli for each study. When the tissue sample
only of superficial glomeruli, cannot exclude the obtained is very small, the nephrologist and the
presence of early FSGS, in which the initial pathologist should consider the differential diag-
involvement is in the juxtamedullary glomeruli. nosis and allocate tissue accordingly. For exam-
Likewise, although nephronophthisis is most ple, in a case of suspected IgA nephropathy, tissue
often diagnosed clinically, a juxtamedullary for IF is most important. IF studies can be
biopsy would be necessary for its morphologic attempted on remaining fixed tissue in the paraffin
diagnosis. block, but are not as reliable as when performed
on frozen nonfixed material. Electron microscopic
studies can be done on other portions of tissue
Allotment of Tissue (as long as mercury-based fixatives have not been
used). When tissue for EM is inadequate, portions
Renal tissue should be studied by light micro- of the paraffin-embedded tissue left after light
scopic techniques with special stains (hematoxy- microscopic examination may be cut out from
lin and eosin, modified silver stain [periodic acid/ the block and processed for EM. Although the
methenamine, also called Jones’ stain], periodic quality is not optimal, diagnostic findings can
acid-Schiff [PAS], and/or Masson trichrome), IF, still be discerned. In special circumstances, when
and EM [34, 57–59]. The tissue is divided so that no tissue remains in the paraffin block and a focal
glomeruli are present in each portion of the sam- lesion in one section must be studied, one may
ple. Optimally, the pathologist or an experienced attempt to process the tissue section from a glass
histotechnologist or other trained person will slide for EM.
attend the biopsy and inspect and allot tissue for
each study. If this is impossible, tissue may be
placed in saline and brought directly to the labo- Light Microscopy
ratory for prompt processing. It is important to
handle the tissue gently so that artifacts do not Numerous fixatives are used for light microscopic
occur. The fresh tissue must not be picked up with examination, and they vary from institute to insti-
forceps because this will crush and distort the tute. Satisfactory results may be obtained with
morphology. The core can be handled carefully Zenker’s, Bouin’s, formalin, Carnoy’s, or parafor-
with a wooden stick or pipette. The core should maldehyde. Material for IF studies may be snap
not be placed on a sponge or gauze pad because frozen immediately at 20 C in solutions of
this may cause a divot-like artifact as the unfixed isopentane, dry ice, acetone, or Freon and embed-
tissue molds to the holes of the underlying sur- ded in Tissue-Tek, OCT, or other compounds for
face. The biopsy specimen is therefore placed on a frozen sections. If tissue cannot immediately be
714 A.B. Fogo

snap frozen, it may be placed in Michel’s tissue Immunofluorescence


media, where it may be stored for up to 1 week
before freezing. This allows tissue to be sent to Immunofluorescence (IF) studies are most com-
reference laboratories for appropriate processing. monly done by direct IF on frozen tissue sections
Tissue for EM may be fixed in glutaraldehyde, with application of fluorescein-conjugated anti-
formaldehyde, or other appropriate non-mercury bodies directed against IgG, IgA, IgM, comple-
fixatives. Tissue placed in glutaraldehyde should ment components C3 and C1q, kappa, lambda,
be promptly processed or, if stored for future and often albumin and fibrinogen. Additional
possible processing, should be transferred to an antisera may be used as clinically indicated. Phos-
appropriate buffer solution within a week to avoid pholipase A2 receptor, (PLA2R), the antigen in
artifacts. Tissue for LM is routinely processed, most cases of primary membranous GP, is
embedded in paraffin, and cut into 2–3-μ thick detected by IF by digestion techniques, followed
sections. Serial sections with multiple levels are by applying antibody to PLA2R to tissue section
then prepared for examination. cut from the paraffin block [60, 61]. Additional
If water-soluble compounds are expected, such immunostaining studies include those for hepati-
as urate or uric acid, the tissue should be fixed in tis B antigen, thyroglobulin, C4, C4d, specific
ethanol. Lipids are best detected in frozen sections viruses (e.g., SV40 for polyoma virus), myoglo-
because they are extracted during xylene bin, and type IV collagen alpha chains. Some of
processing for paraffin sections. Hematoxylin these antigens are recognized by antisera even
and eosin stains are most useful for overall assess- after fixation and can be detected by immunohis-
ment of the interstitium and crystals. This stain tochemical techniques (e.g., immunoperoxidase)
also allows particularly good visualization of on the formalin-fixed tissue sections and then
infiltrating cells, especially eosinophils. In addi- studied with a light microscope. This technique
tion, fibrin may be easily visualized by this requires enzyme pretreatment of tissue depending
stain. Periodic acid-Schiff (PAS) stains glycopro- on the antigen to be studied, which must be tai-
teins and accentuates basement membranes and lored exactly depending on length and type of
matrix material and allows definition of the brush fixation and section thickness. Direct observation
border of proximal tubular cells. Areas of of the digestion process, stopping when all plasma
hyalinosis and protein precipitation, including is removed from capillary loops, has been used to
cryoglobulin (which usually is dominantly IgM), achieve reliable results [62]. These challenges
are also accentuated with PAS. Silver stains, such have prevented widespread use of this technique
as Jones’ stain, stain basement membrane in the USA. In general, IF on frozen tissue for
material but not deposits, thus allowing distinc- detection of immunoglobulins and complements
tion of these components. Masson’s trichrome is more sensitive than IHC on fixed tissue.
stain detects areas of collagen deposition by Frozen tissue sections stained by the com-
staining bluish. Sirius red stains all collagens monly used method of fluorescein-conjugated
and can also be visualized and quantified under antibodies are viewed by IF microscopy, evaluat-
polarized light to assess fibrosis. Other special ing staining in glomeruli, vessels, tubules, and
stains, such as immunohistochemistry for interstitium. The pattern of glomerular staining is
specific molecules, may be indicated. Congo red assessed to define granular or linear capillary wall
stain detects amyloid. Special stains can detect staining or mesangial deposits. Arteriolar staining
bacterial or fungal organisms and acid-fast may be of diagnostic significance to detect, e.g.,
bacilli. Special techniques may also be used on deposits in lupus. Tubules may show granular
the light microscopic material. These include deposits in lupus nephritis, pseudolinear deposits
polarization to detect crystals or foreign bodies in light chain deposition disease, or linear staining
and morphometry to assess glomerular size (see in the rare anti-TBM antibody disease. Nuclear
below) and severity of interstitial fibrosis staining can be seen in lupus or lupus-like diseases
quantitatively. as a tissue manifestation of the patient’s positive
24 Pediatric Renal Pathology 715

ANA. IF is more sensitive than EM in identifying fingerprinting, are present in some cases of lupus
immune deposits, although EM provides more nephritis. Reticular aggregates (also called tubular
detailed information on the exact localization of arrays) of organelle membrane material in endo-
those deposits [58, 59]. Diagnostic lesions of thelial cell cytoplasm throughout the body are
interest should be photographed because fluores- characteristically seen in large numbers in patients
cence fades on storage and with light exposure. with SLE or HIV infection, or those treated with
exogenous interferon, thought to reflect a
response to high levels of interferon [59, 63].
Electron Microscopy EM also delineates specific basement mem-
brane abnormalities. For instance, small
Tissue for EM is processed with postfixation in 1 % subepithelial deposits without surrounding new
osmium tetroxide, which enhances contrast of the basement membrane material do not result in
tissue, and then dehydrated and embedded. With spikes, and therefore cannot be visualized by
new, more rapidly polymerizing embedding media, Jones’ stain on LM, but can still be detected
such as Spurr, tissue may be ready for examination directly by EM. The lamina densa of the GBM is
within 1 day. Electron microscopic study was markedly thickened in diabetic nephropathy. Cir-
found to add information in 6–11 % of renal biop- cumferential cellular interposition is defined by
sies in a study from 1983 [32]. In a later study, EM extension of monocytes and mesangial cell cyto-
was needed to make a diagnosis in 21 % of cases plasm into the subendothelial space, with newly
and provided important confirmatory data in formed basement membrane interpositioning
approximately 20 % of cases [58]. EM showed between the advancing infiltrating cells and the
diagnostic pathological abnormalities in 18 % of endothelium with intervening basement membrane
patients with “normal” light microscopic findings. material, thus giving rise to the classic double
Larger, so-called thick scout sections (1 μ) are contours seen with silver stain by LM in MPGN.
stained with toluidine blue to select smaller areas Increased lucent material is present in the expanded
for thin sectioning for the electron microscope. lamina rara interna in transplant glomerulopathy,
Usually, the glomerulus containing the most repre- HUS, eclampsia/preeclampsia, and other diseases
sentative lesion is chosen, but sclerotic glomeruli presumed to involve endothelial injury/
are not useful to examine by EM. If there are coagulopathy. In these conditions, deposition of
irregular or focal proliferative lesions, several glo- fibrin, fibrinogen, and their degradation products
meruli may be sampled, including the proliferative may also occur. Fibrin is recognizable by its dense,
ones. The 60–90 Å thin sections are stained with coarse sheaflike structure by EM, with periodicity
uranyl acetate and lead citrate to enhance contrast observed in favorable sections. Morphometry of
before viewing in the EM scope. the GBM from EM prints is used to diagnose thin
Immune complex deposits are nonmembrane basement membranes in hereditary nephritides.
bound and denser than basement membrane or EM also allows structural assessment of changes
matrix materials. In specific diseases, such as of specific cells (see below). Diagnostic inclusions
cryoglobulinemia, amyloid, immunotactoid or are seen in various storage/metabolic disease, such
fibrillary glomerulopathies, or lupus nephritis, as Fabry’s disease.
specific substructure of deposits may be seen.
Specific localization of immune complexes is
done by EM examination, indicating whether Basic Renal Lesions
deposits are subendothelial, subepithelial,
mesangial, or in all of the above compartments. Normal
In some diseases, such as light chain deposition
disease or lupus nephritis, deposition may also be During fetal maturation, the glomerular capillary
seen in vessels and tubules. The so-called finger- tufts are initially covered by large, cuboidal,
print deposits, with substructure reminiscent of darkly staining epithelial cells with only small
716 A.B. Fogo

Fig. 1 Schematic illustration of glomerulus, with glomer- the epithelial cell and its foot processes (Ep). The
ular capillary attached to a mesangial stalk area. The glo- mesangial cell (M ) is embedded within the mesangial
merular endothelium (E) is fenestrated and lines the matrix (MM), with processes connecting to the GBM (Pro-
glomerular basement membrane (GBM), which covers vided courtesy of Professor Wilhelm Kriz with permission
the mesangium. The outside of the GBM is covered by from [56])

Fig. 2 Immature
glomerulus with plump,
dark epithelial cells from
biopsy of a 29-week
gestation baby (PAS, 670)

lumina visible (Fig. 1). The cells lining Bowman’s than 5 years old (average age 2.2 years) and
space undergo similar change from initial tall 140–160 μ in adulthood [64, 65].
columnar to cuboidal to flattened epithelial cells, The normal mature glomerulus consists of a
except for those located at the opening of the complex branching network of capillaries origi-
proximal tubule, where cells remain taller. Imma- nating at the afferent arteriole and draining into
ture nephrons may occasionally be seen in the the efferent arteriole. The glomerulus contains
superficial cortex of children up to 1 year of age. three resident cell types: mesangial, endothelial,
Glomerular growth continues until adulthood, and epithelial cells (Fig. 2). The visceral epithelial
with average normal glomerular diameter approx- cells (podocytes) cover the urinary surface of the
imately 95 μ in a group of patients less GBM with pseudopodlike extensions called foot
24 Pediatric Renal Pathology 717

processes, with intervening filtration slits. Endo- Overall Pattern


thelial cells are opposed to the inner surface of the
GBM and are fenestrated. At the stalk of the Assessment of the biopsy specimen must include
capillary, the endothelial cell is separated from inspection of all sections from different levels
the mesangial cells by intervening mesangial because additional glomeruli may be sampled on
matrix. The term endocapillary is used to describe deeper cuts of the biopsy core and many diseases
hypercellularity/proliferation filling up the capil- are characterized by focal lesions [68]. The sever-
lary lumen, contributed to by proliferation of ity and patterns of lesions are assessed, and nor-
mesangial, endothelial, and infiltrating inflamma- mal and affected glomeruli are counted. Lesions
tory cells. In contrast, extracapillary proliferation are classified as focal if only some (less than half)
refers to proliferation of the parietal epithelial glomeruli are involved, diffuse if all (or most)
cells that line Bowman’s capsule. glomeruli are involved, segmental if only portions
The mesangial cell is a contractile cell that also of glomeruli are involved, and global if entire
has phagocytic properties. It lies embedded in the glomerular tufts are involved. Characteristic glo-
mesangial matrix in the stalk region of the capil- merular disease patterns include lobular prolifer-
lary loops, attached to anchor sites at the ends of ation in MPGN, nodular proliferation of
the loop by thin extensions of its cytoplasm. Nor- mesangial cells, and abundant matrix material in
mally, up to three mesangial cell nuclei per lobule characteristic Kimmelstiel-Wilson lesions in dia-
are present. The basement membrane consists of betic nephropathy, focal and segmental sclerosis,
three layers distinct by EM, the central broadest intraglomerular/arteriolar fibrin thrombi, necrotiz-
lamina densa and the less electron-dense zones of ing lesions, and crescent formation (Table 1). The
lamina rara externa and interna. Thickening crescent, a lesion due to proliferation of mostly
occurs with maturational growth. Most investiga- parietal epithelial cells, owes its name to its shape
tors have found thicker basement membranes in in well-established lesions.
boys, with normal range from 220 to 260 nm at Glomeruli are assessed for alterations in size
1 year of age, 280 to 327 nm at age 5 years, 329 to (see below). It is important to compare with a
370 nm at age 10 years, and 358 to 399 nm at age normal control for a given age group because
15 years [66, 67]. In our laboratory, we found a marked glomerular maturational growth occurs
range of GBM thickness in children with normal in children. Glomerular hypertrophy may be an
kidneys from approximately 110 nm at age 1 year important predictor of increased risk for ultimate
to 222  14 nm at 7 years of age. development of FSGS in children with apparent
The glomerulus is surrounded by Bowman’s MCD (see below). Maturational pattern of glo-
capsule, which is lined by parietal epithelial cells. meruli (see above) should be noted. Occasional
These are continuous with the proximal tubule, fetal glomeruli may be found in children beyond
identifiable by its PAS-positive brush border. The infancy and are not of specific diagnostic
efferent and afferent arterioles can be distinguished significance.
morphologically in favorably oriented sections or Glomeruli are assessed for glomerulosclerosis,
by tracing their origins on serial sections. Segmen- that is, the presence of segmental obliteration and
tal, interlobular, and arcuate arteries may also be scarring of glomerular capillary tufts. Sclerosis
present in the renal biopsy specimen. The cortical may be in a segmental or global pattern
biopsy also allows assessment of the tubules and (Table 2). Previous studies suggested that up to
interstitium. Proximal tubules are readily identified 10 % of glomeruli may be normally globally
by their PAS-positive brush border, lacking in the sclerosed in people younger than 40 years of age
distal tubules. Collecting ducts show cuboidal, [69]. This number may be even smaller in chil-
cobblestone-like epithelium. Tubules are normally dren, with less than 1–3 % global sclerosis
back to back with minimal interstitial cells and the expected normally up to age 40 or 56, respectively
peritubular capillaries intervening. The medulla [70, 71]. These occasional globally sclerotic glo-
may also be included in the biopsy. meruli are thought to represent errors of
718 A.B. Fogo

Table 2 Definitions of common morphological terms Table 2 (continued)


Light microscopy Chronicity Extent of probable irreversible
Focal Involving some glomeruli lesions, based on, e.g., extent
Diffuse Involving all glomeruli of tubular atrophy, interstitial
fibrosis, fibrous crescents,
Segmental Involving part of glomerular
glomerulosclerosis
tuft
Immunofluorescence microscopy
Global Involving total glomerular tuft
Granular Discontinuous flecks of
Lobular Simplified, lobular
staining along capillary loop
appearance of capillary loop
producing granular pattern
architecture (MPGN)
Linear Smooth continuous staining
Nodular Relatively acellular areas of
along capillary loop
mesangial matrix (diabetic
nephropathy) Electron microscopy
Sclerosis Obliteration of capillary loop Foot process Flattening of foot processes of
by increased matrix (scarring) effacement podocytes so that they cover
the basement membrane
Crescent Proliferation of parietal
epithelial cells Microvillous Small extensions of podocytes
transformation with villus-like appearance
Spikes Projections of glomerular
basement membrane Interposition Extension of cell cytoplasm
intervening between with interposition between
subepithelial immune endothelial cell cytoplasm and
deposits (membranous GP) basement membrane and
underlying new basement
Endocapillary Increase in mesangial and/or
membrane formation
proliferation/ endothelial cells, +/
hypercellularity infiltrating inflammatory cells Reticular aggregates Organized arrays of organelle
membrane particles within
Hyaline Descriptive of glassy, smooth
endothelial cells
appearing material
Immunotactoid GP Large, organized
Hyalinosis Hyaline-appearing insudation
microtubular deposits,
of plasma proteins (in, e.g.,
>30 nm diameter
glomeruli in focal segmental
glomerulosclerosis or in Fibrillary GP Fibrils 14–20 nm diameter
arterioles in, e.g., without organization
hypertension or diabetes) GP glomerulopathy
Mesangium Stalk region of capillary loop
with mesangial cells
surrounded by matrix
Subepithelial Between podocyte and nephrogenesis. The percentage of global sclerosis
glomerular basement increases even with normal aging, up to half the
membrane patient’s age, minus 10 [71]. Globally, sclerosed
Subendothelial Between endothelial cell and glomeruli in greater percentage indicate the possi-
glomerular basement bility of renal disease (focal global sclerosis) [72].
membrane
The pattern of tubulointerstitial fibrosis, whether
Tram-track Double contour of glomerular
basement membrane due to proportional to glomerular sclerosis or not and
deposits and/or interposition whether diffuse or present in a “striped” pattern
of cells (see below) following the medullary rays or in broad patchy
Wire loop Thick, rigid appearance of zones, has diagnostic significance (see below).
capillary loop due to
subendothelial deposits
Activity Extent of possible treatment
sensitive lesions, based on, e. Specific Glomerular Cells
g., extent of crescents, cellular
infiltrate, necrosis, Podocytes
proliferation
The podocytes (glomerular visceral epithelial
(continued)
cells) may show vacuolization in various diseases
24 Pediatric Renal Pathology 719

with severe proteinuria. Although more extensive mesangial region, in areas away from the vascular
vacuolization of podocytes has been seen in FSGS pole. Increased mesangial prominence may be due
compared with patients with MCD [73], these to increased cellularity, increased matrix,
changes are only seen after established sclerotic deposits, or a combination. Large mesangial
lesions are identifiable by LM and do not permit deposits appear on Jones’ stain as pinkish areas
distinction of these two disease processes in the surrounded by the light silver-staining areas of
early phase in which segmental sclerosis may be mesangial matrix. The so-called interposition
undetected. Effacement of the foot processes of results when monocyte or mesangial cell cyto-
the podocytes by EM is common to any disease plasm extends outward between basement mem-
with marked proteinuria, and the podocyte may brane and endothelial cells and new matrix
also show microvillous transformation, with long, accumulates between the mesangial and endothe-
attenuated pseudopods. Podocytes are limited in lial cell bodies.
their ability to proliferate. However, the early
sclerotic lesion of FSGS is characterized by prom- Endothelial Cells
inence and hypertrophy of the overlying Extreme proliferation and swelling of endothelial
podocytes (“capping” lesion), often associated cells can obliterate capillary lumina in conditions
with endocapillary foam cells. This cellular vari- characterized by abnormalities of coagulation.
ant of FSGS may be more common in children Endothelial cells usually contain characteristic
than in adults with FSGS [74]. The idiopathic reticular aggregates in lupus nephritis and
collapsing variant of FSGS and HIV-associated HIV-associated nephropathy or after exogenous
nephropathy both show prominent hyperplasia interferon therapy [63]. Endocapillary cell
and protein droplets of the visceral epithelial hypercellularity/proliferation is characteristic of,
cells, with overlying segmental collapse of the for example, diffuse proliferative lupus nephritis
glomerular capillary tuft. These activated cells and MPGN.
have loss of podocyte markers and demonstrate
instead parietal epithelial cell (PEC) markers by Crescents
immunostaining [75]. In the situation of recurrent Crescents consist primarily of proliferating parie-
FSGS in the renal transplant, NS and foot process tal epithelial cells with some infiltrating macro-
effacement may be seen within weeks after phages and are a manifestation of severe
biopsy, with sclerosis becoming apparent at a glomerular injury. The name reflects the often
later date [76]. Even at this early stage, there are crescent-shaped sheet of cells filling up part or
increased activated PECs on the tuft in recurrent nearly all of Bowman’s space. Crescents result
FSGS, but not in native kidney biopsies of mini- from injuries that break the GBM, leading to
mal change disease [77]. In Fabry’s disease, there exudation of plasma protein and formation of
is accumulation of glycosphingolipid because of fibrin within Bowman’s space, which then
deficiency of alpha-galactosidase. Podocytes induces proliferation of the parietal epithelial
show marked vacuolization by LM with charac- cells and infiltration of macrophages. When cres-
teristic whorled, laminated electron-dense myelin cents are a prominent histologic feature, the
bodies by EM. In Fabry’s disease, these inclusions patient most often presents clinically with a rap-
may also be present in endothelial cells, tubular idly progressive glomerulonephritis.
epithelial cells, some interstitial cells, and the Crescents may occur in a variety of diseases.
vessels in early lesions in children [59, 78]. Diseases with crescents as a primary manifesta-
These inclusions decrease after treatment with tion include antibody-mediated injury (anti-GBM
enzyme replacement [79]. antibody disease), severe immune complex dis-
eases (e.g., lupus nephritis) and pauci-immune
Mesangial Cells diseases. The latter are often, but not invariably,
Hyperplasia of mesangial cells is defined as more associated with positive ANCA tests and may be
than three mesangial cell nuclei present per associated with systemic disease or be renal
720 A.B. Fogo

limited. The p-ANCA (anti-myeloperoxidase) The basement membrane may show double
pattern is most often associated with microscopic contours on silver stain by LM in diseases other
polyangiitis, whereas the c-ANCA (anti-protein- than MPGN, C3GN, or dense deposit disease. In
ase-3) pattern is typical in granulomatosis with transplant glomerulopathy, the double contour
polyangiitis (Wegener’s granulomatosis). Of appearance results from varying degrees of cellu-
note, positive ANCA tests are not sensitive in lar interposition and widening, with increased
distinguishing these categories [18–20]. Renal lucent material in the lamina rara interna. This is
biopsy is therefore critical for accurate diagnosis. also a characteristic finding in preeclampsia,
Diagnosis and appropriate treatment must occur transplant glomerulopathy, and chronic HUS or
rapidly in this clinical situation to optimize other chronic thrombotic microangiopathies [59].
chances of recovery of renal function. The early
lesion of cellular crescents is responsive to cyto-
toxic therapy. Biopsy indications of irreversible Tubules
renal damage include breaks of Bowman’s cap-
sule and fibrous transformation of the cellular Morphologically evident acute tubular injury or
crescents, periglomerular fibrosis, and scarred necrosis correlates poorly with the clinical extent
glomeruli and tubulointerstitium. of acute kidney injury (AKI). The changes vary
from nondiagnostic vacuolization to frank necro-
sis with sloughing of tubular epithelial cells (toxic
Glomerular Basement Membrane type) and flattened epithelium characteristic of
regeneration (ischemic type). In cortical necrosis,
GBM abnormalities are best evaluated by zones of cortex, including glomeruli, are necrotic.
EM. The basement membrane is abnormally In chronic kidney disease, tubules are atrophied
thick in diabetic nephropathy [59]. Diffuse abnor- with dilation and flattened epithelium, presum-
mally thin GBMs, less than 250 nm in adults, are ably secondary to lesions affecting the glomeru-
seen in familial hematuria [66, 67]. GBM thinning lus, although primary tubular and interstitial
cannot be accurately from EM processed from injury mechanisms may also be involved in
paraffin tissue, as this back-up technique causes these changes. Tubular atrophy is also present in
artifactual and variable thinning of GBMs [80]. primary tubulointerstitial diseases. Tubuloin-
In children, the diagnosis of thin basement mem- terstitial fibrosis is an important manifestation of
branes is more difficult than in adults because calcineurin inhibitor toxicity. The fibrosis occurs
GBM increases in thickness with normal matura- along the medullary rays due to the more severe
tion. Glomerular basement membrane thickness ischemia occurring in these areas, resulting in a
should be compared with normal for age and sex striped, rather than diffuse, pattern of fibrosis,
[see above; [66, 81, 82]]. In Alport syndrome, the with intervening preserved tubules.
basement membrane in established lesions is char- Nonspecific casts of Tamm-Horsfall
acterized by irregular areas of very thin and very (uromodulin) protein are seen in chronic kidney
thick, with splitting and splintering of the base- disease. Other casts may have a diagnostic appear-
ment membrane [30, 83]. The GBM in nail-patella ance, such as the giant cells surrounding tubular
syndrome is irregular, thickened, and split, with casts in light chain cast nephropathy (the so-called
electron-lucent areas containing banded collagen myeloma kidney). Casts of myoglobin with char-
type I fibers [59]. acteristic reddish-brown appearance are seen in
Immune deposits may localize on either side rhabdomyolysis, often with associated acute tubu-
of the GBM. Subepithelial immune deposits lar necrosis [84]. Crystals, for example, oxalate,
are characteristically seen in membranous dihydroxyadenine (DHA), or cysteine, may be
glomerulopathy. Subendothelial immune deposits identified by examination under polarized light
are seen, for example, in proliferative lupus [85, 86]. Tubules contain characteristic inclusions
nephritis or MPGN. in Fabry’s disease [87].
24 Pediatric Renal Pathology 721

Polymorphonuclear neutrophils (PMNs) suggestive of drug-induced interstitial nephritis,


within collecting ducts and proximal tubules are although eosinophils are also present in some
diagnostic of acute pyelonephritis. In chronic cases of idiopathic interstitial nephritis [91].
pyelonephritis, there is tubular atrophy and inter- Eosinophils can also be part of acute cellular
stitial fibrosis, characteristically in a patchy, rejection in the transplant. Nonnecrotizing granu-
regional distribution (geographic or “jigsaw” pat- lomas, with or without eosinophils, most often
tern). The combination of segmental glomerular reflect drug-induced hypersensitivity reaction. A
sclerosis with ischemic changes of corrugation pleomorphic infiltrate of lymphocytes, plasma
and thickening of the GBM and periglomerular cells, and PMNs suggests possible polyoma
fibrosis and patchy, regional interstitial fibrosis virus nephropathy in the transplant, confirmed
and tubular atrophy is also characteristic of reflux by finding viral nuclear inclusions and positive
nephropathy [88]. immunostaining in tubules (see above)
Cysts may be demonstrated by biopsy, [90]. Fibrosis results in increased spacing of
although the diagnosis of specific cystic diseases tubules because of the accumulation of
is usually made by combination of clinical and PAS-positive collagenous material. The collagen
ultrasound findings. The segment of the nephron also stains specifically blue with Masson’s
giving rise to the cysts can be identified by histo- trichrome stain. Fibrosis in a striped pattern sug-
chemical stains [89]. Areas of low cuboidal gests calcineurin inhibitor toxicity [92, 93]. Occa-
epithelial-lined structures surrounded by a cuff sionally, the interstitium is infiltrated by
of immature mesenchyme are present in the dys- malignancy. Hematopoietic neoplasms are espe-
plastic kidney, often with cartilage, fat, or abnor- cially prone to involve the kidney. In cystinosis,
mal blood vessels in the interstitium. The deep characteristic rectangular or trapezoid crystals are
medullary cystic dilation with thickened, present mostly in monocyte/macrophages and can
lamellated TBM characteristic of be recognized by EM or when polarized on frozen
nephronophthisis can be identified with a deep sections [85].
biopsy. Dilation of proximal tubules with
microcyst formation in conjunction with exten-
sive foot process effacement by EM is character- Vessels
istic of congenital nephrotic syndrome of Finnish
type. FSGS in a collapsing pattern with visceral Arterioles and larger segmental and interlobular
epithelial cell hyperplasia, numerous reticular arteries are evaluated for changes in the intima and
aggregates by EM, and tubular cystic dilation media; the presence of deposits, fibrin, hyaline,
and interstitial fibrosis out of proportion to the amyloid, or other material; or the presence of
severity of glomerular lesions are highly sugges- vasculitis. Arterioles show inclusions early in
tive of HIV-associated nephropathy [63]. children with Fabry’s [78]. Larger vessels typi-
Viral infections, including polyoma virus and cally are not sampled by a biopsy, and diseases
CMV, result in characteristic nuclear inclusions in such as classic polyarteritis nodosa that affect
tubular epithelia. Specific diagnosis is made by these large vessels are therefore best evaluated
immunostaining for viral proteins. Characteristic by other methods (e.g., arteriography). Intimal
viral particles may also be detected by EM [90]. fibrosis and medial thickening with hyperplasia
and hypertrophy of media are characteristic of
hypertension-attributable injury. Concentric
Interstitium medial necrosis with nodular protein deposition
suggests acute calcineurin inhibitor nephrotoxi-
Interstitial edema is a nonspecific change, present, city [93]. Fibrin thrombi, when present in glomer-
for example, in early acute transplant rejection, uli and/or arterioles, are the essential lesion of the
renal vein thrombosis, or inflammatory processes. thrombotic microangiopathies caused by, e.g.,
Identification of eosinophils in an infiltrate is HUS [59]. Fibrin localizes predominantly within
722 A.B. Fogo

glomerular lumina in disseminated intravascular antihypertensive agents that affect glomerular fil-
coagulation and hyperacute rejection. tration rate (e.g., ACE inhibitors, which preferen-
tially dilate efferent arterioles) may actually
increase serum creatinine levels in the short run.
Clinical Pathological Correlations Compensation by remaining nephrons may mask
ongoing severe disease processes such that creat-
After evaluation of the structural changes of the inine levels may remain near normal until late in
renal biopsy in conjunction with the clinical his- the course of disease when therapy is less likely to
tory, a diagnosis may be obvious. In some cases, have an impact on chronic progressive injury.
the biopsy specimen may show overlap features,
or there may be elements that do not correlate
clearly with the clinical setting. Close collabora- Diagnostic Findings in Selected Renal
tion by nephrologists and pathologists is essential Diseases
in arriving at the diagnosis. The pathologist must
be familiar with clinical manifestations of renal Minimal Change Disease/Focal
disease, and the nephrologist should be familiar Segmental Glomerulosclerosis
with the terminology used by the pathologist to MCD is diagnosed only after the exclusion of
describe the biopsy findings (Table 2). abnormal findings at the light microscopic level,
When lesions are evaluated, the balance of all with diffuse foot process effacement as the only
elements must be considered. When a typical dis- abnormality by EM. The disease is characteristi-
ease pattern is not present, one must consider cally sensitive to glucocorticoid therapy. How-
whether more than one process is taking place. ever, repeated renal biopsies in patients with
For instance, drug-induced interstitial nephritis apparent MCD initially have shown overt FSGS,
may be superimposed on other glomerular dis- which has a high incidence of progression to end-
ease. This is especially true in the transplant set- stage renal disease (ESRD) [64, 97, 98]. As
ting, where multiple disease processes may occur discussed above, a small sample may not include
at one time. In some instances, the biopsy findings the segmentally sclerotic glomerulus, diagnostic
do not correlate with the patient’s renal function. of FSGS. In FSGS, there is often also hyalinosis,
When such apparent discrepancies are found, one an insudation of plasma proteins and lipids with a
possibility is that the biopsy specimen is not rep- glassy, smooth hyaline appearance on LM. There
resentative of all the nephrons of the kidney. The are no immune deposits, and foot process efface-
number of glomeruli necessary to estimate the ment is present in all glomeruli by EM (Fig. 3).
severity of diseases that show focal distribution The presence of IgM by IF without deposits by
has been discussed above. However, the extent of EM in a biopsy that otherwise appears to be MCD
glomerulosclerosis may in and of itself not corre- (so-called IgM nephropathy) does not have prog-
late with renal function. Tubular atrophy and nostic value [99]. Some variants of FSGS may
interstitial fibrosis may be more closely correlated have prognostic value [100]. A working classifi-
with extent of renal damage and renal function cation of morphological patterns of FSGS has
[94–96]. One must also consider the elements been shown to have prognostic implications
other than structure that influence the patient’s (Table 3) [101]. The usual type is diagnosed
renal function (i.e., blood pressure, filtration prop- when no special features are present. The collaps-
erties of the GBM, and the glomerular filtering ing type of FSGS, characterized by collapse of the
surface area). Patients with enlarged glomeruli, glomerular tuft, either segmental or global with
either caused by compensatory hypertrophy or associated podocyte hypertrophy/hyperplasia,
by a primary pathological process, may show shows a rapid progression to end-stage disease
less deterioration of renal function than expected [102]. The cellular lesion, with endocapillary pro-
based on the extent of glomerular scarring. liferation with frequent foam cells and often with
Similarly, treatment of the patient with podocyte hyperplasia, may represent an early
24 Pediatric Renal Pathology 723

Fig. 3 Minimal change


disease. The foot processes
are effaced (11,000)

Table 3 Working classification of FSGS FSGS studied steroid-resistant children and adults
Possible with FSGS, who were then randomized to cyclo-
prognostic sporine or mycophenolate mofetil and dexameth-
Type Key histologic feature implication asone, in addition to usual ACEI/ARB
FSGS, nos Segmental sclerosis Typical course [107]. Patients with tip lesion had better out-
Collapsing Collapse of tuft, Poor prognosis comes, even in this study where steroid respon-
FSGS GVEC hyperplasia
sive patients, expected to include larger numbers
Cellular Endocapillary ?Early stage
FSGS proliferation, often lesion of tip FSGS, were excluded. Conversely, collaps-
GVEC hyperplasia ing FSGS had worse prognosis, and FSGS nos
Tip lesion Sclerosis of tuft at Better was intermediate [107].
proximal tubule pole prognosis C1q nephropathy is characterized by either no
Perihilar Sclerosis and ?May reflect a sclerosis or segmental glomerulosclerosis or even
variant hyalinosis at vascular secondary type
pole of FSGS mesangial or endocapillary proliferation and
nos not otherwise specified, GVEC glomerular visceral
occasional crescents by LM, with mesangial C1q
epithelial cell deposits and lesser immunoglobulin components
[108]. EM shows mesangial and paramesangial
dense deposits but a lack of reticular aggregates.
Patients typically are adolescents and have
stage of FSGS and appears to occur more often in steroid-resistant NS and do not have clinical evi-
children with FSGS than adults [74, 103]. The tip dence of SLE. A recent study found this lesion in
lesion, that is, adhesion, sclerosis, or 1.9 % of all native kidney biopsies and in 9.2 % of
endocapillary foam cell lesion localized to the pediatric biopsies. Tubular atrophy and interstitial
proximal tubular pole, appears to have better fibrosis were the best correlates of renal insuffi-
prognosis [104–106]. The perihilar variant, with ciency at biopsy and at follow-up. Most patients
sclerosis and hyalinosis localized to the vascular with minimal change-like lesions at biopsy
pole, likely more often represents a secondary achieved remission versus only a third of patients
sclerosing process [101]. Several studies have with segmental sclerosis lesions. Progression to
validated the better prognosis of tip FSGS, the ESRD has occurred in some patients with sclero-
worse prognosis of collapsing FSGS, and the sis at biopsy, but long-term outcome of those
intermediate outcomes of FSGS, not otherwise without sclerosis at presentation has not yet been
specified [101–106]. The NIH Clinical Trial of established [109].
724 A.B. Fogo

Fig. 4 Apparent minimal change disease (MCD) with typical MCD with subsequent benign clinical course (top
subsequent overt focal segmental glomerulosclerosis panel). The patient’s later biopsy, bottom panel, 50 months
(FSGS). The first biopsy from this 5-year-old girl, middle later, showed segmental sclerosis, diagnostic of FSGS
panel, was indistinguishable from MCD, except for (Jones’ stain, 160)
marked glomerular hypertrophy versus age-matched

Because therapy and prognosis are different for with glomerular area greater than 1.5 times that of
MCD versus FSGS, early distinction of these two normal age-matched controls. On the other hand,
entities is of primary interest. We studied pediatric glomerular size equal to or less than normal con-
patients with steroid-resistant nephrotic syndrome trols in this group of patients indicated a good
and MCD on renal biopsies and compared to prognosis. Calculated glomerular diameter for
patients with apparent MCD on biopsy who sub- increased risk of FSGS in these patients less than
sequently progressed to overt FSGS [64]. Mor- 5 years old was more than 118 μ versus 95 μ
phometric analysis of initial biopsies showed that glomerular diameter in age-matched controls.
glomerular size at the onset of disease, before Depending on processing and fixation, these values
sclerosis was apparent, was remarkably larger in may vary (our values are based on paraffin-
patients who subsequently progressed to FSGS embedded tissue fixation in formalin or Zenker’s
(Fig. 4) [64, 110, 111]. There was a higher risk fixative). Normal ranges should be established in
for development of FSGS in patients <5 years old each laboratory assessing glomerular size.
24 Pediatric Renal Pathology 725

From these studies, abnormal glomerular stage is caused by mutations in NPHS2, which
enlargement suggests a high probability of devel- encodes podocin, a key component of the slit
opment of FSGS in pediatric patients with appar- diaphragm [117]. Mutation in ACTN4, which
ent MCD. Causes of abnormal glomerular encodes alpha-actinin-4, causes adult onset auto-
enlargement other than idiopathic FSGS, such as somal dominant FSGS [118]. The prognosis is
diabetes mellitus, cyanotic cardiovascular dis- poor, with progression to renal disease in 50 %
ease, and massive obesity, must be excluded of patients by age 30. Recurrence of NS in the
before such inferences can be made. Interestingly, transplant in patients with genetic FSGS has been
the incidence of FSGS may be increased in these very rare, presumably related to immune events,
settings. The association of abnormal glomerular as the transplant does not carry the mutated gene
growth with development of glomerulosclerosis [118]. TRPC6, a receptor, is mutated in some
may reflect a pathogenic linkage, in that processes kindreds with FSGS, with variable penetrance
leading to excess matrix and sclerosis may be and adult onset [119]. Phospholipase C epsilon
manifested as glomerular growth. This view is mutation (PLCE1) is present in some cases of
supported by the coexistence of these two pro- DMS or rare cases of FSGS, with rare steroid
cesses in many other diseases, including sickle response [120–122]. Other rare mutations have
cell diseases, HIV infection, and reflux been found in both familial and nonfamilial
nephropathy [112]. FSGS [123, 124]. These familial forms do not
Alterations of dystroglycans, specific proteins have specific morphological features of the seg-
that are expressed along the GBM, may be of use mental sclerosis. Children with steroid-resistant
in differentiating MCD in FSGS. α- and NS without family history of FSGS also have
β-dystroglycans were decreased in MCD, but pre- high incidence of abnormalities in crucial
served in the nonsclerotic areas of FSGS in a small podocyte-related genes. Mutations in NPHS2
study [113]. However, we have also observed loss may occur in up to 25 % of pediatric patients
of dystroglycans in the proliferating visceral epi- with sporadic steroid-resistant NS [11, 12].
thelial cells in collapsing FSGS, thus indicating Genetic screening for a panel of these podocyte-
caution in using this marker for definitive diag- related structural genes may prove useful in
nostic distinction of MCD versus FSGS. Expres- identifying those whose NS will prove steroid
sion of CD44, a marker of activated PECs, on the resistant and who have less risk of NS after
glomerular tuft was present in the early recurrence transplant [12].
of FSGS in the transplant with only foot process Congenital nephrotic syndrome (CNS) of
effacement, preceding overt sclerosis, but not in Finnish type is caused by mutation in nephrin.
native kidney MCD [77]. This marker may thus Renal biopsy shows mesangial hypercellularity
also be of use in diagnosing native kidney FSGS or no glomerular lesions and dilated proximal
in its early stages. Recently, molecular studies also tubules by light microscopy [125]. Nephrin local-
indicate distinct gene expression profiles in MCD izes to the slit diaphragm of the podocyte and is
versus FSGS, with much higher ratio of podocin tightly associated with CD2-associated protein
to synaptopodin mRNA in the former, and (CD2AP). Nephrin is thought to function as a
increased podocyte loss in severe proteinuric con- zona occludens-type junction protein. CD2AP
ditions [114–116]. plays a crucial role in receptor patterning and
Specific gene mutations of podocyte-specific cytoskeletal polarity, and its absence resulted in
genes have been identified in patients with famil- sclerosis and foot process effacement in mice,
ial FSGS, and also in pediatric patients with supporting a role for CD2AP in the function of
nonfamilial FSGS. The slit diaphragms are crucial the slit diaphragm. Rare case reports of CD2AP
for regulation of permselectivity, and mutations of mutation in human FSGS exist [126]. Laminin-β2
several slit diaphragm genes cause proteinuria and (LAMB2) is mutated in Pierson syndrome, often
FSGS. Autosomal recessive FSGS with early with microcoria and CNS abnormalities, with
childhood onset and rapid progression to end mesangial sclerosis [127, 128].
726 A.B. Fogo

Fig. 5 Arteriolar fibrin


thrombi with congestion in
capillary loops in
hemolytic-uremic
syndrome (Jones’ stain,
270)

FSGS associated with mitochondrial cytopathy, significance. Generally, both the long-term prog-
due to a mutation of mitochondrial DNA in nosis and the clinical presentation are more severe
tRNAleu(UUR), may show multinucleated if larger vessels are involved. Arterial involve-
podocytes and have abnormal mitochondria by ment was not seen in biopsies performed during
electron microscopic examination. Patients also the first 2 weeks of hospitalization [131]. The
have unusual hyaline lesions in the arterioles. glomerular endothelium is markedly swollen,
Some patients with FSGS without full-blown fea- nearly occluding capillary lumina. Fibrin thrombi
tures of mitochondrial cytopathy (i.e., myopathy, are visualized easily. With chronicity, these areas
stroke, encephalopathy, occasionally diabetes may progress to segmental ischemic collapse with
mellitus, hearing problems, cardiomyopathy) sclerosis, especially when arterioles are involved.
have also been reported to have this mitochondrial The arterioles can become completely occluded
mutation [129]. For further discussion of MCD by thrombi, with necrosis of vessel walls. IF
and FSGS, see ▶ Chap. 27, “Idiopathic Nephrotic shows occasional nonspecific entrapment of C3
Syndrome in Children: Genetic Aspects.” and IgM in injured areas with fibrin and fibrino-
gen. EM shows extreme swelling of the glomeru-
Hemolytic-Uremic Syndrome lar endothelium with increased lucent material in
HUS is the most common disease in children that the lamina rara interna of the basement membrane
is manifested by injury to the microvasculature with entrapped platelets, fibrin, and red cell frag-
and is typically due to E. coli 0:157 verotoxin ments, without immune deposits (Fig. 6). De novo
and associated with diarrhea. The characteristic thrombotic microangiopathy in the renal trans-
lesion of these conditions is thrombotic plant is indistinguishable morphologically from
microangiopathy (TMA). By LM, thrombi in glo- HUS in the native kidney. Cyclosporine and
meruli and arterioles are present (Fig. 5). The FK506 have both been implicated in its pathogen-
renal biopsy findings, rather than clinical param- esis [132, 133]. Mutations of complement regula-
eters, predict long-term prognosis [130]. Patients tory genes are implicated in familial and some
with cortical necrosis have a particularly ominous diarrhea-negative sporadic cases. In adults, addi-
prognosis. The extent of glomerular versus arteri- tional causes of include thrombotic thrombocyto-
olar involvement is also of prognostic penic purpura, related to ADAMTS13 deficiency,
24 Pediatric Renal Pathology 727

Fig. 6 Hemolytic-uremic
syndrome. The lamina rara
interna (endothelial side of
the glomerular basement
membrane) is widened with
increased lucent material.
No immune deposits are
present (3,300)

postpartum renal failure, and various drugs. The associated with more severe glomerular lesions,
morphologic characteristics of the TMA lesion are including crescents, and worse outcome [135].
largely the same regardless of etiology. For further Classification schemas analogous to those for
discussion of HUS, see ▶ Chap. 47, “Renal lupus nephritis have been proposed, but have not
Involvement in Children with HUS.” had widespread use [137, 138]. Therefore, the
International Study Group of IgA Nephropathy
Henoch-Schönlein Purpura/IgA composed of nephrologists and pathologists
Nephropathy reviewed a large number of archival IgA nephrop-
Henoch-Schönlein purpura is often viewed as the athy cases to determine which biopsy lesions cor-
systemic variant of IgA nephropathy (Berger’s dis- related with prognosis [139, 140]. Additional
ease) [4, 134]. The glomerular manifestations are studies have validated these findings, including in
similar, and vary in both, likely depending on children [141–145]. Mesangial hypercellularity,
extent and location of deposits. In some cases, i.e., more than half the glomeruli with more than
there is only focal or even diffuse mild to moderate three nuclei in a mesangial area, endocapillary
mesangial proliferation (Fig. 7) [135]. In more hypercellularity/proliferation, segmental sclerosis,
severe cases, there is focal or even diffuse and significant (>25 %) interstitial fibrosis were
endocapillary hypercellularity/proliferation. In each associated with worse long-term outcome.
severe cases, there may also be necrosis of glomer- Cases with crescents usually had received more
ular tufts with crescents in Bowman’s space. IF by intense immunosuppression and had responded to
definition shows predominance or codominance of this therapy. More recent studies suggest that cel-
mesangial IgA, with capillary loop deposits in lular crescents, if left untreated, also may have
more severe cases. IgG, IgM, and C3 deposits worse prognosis [141]. For further discussion of
may also be detected. The immunoglobulin Henoch-Schönlein purpura/IgA nephropathy, see
deposits are present diffusely, even in glomeruli ▶ Chap. 32, “Immunoglobulin A Nephropathies
that appear normal by LM. By EM, electron- in Children (Includes HSP).”
dense mesangial deposits are present, with occa-
sional “spillover” of deposits to subendothelial Membranoproliferative
regions in the regions adjacent to the mesangium, Glomerulonephritis and C3
particularly in cases with endocapillary hypercel- Glomerulopathies
lularity/proliferative lesions (Fig. 8). Deposits are The MPGN pattern of injury consists of
decreased when clinical remission occurs [136]. In endocapillary/mesangial proliferation and/or
Henoch-Schönlein purpura, deposits are often pre- hypercellularity with double contours of the
sent in subendothelial as well as mesangial areas, GBM. This pattern may be due to chronic
728 A.B. Fogo

Fig. 7 (a) Mesangial and


endocapillary
hypercellularity and small
cellular crescent in Henoch-
Schönlein purpura (Jones’
stain, 430). (b)
Immunofluorescence
demonstrated IgA
mesangial deposits

endothelial injury and thus have no immune com- to distinguish the cause of MPGN pattern lesions.
plex deposits, such as in chronic TMA (see “MPGN” as a diagnosis should only be used when
above). The same light microscopic pattern is the lesion is caused by immune complexes.
seen when immune complexes or monoclonal MPGN is characterized by a tram-track appear-
proteins or cryoglobulins are deposited in the ance of the GBM on silver stain, because of dupli-
subendothelial and mesangial areas, causing a cation around intramembranous/subendothelial
similar LM response. Deposit of C3 only, or dom- deposits and interposition of mesangial cells
inant C3, indicates the underlying etiology and/or macrophages (Fig. 9). The glomeruli are
involves complement dysregulation [13]. This lat- enlarged and hypercellular with a lobular appear-
ter group of disease is called C3 glomerulopathy ance by LM (Fig. 10). There is marked mesangial
and includes dense deposit disease (DDD), where and endocapillary hypercellularity, and occasional
the deposits are characteristically very dense by PMNs and mononuclear cells may be present. By
EM, or C3-dominant glomerulonephritis, where IF, IgG and C3 are present in a coarse granular
deposits have density similar to usual immune pattern along basement membranes, usually with
complexes by EM. Thus, IF and EM are crucial lesser IgM. Subendothelial and mesangial
24 Pediatric Renal Pathology 729

Fig. 8 Immune complex


deposits surrounding
mesangial cell in Henoch-
Schönlein purpura
(3,400)

Fig. 9 Lobular appearance


of glomeruli in MPGN
(200) (Jones’ stain, 400)

immune deposits are seen by EM [4, 59]. MPGN common in adults, more often demonstrates a focal
may be idiopathic or secondary to any of numer- segmental pattern of proliferation, contrasting
ous chronic infections. Hepatitis C positivity, the more diffuse involvement seen in idiopathic
often with associated cryoglobulins, was present MPGN, more common in children.
in about one-fourth of adult cases of MPGN
pattern lesions in adults in Japan and the USA C3 Glomerulopathy: Dense Deposit
[146, 147]. This association has not been demon- Disease and C3 Glomerulonephritis
strated in children with apparent idiopathic MPGN In dense deposit disease (DDD), the glomeruli
[148]. MPGN recurs in 20–30 % of grafts and may may appear similar by LM to those of immune
lead to graft loss [16]. Secondary MPGN, more complex-type MPGN, and this disease has
730 A.B. Fogo

Fig. 10 (a) Double


contours of the glomerular
basement membrane
(“tram-tracking”) in
MPGN, caused by
subendothelial/
intramembranous deposits
and cellular interposition
(Jones’ silver stain, 400).
(b) Chunky irregular
capillary wall and
mesangial deposits in
MPGN, caused by
subendothelial/
intramembranous and
mesangial deposits and
cellular interposition
(anti-IgG, 200)

therefore also previously been called type II electron-dense deposits within the basement
MPGN. However, the pathogenesis is entirely membrane, comprised of complement degrada-
different. These patients often show circulating tion products. A large international study of
IgG autoantibodies, also known as C3 nephritic 69 cases of DDD by the Renal Pathology Society
factor, or have defects in key complement regula- showed mesangial proliferation was most com-
tory molecules or both [13, 15, 149, 150]. Therapy mon, followed in order by membranoproliferative
directed at abnormal complement regulation has appearance with endocapillary proliferation and
therefore been attempted. crescentic or acute proliferative patterns [151,
The basement membranes are deeply eosino- 152]. Renal survival may be worse in DDD than
philic, often with a ribbon garland or sausage- in immune complex-type MPGN (median sur-
shaped contour. By IF, discontinuous smooth lin- vival 8.7 vs. 15.3 years) [153]. The distinction
ear deposits of C3 along the GBM and round between these two diseases is also important
globular mesangial deposits are found, typically since dense deposit disease invariably recurs in
without immunoglobulin staining. The disease is renal transplantation, with worse graft outcome
named dense deposit disease because of the char- [15, 154]. C3-dominant glomerulonephritis
acteristic appearance by EM with strongly (C3GN) also had dominant or only C3 by IF
24 Pediatric Renal Pathology 731

Fig. 11 Diffuse lupus


nephritis (ISN/RPS Class
IV) with massive dense
mesangial and
subendothelial deposits and
fewer deposits in
subepithelial areas
(5,600)

Fig. 12 Membranous
glomerulopathy with
subepithelial deposits and
intervening lamina densa
(seen as spikes by silver
stain on LM) (15,580)

(2 steps more intense than Ig), but usual density of SLE. Most patients with SLE will have mor-
of deposits by EM, and often with underlying phologic manifestation of renal immune deposi-
complement regulatory molecules defects rather tion. However, patients who undergo renal biopsy
than C3 nephritic factor [13]. There is high recur- most often have clinical renal manifestations and
rence in the transplant, about 75 % by 10 years, will have more pronounced changes [27]. Lupus
with worse outcome [14, 155]. Some patients nephritis is characterized by deposits in all ana-
presenting after infection with apparent tomic compartments of the glomerulus (i.e.,
postinfectious GN clinically and morphologically mesangial, subepithelial, and subendothelial
have underlying mutations in complement regu- regions) [23–25, 157] (Figs. 11 and 12). All
latory genes [156], with classic C3GN on follow- immunoglobulin classes with dominant IgG, C3,
up biopsy, and have worse outcome than typical and smaller amounts of C4/C1q are usually
postinfectious GN. found in lupus nephritis deposits (Fig. 13), and
immune complex deposits are seen by
Lupus Nephritis EM. Reticular aggregates (see above) are typi-
Lupus nephritis is not a single disease, but rather cally seen in endothelial cells in any patient with
there is a spectrum of severity of involvement of SLE, regardless of presence of lupus nephritis
the kidney by the immune complex characteristic [4, 59, 157] (Fig. 14).
732 A.B. Fogo

Fig. 13 Immunofluor-
escence of chunky capillary
and small mesangial IgG
deposits in diffuse lupus
nephritis (ISN/RPS Class
IV). The larger segments of
capillary loop staining
correspond to
subendothelial deposits,
with a smooth outer edge
where deposits are molded
underneath the GBM
(4,000)

Fig. 14 Endothelial cell


containing tubular-shaped
reticular aggregates in lupus
nephritis (20,000)

The WHO classifications, either the original or involve less than 50 % of glomeruli. The
modified, were previously most commonly used subendothelial deposits can result in thick, rigid-
[22, 23]. However, the International Society of appearing capillary basement membranes by LM,
Nephrology (ISN) and the Renal Pathology Soci- the so-called wire-loop lesions. “Hyaline”
ety (RPS) put forth a revised lupus nephritis clas- thrombi (aggregates of immune complexes) may
sification to clarify some areas of difficulty in the fill capillary lumina. When the above lesions
previous versions [24, 158, 159]. Greater affect at least 50 % of glomeruli, lupus nephritis
interobserver reproducibility occurs with the is classified as class IV diffuse lupus nephritis.
ISN/RPS classification [158]. In the ISN/RPS The proliferative lesions of both class III and IV
classification, Class I has minimal mesangial are typically associated with subendothelial
deposits with normal LM. Class II is characterized deposits in addition to mesangial deposits, with
by mesangial expansion visible by LM and only rare or scattered subepithelial deposits. IF
mesangial deposits, with only scattered peripheral demonstrates widespread distribution of immune
loop deposits. In class III, focal lupus nephritis, complexes. EM confirms the massive and exten-
deposits are present in mesangial areas, and there sive immune complex deposition (Fig. 11). For
are lesions of either active endocapillary prolifer- class III and IV, the extent of active versus
ation, necrosis, cellular crescents, or chronic chronic lesions (A vs. C) is specified. The seg-
lesions with sclerosis, fibrocellular, or fibrous mental necrotizing lesions in class IV lupus
crescents. These focal lesions, by definition, nephritis may have worse prognosis and are
24 Pediatric Renal Pathology 733

Fig. 15 Anti-glomerular
basement membrane
antibody disease with linear
staining of GBMs by
immunofluorescence for
IgG (125)

often associated with necrosis, and thus the pres- account in part for the tubulointerstitial injury.
ence of these segmental lesions versus global Vascular lesions include immune deposits, or
(S vs. G) endocapillary proliferation is noted thrombotic microangiopathy, often related to
[24, 159, 160]. antiphospholipid antibodies. Vasculitis occurs
Class V membranous lupus nephritis is char- rarely. For further discussion of lupus nephritis,
acterized by predominance of subepithelial see ▶ Chap. 46, “Renal Involvement in Children
deposits in a pattern similar to that of idiopathic with Systemic Lupus Erythematosus.”
membranous glomerulopathy, with added
mesangial deposits (Fig. 12). Primary membra-
nous glomerulopathy, most often due to anti- Anti-glomerular Basement Membrane
bodies to phospholipase A2 receptor (PLA2R), Antibody Disease
is rare in children [60]. Renal biopsy staining
can determine the presence of PLA2R in the Light microscopic examination shows crescentic
deposits, and this staining is negative in cases of glomerulonephritis with focal necrotizing lesions.
membranous lupus nephritis [60, 61]. In some Patients may not always show detectable serum
patients with membranous GP associated with levels of anti-GBM antibodies, especially after the
hepatitis B, PLA2R antibodies may also be pre- acute phase of illness. Serology is positive in 95 %
sent, suggesting perhaps epitope spreading of patients in the first 6 months after onset. However,
[161]. Serum ELISA assays for detection of in all patients, even those with negative serology,
PLA2R autoantibodies are now also available. linear IgG staining by IF of glomerular basement
Subendothelial deposits are minor components membranes is present (Fig. 15). Standard EM does
in class V lupus membranous nephritis. When not visualize the immune deposits, perhaps because
there are superimposed focal or diffuse prolifera- of the diffuse distribution of the antigen (α3 type IV
tive or sclerosing lesions in addition to membra- collagen NC-domain). Patients with more than 50 %
nous changes, both processes are diagnosed, e.g., crescents have a worse prognosis [162].
combined focal or diffuse lupus nephritis and
membranous lupus nephritis, ISN/RPS Class III Granulomatosis with Polyangiitis
+ V or Class IV + V, respectively. Widespread By LM, the appearance of granulomatosis with
chronic sclerosing lesions in a nonspecific pattern polyangiitis is the same as for other nonimmune
in a case of lupus nephritis are defined as Class complex crescentic necrotizing glomerulonephrit-
VI. Tubular basement membrane deposits can ides, such as microscopic polyangiitis or anti-
occur in any class of lupus nephritis and may GBM antibody disease (Fig. 16). The lesions are
734 A.B. Fogo

Fig. 16 (a) Segmental necrosis and crescent in and cellular crescent in granulomatosis with polyangiitis,
granulomatosis with polyangiitis, with irregular focal dis- without glomerular hypercellularity in noninvolved seg-
tribution of lesions. Immunofluorescence was negative ment of the glomerulus. Immunofluorescence was negative
(Jones’ stain, 100). (b) Segmental GBM breaks, necrosis, (Jones’ stain, 430)

focal and segmental. Granulomas are rare in the or when the initial disease is severe, renal biopsy
kidney, and arteritis is rarely found in the small may be done. Glomeruli are enlarged and
sample inherent to the renal needle biopsy. IF stud- hypercellular with prominent endocapillary pro-
ies allow differentiation of the lesion from liferation and infiltration by neutrophils and
anti-GBM antibody disease. It shows fibrin and mononuclear cells (Fig. 17) [165]. In severe dis-
fibrinogen in areas of necrosis, and nonspecific ease, crescents are present. Occasionally, large
trapping of immunoglobulin, especially IgM. By subepithelial deposits (“humps”) can be visual-
EM, immune deposits are absent or very rare ized by LM. These differ from those typical of
(hence the name “pauci”-immune). Distinction membranous glomerulopathy in being more
from microscopic polyangiitis cannot usually be unevenly distributed along the capillary basement
made by renal biopsy findings. Clinical manifesta- membrane and larger in size. The deposits lie on
tions must be used to distinguish between these top of the basement membrane, rather than being
two disorders. For further discussion of embedded within it (as in membranous
granulomatosis with polyangiitis, see ▶ Chap. 45, glomerulopathy), and therefore spikes are not usu-
“Renal Involvement in Children with Vasculitis.” ally present. By IF, there are coarsely granular,
A classification of these lesions shows prognostic discontinuous areas of IgG and prominent C3
value based on the extent of crescents versus scle- along the capillary wall and in the mesangium.
rosis, with those with more than 50 % of glomeruli Postinfectious glomerulonephritis due to
affected with sclerosis having worst prognosis and staphylococcal infection may have dominant IgA
best outcome in those with focal, i.e., <50 % of rather than IgG and can be distinguished from
glomeruli affected, lesions [163]. IgA nephropathy by EM appearance of
deposits [166]. Electron-dense subepithelial
Postinfectious Glomerulonephritis deposits are large, variegated, hump or dome
Patients with typical postinfectious glomerulone- shaped, and irregularly spaced (Fig. 18).
phritis due to streptococcal infection do not usu- Occasional mesangial deposits are present in
ally undergo renal biopsy. Infectious agents other many biopsies. For further discussion of
than streptococci can also cause postinfectious postinfectious glomerulonephritis, see ▶ Chap.
glomerulonephritis [164]. When the diagnosis 31, “Acute Postinfectious Glomerulonephritis in
remains in question, when abnormalities persist, Children.”
24 Pediatric Renal Pathology 735

Fig. 17 Postinfectious
glomerulonephritis with
endocapillary
hypercellularity/
proliferation and PMN
infiltration (PAS, 430)

Fig. 18 Electron
micrograph of subepithelial
large, irregularly spaced,
hump-shaped subepithelial
deposits in postinfectious
glomerulonephritis. The
deposits are variegated and
lie on top of the GBM
(small arrows). There is
endocapillary proliferation
with PMN (long arrow)
infiltration (7,000)

Diabetic Nephropathy efferent arteriolar hyalinization was present in


Diabetic nephropathy affects 30–40 % of patients several of these young patients. A classification
with diabetes mellitus, either type 1 or type 2, with of diabetic nephropathy lesions has been proposed
overt clinical nephropathy manifest 15–20 years to allow stratification of patients with similar
after onset of diabetes. Therefore, diabetic lesions for further study, ranging from just GBM
nephropathy has been considered a disease of thickening to advanced nodular lesions in most
adults. However, adolescents may have diabetic glomeruli [168]. For further discussion of diabetic
renal lesions even after short duration of disease nephropathy, see ▶ Chap. 49, “Diabetic Nephrop-
[167]. In addition, obesity and type 2 diabetes athy in Children.”
mellitus are increasing in children. The structural
changes in these diabetic children include GBM Alport Syndrome/Thin Basement
thickening and mesangial expansion and were Membrane Lesion
associated with proteinuria, hypertension, and Early in life in males with classic Alport syn-
decline in GFR. Overt diabetic nephropathy with drome, and in female carriers, the renal biopsy
nodular glomerulosclerosis and afferent and may show no significant light microscopic
736 A.B. Fogo

abnormalities. At later stages, glomerulosclerosis, normal expression of α5 type IV collagen in


interstitial fibrosis, and prominent interstitial foam Bowman’s capsule, distal tubular basement mem-
cells are typical. These foam cells are not specific brane, and skin, where the α5 collagen chain is
for this disease and are found in numerous part of other heterotrimers. Thus, skin biopsy is
proteinuric states. Glomeruli can show segmental not useful for diagnosis of ARAS, since the
sclerosis. Immunofluorescence may show mutated alpha chains are not normally present in
nonspecific trapping of IgM. By electron micros- the skin. Female heterozygotes for X-linked Alport
copy, the diagnostic lesion consists of irregular syndrome show mosaic staining of GBM and distal
thinned and thickened areas of the glomerular TBM for α3, α4, and α5 type IV collagen chains
basement membranes with splitting and irregular and skin mosaic staining for α5 type IV collagen
multilaminated appearance of the lamina densa, due to the lyonization effect. Patients with autoso-
so-called basket weaving. In between these lami- mal dominant Alport syndrome have not been
nae, a granular, mottled material is present. Sim- studied immunohistochemically. Of note, occa-
ilar GBM changes by EM, but with normal sional cases with Alport syndrome clinically and
collagen chain staining (see below), occur in by renal biopsy showed apparent normal α5 type
Frasier syndrome. This entity is due to WT-1 IV pattern of skin IF staining, and about 20 % of
mutation and manifests as NS with FSGS by male X-linked Alport patients show faint or even
LM, no deposits, pseudohermaphroditism, and normal staining of the GBM for α3 and α5, likely
increased risk of gonadoblastoma [169]. At early because the antigenic site recognized by the anti-
stages of Alport, i.e., in children or carrier women, body has not been altered by the mutation [170].
the glomerular basement membrane shows only Thinning of the GBM is also the characteristic
thinning. Some males with classic Alport syn- finding in benign familial hematuria [81, 82]. Most
drome only have glomerular basement membrane familial benign hematuria is caused by a carrier state
thinning even at advanced clinical stages [39, of ARAS, with mutation of either α3 or α4, causing
170]. Rarely, Alport patients may have prolifera- thin basement membranes and hematuria [172]. The
tive lesions with IF and EM deposits within diagnosis of thin basement membranes is based on
lamellated GBM areas, suggesting trapping of morphometric measurements from electron micro-
immunoglobulins rather than a superimposed scopic prints and was present in 1.9 % of a large
immune complex disease [171]. However, we native kidney biopsy series [173, 174]. LM and
have also observed superimposed glomerulone- standard IF are normal. The GBM thickness nor-
phritides in Alport patients. mally increases with age. Normal thickness in
Immunostaining for type IV collagen chains adults in one series was 373  42 nm in men versus
can aid in the interpretation of thin basement 326  45 nm in women. GBM thickness <250 nm
membranes [170]. Heterotrimers of α3, α4, and has been used as a cutoff in many series [175]. In
α5 type IV collagen are a key normal component another series, average was 330  50 nm in males
of the GBM. Mutation of α5 type IV collagen in and 305  45 nm in women [173]. In children, the
X-linked Alport syndrome, or of α3 or α4 diagnosis of thin basement membranes must
subchains in autosomal forms, prevents incorpo- be made with caution, establishing normal
ration of the other chains into this heterotrimer age-matched controls within each laboratory. In
of the GBM. Thus, in kidney biopsies, about our laboratory, we found a range of GBM thickness
70–80 % of males with X-linked Alport syndrome in normal children, from approximately 110 nm at
lack staining of GBM, distal tubular basement age 1 year to 222  14 nm in seven year olds. As
membrane, and Bowman’s capsule for α3, α4, mentioned above, thin GBM (without lamellation)
and α5 (IV) chains [170]. In autosomal recessive is also the early, or may be the only, manifestation in
Alport syndrome (ARAS), due to mutations of some kindreds with Alport syndrome and in female
either α3 or α4, the GBMs also usually show no carriers of X-linked Alport. Thus, the presence of
expression of α3, α4, or α5 type IV collagen. In thin GBM cannot per se be taken to categorically
contrast to X-linked cases, ARAS patients have indicate a benign prognosis.
24 Pediatric Renal Pathology 737

Prognostic Implications of Biopsy controversial [27, 159]. Lesions of activity in


Findings lupus nephritis include endocapillary prolifera-
tion, necrosis, cellular crescents, and interstitial
When the biopsy sample is adequate, extensive, inflammatory cells. Lesions that indicate chronic-
severe, and irreversible lesions signify a dismal ity include tubular atrophy, interstitial fibrosis,
prognosis for the patient. Globally sclerotic glo- glomerular sclerosis, and fibrous crescents.
meruli are not amenable to treatment, although Although assessment of activity and chronicity
evidence from human diabetic nephropathy and indices is useful for population groups, these
animal studies indicates that the earlier stages of appear to have less absolute information to guide
sclerosis may be affected by some therapeutic assessment in individual patients. Nonetheless, in
interventions and may even be reversible large series, assessment of indices of activity and
[176–179]. Similarly, active lesions with ongoing severity in patients with lupus nephritis or other
cellular crescents, necrosis, and inflammatory diseases has shown some correlation with prog-
infiltrate are potentially dramatically modulated nosis and response to therapy. Diffuse prolifera-
by therapy, allowing subsequent healing. There tive lesions, extensive crescents, segmental
may be minimal irreversible damage to glomeru- necrosis, and tubulointerstitial fibrosis are associ-
lar structures when intervention occurs early. ated with progression to ESRD [27, 180,
Although the renal biopsy may yield a diagno- 181]. The best prognostic indicator in one study
sis, there is less information of prognostic indica- was the proportion remaining of intact glomeruli
tors in diseases that have a variable course. in follow-up biopsies [182].
Extensive analysis aimed at determining histo- IgA nephropathy was previously thought to
logic features associated with poor prognosis has have a benign prognosis. In a large series of adult
been done in some diseases discussed below. patients with IgA nephropathy, poor prognosis was
Classification schemes, especially for lupus indicated by segmental glomerulosclerosis, adhe-
nephritis and membranous glomerulopathy, sions or crescents, and tubulointerstitial fibrosis
imply progression from one stage of disease to [183]. Progression occurs in 11–15 % of pediatric
the next. Although sequential biopsies have patients [4, 112, 135, 184]. Scoring of activity and
illustrated progression from focal to diffuse chronicity of lesions has been correlated with clin-
proliferative glomerulonephritis in lupus nephri- ical course. Activity was assessed by degrees of
tis, patients may change from any class to any crescent formation, mesangial proliferation, and
other, with or without treatment, and recurrence interstitial infiltrate. Chronicity was scored by
in the transplant may be a different ISN/RPS class degrees of fibrous crescents, segmental and global
than that observed in the native kidney [22, 23, 25, sclerosis, tubular atrophy, and interstitial fibrosis
27, 159]. In lupus nephritis, patients with less [185]. The Oxford IgA nephropathy study identi-
severe proliferative disease appear to have better fied four lesions associated with worse long-term
prognoses than those with extensive involvement outcome, namely, mesangial hypercellularity (M),
of glomeruli, especially with necrosis. i.e., more than half the glomeruli with more than
Although the presence of cellular crescents is three nuclei in a mesangial area, endocapillary
associated with activity of disease clinically, the hypercellularity/proliferation (E), segmental scle-
renal biopsy offers additional prognostic informa- rosis (S), and significant (25 %) tubulointerstitial
tion beyond that gleaned from the clinical presen- fibrosis (T), also validated in children with IgA
tation [27, 180]. Focal and diffuse proliferative nephropathy [139–145]. Extension of deposits to
lesions (ISN/RPS III and IV) may present very peripheral capillary wall areas has also been
similarly clinically, but only the latter appears to reported as a poor prognostic indicator, but did
require intense, long-term immunosuppression. not add independent risk beyond the four MEST
Whether the segmental, more often necrotizing criteria of the Oxford IgA nephropathy scoring
lupus nephritis lesions have worse outcome than [141]. Crescents may also have prognostic
more diffuse global proliferative lesions is implications [141].
738 A.B. Fogo

Fig. 19 Acute rejection,


classified as type I by
Cooperative Clinical Trials
in Transplantation criteria.
There is interstitial
lymphocytic infiltrate with
tubulitis, activated
lymphocytes, tubular cell
injury, and interstitial
edema (Jones’ stain, 220)

Fig. 20 Acute vascular


rejection, classified as type
II by Cooperative Clinical
Trials in Transplantation or
also by Banff criteria. There
is subendothelial infiltration
by lymphocytes in this
artery, the so-called
endothelialitis (Jones’ stain,
220)

Focal glomerulosclerosis superimposed on disease, and the procedure itself, such as acute
membranous glomerulopathy has been associated tubular injury/necrosis.
with more severe tubulointerstitial nephritis and a
worse outcome. This lesion was present in 20 % of Rejection
children with hepatitis B-associated membranous Acute rejection is diagnosed by the presence of
glomerulopathy [186]. either interstitial inflammation with lymphocytes
and plasma cells infiltrating tubules (tubulitis, the
hallmark of acute interstitial type rejection;
Renal Transplant Biopsy Fig. 19) or, when more severe, by extension of
this process to vessels, with subendothelial arte-
The primary use of biopsy in the renal transplan- rial or arteriolar infiltration by lymphocytes
tation is to uncover the reason for altered renal (endothelialitis, the hallmark of acute vascular
function. Causes of renal dysfunction in the trans- rejection; Fig. 20). The interstitial changes of
plant can be broadly divided into those related to acute rejection are not pathognomonic. In con-
rejection, drug toxicity, recurrent or de novo trast, the finding of endothelialitis is highly
24 Pediatric Renal Pathology 739

specific for acute vascular rejection. Appropriate


stains, such as PAS, must be used to allow visu-
alization of the tubular basement membrane and
identification of tubulitis. An adequate specimen
for evaluation of possible rejection should contain
at least two cores, with at least seven glomeruli
and two arteries [187].
Several schemes have been used to diagnose
and classify rejection: the Banff scoring system,
based on detailed scoring of various components
of injury, and the Cooperative Clinical Trials in
Transplantation (CCTT) criteria [187–189]. In
both classification schemes, acute rejection is
based on the presence of tubulitis or
endothelialitis, i.e., lymphocytes in the tubule
under the tubular basement membrane or under-
neath the endothelium of arteries. Other inflam-
matory cells (e.g., eosinophils, neutrophils, and
plasma cells), although much fewer in number
than T lymphocytes, may also contribute to the
infiltrate in acute rejection. Type I rejection in
both schemas is diagnosed when interstitial lym-
phocytic infiltrate and tubulitis are present
(>25 % of parenchyma infiltrated in Banff,
>5 % in CCTT) (Fig. 20). Infiltrate and tubulitis Fig. 21 Antibody-mediated rejection is characterized by
C4d positivity in peritubular capillaries (200)
less than specified for type I is called “borderline”
by Banff criteria [188, 189]. Type II acute vascular
rejection is diagnosed in both schemas when there follow-up. In contrast, treatment of such subclin-
is mild or moderate endothelialitis (arteritis). ical rejection in the early time period after trans-
Severe acute vascular rejection, type III, is diag- plantation resulted in better preserved renal
nosed when there is transmural vascular inflam- function at 24 months [191].
mation and/or fibrinoid necrosis. These types are There are no specific IF or electron micro-
differentiated not only based on histologic pattern scopic immune complexes associated with acute
but also on differences in underlying mechanisms rejection. Molecular studies indicate the possibil-
and response to therapies: type I and II are likely ity of earlier, more sensitive, and specific diagno-
T-cell-dependent processes and are separated sis of acute rejection using these techniques (see
based on the likely greater clinical severity of below) [192]. In particular, the presence of C4d, a
any rejection when endothelialitis is present, complement breakdown product which binds
whereas antibody-mediated mechanisms contrib- covalently to tissue, in peritubular capillaries is
ute to type III changes. Isolated type II highly associated with anti-donor antibodies
endothelialitis lesions are often associated with (humoral rejection) [193] (Fig. 21). Diagnosis of
antibody-mediated rejection [190]. humoral, antibody-mediated rejection has impor-
Identification of acute rejection at earlier stages tant therapeutic and prognostic implications. C4d
and thus initiation of treatment at milder levels of staining can be done on frozen or fixed paraffin-
injury appear to be clinically important. Thus, processed tissue, although the latter is less
mild tubulitis that is borderline by Banff criteria, sensitive [194].
even in normally functioning grafts, was found The changes of chronic rejection include inti-
to be predictive of higher serum creatinine at mal fibrosis of arteries, interstitial fibrosis, and
740 A.B. Fogo

transplant glomerulopathy [188, 195]. A previous striped distribution of interstitial fibrosis caused
or baseline biopsy is necessary to prove that inti- by injury along the medullary rays [195]. This
mal fibrosis is de novo and potentially represents pattern often cannot be gleaned in small needle
chronic rejection, rather than a preexisting, biopsies. FSGS with ischemic, corrugated GBMs
nonspecific change in the graft. Interstitial fibrosis in remaining glomeruli may also result from
is also a nonspecific finding and may result from calcineurin inhibitor toxicity and can be associ-
various injuries. Transplant glomerulopathy is a ated with significant proteinuria [197].
more specific lesion indicative of chronic rejec- Calcineurin inhibitors have also been associ-
tion and is particularly linked to previous ated with thrombotic microangiopathy lesions
antibody-mediated rejection. By LM, the glomer- (see above) [197]. Of note, thrombotic
uli show basement membrane double contours microangiopathy can occur in patients who are
and corrugation and even segmental sclerosis recipients of transplants of kidneys or other
with hyalinosis. The latter lesion likely resulted organs and following radiation, with or without
in reports of de novo “idiopathic FSGS” in calcineurin inhibitor treatment. In some patients,
the transplant. However, in transplant collapsing-type glomerulosclerosis may be asso-
glomerulopathy, there is widening of the lamina ciated with cyclosporine toxicity, likely
rara interna of the GBM with cellular interposition representing a response to severe vascular injury
and new basement membrane formation by and ischemia and usually with less complete foot
EM. Reduplication of basal lamina of peritubular process effacement than in idiopathic collapsing
capillaries is suggested to be more specific for glomerulopathy in the native kidney, and with
transplant glomerulopathy but may also occur in improvement on cessation of CNI therapy [198].
some other glomerular diseases and HUS [196].
Recurrent and De Novo Disease
Calcineurin Inhibitor Toxicity Recurrent and de novo diseases are important
Calcineurin inhibitor toxicity may manifest in causes of renal allograft injury, affecting approx-
various ways. Tacrolimus (FK506) has much the imately 10 % of renal allografts [199]. Of all graft
same spectrum of toxicity as cyclosporine loss, 2–4 % is due to recurrence of disease. IF
[197]. The most common morphologic lesion in microscopy should be performed in all transplant
patients with a clinical diagnosis of cyclosporine biopsies to rule out this possibility. When IF or
toxicity, as verified by clinical follow-up, is that of LM findings in conjunction with the clinical set-
a normal kidney biopsy morphologically. In these ting indicate an undetermined lesion, electron
patients, renal dysfunction is due to reversible, microscopic study should also be performed [68].
calcineurin inhibitor-induced vasoconstriction In children, the most common recurrent dis-
and hypofiltration. Morphologic changes sugges- eases include IgA nephropathy and Henoch-
tive, but not pathognomonic, of calcineurin inhib- Schönlein purpura, MPGN, dense deposit disease,
itor toxicity include arteriolopathy with injury to and FSGS [200]. Although SLE has been reported
the endothelium and vascular smooth muscle to recur only rarely, our experience indicates a
cells. In its classic form, this injury results in recurrence rate of approximately 30 %
nodular IgM IF positivity along the apical side [201]. However, morphologic recurrence of dis-
of the arteriole, with necrosis and smooth muscle ease does not necessarily lead to graft loss
cell injury demonstrated by EM [197]. By LM, [200]. Dense deposit disease recurs morphologi-
concentric hyalinosis is present, whereas typically cally in nearly all patients, with worse graft out-
eccentric, more segmental hyalinosis is associated come [14]. In contrast, HUS has an overall
with hypertension. Isometric tubular recurrence rate of 15–25 %, reflecting largely
vacuolization in a patchy distribution, although invariable recurrence in those with atypical HUS
not specific, is also indicative of cyclosporine due to inherited complement dysregulation, with
toxicity; calcineurin inhibitor toxicity results in a high rates of graft loss. Although IgA
24 Pediatric Renal Pathology 741

nephropathy recurs morphologically in approxi- occasional eosinophils, with enlarged tubular


mately 50 % of patients, only 10 % of grafts cells with smudgy nuclei. Early lesions (stage A)
with recurrent disease are lost. MPGN recurs in with minimal inflammation, fibrosis, or injury
20–30 %, with 10–40 % graft loss. FSGS recurs have better prognosis than stage B with marked
in 20–30 % of cases, resulting in graft loss in viral changes and inflammation and moderate
30–50 % of these. Of note, in recurrent FSGS, fibrosis, or stage C, with extensive fibrosis
the only morphologic change found in the first [90]. Polyoma virus infection is confirmed by
weeks after recurrence of proteinuria is foot pro- immunostaining for SV40 large T antigen present
cess effacement, with early segmental sclerosis in replicating polyoma virus (includes BK, JC and
detectable at 6–8 weeks after recurrent NS. SV40). In some polyoma virus nephropathy
De novo disease may also affect the transplant. cases, there may be associated granular tubular
Membranous glomerulopathy is the most com- basement membrane deposits staining with IgG
mon de novo glomerulonephritis in the transplant and C3 and visualized by EM [207]. Polyoma
and is linked to antibody-mediated rejection virus nephropathy can respond to decreased or
[202]. Glomerulonephritis related to infections, changed immunosuppression and antiviral
such as hepatitis C-related MPGN, also can therapy.
occur in the transplant. Early changes of diabetic
nephropathy develop much more rapidly in the
transplant than in the native kidney and New Methods for the Future
may occur within a few years, whether diabetes
preexisted or is corticosteroid induced [203, 204]. With the recent surge of application of molecular
Thrombotic microangiopathy may be related to biology techniques to the study of renal disease,
drug toxicity (see above) or be idiopathic in the candidate factors involved in pathogenesis and
transplant. progression of disease are being studied in animal
Posttransplant lymphoproliferative disease models. Studies in human beings have also com-
(PTLD) is due to the unrestrained proliferation menced. With further development of such stud-
of B lymphocytes, most often because of transfor- ies, we may identify specific abnormal processes
mation by Epstein-Barr virus, and is an aggressive and thus target therapy more specifically.
process, which, if untreated, disseminates and Research techniques that have been advanta-
may cause death [205]. PTLD may respond to geously applied to elucidate pathogenesis of
decreased immunosuppression. An expansile disease include immunostaining; identifying spe-
lymphoid infiltrate with atypical, transformed cific antigen in deposits of membranous
lymphocytes and serpiginous necrosis are features glomerulopathy, e.g., PLA2R in primary membra-
suggestive of PTLD [205]. Immunohistochemical nous GP or causes of secondary membranous GP
studies can be used to detect Epstein-Barr virus to (thyroglobulin with Hashimoto’s disease and hep-
further support this diagnosis. Typing studies of atitis B and C antigen), and light chains or
the lymphocytic infiltrate are not often helpful paraproteins in plasma cell dyscrasia-associated
because most PTLD is polytypic, rather than diseases; identification of specific type IV colla-
clonal. Of note, acute rejection and PTLD may gen abnormality in Alport syndrome, C4d as a
be present concurrently. marker of humoral rejection, and novel insights
Polyoma virus nephropathy (PVN), most often into C3 glomerulopathies; and elucidation of
due to the BK virus, has increased in the last years pathogenesis of specific E. coli-associated toxins
in the transplant, perhaps related to increased in some forms of HUS.
immunosuppression [90]. PVN occurs in both Current studies are aimed at understanding
adult and pediatric transplant recipients disease etiologies and mechanisms at a molecular
[206]. The biopsy shows a pleomorphic infiltrate and proteomic level, studying renal biopsies by
with lymphocytes, plasma cells, PMNs, and laser capture microdissection (LCM), with mass
742 A.B. Fogo

spectrometry, real-time reverse transcription poly- Acknowledgments The author wishes to thank Drs. Tina
merase chain reaction (RT-PCR), and in situ Kon, Kathy Jabs, and Tray Hunley for their suggestions.
hybridization techniques [114–116, 208, 209].
Recent efforts have expanded use of these tech-
niques to study mechanisms of injury and progres- References
sion. Competitive and real-time RT-PCR have
been used successfully on small cores and even 1. Hisano S, Kwano M, Hatae K, et al. Asymptomatic
single isolated glomeruli from human biopsies isolated microhaematuria: natural history of 136 chil-
dren. Pediatr Nephrol. 1991;5:578–81.
[114, 116]. Modulation of growth factors, colla-
2. Turi S, Visy M, Vissy Á, et al. Long-term follow-up of
gens, cytokines, chemokines, and their receptors patients with persistent/recurrent, isolated
has been investigated molecularly in renal biop- haematuria: a Hungarian multicentre study. Pediatr
sies. Cytokines and chemokines and their recep- Nephrol. 1989;3:235–9.
tors were upregulated in diseases with 3. Trachtman H, Weiss RA, Bennett B, et al. Isolated
hematuria in children: indications for a renal biopsy.
macrophage influx and mesangial cell prolifera- Kidney Int. 1984;25:94–9.
tion, supporting an important role in initiating and 4. Silva FG. Overview of pediatric nephropathology.
perpetuating injury. Such approaches can offer Kidney Int. 1988;33:1016–32.
exciting new mechanistic insights into renal 5. Sibley RK, Mahan J, Mauer SM, et al. A clinicopath-
ologic study of forty-eight infants with nephrotic syn-
diseases. drome. Kidney Int. 1985;27:544–52.
In parallel, genetic studies are targeted both at 6. Habib R, Kleinknecht C. The primary nephrotic syn-
diagnosis and identifying patients at risk for pro- drome of childhood. Classification and clinicopatho-
gression in diseases with a variable course, such logic study of 406 cases. In: Sommers SC, editor.
Pathology annual. New York: Appleton-Century-
as diabetes and IgA nephropathy. Genetic screen- Crofts; 1971. p. 417–74.
ing for key podocyte-related mutations is often 7. Cameron JS. Histology, protein clearances, and
done in children with FSGS. Noninvasive urinary response to treatment in the nephrotic syndrome.
proteomic and microvesicle studies are also BMJ. 1968;4:352–6.
8. White RHR, Glasgow EF, Mills RJ. Clinicopatholog-
explored to identify particular signature patterns ical study of nephrotic syndrome in childhood.
indicative or predictive of specific lesions or risks Lancet. 1970;1:1353–9.
[210–212]. 9. Rapola J. Congenital nephrotic syndrome. Pediatr
Diagnostic use of RT-PCR and in situ Nephrol. 1987;1:441–6.
10. Antignac C. Molecular basis of steroid-resistant
hybridization have focused on detection of nephrotic syndrome. Nefrologia. 2005;25 Suppl
viruses, including hepatitis B and C, cytomegalo- 2:25–8.
virus, polyoma virus, and Epstein-Barr virus. 11. Ruf RG, Lichtenberger A, Karle SM, et al. Patients
Endothelial injury transcripts in kidney with mutations in NPHS2 (podocin) do not respond to
standard steroid treatment of nephrotic syndrome. J
biopsies may allow diagnosis and earlier Am Soc Nephrol. 2004;15:722–32.
detection of specific types of injury in the 12. Giglio S, Provenzano A, Mazzinghi B, et al.
transplant [213]. Heterogeneous genetic alterations in sporadic
Together, these approaches promise to map nephrotic syndrome associate with resistance to
immunosuppression. J Am Soc Nephrol. 2015;26
risks and mechanisms of disease initiation and (1):230–6.
progression and point to targets to achieve resolu- 13. Pickering MC, D’Agati VD, Nester CM, et al. C3
tion of injury. Sequential biopsies with evaluation glomerulopathy: consensus report. Kidney Int.
of changes in structure and patterns of abnormal 2013;84:1079–89.
14. Zand L, Lorenz EC, Cosio FG, et al. Clinical findings,
factors and modulation by therapy may be neces- pathology, and outcomes of C3GN after kidney trans-
sary to fully understand pathogenesis. Instead of plantation. J Am Soc Nephrol. 2014;25:1110–7.
diagnoses of morphologic patterns recognized by 15. Habib R, Gubler M-C, Loirat C, et al. Dense deposit
current techniques in the renal biopsy specimen, disease: a variant of membranoproliferative glomeru-
lonephritis. Kidney Int. 1975;7:204–15.
molecular techniques may allow more precise 16. Cameron JS. Recurrent primary disease and de novo
diagnosis of the specific diseases and identifica- nephritis following renal transplantation. Pediatr
tion of injury mechanisms. Nephrol. 1991;5:412–21.
24 Pediatric Renal Pathology 743

17. Mustonen J, Pasternack A, Helin H, et al. Renal 32. Gault MH, Muehrcke RC. Renal biopsy: current
biopsy in acute renal failure. Am J Nephrol. views and controversies. Nephron. 1983;34:1–34.
1984;4:27–31. 33. Madaio MP. Renal biopsy. Kidney Int. 1990;38:
18. Jennette JC, Wilkman AS, Tuttle RH, et al. Frequency 529–43.
and pathologic significance of anti-proteinase 3 and 34. Walker PD, Cavallo T, Bonsib SM, Ad Hoc Commit-
anti-myeloperoxidase antineutrophil cytoplasmic tee on Renal Biopsy Guidelines of the Renal Pathol-
autoantibodies (ANCA) in immune complex ogy Society. Practice guidelines for the renal biopsy.
glomerulonephritis (Abstract). Lab Invest. 1996;74: Mod Pathol. 2004;17:1555–63.
167A. 35. de Chadarevian J-P, Kaplan BS. The kidney biopsy.
19. Rao JK, Weinberger M, Oddone EZ, et al. The role of In: Barakat AY, editor. Renal disease in children,
antineutrophil cytoplasmic antibody (c-ANCA) test- Clinical evaluation and diagnosis. New York:
ing in the diagnosis of Wegener granulomatosis. A Springer; 1990. p. 117–32.
literature review and meta-analysis. Ann Intern Med. 36. Greenbaum LA, Simckes AM, McKenney D,
1995;123:925–32. et al. Pediatric biopsy of a single native kidney.
20. Jennette JC. Antineutrophil cytoplasmic Pediatr Nephrol. 2000;15:66–9.
autoantibody-associated disease: a pathologist’s per- 37. Shidham GB, Siddiqi N, Beres JA, Logan B, Nagaraja
spective. Am J Kidney Dis. 1991;18:164–70. HN, Shidham SG, Piering WF. Clinical risk factors
21. Jennette JC, Thomas DB. Pauci-immune and associated with bleeding after native kidney biopsy.
antineutrophil cytoplasmic autoantibody-mediated Nephrology. 2005;10:305–10.
crescentic glomerulonephritis and vasculitis. In: 38. Wiseman DA, Hawkins R, Numerow LM,
Jennette JC, Olsson JL, Silva FG, D’Agati VD, edi- et al. Percutaneous renal biopsy utilizing real time,
tors. Heptinstall’s pathology of the kidney. 7th ultrasonic guidance and a semiautomated biopsy
ed. Philadelphia: Wolters Kluwer; 2015. device. Kidney Int. 1990;38:347–9.
22. McCluskey RT. Lupus nephritis. In: Sommers SC, 39. Donovan KL, Thomas DM, Wheeler DC, et al.
editor. Kidney pathology decennial 1966–1975. East Experience with a new method for
Norwalk: Appleton-Century-Crofts; 1975. p. 435. percutaneous renal biopsy. Nephrol Dial Transplant.
23. Churg J, Sobin LH. Monograph: renal disease: clas- 1991;6:731–3.
sification and atlas of glomerular diseases. New York: 40. Oberholzer M, Trohorst E, Perret E, et al. Minimum
Igaku-Shoin/World Health Organization (WHO); sample size of kidney biopsies for semiquantitative
1982. and quantitative evaluation. Nephron. 1983;34:
24. Weening JJ, D’Agati VD, Schwartz MM, et al. The 192–5.
classification of glomerulonephritis in systemic lupus 41. Simckes AM, Blowey DL, Gyves KM, Alon US.
erythematosus revisited. J Am Soc Nephrol. 2004;15: Success and safety of same-day kidney biopsy in
241–50. children and adolescents. Pediatr Nephrol.
25. Rush PJ, Baumal R, Shore A, et al. Correlation of 2000;14:946–52.
renal histology with outcome in children with lupus 42. Sinha MD, Lewis MA, Bradbury MG, Webb NJA.
nephritis. Kidney Int. 1986;29:1066–71. Percutaneous real-time ultrasound-guided
26. Schwartz MM, Bernstein J, Hill GS, Lupus Nephritis renal biopsy by automated biopsy gun in
Collaborative Study Group, et al. Predictive value of children: safety and complications. J Nephrol.
renal pathology in diffuse proliferative lupus glomer- 2006;19:41–4.
ulonephritis. Kidney Int. 1989;36:891–6. 43. Sweeney C, Geary DF, Hebert D, Robinson L,
27. Giannico G, Fogo AB. Lupus nephritis: is the kidney Langlois V. Outpatient pediatric renal transplant
biopsy currently necessary in the management of biopsy – is it safe? Pediatr Transplant. 2006;10:
lupus nephritis? Clin J Am Soc Nephrol. 2013;8: 159–61.
138–45. 44. Häyry P, von Willebrand E. Fine needle aspiration in
28. Remuzzi G, Ruggenenti P, Perico N. Chronic renal transplantation pathology. In: Sabe GE, editor. The
diseases: renoprotective benefits of renin-angiotensin pathology of organ transplantation. Boston:
system inhibition. Ann Intern Med. 2002;136: Butterworths; 1990. p. 285–301.
604–15. 45. Yussim A, Shapira Z, Shmueli D, et al. Use of mod-
29. van de Heuvel LPWJ, Schröder CH, Savage COS, ified fine needle aspiration for study of glomerular
et al. The development of anti-glomerular basement pathology in human kidneys. Kidney Int.
membrane nephritis in two children with Alport syn- 1990;37:812–7.
drome after renal transplantation: characterization of 46. Al Rasheed SA, Al Mugeiren MM, Abdurrahman
the antibody target. Pediatr Nephrol. 1989;3:406–13. MB, et al. The outcome of percutaneous renal biopsy
30. Savige J, Gregory M, Gross O, Kashtan C, Ding J, in children: an analysis of 120 consecutive cases.
Flinter F. Expert guidelines for the management of Pediatr Nephrol. 1990;4:600–3.
Alport syndrome and thin basement membrane 47. Edelmann Jr CM, Greifer I. A modified technique for
nephropathy. J Am Soc Nephrol. 2013;24:364–735. percutaneous needle biopsy of the kidney. J Pediatr.
31. Kark RM. Renal biopsy. JAMA. 1968;205:220–6. 1967;70:81–6.
744 A.B. Fogo

48. Abdurraman MB. Percutaneous renal biopsy in a 65. Hurley RM, Drummond KN. Glomerular enlarge-
developing country: experience with 300 cases. Ann ment in primary renal disease. A quantitative study.
Trop Paediatr. 1984;4:25–30. Arch Pathol. 1974;97:389–91.
49. Karafin L, Kendall AR, Fleisher DS. Urologic com- 66. Shindo S, Yoshimoto M, Kuriya N, et al. Glomerular
plications in percutaneous renal biopsy in children. basement membrane thickness in recurrent and per-
J Urol. 1970;103:332–5. sistent hematuria and nephrotic syndrome: correlation
50. Carvajal HF, Travis LB, Srivastava RN, et al. with sex and age. Pediatr Nephrol. 1988;2:196–9.
Percutaneous renal biopsy in children: an analysis of 67. Morita M, White RHR, Raafat F, et al. Glomerular
complications in 890 consecutive biopsies. Tex Rep basement membrane thickness in children. A mor-
Biol Med. 1971;29:253–64. phometric study. Pediatr Nephrol. 1988;2:190–5.
51. Colodny AH, Reckler JM. A safe, simple and reliable 68. Chang A, Gibson IW, Cohen AH, Weening JJ,
method for percutaneous (closed) renal biopsies in Jennette JC, Fogo AB, Renal Pathology Society. A
children: results in 100 consecutive patients. J Urol. position paper on standardizing the nonneoplastic
1975;113:222–4. kidney biopsy report. Clin J Am Soc Nephrol.
52. Welt L. Questionnaire on renal biopsies. JAMA. 2012;7:1365–8.
1968;205:226. 69. Kaplan C, Pasternack B, Shah H, et al. Age-related
53. McVicar M, Nicastri AD, Gauthier B. Improved renal incidence of sclerotic glomeruli in human kidneys.
biopsy technique in children. NY State J Med. Am J Pathol. 1975;80:227–34.
1974;74:830–1. 70. Kappel B, Olsen S. Cortical interstitial tissue and
54. White RHR. Observations on percutaneous renal sclerosed glomeruli in the normal human kidney,
biopsy in children. Arch Dis Child. 1963;38:260–6. related to age and sex. A quantitative study. Virchows
55. Burstein DM, Korbet SM, Schwartz MM. The use of Arch (Pathol Anat). 1980;387:271–7.
the automatic core biopsy system in percutaneous 71. Smith SM, Hoy WE, Cobb L. Low incidence of
renal biopsies: a comparative study. Am J Kidney glomerulosclerosis in normal kidneys. Arch Pathol
Dis. 1993;22:545–52. Lab Med. 1989;113:1253–6.
56. Corwin HL, Schwartz MM, Lewis EJ. The impor- 72. Nash MA, Greifer I, Olbing H, et al. The significance
tance of sample size in the interpretation of the renal of focal sclerotic lesions in glomeruli in children.
biopsy. Am J Nephrol. 1988;8:85–9. J Pediatr. 1976;88:806–13.
57. Cohen AH, Nast CC, Adler SG, et al. Clinical utility 73. Chiang ML, Hawkins EP, Berry PL, et al. Diagnostic
of kidney biopsies in the diagnosis and management and prognostic significance of glomerular epithelial
of renal disease. Am J Nephrol. 1989;9:309–15. cell vacuolization and podocyte effacement in chil-
58. Haas M. A reevaluation of routine electron micros- dren with minimal lesion nephrotic syndrome and
copy in the examination of native renal biopsies. J Am focal segmental glomerulosclerosis: an ultrastructural
Soc Nephrol. 1997;8:70–6. study. Clin Nephrol. 1988;30:8–14.
59. Silva FG, Pirani CL. Electron microscopic study of 74. Silverstein DM, Craver R. Presenting features and
medical diseases of the kidney: update 1988. Mod short-term outcome according to pathologic variant
Pathol. 1988;1:292–315. in childhood primary focal segmental glomerulo-
60. Beck Jr LH, Bonegio RG, Lambeau G, Beck DM, sclerosis. Clin J Am Soc Nephrol. 2007;2:700–7.
Powell DW, Cummins TD, Klein JB, Salant DJ. 75. Shankland SJ, Smeets B, Pippin JW, Moeller MJ. The
M-type phospholipase A2 receptor as target antigen emergence of the glomerular parietal epithelial cell.
in idiopathic membranous nephropathy. N Engl J Nat Rev Nephrol. 2014;10:158–73.
Med. 2009;361:11–21. 76. Verani RR, Hawkins EP. Recurrent focal
61. Larsen CP, Messias NC, Silva FG, Messias E, segmental glomerulosclerosis. Am J Nephrol. 1986;
Walker PD. Determination of primary versus second- 6:263–70.
ary membranous glomerulopathy utilizing phospholi- 77. Fatima H, Moeller MJ, Smeets B, Yang HC, D’Agati
pase A2 receptor staining in renal biopsies. Mod VD, Alpers CE, Fogo AB. Parietal epithelial cell
Pathol. 2013;26:709–15. activation marker in early recurrence of FSGS in the
62. Furness PN, Boyd S. Electron microscopy and immu- transplant. Clin J Am Soc Nephrol. 2012;7:1852–8.
nocytochemistry in the assessment of renal biopsy 78. Tøndel C, Bostad L, Hirth A, Svarstad E. Renal
specimens: actual and optimal practice. J Clin Pathol. biopsy findings in children and adolescents with
1996;49:233–7. Fabry disease and minimal albuminuria. Am J Kidney
63. D’Agati V, Suh J-I, Carbone L, et al. Pathology of Dis. 2008;51:767–76.
HIV-associated nephropathy: a detailed morphologic 79. Tøndel C, Bostad L, Larsen KK, Hirth A, Vikse BE,
and comparative study. Kidney Int. 1989;35: Houge G, Svarstad E. Agalsidase benefits renal his-
1358–70. tology in young patients with Fabry disease. J Am Soc
64. Fogo A, Hawkins EP, Berry PL, et al. Glomerular Nephrol. 2013;24:137–48.
hypertrophy in minimal change disease predicts sub- 80. Nasr SH, Markowitz GS, Valeri AM, Yu Z, Chen L,
sequent progression to focal glomerular sclerosis. D’Agati VD. Thin basement membrane nephropathy
Kidney Int. 1990;38:115–23. cannot be diagnosed reliably in deparaffinized,
24 Pediatric Renal Pathology 745

formalin-fixed tissue. Nephrol Dial Transplant. 96. Schainuck LI, Striker GE, Cutler RE, et al. Structural-
2007;22:1228–32. functional correlations in renal disease. II. The corre-
81. Yoshikawa N, Matsuyama S, Iijima K, et al. Benign lations. Hum Pathol. 1970;1:631–41.
familial hematuria. Arch Pathol Lab Med. 1988;112: 97. Trainin EB, Gomez-Leon G. Development of renal
794–7. insufficiency after long-standing steroid-responsive
82. Steffes MW, Barbosa J, Basgen JM, et al. Quantitative nephrotic syndrome. Int J Pediatr Nephrol.
glomerular morphology of the normal human kidney. 1982;3:55–8.
Lab Invest. 1983;49:82–6. 98. Southwest Pediatric Nephrology Study Group. Focal
83. Yoshioka K, Hino S, Takemura T, et al. Type IV segmental glomerulosclerosis in children with idio-
collagen α5 chain: normal distribution and abnormal- pathic nephrotic syndrome. A report of the Southwest
ities in X-linked Alport syndrome revealed by mono- Pediatric Nephrology Study Group. Kidney Int.
clonal antibody. Am J Pathol. 1994;144:986–96. 1985;27:442–9.
84. Najafian B, Franklin DB, Fogo AB. Acute renal fail- 99. Al-Eisa A, Carter JE, Lirenmann DS, et al. Childhood
ure and myalgia in a transplant patient. J Am Soc IgM nephropathy: comparison with minimal change
Nephrol. 2007;18:2870–4. disease. Nephron. 1996;72:37–43.
85. Schaefer HM, Helderman JH, Fogo AB. Slow decline 100. Fogo A, Ichikawa I. Focal segmental glomerulo-
in allograft function in a renal transplant patient. Am J sclerosis: a view and review. Invited Editorial. Pediatr
Kidney Dis. 2006;48:335–8. Nephrol. 1996;10:374–91.
86. Nasr SH, Sethi S, Cornell LD, Milliner DS, 101. D’Agati VD, Fogo AB, Bruijn JA, Jennette JC.
Boelkins M, Broviac J, Fidler ME. Crystalline Pathologic classification of focal segmental
nephropathy due to 2,8-dihydroxyadeninuria: an glomerulosclerosis: a working proposal. Am J Kidney
under-recognized cause of irreversible renal failure. Dis. 2004;43:368–82.
Nephrol Dial Transplant. 2010;25:1909–15. 102. Detwiler RK, Falk RF, Hogan SL, et al. Collapsing
87. Fogo AB, Bostad L, Svarstad E, All Members of the glomerulopathy: a clinically and pathologically dis-
International Study Group of Fabry Nephropathy tinct variant of focal segmental glomerulosclerosis.
(ISGFN), et al. Scoring system for renal pathology Kidney Int. 1994;45:1416–24.
in Fabry disease: report of the International Study 103. Stokes MB, Valeri AM, Markowitz GS, D’Agati VD.
Group of Fabry Nephropathy (ISGFN). Nephrol Cellular focal segmental glomerulosclerosis: clinical
Dial Transplant. 2010;25:2168–77. and pathologic features. Kidney Int. 2006;70:
88. Heptinstall RH. Pyelonephritis: pathologic features. 1783–92.
In: Heptinstall RH, editor. Pathology of the kidney. 104. Howie AJ, Brewer DB. Further studies on the glo-
4th ed. Boston: Brown, Little; 1992. p. 1489–561. merular tip lesion: early and late stages and life table
89. Verani R, Walker P, Silva FG. Renal cystic disease of analysis. J Pathol. 1985;147:245–55.
infancy: results of histochemical studies. A report of 105. Stokes MB, Markowitz GS, Lin J, Valeri AM,
the Southwest Pediatric Nephrology Study Group. D’Agati VD. Glomerular tip lesion: a distinct entity
Pediatr Nephrol. 1989;3:37–42. within the minimal change disease/focal segmental
90. Nickeleit V, Mihatsch MJ. Polyomavirus nephropathy glomerulosclerosis spectrum. Kidney Int. 2004;65:
in native kidneys and renal allografts: an update on an 1690–702.
escalating threat. Transpl Int. 2006;19:960–73. 106. Thomas DB, Franceschini N, Hogan SL, Ten
91. Hawkins EP, Berry PL, Silva FG. Acute tubuloin- Holder S, Jennette CE, Falk RJ, Jennette JC. Clinical
terstitial nephritis in children: clinical, morphologic, and pathologic characteristics of focal segmental
and lectin studies. A report of the Southwest Pediatric glomerulosclerosis pathologic variants. Kidney Int.
Nephrology Study Group. Am J Kidney Dis. 1989; 2006;69:920–6.
14:466–71. 107. D’Agati VD, Alster JM, Jennette JC, et al. Associa-
92. Myers BD, Ross J, Newton L, et al. Cyclosporine- tion of histologic variants in FSGS clinical trial with
associated chronic nephropathy. N Engl J Med. presenting features and outcomes. Clin J Am Soc
1984;311:699–705. Nephrol. 2013;8:399–406.
93. Mihatsch MJ, Thiel G, Basler V, et al. Morphological 108. Iskandar SS, Browning MC, Lorentz WB. C1q
patterns in cyclosporine-treated renal transplant recip- nephropathy: a pediatric clinicopathologic study.
ients. Transplant Proc. 1985;17 Suppl 1:101–16. Am J Kidney Dis. 1991;18:459–65.
94. Böhle A, Mackensen-Haen S, Gise H. Significance of 109. Vizjak A, Ferluga D, Rozic M, Hvala A, Lindic J,
tubulointerstitial changes in the renal cortex for the Levart TK, Jurcic V, Jennette JC. Pathology, clinical
excretory function and concentration ability of the presentations, and outcomes of C1q nephropathy.
kidney: a morphometric contribution. Am J Nephrol. J Am Soc Nephrol. 2008;19:2237–44.
1987;7:421–33. 110. Suzuki J, Yoshikawa N, Nakamura H. A quantitative
95. Striker GE, Shainuck LI, Cutler RE, et al. Structural- analysis of the glomeruli in focal segmental
functional correlations in renal disease. I. A method glomerulosclerosis. Pediatr Nephrol. 1994;8:416–9.
for assaying and classifying histopathologic changes 111. Nyberg E, Bohman SO, Berg U. Glomerular volume
in renal disease. Hum Pathol. 1970;1:615–30. and renal function in children with different types of
746 A.B. Fogo

the nephrotic syndrome. Pediatr Nephrol. 1994;8: 127. Hasselbacher K, Wiggins RC, Matejas V,
285–9. et al. Recessive missense mutations in LAMB2
112. Fogo AB. Glomerular hypertension, abnormal glo- expand the clinical spectrum of LAMB2-associated
merular growth and progression of renal diseases. disorders. Kidney Int. 2006;70:1008–12.
Kidney Int. 2000;57 Suppl 75:S15–21. 128. Zenker M, Aigner T, Wendler O, et al. Human laminin
113. Regel HM, Fillipovic E, Langer B, Poczewki H, beta2 deficiency causes congenital nephrosis with
Kraxberger I, Bittner RE, Kerjaschki D. Glomerular mesangial sclerosis and distinct eye abnormalities.
expression of dystroglycans is reduced in minimal Hum Mol Genet. 2004;13:2625–32.
change nephrosis but not in focal segmental 129. Doleris LM, Hill GS, Chedin P, et al. Focal segmental
glomerulosclerosis. J Am Soc Nephrol. 2000;11: glomerulosclerosis associated with mitochondrial
403–12. cytopathy. Kidney Int. 2000;58:1851–8.
114. Schmid H, Henger A, Cohen CD, Frach K, Gröne HJ, 130. Gagnadoux MF, Habib R, Gubler MC, et al. Long-
Schlöndorff D, Kretzler M. Gene expression profiles term (15–25 years) outcome of childhood hemolytic-
of podocyte-associated molecules as diagnostic uremic syndrome. Clin Nephrol. 1996;46:39–41.
markers in acquired proteinuric diseases. J Am Soc 131. Argyle JC, Hogg RJ, Pysher TJ, et al. A clinicopath-
Nephrol. 2003;14:2958–66. ological study of 24 children with hemolytic uremic
115. Fukuda A, Wickman LT, Venkatareddy MP, Wang syndrome. Pediatr Nephrol. 1990;4:52–8.
SQ, Chowdhury MA, Wiggins JE, Shedden KA, 132. Van Buren D, Van Buren CT, Flechner SM, et al. De
Wiggins RC. Urine podocin: nephrin mRNA ratio novo hemolytic uremic syndrome in renal transplant
(PNR) as a podocyte stress biomarker. Nephrol Dial recipients immunosuppressed with cyclosporine. Sur-
Transplant. 2012;27:4079–87. gery. 1985;98:54–62.
116. Wickman L, Afshinnia F, Wang SQ, et al. Urine 133. Schwarz A, Krause P-H, Offerman G, et al. Recurrent
podocyte mRNAs, proteinuria, and progression in and de novo renal disease after kidney transplantation
human glomerular diseases. J Am Soc Nephrol. with or without cyclosporine A. Am J Kidney Dis.
2013;24:2081–95. 1991;17:524–31.
117. Boute N, Gribouval O, Roselli S, et al. 134. Lévy M, Gonzalez-Burchard G, Broyer M, et al.
NPHS2, encoding the glomerular protein Berger’s disease in children: natural history and out-
podocin, is mutated in autosomal recessive steroid- come. Medicine. 1985;64:157–80.
resistant nephrotic syndrome. Nat Genet. 2000;24: 135. Yoshikawa N, Ito H, Nakamura H. IgA nephropathy
349–54. in children from Japan. Child Nephrol Urol.
118. Kaplan JM, Kim SH, North KN, et al. Mutations in 1989;9:191–9.
ACTN4, encoding alpha-actinin-4, cause familial 136. Yoshikawa N, Iijima K, Matsuyama S, et al. Repeat
focal segmental glomerulosclerosis. Nat Genet. renal biopsy in children with IgA nephropathy. Clin
2000;24:251–6. Nephrol. 1990;33:160–7.
119. Winn MP. 2007 Young Investigator Award: TRP’ing 137. Lee SM, Rao VM, Franklin WA, Schiffer MS,
into a new era for glomerular disease. J Am Soc Aronson AJ, Spargo BH, Katz AI. IgA nephropathy:
Nephrol. 2008;19(6):1071–5. morphologic predictors of progressive renal disease.
120. Hinkes B, Wiggins RC, Gbadegesin R, et al. Posi- Hum Pathol. 1982;13:314–22.
tional cloning uncovers mutations in PLCE1 respon- 138. Haas M. Histologic subclassification of IgA nephrop-
sible for a nephrotic syndrome variant that may be athy: a clinicopathologic study of 244 cases. Am J
reversible. Nat Genet. 2006;38:1397–405. Kidney Dis. 1997;29:829–42.
121. Hinkes BG. NPHS3: new clues for understanding 139. Working Group of the International IgA Nephropathy
idiopathic nephrotic syndrome. Pediatr Nephrol. Network and the Renal Pathology Society, Roberts
2008;23(6):847–50. IS, Cook HT, Troyanov S, et al. The Oxford classifi-
122. Gbadegesin R, Hinkes BG, Hoskins BE, et al. Muta- cation of IgA nephropathy: pathology definitions,
tions in PLCE1 are a major cause of isolated diffuse correlations, and reproducibility. Kidney Int.
mesangial sclerosis (IDMS). Nephrol Dial Transplant. 2009;76:546–56.
2008;23:1291–7. 140. Working Group of the International IgA Nephropathy
123. D’Agati VD, Kaskel FJ, Falk RJ. Focal segmental Network and the Renal Pathology Society, Cattran
glomerulosclerosis. N Engl J Med. 2011;365: DC, Coppo R, Cook HT, et al. The Oxford classifica-
2398–411. tion of IgA nephropathy: rationale, clinicopathologi-
124. Fogo AB. Causes and pathogenesis of focal segmen- cal correlations, and classification. Kidney Int.
tal glomerulosclerosis. Nat Rev Nephrol (in press). 2009;76:534–45.
125. Khoshnoodi J, Tryggvason K. Congenital 141. Roberts IS. Pathology of IgA nephropathy. Nat Rev
nephrotic syndromes. Curr Opin Genet Dev. Nephrol. 2014;10:445–54.
2001;11:322–7. 142. Zeng CH, Le W, Ni Z, et al. A multicenter application
126. Kim JM, Wu H, Green G, et al. CD2-associated and evaluation of the Oxford classification of IgA
protein haploinsufficiency is linked to glomerular nephropathy in adult Chinese patients. Am J Kidney
disease susceptibility. Science. 2003;300:1298–300. Dis. 2012;60:812–20.
24 Pediatric Renal Pathology 747

143. Le W, Zeng CH, Liu Z, et al. Validation of the 157. Anders HJ, Fogo AB. Immunopathology of lupus
Oxford classification of IgA nephropathy for nephritis. Semin Immunopathol. 2014;36:443–59.
pediatric patients from China. BMC Nephrol. 158. Furness PN, Taub N. Interobserver reproducibility
2012;13:158. and application of the ISN/RPS classification of
144. Coppo R, Troyanov S, Bellur S, on Behalf of the lupus nephritis-a UK-wide study. Am J Surg Pathol.
VALIGA Study of the ERA-EDTA Immunone- 2006;30:1030–5.
phrology Working Group, et al. Validation of the 159. Schwartz MM, Korbet SM, Lewis EJ, Collaborative
Oxford classification of IgA nephropathy in cohorts Study Group. The prognosis and pathogenesis of
with different presentations and treatments. Kidney severe lupus glomerulonephritis. Nephrol Dial Trans-
Int. 2014;86(4):828–36. plant. 2008;23:1298–306.
145. Working Group of the International IgA Nephropathy 160. Markowitz GS, D’Agati VD. The ISN/RPS 2003
Network and the Renal Pathology Society, Coppo R, classification of lupus nephritis: an assessment at
Troyanov S, Camilla R, et al. The Oxford IgA 3 years. Kidney Int. 2007;71:491–5.
nephropathy clinicopathological classification is 161. Svobodova B, Honsova E, Ronco P, Tesar V,
valid for children as well as adults. Kidney Int. Debiec H. Kidney biopsy is a sensitive tool for retro-
2010;77:921–7. spective diagnosis of PLA2R-related membranous
146. Yamabe H, Johnson RJ, Gretch DR, et al. Hepatitis C nephropathy. Nephrol Dial Transplant. 2013;28:
virus infection and membranoproliferative glomeru- 1839–44.
lonephritis in Japan. J Am Soc Nephrol. 1995;6: 162. Merkel F, Pullig O, Marx M, et al. Course and prog-
220–3. nosis of anti-basement membrane antibody (anti-BM-
147. Johnson RJ, Gretch DR, Yamabe H, et al. Ab)-mediated disease: report of 35 cases. Nephrol
Membranoproliferative glomerulonephritis associ- Dial Transplant. 1994;9:372–6.
ated with hepatitis C virus infection. N Engl J Med. 163. Berden AE, Ferrario F, Hagen EC, et al. Histopatho-
1993;328:465–70. logic classification of ANCA-associated glomerulo-
148. Nowicki MJ, Welch TR, Ahmad N, et al. Absence of nephritis. J Am Soc Nephrol. 2010;21:1628–36.
hepatitis B and C viruses in pediatric idiopathic 164. Kanjanabuch T, Kittikowit W, Eiam-Ong S. An
membranoproliferative glomerulonephritis. Pediatr update on acute postinfectious glomerulonephritis
Nephrol. 1995;9:16–8. worldwide. Nat Rev Nephrol. 2009;5:259–69.
149. Galle P, Mahieu P. Electron dense alteration of kidney 165. Nasr SH, Markowitz GS, Stokes MB, Said SM, Valeri
basement membranes. A renal lesion specific of a AM, D’Agati VD. Acute postinfectious glomerulone-
systemic disease. Am J Med. 1975;58:749–64. phritis in the modern era: experience with 86 adults
150. Churg J, Duffy JL, Bernstein J. Identification of dense and review of the literature. Medicine (Baltimore).
deposit disease: a report for the international study of 2008;87:21–32.
kidney diseases in children. Arch Pathol Lab Med. 166. Nasr SH, Share DS, Vargas MT, D’Agati VD, Mar-
1979;103:67–72. kowitz GS. Acute poststaphylococcal glomerulone-
151. Walker PD, Ferrario F, Joh K, Bonsib SM. phritis superimposed on diabetic glomerulosclerosis.
Dense deposit disease is not a membranoproliferative Kidney Int. 2007;71:1317–21.
glomerulonephritis. Mod Pathol. 2007;20:605–16. 167. Ellis EN, Pysher TJ. Renal disease in adolescents with
152. Walker PD. Dense deposit disease: new insights. Curr type I diabetes mellitus: a report of the Southwest
Opin Nephrol Hypertens. 2007;16:204–12. Pediatric Nephrology Study Group. Am J Kidney
153. Schwertz R, de Jong R, Gretz N, et al. Outcome of Dis. 1993;22:783–90.
idiopathic membranoproliferative glomerulonephritis 168. Tervaert Tervaert TW, Mooyaart AL, Amann K,
in children. Acta Paediatr. 1996;85:308–12. Renal Pathology Society, et al. Pathologic classifica-
154. Braun MC, Stablein DM, Hamiwka LA, Bell L, tion of diabetic nephropathy. J Am Soc Nephrol.
Bartosh SM, Strife CF. Recurrence of membranopro- 2010;21:556–63.
liferative glomerulonephritis type II in renal allo- 169. Ito S, Hataya H, Ikeda M, et al. Alport syndrome-like
grafts: the North American Pediatric Renal basement membrane changes in Frasier syndrome: an
Transplant Cooperative Study experience. J Am Soc electron microscopy study. Am J Kidney Dis.
Nephrol. 2005;16:2225–33. 2003;41:1110–5.
155. Sethi S, Fervenza FC, Zhang Y, Nasr SH, Leung N, 170. Pirson Y. Making the diagnosis of Alport syndrome.
Vrana J, Cramer C, Nester CM, Smith RJH. Prolifer- Kidney Int. 1999;56:760–75.
ative glomerulonephritis secondary to dysfunction of 171. Nasr SH, Markowitz GS, Goldstein CS, Fildes RD,
the alternative pathway of complement. Clin J Am D’Agati VD. Hereditary nephritis mimicking immune
Soc Nephrol. 2011;6:1009–17. complex-mediated glomerulonephritis. Hum Pathol.
156. Sethi S, Fervenza FC, Zhang Y, Zand L, Meyer NC, 2006;37:547–54.
Borsa N, Nasr SH, Smith RJ. Atypical postinfectious 172. Buzza M, Wang YY, Dagher H, et al. COL4A4 muta-
glomerulonephritis is associated with abnormalities in tion in thin basement membrane disease previously
the alternative pathway of complement. Kidney Int. described in Alport syndrome. Kidney Int.
2013;83:293–9. 2001;60:480–3.
748 A.B. Fogo

173. Haas M. Thin glomerular basement membrane 189. Haas M, Sis B, Racusen LC, Banff meeting report
nephropathy: incidence in 3471 consecutive renal writing committee, et al. Banff meeting report writing
biopsies examined by electron microscopy. Arch committee. Banff 2013 meeting report: inclusion of
Pathol Lab Med. 2006;130:699–706. C4d-negative antibody-mediated rejection and
174. Haas M. Alport syndrome and thin glomerular base- antibody-associated arterial lesions. Am J Transplant.
ment membrane nephropathy: a practical approach to 2014;14:272–83.
diagnosis. Arch Pathol Lab Med. 2009;133:224–32. 190. Lefaucheur C, Loupy A, Vernerey D, et al. Antibody-
175. Gauthier B, Trachtman H, Frank R, et al. Familial thin mediated vascular rejection of kidney allografts: a
basement membrane nephropathy in children with population-based study. Lancet. 2013;381:313–9.
asymptomatic microhematuria. Nephron. 1989;51: 191. Rush D, Jeffery J, Trpkov K, et al. Effect of subclin-
502–8. ical rejection on renal allograft histology and function
176. Fioretto P, Steffes MW, Sutherland DE, et al. Reversal at 6 months. Transplant Proc. 1996;28:494–5.
of lesions of diabetic nephropathy after pancreas 192. Strehlau J, Pavlakis M, Lipman M, et al. Quantitative
transplantation. N Engl J Med. 1998;339:69–75. detection of immune activation transcripts as a diag-
177. Ikoma M, Kawamura T, Fogo A, et al. Cause of nostic tool in kidney transplantation. Proc Natl Acad
variable therapeutic efficiency of angiotensin Sci U S A. 1997;94:695–700.
converting enzyme inhibitor on the glomerular 193. Mauiyyedi S, Crespo M, Collins AB, et al. Acute
mesangial lesions. Kidney Int. 1991;40:291–301. humoral rejection in kidney transplantation: II.
178. Yang HC, Liu SJ, Fogo AB. Kidney regeneration in Morphology, immunopathology, and pathologic clas-
mammals. Nephron Exp Nephrol. 2014;126:50–3. sification. J Am Soc Nephrol. 2002;13:779–87.
179. Pichaiwong W, Hudkins KL, Wietecha T, et al. 194. Bohmig GA, Exner M, Habicht A, et al. Capillary
Reversibility of structural and functional damage in C4d deposition in kidney allografts: a specific marker
a model of advanced diabetic nephropathy. J Am Soc of alloantibody-dependent graft injury. J Am Soc
Nephrol. 2013;24:1088–102. Nephrol. 2002;13:1091–9.
180. Austin III HA, Boumpas DT, Vaughan EM, 195. Haas M. Pathologic features of antibody-mediated
et al. High-risk features of lupus nephritis: importance rejection in renal allografts: an expanding spectrum.
of race and clinical and histological factors in Curr Opin Nephrol Hypertens. 2012;21:264–71.
166 patients. Nephrol Dial Transplant. 1995;10: 196. Drachenberg CB, Steinberger E, Hoehn-Saric E,
1620–8. et al. Specificity of intertubular capillary changes:
181. Baqi N, Moazami S, Singh A, et al. Lupus nephritis in comparative ultrastructural studies in renal allografts
children: a longitudinal study of prognostic factors and native kidneys. Ultrastruct Pathol. 1997;21:
and therapy. J Am Soc Nephrol. 1996;7:924–9. 227–33.
182. Hill GS, Delahousse M, Nochy D, Tomkiewicz E, 197. Mihatsch MJ, Ryffel B, Gudat F. The differential
Remy P, Mignon F, Mery JP. A new morphologic diagnosis between rejection and cyclosporine toxic-
index for the evaluation of renal biopsies in lupus ity. Kidney Int. 1995;48 Suppl 52:S63–9.
nephritis. Kidney Int. 2000;58:1160–73. 198. Meehan SM, Pascual M, Williams WW, et al. De
183. Katafuchi R, Oh Y, Hori K, et al. An important role of novo collapsing glomerulopathy in renal allografts.
glomerular segmental lesions on progression of IgA Transplantation. 1998;65:1192–7.
nephropathy: a multivariate analysis. Clin Nephrol. 199. Ponticelli C, Glassock RJ. Posttransplant recurrence
1994;41:191–8. of primary glomerulonephritis. Clin J Am Soc
184. Hogg RJ, Silva FG, Wyatt RJ, et al. Prognostic indi- Nephrol. 2010;5:2363–72.
cators in children with IgA nephropathy: report of the 200. Habib R, Gagnadoux M-F, Broyer M. Recurrent glo-
Southwest Pediatric Nephrology Study Group. merulonephritis in transplanted children. Contrib
Pediatr Nephrol. 1994;8:15–20. Nephrol. 1987;55:123–35.
185. Andreoli SP, Yum MN, Bergstein JM. IgA nephropa- 201. Goral S, Ynares C, Shappell SB, Snyder S, Feurer ID,
thy in children: significance of glomerular basement Kazancioglu R, Fogo AB, Helderman JH. Recurrent
membrane deposition of IgA. Am J Nephrol. lupus nephritis in renal transplant recipients revisited:
1986;6:28–33. it is not rare. Transplantation. 2003;75:651–6.
186. Hsu H-C, Wu C-Y, Lin C-Y, et al. Membranous 202. Patel K, Hirsch J, Beck L, Herlitz L, Radhakrishnan
nephropathy in 52 hepatitis B surface antigen J. De novo membranous nephropathy in renal allo-
(HBsAg) carrier children in Taiwan. Kidney Int. graft associated with antibody-mediated rejection and
1989;36:1103–7. review of the literature. Transplant Proc. 2013;45:
187. Colvin RB, Cohen AH, Saiontz C, et al. Evaluation of 3424–8.
pathologic criteria for acute renal allograft rejection: 203. Hariharan S, Smith RD, Viero R, First MR. Diabetic
reproducibility, sensitivity, and clinical correlation. nephropathy after renal transplantation. Clinical
J Am Soc Nephrol. 1997;8:1930–41. and pathologic features. Transplantation. 1996;62:
188. Racusen LC, Solez K, Colvin RB, et al. The Banff 632–5.
97 working classification of renal allograft pathology. 204. Bhalla V, Nast CC, Stollenwerk N, Tran S, Barba L,
Kidney Int. 1999;55:713–23. Kamil ES, Danovitch G, Adler SG. Recurrent and de
24 Pediatric Renal Pathology 749

novo diabetic nephropathy in renal allografts. Trans- 209. Sethi S, Vrana JA, Theis JD, Dogan A. Mass spec-
plantation. 2003;75:66–71. trometry based proteomics in the diagnosis of kidney
205. Randhawa PS, Magnone M, Jordan M, et al. Renal disease. Curr Opin Nephrol Hypertens. 2013;22:
allograft involvement by Epstein-Barr virus associ- 273–80.
ated with post-transplant lymphoproliferative disease. 210. Schaub S, Rush D, Wilkins J, et al. Proteomic-based
Am J Surg Pathol. 1996;20:563–71. detection of urine proteins associated with acute renal
206. Smith JM, Dharnidharka VR, Talley L, Martz K, allograft rejection. J Am Soc Nephrol. 2004;15:
McDonald RA. BK virus nephropathy in pediatric 219–27.
renal transplant recipients: an analysis of the North 211. Devarajan P. Proteomics for biomarker discovery
American Pediatric Renal Trials and Collaborative in acute kidney injury. Semin Nephrol. 2007;27:
Studies (NAPRTCS) registry. Clin J Am Soc Nephrol. 637–51.
2007;2:1037–42. 212. Wang D, Sun W. Urinary extracellular microvesicles:
207. Bracamonte E, Leca N, Smith KD, et al. Tubular isolation methods and prospects for urinary proteome.
basement membrane immune deposits in association Proteomics. 2014;14:1922–32.
with BK polyomavirus nephropathy. Am J Trans- 213. Sis B, Halloran PF. Endothelial transcripts uncover a
plant. 2007;7:1552–60. previously unknown phenotype: C4d-negative anti-
208. Barnes JL, Milani S. In situ hybridization in the study body-mediated rejection. Curr Opin Organ Trans-
of the kidney and renal disease. Semin Nephrol. plant. 2010;15:42–8.
1995;15:9–28.
Part V
Glomerular Disease
Congenital Nephrotic Syndrome
25
Hannu Jalanko and Christer Holmberg

Contents Secondary Nephrotic Syndromes . . . . . . . . . . . . . . . . . . 764


Infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 764
Glomerular Filtration Barrier . . . . . . . . . . . . . . . . . . . . . 755 Immune Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
Glomerular Capillary Wall and Podocytes . . . . . . . . . . 755
Slit Diaphragm Components . . . . . . . . . . . . . . . . . . . . . . . . 755 Diagnosis of CNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
Clinical Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
Primary Nephrotic Syndrome . . . . . . . . . . . . . . . . . . . . . 756 Kidney Biopsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 765
CNS of the Finnish Type (CNF, NPHS1) . . . . . . . . . . . 756 Gene Testing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 766
Podocin Gene Mutations (NPHS2) . . . . . . . . . . . . . . . . 760 Management of CNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 766
Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760 Immunosuppressive Medication . . . . . . . . . . . . . . . . . . . . . 766
Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 760 Antiproteinuric Medication . . . . . . . . . . . . . . . . . . . . . . . . . . 767
WT1 Gene Mutations (Denys–Drash, Albumin Substitution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 767
Isolated CNS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 761 Nutrition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 767
Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 761 Additional Medication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 761 Unilateral Nephrectomy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 768
Dialysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
Laminin β2 Gene Mutations Kidney Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
(Pierson Syndrome) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 762
Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 769
PLC«1 Gene Mutations (NPHS3) . . . . . . . . . . . . . . . . . 763
Other Forms of Primary CNS . . . . . . . . . . . . . . . . . . . . . . . 763 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 770

H. Jalanko (*) • C. Holmberg


Hospital for Children and Adolescents, University of
Helsinki, Helsinki, Finland
e-mail: hannu.jalanko@hus.fi; christer.holmberg@hus.fi

# Springer-Verlag Berlin Heidelberg 2016 753


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_78
754 H. Jalanko and C. Holmberg

Renal diseases associated with nephrotic syn- clinical diagnosis, it is arbitrary in the sense that
drome (NS) in the first year of life are uncommon NS caused by a specific gene mutation can
and make up a heterogeneous group of disorders manifest soon after birth or later in life. However,
[73]. Congenital nephrotic syndrome (CNS) is since the management of CNS is often
defined as proteinuria manifesting in the first different from the more common forms of child-
3 months of life. NS appearing later during the hood NS, the terminology still seems warranted
first year (4–12 months) is defined infantile, and (Table 1).
NS manifesting thereafter is called childhood Primary CNS is typically caused by mutations
NS. While this classification is used to help the in genes encoding for components of the glomer-
ular filtration barrier [100, 147, 203, 216]. The
classical form is the Finnish type of CNS (CNF,
Table 1 Classification of congenital nephrotic syndrome NPHS1), which is caused by mutations in nephrin
(CNS) gene (NPHS1) leading to massive proteinuria,
Primary CNS hypoproteinemia, and edema in the newborn
Nephrin (NPHS1) gene mutations (NPHS1, CNF, period. Other important genes causing CNS are
isolated NS) podocin gene (NPHS2), Wilms’ tumor factor
Podocin (NPHS2) gene mutations (NPHS2, isolated 1 gene (WT1), laminin β2-gene (LAMB2), and
NS)
PLCe1 gene (PLCe1, NPHS3). These disorders
Phospholipase C epsilon 1 (PLCE1) gene mutations
(NPHS3, isolated NS)
have a more widespread age of onset and
Wilms tumor suppressor 1 (WT1) gene mutations also more variable clinical manifestations.
(Denys–Drash, isolated NS) Mutations in these five genes cover the
Laminin β2 (LAMB2) gene mutations (Pierson majority (>80 %) of the genetic defects observed
syndrome, isolated NS) in CNS patients (Table 2) [83, 85, 133, 183]. Even
Laminin β3 (LAMB3) gene mutations (Herlitz though CNS is a rare disorder, new genetic forms
junctional epidermolysis bullosa)
are repeatedly discovered. These include NS
Lim homeobox transcription factor 1b (LMXB1)
mutations (nail–patella syndrome)
caused by mutations in ARHGDIA gene [57, 70]
OCRL gene defect (Lowe) and in genes encoding for enzymes taking part
ARHGDIA gene mutations (RhoGDIA defect) in mitochondrial coenzyme Q10 (ubiquinone)
Mutations in genes encoding mitochondrial coenzyme biosynthesis [45, 186]. In the genetic forms of
Q10 synthesis (COQ2, COQ6, ADCK4) CNS, proteinuria can be an isolated disorder or
Galloway–Mowat (gene defect not yet known) part of more generalized syndrome. It is important
Secondary CNS to notice that CNS can also be caused by autoim-
Congenital syphilis mune disorders and neonatal infections, espe-
Toxoplasmosis, malaria cially in Africa and Asia. Thus, the diagnosis of
Cytomegalovirus, rubella, hepatitis B, HIV a patient with early manifestations of NS must be
Maternal systemic lupus erythematosus (SLE) based on several criteria including clinical presen-
Neonatal antibodies against neutral endopeptidase
tation, family history, laboratory findings, genetic
Maternal steroid–chlorpheniramine treatment
testing, and renal histology.

Table 2 Prevalence of genetic findings of CNS patients in four international studies


Study No NPHS1 (%) NPHS2 (%) WT1 (%) LAMB2 PLCE1
Hinkess (6) 46 39 39 2.2 4.4 % n.a.
Heeringa (7) 42 40 17 8 n.a. 8%
Machuca (8) 117 54 15 1.7 1.7 % 1.7 %
Santini (9) 15 80 7 13 n.a. 0%
25 Congenital Nephrotic Syndrome 755

Fig. 1 A cross-sectional image of one glomerular capil- pathogenesis of CNS are also depicted. WT1 is a transcrip-
lary (left) and glomerular capillary wall (right), which tion factor important for podocyte functions. PLCε1 is a
comprises three layers: fenestrated endothelium, glomeru- cytoplasmic enzyme involved in cell signaling. Nephrin is
lar basement membrane (GBM), and epithelial cell layer a major component of SD and podocin is an adapter and
with podocyte foot processes and interposing slit dia- scaffold protein for the SD components
phragms (SDs). The four most important molecules in the

in a GBM component, laminin, results in NS [35,


Glomerular Filtration Barrier 103]. Also, mutations a gene encoding integrin α3
(ITGA3) may lead to nephrotic syndrome indicat-
Glomerular Capillary Wall ing that the binding of the GBM to podocyte
and Podocytes (cytoskeleton) is important for the glomerular fil-
tration barrier [149].
The main feature of primary CNS is the extensive Podocytes and SD are, however, even more
leakage of plasma proteins into urine through the important in preventing proteinuria than the GBM
kidney filtration barrier (Fig. 1) [42, 102]. This [40, 124, 203]. Podocytes are specialized cells that
barrier is located in the glomerular capillary wall consist of the cell body and primary, secondary, and
and comprises three layers: fenestrated endothe- tertiary processes. The cytoskeleton of the cell body
lium, glomerular basement membrane (GBM), and major processes consist of microtubules and
and epithelial cell (podocyte) layer with distal intermediate filaments, whereas actin is the major
foot processes and interposed slit diaphragms cytoskeletal component of foot processes. Actin
(SDs) (Fig. 1). The filter is a highly sophisticated network and the interacting proteins, such as
size- and charge-selective molecular sieve, and α-actinin-4, inverted formin-2, myosin-9, myosin-
normally only water and small plasma solutes 1E, and ARHGDIA, maintain the architecture of the
pass into Bowman’s space [40, 111]. As foot processes and respond to signals from outside,
100–200 l of protein free filtrate (primary urine) modifying the cellular functions accordingly.
is formed daily, one can calculate that 3–6 kg of Genetic defects in these five interacting proteins
albumin has to be prevented from crossing the cause NS manifesting in older children and adults
glomerular filtration barrier each day. [11, 24, 27, 108, 115, 142, 181, 192, 212]. Some-
The flow of glomerular filtrate follows the what surprisingly, there have so far not been reports
extracellular route, passing across the GBM and on CNS cases caused by these five genes.
SD, which bridge adjacent foot processes just
above the GBM. The GBM is a protein network
formed by type IV collagen, laminin, nidogen, Slit Diaphragm Components
and negatively charged proteoglycans. The role
of GBM in glomerular permselectivity has been The precise molecular structure of SD is still
debated, but it is now known that a primary defect unresolved. The first SD component identified
756 H. Jalanko and C. Holmberg

Fig. 2 The nephrin molecule is a transmembrane cell part contains three cysteine residues taking part in molec-
adhesion protein with small intracellular and transmem- ular interactions. The intracellular part contains several
brane domains and a larger extracellular domain. The tyrosine residues phosphorylated by protein kinases,
extracellular domain contains eight Ig-like modules and which is important for cellular signaling
one fibronectin type module adjacent. The extracellular

was nephrin, which is a 1,241-residue cell adhe- are located in the cytosolic part of the podocyte
sion protein of the immunoglobulin family foot process [23, 190, 208]. Podocin is encoded
[92, 112, 174, 206]. The extracellular part of by the NPHS2 gene and it is a small hairpin-like
nephrin contains eight immunoglobulin-like mod- protein belonging to a raft-associated stomatin
ules and one type III fibronectin domain (Fig. 2). family. It is a scaffolding protein and serves in
The intracellular domain contains nine tyrosine the structural organization of the SD as well as in
residues, which may become phosphorylated by the signal transduction from the SD into
protein kinases. This phosphorylation modulates podocytes [94, 95]. Genetic mutations in podocin
the interaction of nephrin with other proteins, such result in CNS as well as NS manifesting later
as adaptor protein Nck and phosphoinositide in life.
3-kinase (PI3K) [94, 106, 217]. These proteins
link nephrin to actin cytoskeleton, suggesting an
important role for nephrin in the regulation of the
actin cytoskeleton and podocyte morphology. Primary Nephrotic Syndrome
Nephrin takes part also in other cell signaling
processes involved in the podocyte cell survival CNS of the Finnish Type (CNF, NPHS1)
[19, 125].
In addition to nephrin, several other podocyte CNF originally denoted to a severe form congen-
proteins have been identified in the SD, such as ital nephrotic syndrome typically seen in the Finn-
Neph1, Neph 2, Neph3, and FAT 1 and FAT 2 [46, ish newborns [75, 154]. After the gene (NPHS1)
97, 173, 194]. According to the present under- responsible for this disorder was isolated, both
standing, nephrins associate with the Neph pro- CNF and NPHS1 have been used as abbreviations
teins extracellularly and form the backbone of SD for the same disorder. The disease is highly
[10, 58, 191]. Neph proteins are structurally enriched in Finland, the incidence being 1:8,200
related to nephrin and they are also involved in live births [96]. However, patients are reported all
the cell signaling [191]. Mice with genetic defects over the world among various ethnic groups
in Neph 1 develop heavy proteinuria, but no [7, 16, 117, 187]. A very high incidence (1/500
humans with mutations in Neph1 have so far live births) of CNF has been reported among the
been identified [46]. Another interesting SD com- Old Order Mennonites in Lancaster County,
ponent is transient receptor potential cation chan- Pennsylvania [22]. In recent reports of genetic
nel 6 (TRPC6), which is a cation channel that findings in CNS patients coming from all over
interacts with nephrin [207]. Mutations in this the world, mutations in the NPHS1 gene
gene lead to autosomal dominant FSGS have accounted for 40–80 % of the cases
manifesting in adulthood but not in infants. (Table 2) indicating that NPHS1 mutations
Nephrin and Neph proteins interact with the are the most important cause of genetic CNS
adapter proteins podocin, CD2AP, and ZO-1 that [83, 85, 133, 183].
25 Congenital Nephrotic Syndrome 757

Genetics misfolding of nephrin protein and defective intra-


CNF is inherited as an autosomal-recessive trait. cellular nephrin transport with absence of nephrin
The NPHS1 gene is located to chromosome in SD as well. This most probably explains why
19q13.1 and exon sequencing analysis has even “mild” mutations can cause heavy protein-
revealed two important mutations in over 90 % uria and a severe form of CNS.
of the Finnish CNF patients (Fin-major and The expression of nephrin was first reported to
Fin-minor) [112]. Both mutations result in a stop be restricted to glomerular podocytes and this was
codon and a truncated nephrin protein not later confirmed [112, 144]. The data obtained
expressed in the SD. The Fin-major and especially from mice, however, suggest that
Fin-minor mutations are rare outside Finland, nephrin may also be expressed in some areas of
but enrichment of other mutations has been the central nervous system, pancreatic beta cells,
reported also in non-Finns. In Mennonites, and testis [165]. No constant extrarenal manifes-
1481delC mutation is common and leads to a tations, however, are observed in CNF patients
truncated protein of 547 residues [22]. On the speaking against a significant role of nephrin in
other hand, a homozygous nonsense mutation other organs than the kidney [119].
R1160X in exon 27 has been found in all Maltese
patients [117]. Six of the 16 cases with this muta- Renal Pathology
tion had an atypically mild disease. The same The CNF kidneys develop quite normally in the
mutation has been reported in six French CNF fetal period [172, 175]. At the 16–22 gestational
patients and two of them had a mild disease. weeks, the glomeruli show little changes in light
Recently enrichment of c.2131C > A missense microscopy, but occasional dilated tubuli may be
mutation was observed in Maori patients from seen. In electron microscopy the mature glomeruli
New Zealand [209]. Over two hundred NPHS1 show effacement of podocyte foot processes, and
mutations have been identified and most the SD image may be completely missing in cases
non-Finns have individual mutations including with severe NPHS1 mutations [159]. It is remark-
deletions, insertions, nonsense, missense, and able that also carriers of NPHS1 mutations can
splicing mutations spanning over the whole have these “proteinuric” changes making the
gene. While most NPHS1 mutations lead to pathological diagnosis difficult [160].
CNS, it has become clear that some “mild” muta- After birth, the CNF kidneys are large with the
tions can lead to NS manifesting in older children mean kidney weight of almost twice that of
or even in adults expanding the phenotype caused age-matched control children [172]. The glomer-
by this gene defect [156, 163, 184]. uli show a slight to moderate increase of
mesangial matrix and mesangial hypercellularity
Pathogenesis (Fig. 5). Degenerative changes such as shrinking
Mutations in the NPHS1 gene result in defective of the glomerular tuft, fibrotic thickening of the
nephrin molecule and disturbed structure and Bowman’s capsule, and glomerular sclerosis
function of the glomerular filtration barrier. The become evident with time [122]. Of note, the
Fin-major and Fin-minor mutations cause a com- same kidney biopsy may show variable
plete absence of nephrin in SD, and these kidneys glomerular changes as depicted in Fig. 5. Electron
lack the filamentous image of SD in electron microscopy shows effacement of foot processes,
microscopy (Figs. 3 and 4) [159]. Similarly, irregularities in the GBM, and some swelling of
nephrin knockout mice lack the slit diaphragm the endothelial cells (peliosis) [110]. Dilated
filaments in electron microscopy and die of severe proximal tubules are few at first, but radial dila-
proteinuria soon after birth [165]. The findings tions of the tubules become obvious with time.
suggest that the absence of nephrin leads to leak- Also, interstitial fibrosis and lymphocyte
age of plasma proteins into urine through the collections especially around sclerotic glomeruli
“empty” podocyte pores. Liu et al. [130] have increase substantially within the first 1–2
shown that many missense mutations cause years [120].
758 H. Jalanko and C. Holmberg

Fig. 3 Electron
microscopy of the
glomerular capillary wall.
(a) Normal kidney showing
the podocyte foot processes,
the glomerular basement
membrane, and the
fenestrate endothelium. (b)
NPHS1 kidney with
irregular podocyte foot
processes (effacement)
typical for all proteinuric
kidney diseases. (c)
Immunoelectron microcopy
of a podocyte pore in a
normal kidney. The
filamentous image of the slit
diaphragm (S) is seen
(arrow). Antibodies against
the intracellular part of
nephrin show the
localization of nephrin in
the S (gold particles). (d):
Immunoelectron
microscopy of the podocyte
slit pore in a NPHS1 kidney.
No SD or nephrin is seen

Fig. 4 Scanning electron microscopy of a capillary wall in structure is distorted. The cell body (arrow) “hang” in a
a normal and a NPHS1 kidney. (a). The podocyte cell body primary process and the ramification of secondary pro-
(arrow) and the primary, secondary, and tertiary processes cesses is absent
are clearly seen. (b). In the NPHS1 kidney, the podocyte
25 Congenital Nephrotic Syndrome 759

Fig. 5 Typical histological findings in CNF kidneys demonstrating the variability of glomerular lesions. The findings
range from normal to severely sclerotic glomeruli indicating that the etiological diagnosis requires genetic testing

Clinical Features features of CNF, such as generalized edema,


The basic problem in CNF is severe renal loss of abdominal distension, ascites, and widened cra-
plasma protein, which may begin already in utero. nial sutures and fontanelles, may develop during
The early signs and symptoms are secondary to the first weeks or months of life.
this protein deficiency [75, 96, 159]. In a survey of Infants with CNF do not have extrarenal
the Finnish patients, over 80 % of the children malformations. Minor functional disorders in the
were found to be born prematurely (<38th week), central nervous system and heart, however, are
with a mean birth weight of 2,600 g quite common during the nephrotic stage. Most
(1,500–3,500). However, only two of the 46 new- children have muscular hypotonia, and mild atro-
borns were small for gestational age (65). Most phic changes in the brain were observed by com-
neonates do not have major pulmonary problems, puter tomography (CT) or magnetic resonance
although amniotic fluid is often meconium imaging (MRI) in a third of the Finnish infants
stained. The placenta is larger than normal and [159]. In 8 % of the Finnish patients, dystonic
almost invariably weighs more than 25 % of the cerebral palsy has been diagnosed; the etiology
baby’s birth weight. The mean ratio of placental to of this is not known. Minor cardiac findings such
infant weight (ISP) is 0.38 in babies with CNF as hypertrophy and mild pulmonary stenosis have
compared with 0.18 in normal babies. The reason been reported in one-fourth of the Finnish patients
for this is not known. [159]. In a report form Malta, pulmonary valve
In typical CNF, edema and abdominal disten- stenosis was found in three cases and a subaortic
sion become evident soon after birth. NS was stenosis in one patient [67].
diagnosed within the first week in 82 % of the
Finnish patients and within 2 months in the rest of Laboratory Findings
the cases. In a survey of European and Turkish The magnitude of proteinuria depends on the
patients, CNF manifested within the first 90 days “severity” of the NPHS1 mutations. Typically,
in all 18 cases [85]. Protein losses lead to severe the first urine analysis already shows proteinuria,
hypoalbuminemia (<10 g/L) before protein sub- microscopic hematuria, and some leukocyturia. If
stitution is started [88]. Without albumin substi- the serum albumin concentration is not low (<15
tution and nutritional support, the classical g/L), the urinary protein concentration usually
760 H. Jalanko and C. Holmberg

exceeds 20 g/L. In addition to albumin, many that identical mutations may result in onset of
other proteins are lost into urine: immunoglobulin SRNS over spectrum of several years suggests
G (IgG), transferrin, apoproteins, lipoprotein that additional modifying factors are involved.
lipase (LPL), antithrombin III (ATIII), ceruloplas- Since podocin is a podocyte adapter protein
min, vitamin D binding protein, and thyroid bind- required for the proper targeting of nephrin into
ing globulin [3]. The serum levels of these and SD, also nephrin expression may be distorted in
their ligands (e.g., thyroxin) are low in serum, CNS caused by NPHS2 mutations [66, 94, 152,
leading to secondary metabolic disturbances. 188]. Coexistence of NPHS1 and NPHS2 muta-
The low thyroxin concentration leads to an tions has been reported in CNS patients, but the
increase in thyroid-stimulating hormone clinical significance of this is not clear [117, 188].
(TSH) [141]. CNS caused by NPHS2 mutations is an autosomal
Low serum albumin and postheparin plasma recessive disease requiring disease-causing muta-
LPL activities and high free fatty acid concentra- tions on both alleles. Interestingly, it was recently
tions lead to hypertriglyceridemia. Total and found that p.Arg229Gln mutation leads to disease
low-density lipoprotein (LDL) cholesterol levels phenotype only when it is associated with some
are high but high-density lipoprotein (HDL) 30 -mutations [201].
levels are low, and the LDL and HDL particles
are enriched with triglycerides [4]. These lipid
abnormalities and arteriolar changes seen already Clinical Features
during the first year of life may lead to an
increased risk of arteriosclerosis. Deficiency in No systematic analysis of the clinical findings in
polyunsaturated fatty acids may be prominent in CNS patients with NPHS2 mutations has been
infants with CNS, which may be regarded as a published. It is, however, obvious that the severity
possible risk factor for the developing brains [1]. of proteinuria and the clinical findings are more
variable than in CNF patients. While nephrotic
range proteinuria is typically detected in the first
Podocin Gene Mutations (NPHS2) few days of life in CNF patients, CNS often man-
ifests at the age of a few weeks in infants with
Genetics NPHS2 mutations [18]. In the study of Machuca
et al., 74 % of all patients presenting NS during
Mutations in the NPHS2 gene, encoding for a the first week of life had NPHS1 mutations,
podocyte protein podocin, are a common cause whereas only 11 % carried NPHS2 mutations.
of a steroid-resistant NS (SRNS) in children and Conversely, the latter gene mutations accounted
adults, accounting for up to 28 % of sporadic and for 37 % of patients in whom NS began after
over 40 % of familial cases of SRNS manifesting 1 month of life, whereas NPHS1 mutations were
at various ages [30, 31, 52, 109, 157, 178]. The less frequent (16 %) at this stage [133]. CNS with
podocin gene mutations, however, are also an NPHS2 mutations develops end stage renal dis-
important cause of CNS accounting for 17–39 % ease (ESRD) on average 6.6 years after diagnosis
of the cases (Table 2). [85, 86]. Patients with p.R138Q mutations
NPHS2 mutations were found in 18 % of over progressed to ESRD at the median age of
400 patients from a worldwide cohort of SRNS 79 months, whereas patients carrying the p.
cases with the disease onset ranging from birth to V260E mutations progressed more rapidly
21 years (88). Severe truncating mutations (non- [133]. As podocin is only expressed in kidney
sense or frameshift) resulted in early onset of NS glomerulus, non-renal manifestations are unlikely
(from birth to 9.1 years of age; mean 1.75 years). as is the case with CNF. The NPHS2 founder
This was also true for homozygous R138Q muta- mutation R138X was associated with cardiac
tion (from birth to 5.4 years; mean 1.77 years), anomalies in six consanguineous Arabs
which is common in European patients. The fact [51]. However, in another cohort of 12 patients,
25 Congenital Nephrotic Syndrome 761

10 had a normal heart condition and two presented tumor, aniridia, genitourinary anomalies, and
mild anomalies with mitral insufficiency and left mental retardation) [148]. Missense mutations
ventricular hypertrophy [32]. mostly cause DDS, donor splice site mutations
In general, most patients with NPHS2 muta- in intron 9 are associated with FS, and constitu-
tions show histology of focal segmental tional deletion of one whole copy of WTI is asso-
glomerulosclerosis (FSGS) in kidney biopsy. ciated with WAGR. FS is characterized by the
However, CNS patients with NPHS2 mutations association of male pseudohermaphroditism and
often show minimal histological changes or glomerulopathy [9, 114]. There is complete male
CNF-type histology, and FSGS is seen in less to female gender reversal in 46, XY patients. FS is
than half of the patients [133]. Thus, it is clear associated with gonadoblastomas but not with
that CNS caused by NPHS1 or NPHS2 mutations Wilms’ tumor. Proteinuria is detected in child-
cannot be differentiated by the renal histology or hood, usually between 2 and 6 years of age, and
clinical findings. kidney biopsy reveals FSGS.
DDS is caused by heterozygous mutations in
WT1 [59, 162]. More than 60 germline mutations
WT1 Gene Mutations (Denys–Drash, (both familial and de novo) have been described in
Isolated CNS) DDS patients. Most are missense mutations
within exons 8 and 9, coding for zinc finger
Genetics domains 2 and 3, which leads to alteration in the
DNA-binding capacity of WT1 [128]. Also dele-
Wilms’ tumor suppressor gene (WT1) is located tions, insertions, and nonsense mutations have
on chromosome 11p13 [29]. It encodes for a been described in DDS patients leading to a trun-
nuclear WT1 protein, which is a transcription cated protein [189]. The mutant protein probably
factor of the zinc finger family presumed to regu- suppresses the normal allele, which explains the
late the expression of numerous target genes more severe phenotype seen in DDS compared
through DNA binding [145]. WT1 plays a crucial with children with complete deletion of one
role in the embryonic development of the kidney WT1 allele. The podocyte function is affected,
and genitalia. In mature kidney, WT1 is abun- and, at the molecular level, upregulation of
dantly expressed in podocytes and it is believed PAX2 and downregulation of nephrin have been
to control the expression the SD proteins, such as reported [146]. Also, the expression of growth
nephrin [198]. The WT1 gene contains 10 exons, factors that regulate glomerular capillary develop-
the first six of which encode a proline-/glutamine- ment is affected in DDS [69].
rich transcriptional regulatory region. Exons 7–10 DDS is a combination of NS showing a histo-
encode the four zinc fingers of the DNA-binding pathologic picture of diffuse mesangial sclerosis
domain. Up to 24 different isoforms of WT1 may (DMS), male pseudohermaphroditism, and
result from the combination of alternative trans- Wilms’ tumor [44, 47, 49]. DDS can be divided
lations sites, alternative RNA splicing, and RNA into three clinical categories: (1) genotypic males
editing. The biological role of all these isoforms is with all three abnormalities, (2) genotypic males
not known. with nephropathy and ambiguous external and/or
internal genitalia only, and (3) genotypic females
with nephropathy and Wilms’ tumor only
Clinical Features [189]. While male pseudohermaphroditism is an
important diagnostic sign, the diagnosis in
A variety of WT1 mutations, which either affect females may be difficult and delayed. The renal
development or induce tumor formation, have tumor and nephropathy may be clinically simul-
been identified. Developmental defects include taneous, but in many cases, nephropathy either
the Denys–Drash syndrome (DDS), Frasier syn- precedes the tumor or remains the only abnormal-
drome (FS), and WAGR syndrome (Wilms’ ity of the disease [8, 38].
762 H. Jalanko and C. Holmberg

The nephropathy begins with proteinuria and encodes for the laminin β2-chain that is specifi-
NS. It is usually discovered at the age of a few cally expressed in the GBM, ocular structures, and
months, sometimes at birth. Overall, WT1 muta- neuromuscular synapses. Laminins are extracel-
tions account for a few percent of CNS cases lular matrix proteins with important roles in cell
(Table 2). Renal failure is often noted concomitantly adhesion, differentiation, and proliferation. The
with NS and progression to ESRD before the age of deficiency of laminin β2-chain results in the
4 years is the rule. The characteristic glomerular GBM alterations and impairment of the function
lesion is DMS and in rare cases FSGS [211]. No of the glomerular filtration barrier. In an animal
correlation between the genotype and the severity of model, lack of laminin β2-chain resulted in
renal disease has been observed. Because the renal ectopic deposition of laminin and mislocalization
disease in DDS is resistant to drug treatment, kidney of anionic sites in the GBM [103]. These changes
transplantation remains the only therapeutic alterna- and proteinuria preceded the lesions in podocytes
tive, and favorable results without recurrence of indicating that the GBM lesions were responsible
DMS have been reported. In order to avoid the for protein leakage.
development of Wilms’ tumor, bilateral nephrec- Pierson syndrome comprises CNS with the
tomy at the onset of terminal renal failure is histological presentation of DMS, associated
recommended. Removal of native kidneys is with complex ocular maldevelopment. Renal dis-
recommended to be performed to all patients with ease may already start prenatally resulting in ter-
nephropathy caused by WT1 mutations [93]. minal renal failure during the first few weeks or
Mutations in WT1 can also cause an isolated months of life [214, 215]. The most characteristic
kidney disease with NS appearing at a various ocular feature in Pierson syndrome is microcoria,
age. In many cases, the kidney biopsy reveals a fixed narrowing of the pupils due to a defect of
DMS histology. Three historical studies reported the dilator muscle. Ocular manifestations also
WT1 mutations in eight of 21 patients with iso- include abnormalities of the lens, cornea, and
lated DMS [74, 98, 99, 104, 177]. These muta- retina, often leading to blindness. Patients who
tions were localized on exons 7, 8, and 9 and on survive the newborn period due to renal replace-
intron 9 in the WT1 gene. The connection of CNS ment therapy develop neurological deficits, con-
and DMS histology was described already in the sistent with the known expression of laminin β2 in
1970s [73, 126]. As the clinical picture appeared the nervous system. These include severe muscu-
to be distinct, DMS was thought to represent a lar hypotonia and psychomotor retardation [210].
specific entity. In the published reports, protein- Pierson syndrome is typically caused by
uria was often moderate and NS was detected biallelic truncating mutations in the LAMB2
soon after birth or later in infancy, often in the gene. Several reports have, however, revealed
second and third years. It is clear now that isolated LAMB2 mutations causing milder variants of
DMS is not a single entity but caused by several Pierson syndrome. Hasselbacher et al. described
gene defects. Besides WT1, mutations in the two children with CNS and early development of
PLCE1 and LAMB2 gene often result in DMS ESRD, but no other primary abnormalities
type histology, as discussed later. It is also proba- [78]. Kagan et al. described a patient with CNS,
ble that new genetic variants causing DMS will be high-grade myopia, and minor structural eye
found in the next few years. anomalies, but no microcoria [107]. Choi
et al. reported a girl with congenital microcoria
and infantile NS, with normal renal function still
Laminin b2 Gene Mutations (Pierson at the age of 6 years [36]. Matejas et al. reported a
Syndrome) large cohort of 51 patients from 39 unrelated fam-
ilies showing that in most patients (90 %) NS
Pierson syndrome is a form of primary CNS started during the first 3 months of age. DMS
caused by loss-of-function mutations in the was the major histological finding in biopsies
LAMB2 gene [137, 205, 214, 215]. This gene with a few samples showing FSGS. ESRD
25 Congenital Nephrotic Syndrome 763

developed within a few weeks or months in most DMS histology in the kidney biopsy. Nine of
cases. Microcoria or other ocular abnormalities these 12 patients progressed to ESRD by the age
were detected in almost all, and severe visual of 5 years. Interestingly, two patients responded to
impairment was noticed in every other patient. cyclosporine A or prednisone and have remained
Microcephaly or neurodevelopmental deficits in remission for many years. Two siblings had
were present in 10 and 50 % of the patients, nontruncating missense mutations and both had
respectively [136]. FSGS histology in kidney biopsy.
Since missense mutations in the LAMB2 pre- Based on the original reports, Gbadegesin
sent with a spectrum of symptoms reaching from et al. analyzed the relative frequency of PLCE1
isolated early-onset NS to patients with CNS and mutations in a worldwide cohort of children with
mild ocular and neurological symptoms, genetic “idiopathic” DMS and found truncating PLCE1
analysis of LAMB2 may be warranted in CNS mutations in 10 of the 35 families (29 %) [61]. The
patients with no mutations in the NPHS1 and age of patients at the onset of the disease varied
NPHS2 genes. LAMB2 mutations, however, are between 1 and 36 months and most patients
much less frequent as indicated in Table 2. progressed to ESRD quite fast, with an age
range of 8–60 months. Importantly, three of the
35 families had mutations in the WT1 gene and
PLCe1 Gene Mutations (NPHS3) none had LAMB2 mutations. Thus, PLCE1 muta-
tions were the most common cause of idiopathic
Mutations in the phospholipase C epsilon DMS in this cohort. PLCE1 mutations are, how-
1 (PLCE1) gene cause an autosomal recessive ever, a rare cause of NS in infants (Table 2) or
form of early-onset NS [87]. PLCe1 is a phospho- older children with steroid-resistant NS. Besides
lipase enzyme and a signaling protein for many G DMS, the kidney biopsy may also show FSGS
protein-coupled receptors, including angiotensin histology [25, 60]. It has also been shown that
II. PLCe1 promotes the downstream activation of even homozygous mutation in PLCE1 gene may
protein kinase C enhancing calcium signaling not be sufficient to cause NS, suggesting that other
events. PLCe1 is abundant in kidney glomerulus factors modify the effect of PLCE1 abnormalities
and localizes to the cytoplasm of the podocyte cell [63]. Accordingly, simultaneous mutation in
body and major and secondary processes. It inter- PLCE1 gene and mitochondrial RC complex defi-
acts with IQ motif-containing GTPase-activating ciency has been reported in an infant with
protein 1 (IQGAP1), which also interacts with NS [13].
nephrin [105]. This suggests that PLCe1 may be
involved in the SD signaling and the function of
the glomerular filter [34]. PLCe1 is highly Other Forms of Primary CNS
expressed in the developing glomerulus and the
absence of PLCe1 may halt kidney development Nail–patella syndrome is caused by mutations in
leading to DMS-type glomerular lesions. Of note, the gene coding for Lim homeobox transcription
this is associated with a major reduction in the factor 1β (LMXB1) [21, 80]. This protein is
expression of nephrin and podocin [84]. expressed in podocytes and regulates the synthe-
Mutations in PLCE1 were originally reported sis of alpha-chains of collagen type IV, podocin,
in 14 patients coming from seven families and CD2AP. Renal symptoms are found in a third
[87]. Most patients were Turkish and the age at of these patients and a single case with CNS has
the onset of the disease ranged from 2 months to been described. NS has also been described in
9 years. The patients did not have extrarenal man- single pediatric and adult patients with genetic
ifestations resembling the situation in patients defect in acting-binding proteins (α-actinin-4,
with NPHS1 and NPHS2 mutations. Twelve inverted formin-2, myosin-9, myosin 1E, and
patients had homozygous truncating mutation, ARHGDIA) [11, 24, 27, 57, 70, 108, 113, 115,
and all developed gross proteinuria and showed 142, 181, 192] and integrin α3-gene [149] as well
764 H. Jalanko and C. Holmberg

as those with Lowe syndrome [150], Herlitz junc- cardiac, and diaphragmatic anomalies. Most of
tional epidermolysis bullosa [79], sialic acid stor- these case reports were published in the 1990s
age disease [196], and congenital disorders of when no genetic analyses of podocyte proteins
glycosylation [195]. were available [54, 143, 153, 182].
During the past few years, mitochondrial dis-
orders have been associated with NS and some of
the patients have been small infants [45, 65, 76, Secondary Nephrotic Syndromes
82, 135, 180, 186]. The metabolic problem in
these patients has been decreased levels of mito- NS has been described secondary to some con-
chondrial coenzyme Q10 (ubiquinone), which is genital and infantile disorders. Syphilis, toxoplas-
an essential component of the mitochondrial mosis, some viral infections, and the infantile
electron-transport chain. Its biosynthesis requires form of SLE are currently the most important
at least 15 genes. Mutations in eight of them secondary causes of congenital and infantile NS.
(PDSS1, PDSS2, COQ2, COQ4, COQ6,
ADCK3, ADCK4, and COQ9) cause primary
CoQ10 deficiency, a heterogeneous group of dis- Infections
orders with variable age of onset (from birth to the
seventh decade) and associated clinical pheno- Congenital syphilis has long been known to cause
types, ranging from a fatal multisystem disease CNS. It may cause a nephritic or nephrotic syn-
to isolated steroid-resistant NS or isolated central drome in the newborn [151, pp. 157–159, 204,
nervous system disease. The interesting question 213]. Proteinuria and hematuria are present, but
here is whether ubiquinone supplementation in severe NS is less common. NS may manifest in
these patients would be helpful, which seems to the newborn but is more often seen between 1 and
be the case. In the report by Heeringa et al., ubi- 4 months of age. Membranous nephropathy is a
quinone treatment (15–30 mg/kg/day) was started common finding in kidney biopsy. Antimicrobial
in a 2-month-old infant with COQ6 mutations and therapy, usually penicillin, is curative, provided
NS. During the follow-up proteinuria decreased that irreversible renal lesions have not
quite dramatically and remained low until the age developed [12].
of 15 months [82]. An association of neonatal cytomegalovirus
In 1968, Galloway and Mowat [53] described (CMV) infection and CNS has also been reported
two siblings showing congenital microcephaly, [14, 20, 170]. In these cases, DMS type histology
NS, and hiatus hernia. Several subsequent reports is often found and the disease clearly responds to
showing various brain malformations and con- ganciclovir therapy. In these infections, manifes-
genital or early-onset NS have been called tations other than NS lead to the correct diagnosis.
Galloway–Mowat syndrome. The classic Toxoplasmosis has been associated with CNS in a
Galloway–Mowat syndrome is an autosomal few cases [193]. Congenital rubella has been
recessive disorder, but the disease-causing gene reported to cause CNS with membranous glomer-
has not been isolated [118, 197]. ulonephritis [50]. The acquired immunodefi-
Since podocytes and neuronal cells share com- ciency syndrome caused by the human
mon morphology and biology with many proteins immunodeficiency virus is associated with
expressed in both types of cells, the association of nephropathy, including NS. This disease usually
CNS and brain problems is not unexpected. In affects children older than 1 year, but some HIV
cases with CNS and brain malformation, the his- infants with nephropathy have been reported. In a
tological finding has often been DMS, but other recent report highly active antiretroviral therapy
histological findings have also been reported. A in combination with angiotensin-converting
wide variety of clinical anomalies have also been enzyme antagonists was shown to be effective in
reported in patients with CNS, including dysmor- decreasing proteinuria and preserving renal func-
phic facial features as well as ocular, limb, tion [171]. Hepatitis B virus may cause
25 Congenital Nephrotic Syndrome 765

membranous glomerulonephritis, and infants with cells and leucocytes are often present in urine.
nephrosis associated with hepatitis B infection Serum creatinine and urea levels are variable.
have been described [64]. Renal function remains quite normal for the first
months in CNF, but in other forms kidney failure
may develop faster. Blood pressure levels can be
Immune Disorders low due to hypoproteinemia and low oncotic pres-
sure, or elevated if renal failure is already present.
Although SLE is rarely diagnosed before 5 years In newborns, the placental weight >25 % of
of age, an infantile form of SLE has been reported. birth weight is present in CNF but may be seen in
NS was the major clinical finding in five infants other forms of CNS [159]. The kidneys may be of
aged 6 weeks to 6 months with SLE [48]. These normal size or larger than normal in ultrasound
patients had elevated antinuclear antibody titers, scanning, and the renal cortex is often
hypocomplementemia, and diffuse proliferative hyperechogenic [6]. Search for possible
glomerulonephritis. Response to the immunosup- non-renal malformations is important, especially
pressive therapy was poor in many cases. since they may give clue to the etiologic diagno-
Nephrotic syndrome due to membranous sis. These include genital abnormalities (WT1),
nephropathy has been observed antenatally in eye defects (LAMB2), and neurological disorders
infants from mothers who have mutations in the (Mowat–Galloway, LAMB2, mitochon-
metallomembrane endopeptidase gene, which driopathies). CNS caused by nephrin, podocin,
encodes the neutral endopeptidase (NEP) or PLCe1 mutations is not associated with
expressed on podocytes. During pregnancy, the extrarenal malformations. Cardiac evaluation,
fetal NEP protein induces a maternal alloimmune however, often reveals ventricular hypertrophy
response. Maternal antibody to NEP fetal protein but structural defects are rare. If infectious etiol-
results in subepithelial immune deposits in the ogy is regarded possible, search for specific anti-
fetus and the neonate. The mothers’ IgG response bodies (e.g., syphilis, toxoplasma) or DNA/RNA
to the fetal NEP determines the severity of the (e.g., CMV, hepatitis B, HIV) in blood samples is
neonatal disease [43, 155]. The nephrotic syn- indicated.
drome is present at birth but proteinuria decreases
with time along with a progressive decrease of the
maternal antibodies and a disappearance of the Kidney Biopsy
immune deposits. However, persistent proteinuria
and chronic renal insufficiency may be observed The different genetic entities of CNS are associ-
due to nephron loss and renal scarring. ated with typical histological findings, as indi-
cated above. However, since a specific gene
defect may cause several types of glomerular
Diagnosis of CNS lesions, such as mesangial expansion, FSGS,
MCNS, and DMS, and the findings in different
Clinical Findings genetic entities show overlapping, renal biopsy
does not reveal the etiology. Also, the non-
In severe forms of CNS, generalized edema, uri- glomerular findings, such as tubular dilatations
nary protein >20 g/L, and serum albumin level and interstitial fibrosis and inflammation, can be
<10 g/L can be detected in the newborn period. seen in all forms of proteinuric diseases. Thus, the
The amount of proteinuria, however, varies in indications for renal biopsy are not quite clear. The
different entities and the clinical signs may not knowledge of severity of glomerular sclerosis and
be evident during the first weeks of life. Also, the interstitial fibrosis may help in the assessment of
true magnitude of proteinuria may be detectable the treatment strategies. On the other hand, the
only after partial correction of hypoproteinemia lesions can be focal and the biopsy findings may
by albumin infusions. Small amounts of red blood be misleading. If immunohistochemistry for
766 H. Jalanko and C. Holmberg

nephrin and podocin is available, analysis of their 20th week also in carriers of the NPHS1 mutation
expression in biopsy sample is useful. Total lack of and the decision on a possible pregnancy termi-
either protein speaks for a severe disorder not nation is difficult (175). Thus, it is highly
responding to antiproteinuric therapy. recommended that the genetic testing of the fetus
or parents for NPHS1 mutations should be
performed before the 20th week whenever clearly
Gene Testing elevated AFP levels are discovered in a pregnancy
with normal ultrasound findings.
Genetic analysis is the method of choice for the
precise diagnosis of CNS. The knowledge of eti-
ology helps in assessing the management and Management of CNS
prognosis, in follow-up for possible associated
symptoms, and in genetic counseling of the fam- In most patients with primary CNS, the therapeu-
ily. Analysis of the NPHS1 and NPHS2 mutations tic decisions usually aim at renal transplantation,
is warranted in all CNS patients without a family and the goals during the first months are to control
history or clear-cut extrarenal findings. If no proteinuria and edema, provide good nutrition,
mutations are detected in NPHS1 or NPHS2 and prevent thrombosis and infections, allowing
genes, analyses of WT1, LAMB, or PLCe1 are the child to reach a weight and body size consis-
indicated according to the clinical and histological tent with successful kidney transplantation
findings. Gene tests are readily available in sev- [77, 88, 116, 185]. With an optimal therapy,
eral laboratories. Updated information on these growth and development are satisfactory and the
laboratories is provided in www.genetests.org. patients can spend most of the time at home. In
Besides the conventional sequencing of one gene most cases, terminal uremia is to be expected
at a time, next-generation sequencing analysis is within the first years of life.
now available for NS genes, which allows simul-
taneous survey of all known genes in 2–4 weeks
[132, 140]. Immunosuppressive Medication
Prenatal diagnosis in families with a known
risk for CNS should be based on genetic testing CNS is a rare problem and no larger study on the
whenever possible. The results can be obtained use of immunosuppressive drugs, prednisone or
fast if the mutations are known in advance. In case cyclosporine A (CsA), has been published. Based
of no family history or if the mutations in the on the clinical experience and the published data
affected child were not identified, prenatal genetic on older children with genetic forms of NS, it is
testing is more challenging but possible. Espe- very unlikely that either drug would bring primary
cially CNF can still be suspected prenatally CNS into remission [28, 62, 176]. Accordingly,
based on elevated alpha-fetoprotein (AFP) levels only one of the 45 European and Turkish CNS
in maternal serum and amniotic fluid [5]. If the patients achieved a continuous remission through
AFP concentration in amniotic fluid is very high the steroid treatment. The genetic defect in this
and the ultrasound examination does not reveal child was not known [85]. There are, however,
fetal anencephaly or other malformations, CNF is reports on solitary patients with “mild” mutations
a probable diagnosis. However, heterozygous in NPHS1, NPHS2, WT1, and PLCE1 who have
fetal carriers of NPHS1 gene mutations may shown improvement after immunosuppressive
have temporarily elevated AFP levels in amniotic therapy, but the mechanism behind this is not
fluid and maternal serum, and repeated measure- known [56, 83, 87, 134]. Both prednisone and
ment of amniotic fluid AFP before the 20th week CsA probably have direct effect of podocytes,
of pregnancy is recommended in cases with high which might explain the positive effect. However,
AFP levels [26, 160]. The problem, however, is CNS children most often have “severe” mutations
that the AFP levels may remain elevated after the not responding to therapy and should be spared
25 Congenital Nephrotic Syndrome 767

from long-lasting, unwarranted immunosuppres- Table 3 Management of infants with primary CNS
sive treatment. In secondary CNS, specific ther- Protein substitution parentally
apy against the infectious agent, of course, is 20 % albumin infusions as required
warranted and often effective. Dosing 3–4 g/kg/day when severe NS
Divided in 1–3 infusions (day or night)
Furosemide (0.5 mg/kg) together with albumin
Antiproteinuric Medication Deep vein catheter needed
Nutrition
A reduction in the protein excretion using an ACE Hypercaloric diet (130 kcal/kg/day)
inhibitor and indomethacin has been reported in Protein supplementation (4 g/kg/day)
infants with primary CNS [68, 81, 164]. Blockade Lipid supplementation
Complete multivitamin preparation
of renin-angiotensin axis not only reduces glo-
Calcium and magnesium (potassium) supplementation
merular perfusion pressure, but also exerts protec-
Nasogastric tube or gastrostomy often required for
tive measures on podocytes [37]. In a few patients proper feeding
with CNS, adding of angiotensin II receptor Medication
(AT II) to ACE inhibitor medication has further Anticoagulation (warfarin, AT III infusions)
reduced proteinuria [129]. Thus, the first drug in Thyroxin substitution
the antiproteinuric medication is captopril 1–5 Parenteral antibiotics when bacterial infection
mg/kg/day, and, if no response is obtained in a suspected
few weeks, losartan (0.4–1.3 mg/kg/day) can be Antiproteinuric drugs (ACE inhibitor, AT II R blocker,
indomethacin)
added to the medication. Follow-up of kidney
Dialysis
function is important. Whether indomethacin
Terminal renal failure
(1–5 mg/kg/day) is added in refractory cases is a
After nephrectomy in cases with early transplantation
matter of opinion. The antiproteinuric treatment is
Transplantation
worth trying in many cases of CNS. In our expe- After dialysis/preemptive
rience, CNF patients with the severe (truncating) Extraperitoneal grafting when >10–15 kg
Fin-major and Fin-minor mutations do not
respond to captopril or indomethacin, but if either
of the mutations leads to a single amino acid
change, a reduction in the protein excretion may age. The substitution is first divided into three 2-h
be observed [159]. infusions (starting dose of 1–5 ml/kg/infusion),
and after a few weeks, albumin is given as one
6–8-h infusion during the night (up to 15–20
Albumin Substitution ml/kg/night, which is 3–4 g/ kg of albumin). In
patients with heavy urinary losses, this substitu-
The magnitude of protein losses into urine is tion increases plasma albumin levels only tempo-
crucial for the therapeutic decisions (Table 3). rarily and no target level can be used in
If proteinuria is modest, only occasional or no determining the albumin dosing. Lack of substan-
albumin infusions are needed. On the other tial edema and stable weight are major goals. In
hand, heavy proteinuria (10–100 g/L) inevitably our hands, additional furosemide boluses are
leads to life-threatening edema, protein malnutri- given during the daytime when the maximal albu-
tion, reduced growth, and secondary complica- min dosing (4 g/kg/day) has been reached.
tions. In these cases protein substitution by
parenteral albumin infusions is mandatory. Our
practice in treating CNF patients is to infuse Nutrition
20 % albumin solution together with a bolus of
intravenous furosemide (0.5 mg/kg), using deep- Infants with severe CNS have traditionally been
vein catheters that are inserted at 2–3 weeks of treated with a high-energy (130 kcal/kg per day)
768 H. Jalanko and C. Holmberg

and a high-protein (3–4 g/kg per day) diet before the procedure and ATIII (50 IU/kg) is
[37, 88]. Breast milk and cow’s milk-based infant given preoperatively to temporarily correct the
formulae are first used and the excess protein is ATIII deficiency. Patients with CNS may also
given as a whey- and casein-based protein prod- loose serum transferrin, transcobalamin, and
uct. Glucose polymers and fat emulsion are given erythropoietin into urine, which is associated
to increase energy intake. The children also with development of anemia [202]. Management
receive vitamin D3 (10–40 μg/day), and alpha- of this anemia with erythropoietin is often unsuc-
calcidiol (0.1–0.3 μg/day) is added when an cessful and repeated blood transfusions are
increase of parathyroid hormone level is noticed. needed. On the other hand, some CNS patients
A complete multivitamin preparation is given show thrombophilia and low-dose aspirin has
according to the recommended dietary allowances been used in individual cases.
for healthy children of the same age. Supplemen- Bacterial infections may be a major problem in
tary magnesium (50 mg/day) and calcium infants with NS. Because of urinary losses of
(500–1,000 mg/day) are also given to keep the gamma globulins and complement factors B and
serum levels within the normal range. The daily D [138], nephrotic children are especially prone to
water allowance is 100–130 mL/kg. Most patients infections caused by capsular bacteria such as
need a nasogastric tube or gastrostomy to guaran- pneumococci, and prophylactic use of penicillin
tee their energy intake. Modifications to this treat- has been recommended. Ljungberg
ment are, of course, necessary with deteriorating et al. retrospectively analyzed the incidence and
renal function. type of infection in 21 infants with CNF during
the first year of life [131]. The infants suffered
from 63 verified and 62 suspected episodes of
Additional Medication sepsis. The use of central venous lines had no effect
on the incidence, and the prophylactic use of anti-
Patients with NS often have low levels of serum biotics or immunoglobulins did not reduce the
thyroid-binding globulin and thyroxin [33, 41, 55]. incidence. Thus, we do not recommend prophylac-
McLean et al. [141] reported an increase in TSH in tic medication or immunoglobulin infusions, but a
four of five patients with CNS and a positive high degree of suspicion for septic infections is
response to thyroxin substitution. We have a similar warranted. The symptoms are often vague and
experience. Although serum thyroxin concentra- masked by signs of focal infections occurring at
tion is always low, TSH may be normal in the the same time. Due to urinary losses, plasma
beginning but increases in most patients during c-reactive protein (CRP) levels remain low. Anti-
the first months. Thus, we have adopted a policy biotic therapy should be started promptly on suspi-
of routinely giving thyroxin from birth, adjusting cion and should cover the major hospital strains of
the dosage according to TSH. bacteria. Response to treatment is usually excel-
Urinary excretion of plasminogen and ATIII lent. Because of the urinary loss of IgG, vaccina-
leads to compensatory protein synthesis, resulting tions are preferably given after nephrectomies.
in increased levels of macroglobulins, fibrinogen,
thromboplastin, and factors II, V, VII, X, and XII,
contributing to hypercoagulopathy [158]. Throm- Unilateral Nephrectomy
boses and severe coagulation problems have been
reported in children with CNS [127], and the use Some centers have adopted a routine of
of low-dose aspirin and dipyridamole therapy has performing unilateral nephrectomy to reduce pro-
been recommended. All Finnish CNF patients are tein losses [39, 139]. This decreases the need and
treated with sodium warfarin from 3 to 4 weeks of frequency of the albumin infusions and may help
age, which has clearly decreased the incidence of in the everyday management so that the com-
severe thrombotic events. Before surgical or vas- mencement of dialysis and renal transplantation
cular procedures, warfarin is stopped 5 days can be postponed to an older age. In infants with
25 Congenital Nephrotic Syndrome 769

massive protein losses (up to 100 g/L), such as The fact that CNS children are often transplanted
those with truncating NPHS1 mutations, removal at 1–3 years of age using adult-size kidneys may
of one kidney does not reduce protein losses dra- sometimes be surgically demanding and increase
matically. Based on this we proceed directly to the risk for thrombotic complications, as com-
bilateral nephrectomy and peritoneal dialysis at an pared to older recipients. Peri- and postopera-
early age in CNF patients to avoid the many tively abundant hydration of the recipient (2,500
problems encountered during the nephrotic ml/m2) is necessary to maintain optimum aortic
stage. Prolonged NS is also accompanied by path- and renal artery blood flow and avoid low-flow
ological lipid status, which leads to vascular states that could damage the graft [179].
changes already during infancy. The use of immunosuppressive medication
should be balanced in order to prevent rejection
episodes, which may be clinically subtle and, on
Dialysis the other hand, avoid the many side effects asso-
ciated with these drugs. Recurrence of NS in the
Although quite normal growth can be achieved with graft is very exceptional but has occurred in some
optimal therapy during the nephrotic stage, the CNF children who have developed anti-nephrin
patients with severe CNS are still malnourished and antibodies after transplantation. Treatment of the
hypoproteinemic and have a risk for septic infections recurrence with cyclophosphamide and plasma-
and thrombotic events, which may lead to permanent pheresis and more recently with anti-CD20 anti-
neurological complications [2]. To optimize treat- bodies normally leads to remission [89, 121, 161].
ment we perform bilateral nephrectomy and com- Recurrent NS has also been reported in a
mence peritoneal dialysis or hemodialysis when the few patients with NPHS2 mutations, but in
infant weighs about 7 kg. The PD catheter is insti- these cases no anti-podocin antibodies were
tuted 2 weeks prior to nephrectomy. CNF children detected [15].
often have inguinal hernias, which are corrected Overall, the results of kidney transplantation in
when the PD catheter is inserted. The diet and med- CNS are good and similar to those obtained in other
ication used for a proteinuric child are switched to etiologies [101]. Patient survival at 5 years is over
that of a uremic infant. The aim is to improve the 90 % and graft survival over 80 % in registry data-
child’s nutritional state before kidney transplantation, bases and in single centers [166]. Also, the quality of
which is performed when the child weighs about life is good in most patients [71, 72]. A great major-
10 kg. The general condition improves, muscle ity (72 %) of those who have reached the school
mass increases, and catch-up growth can be attend a normal class. The early CNF patients who
documented while receiving PD [90, 91, 123]. It suffered from thrombotic complications have neu-
must be emphasized that different centers have vari- rologic handicaps [17, 168, 169]. Growth and puber-
able practices in the management of CNS infants. In tal development of CNS patients, who were
contrast to our practice, early (intraabdominal) trans- transplanted as infants, is quite normal [200], but
plantation can be performed in a proteinuric infant fertility in males who have reached the adulthood is
without dialysis, and the results are satisfactory. On reduced [199]. Chronic allograft nephropathy is a
the other hand, in CNS patients with moderate pro- major problem also in these patients and the second
teinuria, the development of ESRD is often waited transplantation is frequent when the patients become
before RTx is performed. So, many patients are more young adults [101, 167].
than 2 years of age at the time of RTx.

Concluding Remarks
Kidney Transplantation
During the past few years, our knowledge on the
Renal transplantation has become an established genetic and molecular basis of CNS has greatly
mode of therapy for most children with CNS. increased. Podocyte proteins play an important
770 H. Jalanko and C. Holmberg

role in the glomerular sieving and mutations in the 14. Batisky F, Roy S, Gaber L. Congenital nephrosis and
nephrin, podocin, WT1, PLCε1, and laminin B2 neonatal cytomegalovirus infection: a clinical associ-
ation. Pediatr Nephrol. 1993;7:741–3.
account for most cases of CNS. It can be expected 15. Becker-Cohen R, Bruschi M, Rinat C, et al. Recurrent
that more genetic defects will be found in CNS nephrotic syndrome in homozygous truncating
patients in the near future. Also, management of NPHS2 mutation is not due to anti-podocin anti-
these infants has improved, so that the outcome of bodies. Am J Transplant. 2007;7:256–60.
16. Beltcheva O, Lenkkeri U, Kestilä M, et al. Mutation
CNS patients is close to other pediatric kidney spectrum in the nephrin gene (NPHS1) in
patients requiring renal transplantation. congenital nephrotic syndrome. Hum Mutat.
2001;17:368–73.
17. Benfield M, McDonald R, Bartosh S, et al. Changing
trends in pediatric transplantation: 2001 Annual
References Report of the North American Pediatric renal trans-
plant cooperative study. Pediatr Transplant.
1. Alamiz-Echevarria L, Vallo A, Aguire M, 2003;7:321–31.
et al. Essential fatty acid deficiency profile in patients 18. Benoit G, Machuca E, Antignac C. Hereditary
with nephrotic-range proteinuria. Pediatr Nephrol. nephrotic syndrome: a systematic approach for
2007;22:533–40. genetic testing and a review of associated podocyte
2. Antikainen M. Protein and lipid metabolism in gene mutations. Pediatr Nephrol. 2010;25:1621–32.
nephrotic infants on peritoneal dialysis. Pediatr 19. Benzing T. Signalling in the slit diaphragm. J Am Soc
Nephrol. 1993;7:428–53. Nephrol. 2004;15:1382–91.
3. Antikainen M, Holmberg C, Taskinen M. Growth, 20. Besbas N, Bayrakei U, Kale G,
serum lipoproteins an apoproteins in infants with et al. Cytomegalovirus-related congenital nephrotic
congenital nephrosis. Clin Nephrol. 1992;38:254–63. syndrome with diffuse mesangial sclerosis. Pediatr
4. Antikainen M, Sariola H, Rapola J, et al. Pathology of Nephrol. 2006;21:740–2.
renal arteries of dyslipidemic children with congenital 21. Bongers E, Gubler M, Knoers N. Nail-patella syn-
nephrosis. APMIS. 1994;102:129–34. drome. Overview on clinical and molecular findings.
5. Aula P, Rapola J, Karjalainen O, et al. Prenatal diag- Pediatr Nephrol. 2002;17:703–12.
nosis of congenital nephrosis in 23 high-risk families. 22. Bolk S, Puffenberger EG, Hudson J, et al. Elevated
Am J Dis Child. 1978;132:984–7. frequency and allelic heterogeneity of congenital
6. Avni E, Vanhdenhoute K, Devriendt A, et al. Update nephrotic syndrome, Finnish type, in the old order
on congenital nephrotic syndromes and contribution Mennonites. Am J Hum Genet. 1999;65:1785–90.
of US. Pediatr Radiol. 2011;41:76–81. 23. Boute N, Gribouval O, Roselli S, et al. NPHS2,
7. Aya K, Tanaka H, Seino Y. Novel mutation in the encoding the glomerular protein podocin, is mutated
nephrin gene of a Japanese patient with congenital in autosomal recessive steroid-resistant nephrotic
nephrotic syndrome of the Finnish type. Kidney Int. syndrome. Nat Genet. 2000;24:349–54.
2000;57:401–4. 24. Boyer O, Benoit G, Gribouval O. Mutations in INF2
8. Barakat A, Papadoupoulo Z, Chana R, are a major cause of autosomal dominant focal seg-
et al. Pseudohermaphroditism, nephron disorder an mental glomerulosclerosis. J Am Soc Nephrol.
Wilms tumor: a unifying concept. Pediatrics. 2011;22:239–45.
1974;54:366–9. 25. Boyer O, Benoit G, Gribouval O, et al. Mutational
9. Barbaux S, Niaudet P, Gubler M-C, et al. Donor analysis f the PLC1 gene in steroid resistant nephrotic
splice-site mutations in WT1 are responsible for syndrome. J Med Genet. 2010;47:445–52.
Frasier syndrome. Nat Genet. 1997;17:467–70. 26. Brady T, Mitra A, Hooks J. Maternal serum alpha-
10. Barletta GM, Kovari I, Verma R, et al. Nephrin and fetoprotein level peak at 19–21 weeks’ gestation and
Neph1 co-localize at the podocyte foot process subsequently decline in a NPHS1 sequence variant
intercellular junction and form cis hetero-oligomers. heterozygote; implications for prenatal diagnosis of
J Biol Chem. 2003;278:19266–71. congenital nephrosis of the Finnish type. Prenat
11. Barua M, Brown E, Charoonratana V, et al. Mutations Diagn. 2014;34:1–3.
in the INF2 gene account for a significant proportion 27. Brown E, Schlondorff J, Becker D, et al. Mutations in
of familial but not sporadic focal and segmental the formin protein INF2 cause focal segmental
glomerulosclerosis. Kidney Int. 2013;83:316–22. glomerulosclerosis. Nat Genet. 2010;42:72–6.
12. Basker M, Agarwal I, Benon K. Congenital nephrotic 28. Buscher A, Kranz B, Buscher R, et al. Immunosup-
syndrome- a treatable cause. Ann Trop Paediatr. pression and renal outcome in congenital and pediat-
2007;27:87–90. ric steroid resistant nephrotic syndrome. Clin J Am
13. Baskin E, Bayracki U, Alehan F, et al. Respiratory Soc Nephrol. 2010;5:2075–84.
chain deficiency presenting as diffuse mesangial scle- 29. Call K, Glaser T, Ito C, et al. Isolation and character-
rosis with NPHS3 mutation. Pediatr Nephrol. ization of a zinc finger polypeptide gene at the human
2011;26:1157–61.
25 Congenital Nephrotic Syndrome 771

chromosome 11 Wilms’ tumor locus. Cell. 46. Donoviel D, Freed D, Vogel H, et al. Proteinuria and
1990;60:509–20. perinatal lethality in mice lacking NEPH1, a novel
30. Caridi G, Bertelli R, Carrea A, et al. Prevalence, protein with homology to nephrin. Mol Cell Biol.
genetics, and clinical features of patients carrying 2001;21:4829–36.
podocin mutations in steroid-resistant nonfamilial 47. Drash A, Sherman F, Hartmann W, Blizzard R. A
focal segmental glomerulosclerosis. J Am Soc syndrome of pseudohermaphroditism, Wilms’
Nephrol. 2001;12:2742–6. tumor, hypertension, and degenerative renal disease.
31. Caridi G, Bertelli R, Di Duca M, et al. Broadening the J Pediatr. 1970;76:585–93.
spectrum of diseases related to podocin mutations. J 48. Dudley J, Fenton T, Unsworth J, et al. Systemic lupus
Am Soc Nephrol. 2003;14:1278–86. erythematosus presenting as congenital nephrotic
32. Caridi G, Dagnino M, Carrea A, et al. Lack of cardiac syndrome. Pediatr Nephrol. 1996;10:752–5.
anomalies in children with NPHS2 mutations. 49. Eddy A, Mauer S. Pseudohermaphroditism,
Nephrol Dial Transplant. 2007;22:1477–8. glomerulopathy, and Wilms’ tumor (Drash syn-
33. Chadha V, Alon U. Bilateral nephrectomy reverses drome): frequency in end-stage renal failure. J Pediatr.
hypothyroidism in congenital nephrotic syndrome. 1985;106:584–7.
Pediatr Nephrol. 1999;13:209–11. 50. Esterly J, Oppenheimer E. Pathological lesions due to
34. Chaib H, Hoskins B, Ashraf S, et al. Identification of congenital rubella. Arch Pathol. 1969;87:380–8.
BRAF as a new interactor of PLCE1, the protein 51. Frishberg Y, Feinstein S, Rinat C, et al. The heart of
mutated in nephrotic syndrome type 3. Am J Physiol children with steroid-resistant nephrotic syndrome; is
Renal Physiol. 2008;294:F93–9. it all podocin? J Am Soc Nephrol. 2006;17:227–31.
35. Chen Y, Zhou Y, Go G, et al. Laminin β2 gene mis- 52. Frishberg Y, Rinat C, Megged O, et al. Mutations in
sense mutation produces endoplasmic reticulum NPHS2 encoding podocin are a prevalent cause of
stress in podocytes. J Am Soc Nephrol. steroid-resistant nephrotic syndrome among Israeli-
2013;24:1223–33. Arab children. J Am Soc Nephrol. 2002;13:400–5.
36. Choi H, Lee B, Kang J, et al. Variable phenotype of 53. Galloway W, Mowat A. Congenital microcephaly
Pierson syndrome. Pediatr Nephrol. with hiatus hernia and nephrotic syndrome in two
2008;23:995–1000. sibs. J Med Genet. 1968;5:319–21.
37. Copelovitch L, Guttenberg M, Pollak M, Kaplan 54. Garty B, Eisenstein B, Sanbank J, et al. Microcephaly
B. Renin-angiotensin axis blockade reduces protein- an congenital nephrotic syndrome owing to diffuse
uria in presymptomatic patients with familial FSGS. mesangial sclerosis: an autosomal recessive syn-
Pediatr Nephrol. 2007;22:1779–84. drome. J Med Genet. 1994;31:121–5.
38. Coppes M, Huff V, Pelletier J. Denys–Drash syn- 55. Gavin L, McMahon F, Castle I, et al. Alterations in
drome: relating a clinical disorder to genetic alter- serum thyroid hormones an thyroxin-binding globulin
ations in the tumor suppressor gene WT1. J Pediatr. in patients with nephrosis. J Clin Endocrinol Metab.
1993;123:673–8. 1978;46:125–30.
39. Coulthard M. Management of Finnish congenital 56. Gellerman J, Stefabidis C, Mitsioni A, Querfeld
nephrotic syndrome by unilateral nephrectomy. U. Successful treatment of steroid-resistant nephrotic
Pediatr Nephrol. 1989;3:451–3. syndrome associated with WT1 mutations. Pediatr
40. Crecka A, Mundel P. Cell biology and Nephrol. 2010;25:2051–3.
pathology of podocytes. Annu Rev Physiol. 57. Gee H, Saisawat P, Ashraf S, et al. ARHDGDIA
2012;74:299–323. mutations cause nephrotic syndrome via defective
41. Dagan A, Cleper R, Krause I, et al. Hypothyroidism in RHO GTPase signaling. J Clin Invest.
children with steroid-resistant nephrotic syndrome. 2013;123:3243–53.
Nephrol Dial Transplant. 2012;27:2171–5. 58. Gerke P, Huber T, Sellin L, et al. Homodimerization
42. Dámico G, Bazi C. Pathophysiology of proteinuria. and heterodimerization of the glomerular podocyte
Kidney Int. 2003;63:809–25. proteins nephrin and NEPH1. J Am Soc Nephrol.
43. Debiecic H, Guigonis V, Mougenot B, et al. Antenatal 2003;14:918–26.
membranous glomerulonephritis due to anti-neutral 59. Gessler M, Poustka A, Cavenee W,
endopeptidase antibodies. N Engl J Med. et al. Homozygous deletion in Wilms tumours of a
2002;346:2053–60. zinc-finger gene identified by chromosome jumping.
44. Denys P, Malvaux P, Van den Berghe H, et al. De Nature. 1990;343:774–8.
pseudohermaphrodisme masculin, d’une tumeur de 60. Gbadegesin R, Bartkowiak B, Lavin P, et al. Exclu-
Wilms, d’une néphropathie parenchymateuse et sion of homozygous PLCE1 (NPHS3) mutations in
d’une mosaicisme XX/XY. Arch Fr Pediatr. 69 families with idiopathic and hereditary FSGS.
1967;24:729–39. Pediatr Nephrol. 2009;24:281–5.
45. Diomedi-Camassei F, Giandomenico S, Santorelli F, 61. Gbadegesin R, Hinkes B, Hoskins B, et al. Mutations
et al. COQ2 Nephropathy: a newly described in PLCE1 are a major cause of isolated diffuse
inherited mitochondriopathy with primary renal mesangial sclerosis (IDMS). Nephrol Dial Trans-
involvement. J Am Soc Nephrol. 2007;18:2773–80. plant. 2008;23:1291–7.
772 H. Jalanko and C. Holmberg

62. Giglio S, Provenzano A, Mazzinghi B. 79. Hata D, Miyazaki M, Seto S, et al. Nephrotic syn-
Heterogeneous genetic alterations in sporadic drome and aberrant expression of laminin isoforms in
nephrotic syndrome associate with resistance to glomerular basement membranes for infant with
immunosuppression. J Am Soc Nephrol. 2014;10: herlitz junctional epidermolysis bullosa. Pediatrics.
e1004262. 2005;116:e601–7.
63. Gilbert R, Turner C, Gibson J, et al. Mutations in 80. Heidet L, Bongers E, Sich M, et al. In vivo expression
phospholipase C epsilon 1 are not sufficient to cause of putative LMX1B targets in nail-patella syndrome
diffuse mesangial sclerosis. Kidney Int. kidneys. Am J Pathol. 2003;163:145–55.
2009;75:415–9. 81. Heaton PA, Smales O, Wong W. Congenital nephrotic
64. Gilbert R, Wiggelinkhuizen J. The clinical course of syndrome responsive to captopril and indomethacin.
hepatitis B virus-associated nephropathy. Pediatr Arch Dis Child. 1999;81:174–5.
Nephrol. 1994;8:11–4. 82. Heeringa S, Chernin G, Chaki M, et al. COQ6 muta-
65. Goldenberg A, Ngoc L, Thouret M, tions in human patients produce nephrotic syndrome
et al. Respiratory chain deficiency presenting as con- with sensorineural deafness. J Clin Invest.
genital nephrotic syndrome. Pediatr Nephrol. 2011;121:2013–24.
2005;20:465–9. 83. Heeringa S, Vlangos C, Chernin G, et al. Thirteen
66. Grahammer F, Schell C, Huber T. The podocyte slit novel NPHS1 mutations in a large cohort of children
diaphragm – from a thin grey line to complex signal- with congenital nephrotic syndrome. Nephrol Dial
ling hub. Nat Rev Nephrol. 2013;9:587–98. Trasnplant. 2008;23:3527–33.
67. Grech V, Chan M, Vella C, et al. Cardiac 84. Hinkes B. NPHS3: new clues for understanding idio-
malformations associated with congenital nephrotic pathic nephrotic syndrome. Pediatr Nephrol.
syndrome. Pediatr Nephrol. 2000;14:1115–7. 2008;23:847–50.
68. Guez S, Giani M, Melzi M, et al. Adequate clinical 85. Hinkes B, Mucha B, Vlangos C, et al. Nephrotic syn-
control of congenital nephrotic syndrome by enala- drome in the first year of life: two thirds of cases are
pril. Pediatr Nephrol. 1998;12:130–2. caused by mutations in 4 genes (NPHS1, NPHS2,
69. Guo JK, Menke A, Gubler M, et al. WT1 is a key WT1, LAMB2). Pediatrics. 2007;119:e907–19.
regulator of podocyte function: reduced expression 86. Hinkes B, Vlangos C, Heeringa S, et al. Specific
levels cause crescentic glomerulonephritis and podocin mutations correlate with age of onset in
mesangial sclerosis. Hum Mol Genet. 2002;11:651–9. steroid-resistant nephrotic syndrome. J Am Soc
70. Gupta I, Baldwin C, Auguste D, et al. ARHGDIA, Nephrol. 2008;19:365–71.
novel gene implicated in nephrotic syndrome. J Med 87. Hinkes B, Wiggins R, Gbadegesin R, et al. Positional
Genet. 2013;50:330–8. cloning uncovers mutations in PLCE1 responsible for
71. Haavisto A, Jalanko H, Sintonen H, et al. Quality of a nephrotic syndrome variant that may be reversible.
life in adult survivors of pediatric kidney transplanta- Nat Genet. 2006;38:1397–405.
tion. Transplantation. 2011;92:1322–6. 88. Holmberg C, Antikainen M, Ronnholm K,
72. Haavisto A, Korkman M, Holmberg C, et al. Management of congenital nephrotic syndrome
et al. Neuropsychological profile of children with of the Finnish type. Pediatr Nephrol. 1995;9:87–93.
kidney transplants. Nephrol Dial Transplant. 89. Holmberg C, Jalanko H. Congenital nephrotic syn-
2012;27:2594–601. drome and recurrence of proteinuria after renal trans-
73. Habib R. Nephrotic syndrome in the 1st year of life. plantation. Pediatr Nephrol. 2014;29:2309–17.
Pediatr Nephrol. 1993;7:347–53. 90. Holtta T, Happonen J, Ronnholm K, et al.
74. Hahn H, Cho Y, Park Y, et al. Two cases of isolate Hypertension, cardiac state, and the role of volume
diffuse mesangial sclerosis with WT1 mutations. J overload during peritoneal dialysis. Pediatr Nephrol.
Korean Med Sci. 2006;21:160–4. 2001;16:324–31.
75. Hallman N, Hjelt L, Ahvenainen EK. Nephrotic syn- 91. Holtta T, Ronnholm K, Jalanko H, Holmberg C.
drome in newborn and young infants. Ann Paediatr Clinical outcome of pediatric patients on peritoneal
Fenn. 1956;2:227–41. dialysis under adequacy control. Pediatr Nephrol.
76. Hameed R, Raafat F, Ramani P, et al. Mitochondrial 2000;11:88–98.
cytopathy presenting with focal segmental 92. Holzman L, St John P, Kovari I, et al. Nephrin local-
glomerulosclerosis, hypoparathyroidism, sensorineu- izes to the slit pore of the glomerular epithelial cell.
ral deafness, and progressive neurological disease. Kidney Int. 1999;56:1481–91.
Postgrad Med J. 2001;77:523–6. 93. Hu M, Zhang G, Arbuckle S, et al. Prophylactic bilat-
77. Hameed R, Shomaf M. Congenital nephrotic syn- eral nephrectomies in two pediatric patients with mis-
drome: a clinico-pathological study of thirty children. sense mutations in the WT1 gene. Nephrol Dial
J Nephrol. 2001;14:104–9. Transplant. 2004;19:223–6.
78. Hasselbacher K, Wiggins R, Matejas V, 94. Huber T, Harteleben B, Kim J, et al. Nephrin and
et al. Recessive missense mutations in LAMB2 CD2AP associate with phosphoinositide 3-OH kinase
expand the clinical spectrum of LAMB2-associated and stimulate AKT dependent signalling. Mol Cell
disorders. Kidney Int. 2006;70:1008–12. Biol. 2003;23:4917–28.
25 Congenital Nephrotic Syndrome 773

95. Huber T, Kottgen M, Schilling B, et al. Interaction protein – nephrin – is mutated in congenital nephrotic
with podocin facilitates nephrin signaling. J Biol syndrome. Mol Cell. 1998;1:575–82.
Chem. 2001;276:41543–6. 113. Khurana S, Chakraborty S, Lam M, et al. Familial
96. Huttunen N-P. Congenital nephrotic syndrome of focal segmental glomerulosclerosis (FSGS)-linked
Finnish type: study of 75 patients. Arch Dis Child. a-actin 4 (ACTN4) protein mutants lose ability to
1976;51:344–8. activate transcription by nuclear hormone receptors.
97. Inoue T, Yaoita E, Kurihara H, et al. FAT is a compo- J Biol Chem. 2012;287:12027–35.
nent of glomerular slit diaphragms. Kidney Int. 114. Klamt B, Koziell A, Poulat F, et al. Frasier syndrome
2001;59:1003–12. is caused by defective alternative splicing of WT1
98. Ismaili K, Verure V, Vanenhoute K, et al. WT1gene leading to an altered ratio of WT1 +/KTS splice
mutations in three girls with nephrotic syndrome. Eur isoforms. Hum Mol Genet. 1998;7:709–14.
J Pediatr. 2008;167:579–81. 115. Kopp J, Smith M, Nelson G, et al. MYH9 is a major-
99. Ito S, Takata A, Hataya H, et al. Isolated diffuse effect risk gene for focal segmental glomerulo-
mesangial sclerosis and Wilms’ tumor suppressor sclerosis. Nat Genet. 2008;40:1175–84.
gene. J Pediatr. 2001;138:425–8. 116. Kovacevic L, Reid C, Ridgen S. Management of
100. Jalanko H. Congenital nephrotic syndrome. Pediatr congenital nephrotic syndrome. Pediatr Nephrol.
Nephrol. 2009;24:2121–8. 2003;18:426–30.
101. Jalanko H, Holmberg C. Renal transplantation in 117. Koziell A, Grech V, Hussain S, et al. Genotype/phe-
infants. Pediatr Nephrol. 2014 (in press) notype correlations of NPHS1 and NPHS2 mutations
102. Jalanko H, Patrakka J, Tryggvason K, et al. Genetic in nephrotic syndrome advocate a functional inter-
kidney diseases disclose the pathogenesis of protein- relationship in glomerular filtration. Hum Mol
uria. Ann Med. 2001;33:526–33. Genet. 2002;11:379–88.
103. Jarad G, Cunningham J, Shaw A, Miner J. Proteinuria 118. Krishnamurthy S, Rajesh N, Ramesh A, Zenker M.
precedes podocyte abnormalities in Lamb2 mice, Infantile nephrotic syndrome with microcephaly and
implicating the glomerular basement membrane global developmental delay: the Galloway Mowat
as an albumin barrier. J Clin Invest. syndrome. Indian J Pediatr. 2012;79:1087–90.
2006;116:2272–9. 119. Kuusniemi AM, Kestilä M, Patrakka J, et al. Tissue
104. Jeanpierre C, Denamur E, Henry I, et al. Identification expression of nephrin in man and pig. Pediatr Res.
of constitutional WT1 mutations, in patients with iso- 2004;55:774–81.
lated diffuse mesangial sclerosis, and analysis of 120. Kuusniemi A, Lapatto R, Holmberg C, et al. Kidneys
genotype/phenotype correlations by use of computer- with heavy proteinuria show fibrosis, inflammation,
ized mutation database. Am J Hum Genet. and oxidative stress, but no tubular phenotypic
1998;62:824–33. change. Kidney Int. 2005;68:121–32.
105. Jefferson J, Shankland S. Familial nephrotic syn- 121. Kuusniemi A-M, Qvist E, Sun Y, et al. Plasma
drome: PLCE1 enters the fray. Nephrol Dial Trans- exchange and retransplantation in recurrent nephrosis
plant. 2007;22:1849–52. of patients with congenital nephrotic syndrome of the
106. Jones N, Blasutig I, Eremina V, et al. Nck adaptor Finnish type. Transplantation. 2007;83:1316–23.
proteins link nephrin to the actin cytoskeleton of 122. Kuusniemi A, Merenmies J, Lahdenkari AT, et al.
kidney podocytes. Nature. 2006;440:818–23. Glomerular sclerosis in kidneys with congenital
107. Kagan M, Cohen A, Matejas V, et al. A milder variant nephrotic syndrome (NPHS1). Kidney Int.
of Pierson syndrome. Pediatr Nephrol. 2006;70:1423–31.
2008;23:323–7. 123. Laakkonen H, Hölttä T, Lönnqvist T, et al. Peritoneal
108. Kaplan JM, Kim SH, North K, et al. Mutations in dialysis in children under two years of age. Nephrol
ACTN4, encoding α-actinin-4 cause familial-focal Dial Transplant. 2008;23:1747–53.
segmental glomerulosclerosis. Nat Genet. 124. Lahdenkari AT, Lounatmaa K, Patrakka J,
2000;24:251–6. et al. Podocytes are firmly attached to glomerular
109. Karle S, Uetz B, Ronner V, et al. Novel mutations in basement membrane in kidneys with heavy protein-
NPHS2 detected in both familial and sporadic steroid- uria. J Am Soc Nephrol. 2004;15:2611–8.
resistant nephrotic syndrome. J Am Soc Nephrol. 125. Lehtonen S. Connecting the interpodocyte slit dia-
2002;13:388–93. phragm and actin dynamics: emerging role for the
110. Kaukinen A, Kuusniemi AM, Lautenschaleger I, nephrin signalling complex. Kidney Int.
et al. Glomerular endothelium in kidneys with con- 2008;73:903–5.
genital nephrotic syndrome (NPHS1). Nephrol Dial 126. Lennert T, Härtling A, Mildenberger E,
Transplant. 2008;23:1224–32. et al. Augenveränderung be diffuser mesangialer
111. Kawachi H, Miyauchi N, Suzuki K, et al. Role of Sklerose (DMS). Monatschr Kinederheilkd.
podocyte slit diaphragm as a filtration barrier. 1997;145:209–12.
Nephrology. 2006;11:274–81. 127. Llach F, Papper S, Massry S. The clinical spectrum
112. Kestilä M, Lenkkeri U, Männikkö M, of renal vein thrombosis. Am J Med.
et al. Positionally cloned gene for a novel glomerular 1980;69:819–27.
774 H. Jalanko and C. Holmberg

128. Little M, Wells C. A clinical overview of WT1 gene 144. Moeller M, Kovari I, Holzman L. Evaluation of a new
mutations. Hum Mutat. 1997;9:209–25. tool for exploring podocyte biology: mouse NPHS1
129. Litwin M, Grena R, Slaowska J, et al. Add-on therapy 5-flanking region drives LacZ expression in
with angiotensin II receptor 1 blocker in children with podocytes. J Am Soc Nephrol. 2000;11:2306–14.
chronic kidney disease already treated with 145. Mrowka C, Schedl A. Wilms’ tumor suppressor gene
angiotensin-converting enzyme inhibitors. Pediatr WT1: from structure to renal pathophysiologic fea-
Nephrol. 2006;21:1716–22. tures. J Am Soc Nephrol. 2000;11:S106–15.
130. Liu L, Done S, Khoshnoodi J, et al. Defective nephrin 146. Natoli T, Liu J, Eremina V, et al. A mutant form of the
trafficking caused by missense mutations in the Wilms’ tumor suppressor gene WT1 observed in
NPHS1 gene: insight into the mechanisms of congen- Denys-Drash syndrome interferes with glomerular
ital nephrotic syndrome. Hum Mol Genet. capillary development. J Am Soc Nephrol.
2001;10:2637–44. 2002;13:2058–67.
131. Ljungberg P, Holmberg C, Jalanko H. Infections in 147. Niaudet P. Genetic forms of nephrotic syndrome.
infants with congenital nephrosis of the Finnish type. Pediatr Nephrol. 2004;19:1313–8.
Pediatr Nephrol. 1997;11:148–52. 148. Niaudet P, Gubler M-C. WT1 and glomerular dis-
132. Lovric S, Fang H, Vega-Warner V, et al. Rapid detec- eases. Pediatr Nephrol. 2006;21:1653–60.
tion of monogenic causes of childhood-onset steroid 149. Nicolaou N, Margadant C, Kevelam S, et al. Gain of
resistant nephrotic syndrome. Clin J Am Soc Nephrol. glycosylation in integrin α3 causes lung disease and
2014;9:1109–16. nephrotic syndrome. J Clin Invest.
133. Machuca E, Benoit G, Nevo F, et al. Genotype- 2012;122:4375–87.
Phenotype correlations in non-Finnish congenital 150. Nielsen K, Steffensen G. Congenital nephrotic syn-
nephrotic syndrome. J Am Soc Nephrol. drome associated with Lowe’s syndrome. Child
2010;21:1209–17. Nephrol Urol. 1990;10:92–5.
134. Malina M, Cinek O, Janda J, Seeman T. Partial remis- 151. Niemsiri S. Congenital syphilitic nephrosis. South-
sion with cyclosporine A in a patient with nephrotic east Asian J Trop Med Public Health.
syndrome due to NPHS2 mutations. Pediatr Nephrol. 1993;24:595–600.
2009;24:2051–3. 152. Nishibori Y, Liu L, Hosoyamada M, et al. Disease-
135. Martin-Hernandez E, Garcia-Silva T, Vara J, causing missense mutations in NPHS2 gene alter
et al. Renal pathology in children with mitochondrial normal nephrin trafficking to the plasma membrane.
diseases. Pediatr Nephrol. 2005;20:1299–305. Kidney Int. 2004;66:1755–65.
136. Matejas V, Hinkes B, Alkandari F, et al. Mutations in 153. Nishikawa M, Ichiyama T, Hayashi T, et al. A case of
the human laminin β2 (LAMB2) gene and the early myoclonic encephalopathy with the congenital
associated phenotype. Hum Mutat. nephrotic syndrome. Brain Dev. 1997;19:144–7.
2010;31:992–1002. 154. Norio R. Heredity in the congenital nephrotic syn-
137. Matejas V, Al-Gazali L, Amirlak I, Zenker M. A drome: a genetic study of 57 Finnish families with a
syndrome comprising childhood-onset glomerular review of reported cases. Ann Paediatr Fenn.
kidney disease and ocular abnormalities with progres- 1966;12:27–32.
sive loss of vision is caused but mutated LAMB2. 155. Nortier J, Debiec H, Tournay Y, et al. Neonatal dis-
Nephrol Dial Transplant. 2006;21:3283–6. ease in neutral endopeptidase alloimmunization: les-
138. Mathias R, Stecklein H, Guay-Woodford L, sons for immunological monitoring. Pediatr Nephrol.
et al. Gamma globulin deficiency in newborns with 2006;21:1399–405.
congenital nephrotic syndrome. N Engl J Med. 156. Ovunc B, Ashraf S, Vega-Warner V, et al. Mutation
1989;320:398–9. analysis of NPHS1 in a worldwide cohort of congen-
139. Mattoo T, Al-Sowailem A, Al-Harbi M. Nephrotic ital nephrotic syndrome patients. Nephron Clin Pract.
syndrome in the 1st year of life and the role of unilat- 2012;120:139–46.
eral nephrectomy. Pediatr Nephrol. 1992;6:16–8. 157. Ozcakar Z, Cenzig F, Cakar N, et al. Analysis of
140. McCarthy H, Bierzynska A, Wherlock M, NPHS2 mutations in Turkish steroid-resistant
et al. Simultaneous sequencing of 24 genes associated nephrotic syndrome patients. Pediatr Nephrol.
with steroid resistant nephrotic syndrome. Clin J Am 2006;21:1093–6.
Soc Nephrol. 2013;8:637–48. 158. Pannicucci F, Sagripanti A, Vispi M,
141. McLean R, Kennedy T, Rosoulpour M, et al. Comprehensive study of hemostasis in
et al. Hypothyroidism in the congenital nephrotic congenital nephrotic syndrome. Nephron.
syndrome. J Pediatr. 1982;101:72–5. 1983;33:9–13.
142. Mele C, Iatropulos P, Donadelli R, et al. MYO1E 159. Patrakka J, Kestilä M, Wartiovaara J, et al. Congenital
mutations and childhood familial focal segmental nephrotic syndrome (NPHS1): features resulting from
glomerulosclerosis. N Eng J med. 2011;365:295–306. different mutations in Finnish patients. Kidney Int.
143. Meyers K, Kaplan P, Kaplan B. Nephrotic syndrome, 2000;58:972–80.
microcephaly, and developmental delay: three sepa- 160. Patrakka J, Martin P, Salonen R, et al. Proteinuria and
rate syndromes. Am J Med Genet. 1999;82:257–60. prenatal diagnosis of congenital nephrosis in fetal
25 Congenital Nephrotic Syndrome 775

carriers of nephrin gene mutations. Lancet. standard steroid treatment of nephrotic syndrome.
2002;359:1575–7. J Am Soc Nephrol. 2004;15:722–32.
161. Patrakka J, Ruotsalainen V, Reponen V, 177. Ruf R, Schultheiss M, Lichtenberger A,
et al. Recurrence of nephrotic syndrome in kidney et al. Prevalence of WT1 mutations in a large cohort
grafts of patients with congenital nephrotic syndrome of patients with steroid-resistant and steroid-sensitive
of the Finnish type: role of nephrin. Transplantation. nephrotic syndrome. Kidney Int. 2004;66:564–70.
2002;73:394–403. 178. Sako M, Nakanishi K, Obana M, et al. Analysis of
162. Pelletier J, Bruening W, Kashtan C, et al. Germline NPHS1, NPHS2, ACTN4, and WT1 in Japanese
mutations in the Wilms’ tumor suppressor gene are patients with congenital nephrotic syndrome. Kidney
associated with abnormal urogenital development in Int. 2005;67:1248–55.
Denys-Drash syndrome. Cell. 1991;67:437–47. 179. Salvatierra O, Sarwal M. Renal perfusion in infant
163. Philippe A, Nevo F, Esquivel E, et al. Nephrin muta- recipients of adult sized kidneys is critical risk factor.
tions can cause childhood-onset steroid resistant Transplantation. 2000;70:412–3.
nephrotic syndrome. J Am Soc Nephrol. 180. Salviati L, Sacconi S, Murer L, et al. Infantile
2008;19:1871–8. encephalomyopathy an nephropathy with CoQ10
164. Pomeranz A, Wolach B, Bernheim J, Korzets efficiency: A CoQ10-responsive condition. Neurol-
Z. Successful treatment of Finnish type congenital ogy. 2005;65:606–8.
nephrotic syndrome with captopril and indomethacin. 181. Sanna-Cherchi S, Burgess K, Nees S, et al. Exome
J Pediatr. 1995;126:140–2. sequencing identified MYO1E and NEIL1 as candi-
165. Putaala H, Soininen R, Kilpelainen P, et al. The murin date genes for human autosomal recessive steroid-
nephrin gene is specifically expressed in kidney, brain resistant nephrotic syndrome. Kidney Int.
and pancreas: inactivation of the gene leads to mas- 2011;80:389–96.
sive proteinuria and neonatal death. Hum Mol Genet. 182. Sano H, Miyanoshita A, Watanabe N,
2001;10:1–8. et al. Microcephaly an early-onset nephrotic syn-
166. Qvist E, Laine J, Rönnholm K, et al. Graft function drome: a case report. Pediatr Nephrol. 1995;9:711–4.
5–7 years after renal transplantation in early child- 183. Santini S, Bullich G, Tazon-Vega B, et al. Clinical
hood. Transplantation. 1999;67:1043–9. utility of genetic testing in children and adults with
167. Qvist E, Krogerus L, Rönnholm K, et al. Course of steroid-resistant nephrotic syndrome. Clin J Am Soc
renal allograft histopathology after transplantation in Nephrol. 2011;6:1139–48.
early childhood. Transplantation. 2000;70:480–7. 184. Santini S, Garcia-Maset R, Ruiz P, et al. Nephrin
168. Qvist E, Marttinen E, Rönnholm K, et al. Growth after mutations cause childhood- and adult-onset focal seg-
renal transplantation in infancy or early childhood. mental glomerulosclerosis. Kidney Int.
Pediatr Nephrol. 2002;17:438–43. 2009;76:1268–76.
169. Qvist E, Pihko H, Fagerudd P, 185. Savage J, Jefferson J, Maxwell A, et al. Improved
et al. Neurodevelopmental outcome in high-risk prognosis for congenital nephrotic syndrome of the
patients after renal transplantation in early childhood. Finnish type in Irish families. Arch Dis Child.
Pediatr Transplant. 2002;6:53–62. 1999;80:466–9.
170. Rahman H, Begum A, Jahan S, et al. Congenital 186. Scaleis E, Chafai R, van Coster R, et al. Early myo-
nephrotic syndrome, an uncommon presentation of clonic epilepsy, hypertrophic cardiomyopathy and
cytomegalovirus infection. Mymesingh Med subsequently a nephrotic syndrome in a patient with
J. 2008;17:210–3. CoQ10 deficiency caused by mutations in para-
171. Ramsuran D, Bhimma R, Randial P, et al. The spec- hydroxybenzoate-polyprenyl transferase (COQ2).
trum of HIV-related nephropathy in children. Pediatr Eur J Pediatr. 2013;17:625–30.
Nephrol. 2012;27:821–7. 187. Schoeb D, Cernin G, Heeringa S, et al. Nineteen novel
172. Rapola J, Savilahti E. Immunofluorescent and mor- NPHS1 mutations in a worldwide cohort of patients
phological studies in congenital nephrotic syndrome. with congenital nephrotic syndrome (CNS). Nephrol
Acta Paediatr. 1971;60:253–63. Dial Transplant. 2010;25:2970–7.
173. Roselli S, Gribouval O, Boute G, et al. Podocin local- 188. Schultheiss M, Ruf R, Mucha B, et al. No evidence
izes in the kidney to the slit diaphragm area. Am J for genotype/phenotype correlation in NPHS1
Pathol. 2002;160:3–5. and NPH2 mutations. Pediatr Nephrol.
174. Ruotsalainen V, Ljungberg P, Wartiovaara J, 2004;19:1340–8.
et al. Nephrin is specifically located at the slit dia- 189. Schumacher V, Scharer K, Wuhl E, et al. Spectrum
phragm of glomerular podocytes. Proc Natl Acad Sci of early onset nephrotic syndrome associated with
U S A. 1999;96:7962–7. WT1 missense mutations. Kidney Int.
175. Ruotsalainen V, Patrakka J, Tissari P, et al. Role of 1998;53:1594–600.
nephrin in cell junction formation in human 190. Schwarz K, Simons M, Reiser J, et al. Podocin, a raft-
nephrogenesis. Am J Pathol. 2000;157:1905–16. associated component of the glomerular slit dia-
176. Ruf R, Lichtenberger A, Karle S, et al. Patients with phragm, interacts with CD2AP and nephrin. J Clin
mutations in NPHS2 (podocin) do not respond to Invest. 2001;108:1621–9.
776 H. Jalanko and C. Holmberg

191. Sellin L, Huber T, Gerke P, et al. NEPH1 defines a and infantile nephrotic syndrome in Thai infants.
novel family of podocin interacting proteins. FASEB Clin Pediatr. 2005;44:169–74.
J. 2003;17:115–7. 205. Van DeVoorde R, Witte D, Kogan J, Goebel J. Pierson
192. Seri M, Pecci A, Di Bari F, et al. MYH9-related syndrome: a novel cause of congenital nephrotic syn-
disease: May-Hegglin anomaly, Sebastian syndrome, drome. Pediatrics. 2006;118:e501–5.
Fechtner syndrome, and Epstein syndrome are not 206. Wartiovaara J, Ofverstedt LG, Khoshnoodi J,
distinct entities but represent a variable expression et al. Nephrin strands contribute to a porous slit dia-
of a single illness. Medicine. 2003;82:203–15. phragm scaffold as revealed by electron tomography.
193. Shahin B, Papaopoulous Z, Jenis E. Congenital J Clin Invest. 2004;114:1475–83.
nephrotic syndrome associate with congenital toxo- 207. Winn M, Conlon P, Lynn K, et al. A mutation in the
plasmosis. J Pediatr. 1997;85:366–72. TRPC6 cation channel causes familial focal segmen-
194. Shih N, Karpitskii V, Nguyen A, et al. Congenital tal glomerulosclerosis. Science. 2005;308:1801–4.
nephrotic syndrome in mice lacking CD2-associated 208. Wolf G, Stahl R. CD2-associated protein and glomer-
protein. Science. 1999;286:312–5. ular disease. Lancet. 2003;362:1746–8.
195. Sinha M, Horsfield C, Komaromy B, et al. Congenital 209. Wong W, Morris M, Kara T. Congenital nephrotic
disorders of glycosylation: a rare cause of nephrotic syndrome with prolonged renal survival without
syndrome. Nephrol Dial Transplant. 2009;24:2591–4. renal replacement therapy. Pediatr Nephrol.
196. Sperl W, Gruber W, Quatacker J, et al. Nephrosis in 2013;28:2313–21.
two siblings with infantile sialic acid storage disease. 210. Wuhl E, Kogan J, Zurowska A,
Eur J Pediatr. 1990;149:477–82. et al. Neurodevelopmental deficits in Pierson
197. Srivastava T, Whiting J, Garola R, et al. Podocyte (microcoria-congenital nephrosis) syndrome. Am J
proteins in Galloway-Mowat syndrome. Pediatr Med Genet. 2007;143:311–9.
Nephrol. 2001;16:1022–9. 211. Yang Y, Jeanpirre C, Dressler G, et al. WT1 and
198. Stoll R, Lee B, Debler E, et al. Structure of the Wilms PAX-2 podocyte expression in Denys-Drash syn-
tumor suppressor protein zinc finger domain bound to drome an isolated diffuse mesangial sclerosis. Am J
DNA. J Mol Biol. 2007;372:1227–45. Pathol. 1999;154:181–92.
199. Tainio J, Jahnukainen K, Nurmio M et al. Testicular 212. Yao J, Le T, Kos C, et al. α-actinin-4-mediated FSGS:
function, semen quality, and fertility in young men an inherited kidney disease caused by an aggregated
after renal transplantation during childhood and ado- and rapidly degraded cytoskeletal protein. PLoS Biol.
lescence. Transplantation 2014;98:987–93. 2004;2:787–94.
200. Tainio J, Qvist E, Vehmas R, et al. Pubertal develop- 213. Xiao H, Liu J, Zhong X. Congenital syphilis
ment is normal an adolescents after renal transplanta- presenting congenital nephrotic syndrome in two chil-
tion in childhood. Transplantation. 2011;92:404–9. dren and related data review. Beijing Da Xue Xue
201. Tory K, Menyhard D, Woerner S, et al. Mutation- Bao. 2011;43:911–3.
dependent recessive inheritance of NPHS2-associated 214. Zenker M, Aigner T, Tralau T, et al. Human laminin
steroid-resistant nephrotric syndrome. Nat Genet. b2 deficiency causes congenital nephrosis with
2014;46:299–304. mesangial sclerosis and distinct eye abnormalities.
202. Toubiana J, Schlageter M, Aoun B, et al. Therapy- Hum Mol Genet. 2004;13:2625–32.
resistant anaemia in congenital nephrotic syndrome of 215. Zenker M, Tralau T, Lennert T, et al. Congenital
the Finnish type-implication of EPO, transferrin and nephrosis, mesangial sclerosis, and distinct eye
transcobalamin losses. Nephrol Dial Transplant. abnormalities with microcoria: an autosomal reces-
2009;23:1338–40. sive syndrome. Am J Med Genet. 2004;130:138–45.
203. Tryggvason K, Patrakka J, Wartiovaara J. Hereditary 216. Zhang A, Huang S. Progress in pathogenesis of pro-
proteinuria syndromes and mechanisms of protein- teinuria. Int J Nephrol 2012;ID314251.
uria. N Engl J Med. 2006;354:1387–401. 217. Zhu J, Sun N, Aoujit L, et al. Nephrin mediates actin
204. Vachvanishsanong P, Mitarnum W, reorganization via phosphoinositide 3-kinase in
Tungsinmunkong K, Disseneewate P. Congenital podocytes. Kidney Int. 2008;73:556–66.
Inherited Glomerular Diseases
26
Michelle N. Rheault and Clifford E. Kashtan

Contents Hereditary Metabolic Disorders


with Secondary Glomerular Involvement . . . . . . . . 792
The Glomerular Basement Membrane . . . . . . . . . . . 778 Familial Amyloidosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 792
Glomerular Basement Membrane Disorders . . . . . 779 Alpha-1 Antitrypsin Deficiency . . . . . . . . . . . . . . . . . . . . . 792
Alagille Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 793
Type IV Collagen Disorders . . . . . . . . . . . . . . . . . . . . . . . 779 Hereditary Lecithin-Cholesterol Acyltransferase
Alport Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 779 (LCAT) Deficiency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 793
Thin Basement Membrane Nephropathy . . . . . . . . . . . . 786 Lipoprotein Glomerulopathy . . . . . . . . . . . . . . . . . . . . . . . . 793
Hereditary Angiopathy with Nephropathy, Familial Juvenile Megaloblastic Anemia . . . . . . . . . . . . 794
Aneurysms, and Cramps (HANAC Syndrome) . . . . . 788
Other Hereditary Diseases with Glomerular
Laminin Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 788 Involvement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 794
Pierson Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 788 Charcot-Marie-Tooth (CMT) Disease . . . . . . . . . . . . . . . 794
Cockayne Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 794
Type III Collagen Nephropathies . . . . . . . . . . . . . . . . . 789
Hereditary Acro-osteolysis with Nephropathy . . . . . . 794
Nail-Patella Syndrome (Hereditary Osteo-
Other Syndromes with Renal Involvement . . . . . . . . . . 794
onychodysplasia) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 789
Collagen Type III Glomerulopathy Hereditary Glomerulopathies Without
(Collagenofibrotic Glomerulopathy) . . . . . . . . . . . . . . . . 791 Extrarenal Symptoms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
Fibronectin Glomerulopathy . . . . . . . . . . . . . . . . . . . . . . . . . 795
Hereditary Nephritis with Thrombocytopenia
Mitochondrial Cytopathies . . . . . . . . . . . . . . . . . . . . . . . . . . 795
and Giant Platelets: Epstein and Fechtner
Syndromes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 791 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
Hereditary Metabolic Disorders
with Primary Glomerular Involvement . . . . . . . . . . . 791
Fabry Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 791
Other Glomerular Lipidoses . . . . . . . . . . . . . . . . . . . . . . . . . 792

M.N. Rheault (*) • C.E. Kashtan


Department of Pediatrics, Division of Pediatric
Nephrology, University of Minnesota Masonic Children’s
Hospital, Minneapolis, MN, USA
e-mail: rheau002@umn.edu; kasht001@umn.edu

# Springer-Verlag Berlin Heidelberg 2016 777


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_79
778 M.N. Rheault and C.E. Kashtan

The Glomerular Basement Membrane While the interstitial collagens, such as type I
collagen, lose their NC1 domains after chain
The structural and functional characteristics of association and form fibrillar networks, type IV
the normal glomerular capillary wall are discussed collagen trimers form open, nonfibrillar networks
in detail elsewhere in this text. For the through NC1-NC1 interactions between two tri-
purposes of this chapter, this discussion will mers and amino-terminal interactions between
focus on the major protein components of four trimers.
glomerular basement membrane (GBM): type IV α1α1α2(IV) trimers are present in all basement
collagen and laminin. The GBM contains membranes, whereas α3α4α5(IV) and α5α5α6
specialized isoforms of these proteins which inter- (IV) trimers are more restricted in their distribu-
act and form networks bridged by entactin/ tions (Fig. 1). In normal, mature human kidneys,
nidogen and heparan sulfate proteoglycans α3α4α5(IV) trimers are found in GBM, Bowman’s
(agrin) [1–4]. capsules, and the basement membranes of distal
Type IV collagen. There are six isoforms of tubules. α5α5α6(IV) trimers are present in
type IV collagen, designated α1(IV)–α6(IV). All Bowman’s capsules and the basement membranes
type IV collagen isoforms share several basic of distal tubules and collecting ducts of
structural features: a collagenous domain of normal kidneys but are not found in GBM [6, 7].
1,400 amino acids containing the repetitive trip- α5α5α6(IV) trimers are also found in normal
let sequence glycine-X-Y (Gly-X-Y, with X and Y epidermal basement membranes; however,
representing other amino acids); a α3α4α5(IV) trimers are not. α3α4α5(IV) trimers
noncollagenous carboxy-terminal (NC1) domain are also found in several ocular and cochlear base-
of 230 amino acids that includes 12 completely ment membranes, as discussed in the section on
conserved cysteine residues; and a “Alport Syndrome” below.
noncollagenous amino-terminal sequence of Each type IV collagen α chain is encoded by
15–20 residues (7S). The collagenous triplet one of six distinct genes, COL4A1–COL4A6,
sequence in each chain contains 20 interruptions. which are arranged in pairs on three chromosomes
The secreted form of type IV collagen is a (Fig. 2). COL4A1 and COL4A2 encode the α1
heterotrimer composed of three α chains, resulting (IV) and α2(IV) chains, respectively, and are
from self-association of NC1 domains and folding located on chromosome 1. The α3(IV) and α4
of the collagenous domains into triple helical struc- (IV) chains are respectively encoded by the
tures. Amino acid sequences within the NC1 COL4A3 and COL4A4 genes on chromosome
domains determine the specificity of chain associ- 2, while the α5(IV) and α6(IV) genes are encoded
ation, resulting in three major trimeric species: by the COL4A5 and COL4A6 genes on the X
α1α1α2(IV), α3α4α5(IV), and α5α5α6(IV) [5]. chromosome. The paired genes are arranged in a

Fig. 1 Immunofluorescence microscopy. Normal distribution of the α(IV) chains in renal (a) and epidermal basement
membranes (b)
26 Inherited Glomerular Diseases 779

3′ 5′ 5′ 3′
13q34
130 bp COL4A1
COL4A2 5′
3′ 5′ 3′
2q35
COL4A4 5′ COL4A3
3′ 5′ 3′

Xq22
450 bp
COL4A6 COL4A5

Fig. 2 Schematic representation of the distribution of type IV collagen gene on chromosomes 13, 2, and X

50 –50 orientation, separated by sequences of vary- Table 1 Type IV collagen disorders


ing length that contain regulatory elements [8, 9]. Gene
Laminin. Laminin also forms networks based Disorder Inheritance Locus product
on heterotrimeric association of α, β, and γ Alport X-linked COL4A5 α5(IV)
isoforms. Each subunit consists of an α, β, and γ syndrome Autosomal COL4A3 or α3(IV) or
recessive COL4A4 α4(IV)
chain. There are 12 known laminin chains (α1–5,
Autosomal COL4A3 or α3(IV) or
β1–4, and γ1–3), each encoded by a distinct gene, dominant COL4A4 α4(IV)
that form at least 15 heterotrimeric isomers. Thin basement Autosomal COL4A3 or α3(IV) or
Laminin-521 has the composition α5β2γ1 and is membrane dominant COL4A4 α4(IV)
the laminin form found in mature GBM [4]. nephropathy
HANAC Autosomal COL4A1 α1(IV)
syndrome dominant

Glomerular Basement Membrane


Disorders

To date, mutations affecting two of the major pro-


teins of GBM, type IV collagen and laminin, have development of proteinuria and renal insuffi-
been identified as causes of inherited glomerular ciency in affected individuals, particularly males
disease. Mutations in the α3(IV), α4(IV), or α5 [11]. In 1927 Alport reported the association of
(IV) isoforms of type IV collagen cause Alport nephritis with deafness in this family [12]. Numer-
syndrome and some cases of thin basement mem- ous descriptions of families with hereditary
brane nephropathy (TBMN), whereas mutations nephritis were published in the 1950s and 1960s,
in the α1(IV) isoform cause HANAC syndrome, leading ultimately to the observations that
comprising hereditary angiopathy, nephropathy, established Alport syndrome as a heritable disor-
aneurysms, and muscle cramps (Table 1). Muta- der of type IV collagen. These seminal events
tions in laminin β2 have been implicated in included the identification of unique ultrastruc-
Pierson syndrome. tural alterations in Alport glomerular basement
membranes by electron microscopy
(EM) [13–15], the observation that Alport GBM
Type IV Collagen Disorders exhibited abnormal reactivity with anti-GBM sera
directed against antigens associated with type IV
Alport Syndrome collagen [16–18], the mapping of the major
Alport locus to the X chromosome [19], the dis-
Familial hematuria was first described as a clinical covery of a type IV collagen gene (COL4A5) on
entity in the early 1900s [10]. Over the next the X chromosome [20], and finally the descrip-
20 years, studies of this condition in successive tion of COL4A5 mutations in families with
generations of a single family described X-linked Alport syndrome [21].
780 M.N. Rheault and C.E. Kashtan

Genetics reasons that are as yet uncertain, some people


Mutations in any of the COL4A3, COL4A4, or with heterozygous mutations in COL4A3 or
COL4A5 genes may adversely affect the forma- COL4A4 have a progressive course leading to
tion and composition of affected basement mem- chronic renal failure or end-stage renal disease
branes. If any of the α3(IV), α4(IV), or α5 and are thus considered to have ADAS.
(IV) chains are absent due to severe mutations, Over 700 different mutations in the COL4A5
then the other type IV collagen isoforms are gene have been identified in patients and families
degraded and no α3α4α5(IV) heterotrimers are with XLAS [28]. Mutations can be found along
deposited in basement membranes [22]. Milder the entire 51 exons of the gene without identified
mutations, generally missense mutations affecting hot spots. About 10–15 % of COL4A5 mutations
the glycine residues involved in triple helix for- occur as spontaneous events; therefore, a family
mation, may lead to the formation of abnormally history of renal disease is not required for a diag-
folded trimers that are either degraded or lead to nosis of XLAS. Reported mutations include large
formation of an abnormal type IV collagen rearrangements ( 20 %), small deletions and
network. insertions ( 20 %), missense mutations that alter
There are three genetic forms of Alport syn- a glycine residue in the collagenous domain of the
drome: X-linked (XLAS), autosomal recessive α5(IV) chains ( 30 %), other missense mutations
(ARAS), and autosomal dominant (ADAS). ( 8 %), nonsense mutations ( 5 %), and splice-
XLAS is caused by mutations in the COL4A5 site mutations ( 15 %) [28–31]. The COL4A5
gene and is the predominant form of the disease, genotype has a powerful effect on the phenotype
accounting for ~80 % of affected patients. of XLAS in affected males [30, 31]. Large dele-
Affected males are hemizygotes carrying a single tions, nonsense mutations and small mutations
mutant COL4A5 allele. Affected females carry a that alter the translational reading frame are asso-
normal COL4A5 allele as well as a mutant allele ciated with a 90 % probability of progression to
and are therefore heterozygotes. Affected females ESRD by age 30 [30, 31]. This risk is 70 % in
are mosaic for expression of the normal and patients with a splice-site mutation and 50 % in
mutant alleles due to X-inactivation [23]. Approx- those with a missense mutation [30, 31]. The posi-
imately 15 % of patients with Alport syndrome tion of a glycine substitution may also affect the
have the recessive form of the disease (ARAS) XLAS phenotype, with 50 glycine missense muta-
due to mutations in both alleles of either COL4A3 tions causing a more severe phenotype than 30
or COL4A4. These patients are either homozy- glycine mutations [31]. The number of side-
gotes, who have the identical mutation in both chain carbon atoms in the substituting amino
alleles of the affected gene (and who may have acid also influence the phenotype associated
consanguineous parents) or compound heterozy- with a glycine substitution [32].
gotes, who have inherited different mutations in A similar variety of mutation types has been
the affected gene from their parents [24]. ADAS described in patients and families with ARAS
caused by heterozygous mutations in COL4A3 or [24, 29, 33–35]. There are also genotype-
COL4A4 was classically thought to be rare, affect- phenotype correlations in patients with ARAS
ing only about 5 % of affected patients [25]. With where nonsense mutations or mutations resulting
the advent of next-generation sequencing, how- in stop codons are associated with early onset renal
ever, recent studies are suggesting a higher per- failure [24].
centage of patients with AS who demonstrate
autosomal dominant inheritance than was previ- Clinical Features
ously recognized [26, 27]. Most individuals with Gender and genotype are the major determinants
heterozygous COL4A3 or COL4A4 mutations are of the severity of renal, cochlear, and ocular dis-
asymptomatic or exhibit isolated, nonprogressive ease in Alport syndrome. Males with XLAS and
microscopic hematuria associated with thin glo- patients of either gender with ARAS inevitably
merular basement membranes (TBMN). For progress to end-stage renal disease (ESRD) and
26 Inherited Glomerular Diseases 781

the majority develop sensorineural deafness asymptomatic hematuria while others develop
[24, 30]. The pacing of these events is influenced ESRD [42]. According to one study of a large
by the nature of the underlying disease mutation. cohort of XLAS females, the risk of ESRD is
While the majority of women with heterozygous 12 % by age 45, 30 % by age 60, and 40 % by
COL4A5 mutations have mild disease manifesta- age 80 [36]. The explanation for the wide vari-
tions, ESRD and severe deafness develop in a ability in outcomes for women with XLAS is
significant minority [36]. Patients with ADAS unclear, but likely multifactorial. Unlike in males
exhibit relatively slow progression of renal and with XLAS, genotype does not correlate with
cochlear dysfunction, and ocular findings are phenotype in females with XLAS, perhaps as a
much less common than in XLAS and ARAS result of the overwhelming influence of random
[26, 37, 38]. X-chromosome inactivation on disease course in
Renal. Persistent microscopic hematuria is the XLAS females [23, 36]. Risk factors for ESRD in
cardinal clinical feature of Alport syndrome, women with XLAS include proteinuria and sen-
occurring in 100 % of males with XLAS, 95 % sorineural deafness [36, 43]. Further studies are
of females with XLAS, and in all patients with required to develop accurate methods to predict
ARAS [30, 36]. Hematuria is likely present from the risk of progressive kidney disease in women
infancy in XLAS males and in patients with with XLAS.
ARAS. Episodic gross hematuria is not unusual, Cochlear. Hearing is normal at birth and dur-
especially during childhood [39]. Some children ing early childhood. Symmetrical deficits in sen-
with Alport syndrome have virtually constant sitivity for high-frequency sounds often become
gross hematuria. detectable by audiometry in late childhood. Over
Overt proteinuria typically appears during later time, the hearing deficit progresses into the fre-
childhood or adolescence in XLAS males and in quency range of conversational speech. Because
ARAS patients. Affected children first demon- the deficit typically does not exceed 60–70 dB and
strate microalbuminuria, which progresses to speech discrimination is preserved, hearing aids
overt proteinuria and even nephrotic syndrome are effective in most affected individuals. In
with time [39–41]. About 75 % of XLAS females XLAS males, the probability of hearing loss is
ultimately develop proteinuria of some degree 50 % by age 15, 75 % by age 25, and 90 % by
[36]. Most children with Alport syndrome have age 40 [30]. COL4A5 genotype influences the
normal blood pressures, but hypertension is com- probability of hearing loss, with 90 % of those
mon in adolescent males with XLAS and teen- with deletions, nonsense mutations, and splice-
aged ARAS patients. site mutations exhibiting deafness before age
In untreated XLAS males, the probability of 30 compared to 60 % of those with missense
ESRD is 50 % by age 25, 80 % by age 40, and mutations [30]. In XLAS females, the probability
100 % by age 60 [30]. COL4A5 genotype has a of hearing loss is 10 % by age 40 and 20 % by age
powerful effect on rate of progression to ESRD in 60 [36]. The majority of patients with ARAS
XLAS males. Large deletions and nonsense muta- develop deafness, although precise data on timing
tions confer a 90 % probability of ESRD before is not available [24].
age 30, compared to a 70 % risk with splice-site Ocular. Anomalies of the lens, retina, and cor-
mutations and a 50 % risk with missense muta- nea are common in Alport syndrome, especially
tions [30]. The epidemiology of ESRD in ARAS among males with XLAS and patients with
is probably similar to XLAS males [24]. It takes ARAS, often becoming apparent during adoles-
50 years for 50 % of ADAS patients to develop cence and young adulthood [38]. About 15 % of
ESRD, twice as long as XLAS males [26, 37]. XLAS males exhibit anterior lenticonus, in which
Women who are heterozygous for COL4A5 the central portion of the lens protrudes into the
mutations (“carriers” of XLAS) demonstrate anterior chamber [30]. While this lesion is often
widely variable disease outcomes with some asymptomatic, it may be associated with reduced
women demonstrating only lifelong visual acuity, and cataracts and even rupture of the
782 M.N. Rheault and C.E. Kashtan

Fig. 3 Electron
micrograph. Lead citrate
and uranyl acetate stain
(4,800). Alport syndrome.
Thin and regular GBM
(arrow) with attenuated
lamina densa. Epithelial
foot processes are
extensively fused

lens may occur. Alteration of retinal pigmentation, Pathology


consisting of whitish-yellowish perimacular The clinical features of Alport syndrome originate
flecks, occurs in about 15 % of XLAS males in changes in basement membrane structure and
[30, 44], often in association with lenticonus. function initiated by absence or abnormalities of
Corneal abnormalities include recurrent corneal α3α4α5(IV) and α5α5α6(IV) networks.
erosions [45, 46] and posterior polymorphous Renal. Light microscopic abnormalities are
dystrophy [47]. Patients with severe COL4A5 unusual in children with Alport syndrome who
mutations are more likely to develop perimacular are less than 5 years of age. Mesangial hypercel-
dot-and-fleck retinopathy [48]. lularity and matrix expansion, and eventually
Other. The association of XLAS with smooth focal segmental glomerulosclerosis, are common
muscle tumors (leiomyomas) of the esophagus, in older children and adolescents, especially boys.
tracheobronchial tree, and, in women, the external Tubular atrophy and interstitial fibrosis develop
genitalia has been described in several dozen fam- progressively after age 10 in boys with Alport
ilies [49–51]. Symptoms such as dysphagia, post- syndrome [55].
prandial vomiting, epigastric or retrosternal pain, Electron microscopy may reveal pathogno-
recurrent bronchitis, dyspnea, cough, and stridor monic changes, depending on the patient’s age
often appear in late childhood. The Alport syn- and gender. The earliest abnormality is diffuse
drome-diffuse leiomyomatosis complex arises attenuation of the GBM; consequently, differenti-
from a contiguous gene deletion on the X chro- ation of Alport syndrome from TBMN by routine
mosome involving exon 1 of COL4A5, the com- renal biopsy processing may be difficult in chil-
mon promoter region that regulates gene dren (Fig. 3). To further complicate matters, in
expression of COL4A5 and COL4A6, and the some families with Alport syndrome due to
first 2 exons of the adjacent COL4A6 gene [49]. COL4A5 mutations, GBM attenuation is the only
Mental retardation, midface hypoplasia, and ultrastructural abnormality. However, the great
elliptocytosis have been described in a small num- majority of boys with XLAS and both boys and
ber of XLAS males who carry deletions that girls with ARAS develop the classic Alport GBM
extend downstream of the 30 end of the COL4A5 lesion, consisting of diffuse thickening accompa-
gene [52, 53]. Finally, abnormalities in arterial nied by “basket-weave” transformation of the
vessels have been described in males with Alport lamina densa, intramembranous vesicles and den-
syndrome including aortic root dilatation and sities, scalloping of the epithelial surface of the
aneurysms of the thoracic and abdominal GBM, and foot process effacement (Fig. 4). The
aorta [54]. percentage of GBM displaying this lesion
26 Inherited Glomerular Diseases 783

Fig. 4 Electron
micrograph. Lead citrate
and uranyl acetate stain
(11,250). Alport
syndrome. Thickened GBM
showing splitting of the
lamina densa and presence
of granulations

Fig. 5 Immunofluorescence microscopy. Distribution of with X-lined Alport syndrome. (c) Absence of epidermal
the α(IV) chains in renal and epidermal basement mem- basement membrane labeling (arrow). (f) Discontinuous
branes of male (a–c) and female (d–f) patients affected epidermal basement membrane labeling (arrows)

increases progressively with age in boys with (IV), and α5(IV) chains is completely absent in
XLAS [56]. Females with XLAS display a range 80 % of XLAS males, and 60–70 % of females
of GBM alteration, from focal GBM attenuation with XLAS exhibit mosaic expression of these
to diffuse thickening and basket-weaving, and chains [57] (Fig. 5). In most patient with ARAS,
there is no consistent correlation of GBM findings GBM is nonreactive with antibodies to the α3(IV),
and age [56]. α4(IV), and α5(IV) chains, and Bowman’s cap-
Routine immunofluorescence is normal or sules and tubular basement membranes are also
shows nonspecific immunoprotein deposition. negative for the α3(IV) and α4(IV) chains [58]
Because disease-causing mutations result in (Fig. 6). However, immunostaining for α5
abnormal expression of α3α4α5(IV) and α5α5α6 (IV) chains in Bowman’s capsules and tubular
(IV) networks in basement membranes in most basement membranes is present, because in these
Alport patients, specific immunostaining for type basement membranes, expression of α5α5α6
IV collagen α chains is useful for both diagnosis (IV) networks is preserved. It is important to
and differentiation of XLAS and ARAS [57] note that altered immunostaining for the α3
(Figs. 5 and 6). Expression of the α3(IV), α4 (IV)–α6(IV) chains in patients with XLAS and
784 M.N. Rheault and C.E. Kashtan

Fig. 6 Immunofluorescence microscopy. Distribution of the α(IV) chains in renal and epidermal basement membranes of
patients affected with autosomal recessive Alport syndrome

ARAS is not age-dependent. Therefore, this although deafness in these models is minimal or
method can provide diagnostic information even absent.
in patients who are too young to display charac- Ocular. The α3(IV), α4(IV), and α5(IV) chains
teristic abnormalities in GBM ultrastructure. are normal components of several basement mem-
Normal epidermal basement membranes (EBM) branes in the eye, including the corneal basement
express the α5α5α6(IV) network but not the membrane, Descemet’s membrane, lens capsule,
α3α4α5(IV) network. EBMs show negative the internal limiting membrane of the retina, and
staining for α5(IV) chains in about 80 % of the retinal pigment epithelium basement mem-
XLAS males, and mosaic expression of α5(IV) is brane [64, 67–69]. The ocular manifestations of
observed in 60–70 % of XLAS females, allowing Alport syndrome likely arise from absence or
diagnosis of XLAS by skin biopsy [59, 60] (Fig. 5). abnormality of α3α4α5(IV) networks in these
Skin biopsy is not useful for diagnosis of ARAS, basement membranes. Absence of the α3α4α5
since expression of the α5α5α6(IV) network in (IV) network leads to increased distensibility in
EBM is normal [58] (Fig. 6). the lens capsule when tested in experimental
Cochlear. The hearing loss of Alport syndrome models of AS [70]. The lens capsules of Alport
arises from cochlear dysfunction [61]. Normal patients with anterior lenticonus exhibit marked
cochleae in mice, dogs, and humans express α3 attenuation and focal areas of dehiscence,
(IV), α4(IV), and α5(IV) chains in the spiral lim- suggesting that the lens capsule lacks the mechan-
bus, in the spiral ligament, and in the basement ical strength to maintain normal lens shape [68,
membrane interposed between the organ of Corti 71, 72].
and the basilar membrane [62–65]. However,
expression of these chains is absent in cochleae Diagnostic Considerations
of ARAS mice [62], XLAS dogs [63], and men Accurate diagnosis of Alport syndrome and dif-
with XLAS [65]. Careful examination of well- ferentiation of this condition from other causes of
preserved cochleae from men with XLAS and familial and sporadic glomerular hematuria are
deafness revealed a zone of separation between based upon careful clinical evaluation, reliable
the organ of Corti and the underlying basilar pedigree data, and thoughtful consideration of
membrane, and cellular infiltration of the tunnel the relative merits of skin biopsy, kidney biopsy,
of Corti and the spaces of Nuel [66]. These and genetic testing.
changes are not observed in similarly well- In a child with isolated hematuria, a positive
preserved cochleae obtained from normal individ- family history of hematuria in the absence of a
uals or patients with other causes of deafness. The history of ESRD suggests a diagnosis of TBMN.
structural changes observed in Alport cochleae Two rare causes of familial hematuria associated
may be associated with defective attuning of bas- with macrothrombocytopenia, Epstein and
ilar membrane motion and hair cell stimulation, Fechtner syndromes, can be excluded if the plate-
resulting in reduced acuity of hearing. Similar let count is normal. Familial IgA nephropathy [73]
changes have not been observed in cochleae and membranoproliferative glomerulonephritis
from mice or dogs with Alport syndrome, [74] are uncommon causes of familial hematuria.
26 Inherited Glomerular Diseases 785

In the absence of a family history of hematuria, for mutations in COL4A3, COL4A4, and COL4A5
the differential diagnosis of glomerular hematuria [27]. Commercially available genetic testing for
includes Alport syndrome, TBMN, IgA nephrop- mutations in COL4A3, COL4A4, and COL4A5 is
athy, membranoproliferative glomerulonephritis, available in many countries.
membranous nephropathy, lupus nephritis,
postinfectious glomerulonephritis, and Henoch- Treatment
Schönlein nephritis. Associated clinical findings The goal of treatment in patients with Alport
(e.g., rash, arthritis) or laboratory findings (e.g., syndrome is to slow the progression of kidney
hypocomplementemia) will suggest diagnoses disease and delay the need for renal replacement
other than Alport syndrome in many of these therapy. There have been no controlled therapeu-
patients. tic trials in human Alport syndrome. Conse-
While results of hearing evaluation are likely to quently, treatment recommendations must be
be normal in young children with Alport syn- derived from animal studies, anecdotal reports,
drome, audiometry may be very useful in children and retrospective registry reviews. In murine
over 6–8 years of age, especially boys. Ophthal- ARAS, several interventions have proven effica-
mologic assessment may also provide valuable cious, including angiotensin antagonism [76–78],
information, although ocular lesions are more TGFb-1 inhibition [79], chemokine receptor
prevalent in Alport patients with advanced dis- 1 suppression [80], administration of bone mor-
ease, and less likely to be present in the population phogenic protein-7 [81], blockade of matrix
of young patients in whom differential diagnosis metalloproteinases [82], and bone marrow trans-
of hematuria may be more difficult. plantation [83, 84].
Tissue studies can complement clinical and Cyclosporine treatment resulted in prolonga-
pedigree information that is insufficient to clearly tion of survival in male XLAS dogs [85]. Cyclo-
differentiate Alport syndrome from other diagno- sporine treatment diminished proteinuria and
ses. Skin biopsy with immunostaining for the α5 appeared to stabilize renal function in a small,
(IV) chain, as described above, may be diagnostic, uncontrolled study of Alport males [86]. However,
especially when clinical and pedigree data apparent acceleration of renal fibrosis was
strongly suggest a diagnosis of XLAS. Normal suggested by the results of other studies of cyclo-
expression of the α5(IV) chain in epidermal base- sporine treatment in Alport patients, and this treat-
ment membrane can be explained in several ways: ment is no longer recommended [87, 88].
(1) the patient has XLAS, but his or her COL4A5 Uncontrolled studies in human Alport syn-
mutation does not abolish α5(IV) expression, drome subjects have shown that angiotensin
(2) the patient has a form of Alport syndrome blockade can transiently reduce proteinuria [89,
(ARAS or ADAS) in which expression of α5 90]. Subgroup analysis of 30 children with Alport
(IV) in epidermal basement membranes is not syndrome in a larger multicenter, randomized,
affected, or (3) the patient does not have Alport double-blind study comparing losartan with pla-
syndrome. Whereas skin biopsy is useful only if it cebo or amlodipine in proteinuric children dem-
provides definitive confirmation of a diagnosis of onstrated a significant reduction in proteinuria in
XLAS, renal biopsy carries the advantage of the losartan treated group [91]. An extension of
enabling the diagnosis of XLAS, ARAS, and this study showed comparable efficacy of either
non-Alport kidney disease, particularly if type enalapril or losartan in reducing proteinuria in
IV collagen immunostaining is applied. children with Alport syndrome [92]. A report
Mutation detection rates of 80–90 % are attain- from the European Alport Registry, which
able in males with XLAS using direct sequencing included 283 patients followed over 20 years,
of COL4A5 [75]. Comparable data for detection compared renal outcomes in Alport syndrome
of COL4A3 and COL4A4 mutations in patients patients treated with ACE inhibition at various
with ARAS are lacking. Next-generation disease time points: microalbuminuria, protein-
sequencing allows for rapid simultaneous analysis uria, or in chronic kidney disease (CKD) stages
786 M.N. Rheault and C.E. Kashtan

III–IV [93]. This retrospective review demon- without hematuria is affected. Given that by age
strated a delay in renal replacement therapy by 60 there is an estimated risk of ESRD of 30 % in
3 years in the treated CKD group and by 18 years women with XLAS [36], female members of
in the treated proteinuric group [93]. Side effects XLAS families who have hematuria should be
of ACE inhibition were rare and included discouraged from kidney donation. One study
hyperkalemia, cough, and hypotension. Based described development of proteinuria and a
on these findings, a prospective, double-blind, decline in renal function in heterozygous females
randomized, placebo controlled trial is under with XLAS after kidney donation [96], highlight-
way to compare outcomes in children with Alport ing the potential risks in this population.
syndrome treated with ramipril versus placebo at Anti-GBM nephritis occurs in 3 % of
an early time point (microalbuminuria or isolated transplanted Alport males [97]. The onset of
hematuria) [94]. anti-GBM nephritis typically occurs during the
Clinical practice guidelines have been devel- first year after transplantation and usually results
oped to guide treatment of patients with Alport in irreversible graft failure within weeks to
syndrome, including heterozygous females months of diagnosis. There is a high rate of recur-
[40]. Treatment with ACE inhibitors or angioten- rence in subsequent allografts. In XLAS males,
sin receptor blockers should be considered for the primary target of anti-GBM antibodies is the
affected individuals with microalbuminuria and α5(IV) chain [98, 99]. Females with XLAS who
either a family history of ESRD at a young age require transplantation are at little or no risk of
(<30 years) or a known severe COL4A5 mutation developing anti-GBM nephritis. However, both
and for all affected individuals with overt protein- males and females with ARAS can develop anti-
uria. Treatment of hypertension and other mani- GBM nephritis after transplantation. The α3
festations of chronic kidney disease are similar to (IV) chain is the primary target of anti-GBM
children with other etiologies of chronic kidney antibodies in ARAS patients [98, 100].
disease. Goodpasture autoantibodies and anti-GBM anti-
bodies from transplanted Alport patients target
Renal Replacement Therapy distinct epitopes on the carboxy-terminal
Patients with Alport syndrome typically have noncollagenous (NC1) domain of the α3
excellent outcomes following renal transplanta- (IV) chain [101].
tion [95]. Two issues require the special attention Renal allograft biopsy with routine immuno-
of transplant physicians involved in the care of fluorescence should be performed early in the
Alport patients. First, evaluation of potential evaluation of Alport patients who develop hema-
related donors must identify affected individuals turia or increased creatinine after transplantation
who may be at risk for development of significant or if circulating anti-GBM antibodies are
renal insufficiency. Second, posttransplant moni- detected. Anti-GBM nephritis after renal trans-
toring must allow early diagnosis of plantation should be treated with cytotoxic ther-
posttransplant anti-GBM nephritis, a complica- apy and plasmapheresis, although such therapy
tion of transplantation that is unique to Alport has been unsuccessful in the majority of reported
syndrome. patients.
Familiarity with the genetics of Alport syn-
drome and the signs and symptoms of the disease
is required for informed donor evaluation. Since Thin Basement Membrane
100 % of males with XLAS have hematuria [30], Nephropathy
the absence of hematuria excludes Alport syn-
drome in male relatives of XLAS patients. Historically, families displaying autosomal domi-
About 95 % of females with XLAS have hematu- nant transmission of isolated, nonprogressive
ria [36], so there is only a 5 % chance that a female hematuria were classified as having “benign
26 Inherited Glomerular Diseases 787

familial hematuria” [102–104]. Affected patients Clinical Features


typically exhibited no renal parenchymal abnor- TBMN is the most common cause of persistent
malities apart from thinning of glomerular base- microscopic hematuria in children and adults
ment membranes (GBM) observed by electron [108]. Persistent microscopic hematuria, associ-
microscopy [105, 106]. The more inclusive term ated with normal blood pressure, renal function,
“thin basement membrane nephropathy” (TBMN) and urine protein excretion, is the characteristic
has gradually displaced benign familial hematuria feature of TBMN in childhood [3], although epi-
as the preferred designation for hematuria associ- sodic gross hematuria may also be observed.
ated with thin GBM, because it encompasses spo- TBMN is not specifically associated with hearing
radic cases of hematuria associated with thin loss, ocular defects, or other extrarenal
GBM; familial or sporadic cases of thin GBM in abnormalities.
which hematuria is accompanied by proteinuria, Proteinuria has been reported in up to 30 % of
hypertension, and/or renal insufficiency; and adults with TBMN [106, 109]. About 5–7 % of
benign familial hematuria. The prevalence of adult patients with TBMN have chronic kidney
TBMN is estimated at 1–2 % of the disease with elevated serum creatinine levels
population [107]. [109–112].
Thinning of GBM is a pathological description
rather than a distinct entity. Depending on the Pathology
timing of renal biopsy, thin basement membranes Diffuse attenuation of the lamina densa and GBM,
may be observed in patients with hemizygous or with preservation of normal podocyte anatomy, is
heterozygous mutations in COL4A5 (XLAS), the characteristic histological abnormality in
biallelic mutations in COL4A3 or COL4A4 TBMN. Glomerular obsolescence or sclerosis
(ARAS), heterozygous mutations in COL4A3 or may be observed in adult patients with TBMN,
COL4A4 (the carrier state of ARAS), and muta- including some with heterozygous COL4A3 or
tions at nontype IV collagen loci [94]. The natural COL4A4 mutations [111, 113].
history of hematuria associated with thin GBM is GBM width is dependent on age and gender.
determined by the underlying mutation, perhaps The lamina densa and GBM increase rapidly in
in combination with remote modifier genes. width between birth and 2 years of age, followed
Hemizygous mutations in COL4A5 and biallelic by a more gradual increase during childhood and
mutations in COL4A3 and COL4A4 result in pro- adolescence [114]. Adult men exhibit greater
gressive GBM thickening, proteinuria, and renal GBM widths than adult women [115].
failure. Heterozygous mutations in COL4A3 or Because different investigators have used differ-
COL4A4 are usually associated with ent techniques to measure GBM width, a standard
persistent GBM attenuation, isolated hematuria, definition of “thin” GBM does not exist. In children
and benign outcomes. Heterozygous mutations in the threshold is 200–250 nm [2, 116, 117],
COL4A5 in women with XLAS are associated while the adult threshold ranges from 250 to
with a wide range of prognostic outcomes, as 330 nm [106, 118].
described in the section on “Alport Syndrome.” Routine immunofluorescence studies of renal
Perhaps this spectrum of outcomes reflects biopsy material from patients with TBMN are
differences in the cellular responses provoked by typically unremarkable. Immunostaining using
complete absence of α3α4α5(IV) networks from specific antibodies against α3(IV), α4(IV), and
GBM (hemizygous XLAS and ARAS), mixed α5(IV) chains yields normal results in patients
α3α4α5(IV)-positive and α3α4α5(IV)-negative with TBMN [105, 119, 120].
GBM (heterozygous XLAS), and homogeneous
reduction in GBM content of α3α4α5 Diagnostic Considerations
(IV) networks (heterozygous COL4A3 or IgA nephropathy, TBMN, and Alport syndrome
COL4A4 mutations). together account for the majority of children with
788 M.N. Rheault and C.E. Kashtan

glomerular hematuria seen in pediatric nephrol- Hereditary Angiopathy


ogy clinics [1–3, 116]. Thorough clinical evalua- with Nephropathy, Aneurysms,
tion, including detailed pedigree analysis, can and Cramps (HANAC Syndrome)
assist in determining which children need tissue
studies and which can be followed prospectively Missense mutations in the COL4A1 gene cause an
without biopsy. Obtaining urinalyses on first- autosomal dominant hereditary angiopathy asso-
degree family members may provide valuable ciated with nephropathy, aneurysms, and muscle
information, since adults with familial hematuria cramps (HANAC syndrome) [123]. The muta-
may be unaware that they are affected [121]. tions affect three highly conserved glycine resi-
When a child has isolated microscopic hema- dues in the collagenous domain of the α1
turia, a family history of dominantly transmitted (IV) chain. Retinal arteriolar tortuosity and retinal
hematuria, and a negative family history for renal hemorrhages are common in affected individuals,
failure, a clinical diagnosis of TBMN is reason- as are intracranial aneurysms [124], leukoence-
able, and renal biopsy is unnecessary. These chil- phalopathy, and elevated creatine kinase levels.
dren should be followed prospectively every 1–2 Some affected individuals have muscle cramps.
years. If proteinuria or hypertension develops, Renal findings in affected individuals include
renal biopsy should be considered. microscopic and gross hematuria, mild renal
In children who have persistent microscopic insufficiency, and renal cysts. Mutations in the
hematuria but do not have affected relatives, COL4A1 and COL4A2 genes have not been
renal biopsy is often informative. In those children found in patients with Alport syndrome or
whose renal biopsy findings are limited to GBM TBMN [125].
attenuation, the clinician’s challenge is to differ- Renal biopsy in affected individuals with
entiate TBMN and Alport syndrome. Audiometry hematuria showed no abnormalities of GBM
and ophthalmologic examination may be helpful structure or type IV collagen expression. How-
in older children, but these studies will usually be ever, basement membranes of Bowman’s cap-
normal in young children with Alport syndrome. sules, tubules, and interstitial capillaries
Abnormal results of immunostaining for the α3 exhibited irregular thickening, splitting into mul-
(IV), α4(IV), and α5(IV) chains suggest a diagno- tiple layers, and focal interruptions.
sis of Alport syndrome. While normal
immunostaining results cannot entirely exclude
Alport syndrome, they can help support a Laminin Disorders
suspected diagnosis of TBMN.
The role of molecular analysis of the COL4A3, Pierson Syndrome
COL4A4, and COL4A5 genes in patients with
suspected TBMN remains to be determined. The association of congenital nephrotic syndrome
Since the finding of a heterozygous mutation in with eye abnormalities in siblings was first
COL4A3 or COL4A4 cannot guarantee a benign described by Pierson in 1963 [126]. Affected
prognosis [26, 27, 113, 122], the need for follow- infants died within the first year of life with
up examination of such patients would not be ESRD. Subsequent reports of additional patients
precluded. described variable abnormalities on fetal ultra-
sound, including kidney enlargement and
Treatment hyperechogenicity, oligohydramnios, placental
Since TBMN usually has a benign outcome, treat- enlargement, and/or pulmonary hypoplasia
ment is rarely indicated. Patients with TBMN who [127, 128]. Infants surviving to term exhibited
have proteinuria are theoretically candidates for congenital nephrotic syndrome, with renal biopsy
angiotensin blockade. findings of diffuse mesangial sclerosis and diffuse
26 Inherited Glomerular Diseases 789

GBM abnormalities, accompanied by ocular nails, hypoplasia or absence of the patellae, dys-
globe enlargement (buphthalmos) with reduction plasia of the elbows and iliac horns, and renal
in the size and reactivity of pupils (microcoria). disease [134, 135]. Affected individuals may
Additional ocular findings in some patients also exhibit glaucoma and sensorineural deafness
included cataract, posterior rupture of the lens [136, 137]. The disorder affects about 1 in 50,000
capsule, and retinal abnormalities. Some affected individuals. The severity of renal involvement,
children have also been found to have muscular which occurs in 30–40 % of patients, determines
hypotonia, central nervous system hemangiomas, prognosis.
and genital abnormalities [129, 130].
The gene for Pierson syndrome was mapped to Genetics
chromosome 3p14-p22 and identified as LAMB2, Targeted disruption of the LIM homeodomain
the gene encoding the laminin β2 chain, by Zenker transcription factor gene Lmx1b in mice resulted
and colleagues in 2004 [127]. Subsequent reports in hypoplastic nails and absent patellae as well as
indicate that LAMB2 mutations may be associated renal disease, suggesting a possible locus for
with isolated congenital nephrotic syndrome and human NPS [138]. LMX1B, the human homo-
with mild variants of Pierson syndrome [131, logue of Lmx1b, and the NPS locus were found
132]. The absence of the laminin β2 chain in to map to chromosome 9q34, and heterozygous
transgenic mice results in massive proteinuria mutations in LMX1B were identified in NPS
along with retinal and neuromuscular abnormali- patients [139, 140]. A variety of LMX1B gene
ties [133]. In these mice, proteinuria precedes the defects have been identified in NPS patients,
appearance of podocyte abnormalities, suggesting including missense, splicing, insertion/deletion,
that the GBM contributes to the barrier function of and nonsense mutations [139–143]. The results
the glomerular capillary wall. Genotype influ- of in vitro experiments in which the transcrip-
ences the phenotype in individuals with LAMB2 tional effects of mixing wild-type and mutant
mutations. Truncating or splice-site mutations are LMX1B protein are measured, along with the
more likely to be associated with complete variety of observed mutation types, suggest that
Pierson syndrome, whereas missense mutations the NPS phenotype results from haploinsuf-
or small deletions are more likely to be associated ficiency rather than a dominant-negative mecha-
with the absence of neurologic abnormalities and nism [141, 144]. Mutations in the homeodomain
higher age of onset of renal disease [132]. of LMX1B protein and female gender may confer
an increased risk of renal involvement [137].
LMX1B protein is specifically expressed in
Type III Collagen Nephropathies glomerular podocytes, beginning at the S-shaped
body stage of glomerular development
In the kidney, type III collagen is normally found [138]. Lmx1b-null mice exhibit marked reduction
in the interstitium and blood vessel walls. Two in GBM expression of the α3 and α4 chains of
rare disorders in which pathological accumulation type IV collagen and the slit diaphragm proteins
of type III collagen is observed in glomeruli are podocin and CD2AP [145–147]. However,
discussed in this section. changes in expression of these putative targets of
LMX1B were not observed in kidneys of patients
with NPS nephropathy [148]. Recently, LMX1B
Nail-Patella Syndrome (Hereditary was found to be essential for the maintenance of
Osteo-onychodysplasia) differentiated podocytes in adult kidneys through
effects on actin cytoskeletal organization
Nail-patella syndrome (NPS) is a rare autosomal [149]. Mutations in LMX1B have also been
dominant disorder characterized by dystrophic described in patients with autosomal dominant
790 M.N. Rheault and C.E. Kashtan

Fig. 7 Electron
microscopy.
Phosphotungstic acid stain
(10,500). Nail-patella
syndrome. Irregular
distribution of fibrillar
collagen within the GBM.
Inset shows the typical
periodicity of interstitial
collagen (48,000)

focal segmental glomerulosclerosis without radius may be associated with mild to severe
extrarenal involvement [150]. limitation in extension, pronation, and supination
of the forearm. About 80 % of patients display
Clinical Features osseous processes projecting posteriorly from the
Renal. Less than half of NPS patients have clinical iliac wings, known as iliac horns, which are patho-
renal disease [151]. Symptoms including mild gnomonic for NPS [152].
proteinuria with or without microscopic hematu-
ria typically appear in adolescence or young adult- Pathology
hood. Occasional patients develop nephrotic The nephropathy of NPS has no specific features
syndrome and hypertension. The nephropathy is by light microscopy or routine immunofluores-
typically mild, with about a 5–10 % risk of pro- cence. Focal segmental glomerulosclerosis and
gression to ESRD. Marked differences in the deposits of IgM and C3 may be observed,
severity of the nephropathy can be observed in depending on the severity of renal involvement.
related patients, suggesting the influence of Characteristic lesions are observed by
superimposed factors. electron microscopy [153, 154]. With standard
Nails. Nail abnormalities occur in over 90 % of staining techniques, the GBM and
patients and are usually apparent from birth mesangium exhibit multiple lucencies, imparting
[152]. Fingernails, especially on the thumb and a “moth-eaten” appearance. Staining with
index finger, are more severely affected than toe- phosphotungstic acid reveals clusters of cross-
nails. The nails may be absent or dystrophic with banded collagen fibrils within these lucent
discoloration, koilonychias, longitudinal ridges, areas (Fig. 7). These collagen fibrils have been
or triangular lunulae. observed in NPS patients with no clinically evi-
Skeletal defects. Over 90 % of NPS patients dent renal disease, and there is no correlation
exhibit aplasia or hypoplasia of the patellae between the extent of GBM changes and patient
[152]. Associated symptoms may include knee age, severity of proteinuria, or degree of renal
pain, effusions, dislocations, and osteoarthritis. functional impairment [141, 142]. Staining of
Dysplasia of the elbows occurs in over 90 % of NPS kidneys with antibodies against type III col-
patients [152]. Anomalies such as hypoplasia of lagen produced irregular, discontinuous labeling
the radial head with dislocation, posterior pro- of GBM in normal-appearing glomeruli and
cesses at the distal ends of the humeri, hypoplasia focally intense staining of sclerotic
of the olecranon, and elongation of the neck of the glomeruli [148].
26 Inherited Glomerular Diseases 791

Treatment Hereditary Nephritis


There is no specific therapy for NPS renal disease. with Thrombocytopenia and Giant
Kidney transplantation has been carried out suc- Platelets: Epstein and Fechtner
cessfully, and recurrence of renal disease appar- Syndromes
ently did not recur. Selection of living donors
must be performed with care, since NPS is an Epstein and Fechtner syndromes are allelic, auto-
autosomal dominant disorder. somal dominant disorders that have some features
in common with Alport syndrome, such as hema-
turia, progressive nephropathy and sensorineural
Collagen Type III Glomerulopathy deafness [159, 160]. However, these conditions
(Collagenofibrotic Glomerulopathy) are distinguished clinically from Alport syndrome
by the invariable presence of thrombocytopenia
Renal disease associated with glomerular deposits and giant platelets. In addition, granulocytes of
of type III collagen has been described in patients patients with Fechtner syndrome display
who have no extrarenal abnormalities [155, 156]. cytoplasmic inclusions known as Dohle-like bod-
In comparison to NPS patients, subjects with col- ies, and these patients may develop cataracts. In
lagen type III glomerulopathy have relatively some patients, ultrastructural changes in GBM
severe renal findings, more extensive histological resemble those of Alport syndrome
changes, and greater glomerular type III collagen [161, 162]. However, glomerular expression of
accumulation. type IV collagen α3, α4, and α5 chains is normal
in these patients [162].
Clinical Features Despite the similarities with Alport syndrome,
Patients with collagen type III glomerulopathy Epstein and Fechtner syndromes are genetically
can present in childhood or in adulthood. Early distinct disorders. Following the mapping of the
presentation is associated with a more severe dis- Epstein and Fechtner loci to chromosome 2q11-
ease course. Proteinuria is common to the juvenile 13 [163, 164], heterozygous mutations in MYH9,
and adult forms of the disease. In affected indi- the gene that encodes nonmuscle myosin heavy
viduals, proteinuria progresses to nephrotic syn- chain IIA (NMMHC-IIA), were identified in
drome and is accompanied by hypertension and patients with these disorders as well as in patients
the development of renal failure in up to 50 % of with two other conditions featuring giant platelets,
affected patients [156, 157]. Microangiopathic Sebastian syndrome and May-Hegglin anomaly,
hemolytic anemia has been described in some and in nonsyndromic hereditary deafness
patients. Patients presenting in adulthood exhibit (DFNA17) [165–168]. NMMHC-IIA is expressed
more gradual increase in proteinuria and loss of in podocytes [169], but the mechanism by which
renal function [158]. mutations in this protein might adversely affect
podocyte function is unknown.
Pathology
Light microscopy shows enlarged glomeruli with
mesangial expansion and glomerular capillary
wall thickening due to subendothelial accumula- Hereditary Metabolic Disorders
tion of poorly staining material. Routine immuno- with Primary Glomerular Involvement
fluorescence studies are unremarkable or show
nonspecific immunoprotein deposits. Electron Fabry Disease
microscopy shows deposits of electron-lucent
material in mesangial matrix and GBM. Staining Anderson-Fabry disease is a rare X-linked
with phosphotungstic acid demonstrates fibrillar disorder of glycosphingolipid metabolism resulting
collagen in these deposits which is identified as from deficiency of the lysosomal hydrolase
type III collagen by specific immunostaining. a-galactosidase A.
792 M.N. Rheault and C.E. Kashtan

Renal tissue from all hemizygous patients Hereditary Metabolic Disorders


demonstrates characteristic glycolipid with Secondary Glomerular
accumulation within every glomerular, vascular, Involvement
and interstitial cell and within distal tubular
cells, regardless of age at biopsy. Two Familial Amyloidosis
intermingled cell populations, normal cells and
cells exhibiting glycolipid accumulation, are Hereditary amyloidosis encompasses a group of
observed in heterozygous females. Degenerative autosomal dominant disorders characterized by
renal changes develop with age. They initially extracellular accumulation of protein fibrils
affect vessels and consist of round arranged in an antiparallel β-pleated sheet config-
fibrinoid deposits resulting from smooth muscle uration [178]. These disorders are classified
cell necrosis. These changes are followed by according to the protein composing amyloid
nonspecific vascular, glomerular, and tubuloin- fibrils and/or the type of mutation in the
terstitial lesions [170]. Enzyme replacement corresponding gene. Transthyretin variants have
appears to result in decreased glycolipid storage been found in most affected families, but variants
in renal cells and stabilization of renal function of cystatin C, gelsolin, apolipoprotein A1, fibrin-
[171–173]. ogen, and lysozyme have been described in other
kindreds. Symptomatic renal involvement is
unusual during childhood.
Familial Mediterranean fever is an autosomal
Other Glomerular Lipidoses recessive disorder described primarily, but not
exclusively, in several ethnic groups originating
Nephrosialidosis is a rare autosomal recessive in the Mediterranean region. The disease is char-
condition caused by neuraminidase deficiency. acterized by recurrent episodes of fever, abdomi-
Clinical and radiologic features include nal pain, joint pain, pleuritis, and pericarditis
dysmorphic facies, visceromegaly, mental retar- [179]. Renal amyloidosis of the AA type may
dation, skeletal anomalies, marrow foam cells, result in proteinuria and eventual renal failure.
and cherry-red spot on fundoscopy. Renal Renal amyloidosis of the AA type may be associ-
involvement consists of proteinuria and ated with the autosomal dominant disorders
progression to ESRD early in life [174]. Renal Muckle-Wells syndrome and tumor necrosis fac-
biopsy findings include podocyte and proximal tor receptor 1-associated periodic syndrome
tubular cell vacuolization [175]. By electron (TRAPS) [180].
microscopy, many of these vesicles appear
empty, but others contain flocculent or
membrane-like electron-dense material. Wheat
germ agglutinin binds to cytoplasmic material in Alpha-1 Antitrypsin Deficiency
podocytes and tubular cells, indicating the pres-
ence of compounds with terminal sialic Chronic liver disease due to deficiency of alpha-1-
(neuraminic) acid moieties [176]. antitrypsin (α1AT) may be associated with glo-
Silent accumulation of glycolipids or muco- merulonephritis in children [181–183]. Renal
polysaccharides has been described in biopsy reveals diffuse or focal segmental
patients with Gaucher disease, Niemann-Pick membranoproliferative glomerulonephritis, type
disease, I-cell disease, and GM1 I in most cases, associated with subendothelial
gangliosidosis [177]. Clinical renal disease during deposits composed of immunoglobulin and com-
childhood is unusual in patients with these plement. Alpha-1-antitrypsin deficiency has also
disorders. been associated with IgA nephropathy and
26 Inherited Glomerular Diseases 793

Fig. 8 Electron
microscopy. Lead citrate
and uranyl acetate stain
(8,600). Alagille
syndrome. Massive
accumulation of lipid
vacuoles within mesangial
cells and matrix. Irregular
distribution of lipid
vacuoles within the GBM

systemic ANCA positive vasculitis [184]. Renal Hereditary Lecithin-Cholesterol


manifestations include proteinuria, hypertension, Acyltransferase (LCAT) Deficiency
and renal failure. Regression of nephrotic syn-
drome and glomerular lesions after liver trans- LCAT deficiency is a rare autosomal recessive
plantation has been observed [185]. disorder characterized by the inability to esterify
plasma cholesterol, leading to deposition of
unesterified cholesterol in tissues, including the
Alagille Syndrome kidney [190]. Clinical manifestations include cor-
neal opacities, anemia, and proteinuria, sometimes
Alagille syndrome is an autosomal dominant dis- in the nephrotic range [191, 192]. Progression to
order with variable penetrance that causes chole- ESRD usually occurs in the fourth or fifth decade.
stasis in childhood, associated with characteristic Glomerular lesions include accumulation of foam
facies, cardiac malformations, vertebral abnor- cells of endothelial and mesangial origin and mas-
malities, posterior embryotoxon, hypogonadism, sive accumulation of lipids within the mesangial
growth retardation, and high-pitched voice. The matrix and subendothelial GBM [190, 191].
disease results from mutations in JAG1, which
encodes a ligand for the Notch1 receptor, or muta-
tions in NOTCH2 [186, 187]. JAG1 and Lipoprotein Glomerulopathy
NOTCH2 are required for glomerular develop-
ment. Renal involvement occurs in ~40 % of Lipoprotein glomerulopathy is characterized by
patients with JAG1 mutations and can include intraglomerular lipoprotein thrombosis and high
renal dysplasia, renal tubular acidosis, plasma concentrations of apolipoprotein E
vesicoureteral reflux, cystic disease, and obstruc- [193]. The disease is usually detected in adults,
tion [188]. Accumulation of lipid vacuoles in but onset of symptoms during childhood has been
mesangial matrix, mesangial cells, and GBM has described [194]. Renal symptoms range from pro-
been observed in some patients [189] (Fig. 8). The teinuria to nephrotic syndrome, and progression
extent of mesangiolipidosis correlates with the to ESRD occurs in some patients [195]. Recur-
severity of cholestasis. Although the glomerular rence of glomerular lesions after renal transplan-
lesions are present early in life, renal symptoms in tation has been observed [196]. Mutations in
childhood are unusual. Progression to ESRD may apolipoprotein E have been found in patients
occur in affected adults. with this disorder [197].
794 M.N. Rheault and C.E. Kashtan

Glomerular lipidosis has also been reported in Cockayne Syndrome


patients with familial hypercholesterolemia
resulting from defects in the LDL receptor, in Cockayne syndrome is an autosomal recessive
type III hyperlipoproteinemia, and in patients disorder characterized by growth retardation, neu-
with cholesterolic polycoria. Renal symptoms rologic abnormalities, premature aging, senile
usually appear in adulthood in patients with facies, sensorineural deafness, cataracts, retinop-
these disorders. athy, sun sensitivity, and dental caries [182]. The
disorder arises from mutations in genes involved
in DNA nucleotide excision repair (ERCC6 and
Familial Juvenile Megaloblastic Anemia ERCC8) [204]. Renal symptoms including hyper-
tension, proteinuria, and renal insufficiency occur
Familial juvenile megaloblastic anemia (Imerslund- in about 10 % of patients, associated with diffuse,
Grasbeck syndrome) is a rare autosomal recessive homogeneous GBM thickening [205].
disorder caused by selective vitamin B12 malabsorp-
tion. Anemia is detected in infancy or early child-
hood and is associated with mild, nonprogressive Hereditary Acro-osteolysis
low-molecular weight proteinuria [198]. Most cases with Nephropathy
arise from mutation in the gene for either cubilin
(chromosome 10) or amnionless (chromosome 14), Hereditary acro-osteolysis is a rare disorder char-
which are components of the intestinal receptor for acterized by arthritic episodes and progressive
the vitamin B12-intrinsic factor complex as well as resorption of carpal and tarsal bones. Familial
the receptor that mediates proximal tubular (dominant or recessive) and sporadic cases have
reabsorption of filtered protein [199]. been reported. Hypertension, proteinuria, and pro-
gressive renal failure occur in some patients
[206]. Renal biopsy findings include arteriolar
Other Hereditary Diseases thickening and sclerosis and focal
with Glomerular Involvement glomerulosclerosis.

Charcot-Marie-Tooth (CMT) Disease


Other Syndromes with Renal
CMT disease is a genetically heterogeneous, Involvement
familial peripheral neuropathy resulting in pro-
gressive symmetric atrophy and weakness of dis- Renal abnormalities, typically cystic renal dyspla-
tal muscles and sensory deficits. Autosomal sia and/or tubulointerstitial lesions, are found in
dominant inheritance is most commonly caused most patients with Bardet-Biedl syndrome (BBS),
by mutations in peripheral myelin protein a genetically heterogeneous, autosomal recessive
22 (PPMP22) and myelin protein zero (MPZ); disorder whose cardinal features included obesity,
however, over 40 genes have been associated with polydactyly, mental retardation, retinal dystrophy,
this disorder [200]. Proteinuria and progression to and hypogonadism [207]. Glomerular symptoms
ESRD associated with focal glomerulosclerosis such as proteinuria and questionable glomerular
(FSGS) occur in some patients [201, 202]. Since changes have been described in a few patients
some of these patients are also deaf, CMT disease [208]. Mutations in a number of genes involved
could potentially be misdiagnosed as Alport syn- in the functioning of primary cilia have been
drome. No specific ultrastructural changes in GBM linked to BBS [209].
have been described in patients with CMT disease. Alstrom syndrome is an autosomal recessive
Recently, mutations in INF2, a cause of autosomal disorder characterized by cone-rod dystrophy,
dominant FSGS, were found in patients with CMT obesity, progressive sensorineural deafness,
and FSGS [203]. dilated cardiomyopathy, insulin resistance
26 Inherited Glomerular Diseases 795

syndrome, and developmental delay due to muta- (mitochondrial encephalopathy with lactic acido-
tions in the ALMS1 gene [210]. Renal tubular sis and stroke-like episodes) [220].
dysfunction and tubulointerstitial lesions have
been observed in some patients [211]. Renal dis-
ease due to vascular lesions and secondary References
glomerulosclerosis occurs in some patients with
familial dysautonomia [212]. 1. Trachtman H, Weiss R, Bennett B, Griefer I. Isolated
hematuria in children: indications for a renal biopsy.
Kidney Int. 1984;25:94–9.
2. Schroder CH, Bontemps CM, Assmann KJM,
Schuurmans-Stekhoven JH, Foidart JM, Monnens
Hereditary Glomerulopathies Without LAH, et al. Renal biopsy and family studies in 65 chil-
Extrarenal Symptoms dren with isolated hematuria. Acta Paediatr Scand.
1990;79:630–6.
3. Piqueras AI, White RH, Raafat F, Moghal N, Milford
Fibronectin Glomerulopathy
DV. Renal biopsy diagnosis in children presenting
with hematuria. Pediatr Nephrol. 1998;12:386–91.
Fibronectin glomerulopathy is associated with 4. Miner JH. The glomerular basement membrane. Exp
massive accumulation of fibronectin derived Cell Res. 2012;318(9):973–8.
5. Hudson BG. The molecular basis of Goodpasture and
mainly from the soluble plasma isoform, transmit-
Alport syndromes: beacons for the discovery of the
ted as an autosomal dominant trait [213–215]. A collagen IV family. J Am Soc Nephrol. 2004;15
substantial fraction of cases arises from mutations (10):2514–27.
in the gene FN1 [216], which is located on chro- 6. Yoshioka K, Hino S, Takemura T, Maki S,
Wieslander J, Takekoshi Y, et al. Type IV Collagen
mosome 2 and encodes fibronectin, but another
a5 chain: normal distribution and abnormalities in
locus appears to exist at chromosome 1q32 X-linked Alport syndrome revealed by monoclonal
[217]. Clinically, the disease is characterized by antibody. Am J Pathol. 1994;144:986–96.
proteinuria of variable magnitude, typically first 7. Peissel B, Geng L, Kalluri R, Kashtan C, Rennke HG,
Gallo GR, et al. Comparative distribution of the a1
observed in early adulthood. Subsequently,
(IV), a5(IV) and a6(IV) collagen chains in normal
patients develop hypertension and renal insuffi- human adult and fetal tissues and in kidneys from
ciency, with gradual progression to ESRD. Recur- X-linked Alport syndrome patients. J Clin Invest.
rence after kidney transplantation has been 1995;96:1948–57.
8. Poschl E, Pollner R, Kuhn K. The genes for the alpha
described [218].
1(IV) and alpha 2(IV) chains of human basement
membrane collagen type IV are arranged head-to-
head and separated by a bidirectional promoter of
Mitochondrial Cytopathies unique structure. EMBO J. 1988;7(9):2687–95.
9. Segal Y, Zhuang L, Rondeau E, Sraer JD, Zhou
J. Regulation of the paired type IV collagen genes
Mitochondrial cytopathies are a heterogeneous COL4A5 and COL4A6. Role of the proximal pro-
group of disorders resulting from genetic defects moter region. J Biol Chem. 2001;276(15):11791–7.
in enzymatic complexes of the respiratory chain, 10. Guthrie LG. “Idiopathic”, or congenital, hereditary
and familial hematuria. Lancet. 1902;1:1243–6.
leading to impaired oxidative phosphorylation
11. Hurst AF. Hereditary familial congenital
and energy production. The most common renal haemorrhagic nephritis occurring in sixteen individ-
defect is the de Toni-Debre-Fanconi syndrome, uals in three generations. Guy’s Hosp Rep.
associated with nonspecific abnormalities in 1923;3:368–70.
12. Alport AC. Hereditary familial congenital
tubules and interstitium on renal biopsy, but glo-
haemorrhagic nephritis. Br Med J. 1927;1:504–6.
merular lesions such as FSGS, collapsing 13. Hinglais N, Grunfeld J-P, Bois LE. Characteristic
glomerulopathy, and crescentic glomerulonephri- ultrastructural lesion of the glomerular basement
tis may also occur [219]. Proteinuria and FSGS membrane in progressive hereditary nephritis
(Alport’s syndrome). Lab Invest. 1972;27:473–87.
have been found in patients with maternally
14. Spear GS, Slusser RJ. Alport’s syndrome: emphasiz-
inherited diabetes and/or deafness and in patients ing electron microscopic studies of the glomerulus.
with complete or incomplete MELAS syndrome Am J Pathol. 1972;69:213–22.
796 M.N. Rheault and C.E. Kashtan

15. Churg J, Sherman RL. Pathologic characteristics of 28. Crockett DK, Pont-Kingdon G, Gedge F, Sumner K,
hereditary nephritis. Arch Pathol. 1973;95:374–9. Seamons R, Lyon E. The Alport syndrome COL4A5
16. Kashtan C, Fish AJ, Kleppel M, Yoshioka K, Michael variant database. Hum Mutat. 2010;31(8):E1652–7.
AF. Nephritogenic antigen determinants in epidermal 29. Lemmink HH, Schroder CH, Monnens LA, Smeets
and renal basement membranes of kindreds with HJ. The clinical spectrum of type IV collagen muta-
Alport-type familial nephritis. J Clin Invest. tions. Hum Mutat. 1997;9(6):477–99.
1986;78:1035–44. 30. Jais JP, Knebelmann B, Giatras I, De Marchi M,
17. McCoy RC, Johnson HK, Stone WJ, Wilson Rizzoni G, Renieri A, et al. X-linked Alport syn-
CB. Absence of nephritogenic GBM antigen(s) in drome: natural history in 195 families and genotype-
some patients with hereditary nephritis. Kidney Int. phenotype correlations in males. J Am Soc Nephrol.
1982;21:642–52. 2000;11(4):649–57.
18. Olson DL, Anand SK, Landing BH, Heuser E, 31. Gross O, Netzer KO, Lambrecht R, Seibold S, Weber
Grushkin CM, Lieberman E. Diagnosis of hereditary M. Meta-analysis of genotype-phenotype correlation
nephritis by failure of glomeruli to bind anti- in X-linked Alport syndrome: impact on clinical
glomerular basement membrane antibodies. J Pediatr. counseling. Nephrol Dial Transplant.
1980;96:697–9. 2002;17:1218–27.
19. Atkin CL, Hasstedt SJ, Menlove L, Cannon L, 32. Tsiakkis D, Pieri M, Koupepidou P, Demosthenous P,
Kirschner N, Schwartz C, et al. Mapping of Alport Panayidou K, Deltas C. Genotype-phenotype correla-
syndrome to the long arm of the X chromosome. Am J tion in X-linked Alport syndrome patients carrying
Hum Genet. 1988;42:249–55. missense mutations in the collagenous domain of
20. Hostikka SL, Eddy RL, Byers MG, Hoyhtya M, COL4A5. Clin Genet. 2012;82(3):297–9.
Shows TB, Tryggvason K. Identification of a distinct 33. Mochizuki T, Lemmink HH, Mariyama M,
type IV collagen alpha chain with restricted kidney Antignac C, Gubler MC, Pirson Y,
distribution and assignment of its gene to the locus of et al. Identification of mutations in the alpha 3
X chromosome-linked Alport syndrome. Proc Natl (IV) and alpha 4(IV) collagen genes in autosomal
Acad Sci USA. 1990;87(4):1606–10. recessive Alport syndrome. Nat Genet. 1994;8
21. Barker DF, Hostikka SL, Zhou J, Chow LT, Oliphant (1):77–81.
AR, Gerken SC, et al. Identification of mutations in 34. Lemmink HH, Mochizuki T, van den Heuvel LP,
the COL4A5 collagen gene in Alport syndrome. Sci- Schroder CH, Barrientos A, Monnens LA,
ence. 1990;248:1224–7. et al. Mutations in the type IV collagen
22. Gunwar S, Ballester F, Noelken ME, Sado Y, alpha 3 (COL4A3) gene in autosomal recessive
Ninomiya Y, Hudson BG. Glomerular basement Alport syndrome. Hum Mol Genet. 1994;3
membrane. Identification of a novel disulfide-cross- (8):1269–73.
linked network of alpha3, alpha4, and alpha5 chains 35. Longo I, Porcedda P, Mari F, Giachino D, Meloni I,
of type IV collagen and its implications for the path- Deplano C, et al. COL4A3/COL4A4 mutations: from
ogenesis of Alport syndrome. J Biol Chem. 1998;273 familial hematuria to autosomal-dominant or reces-
(15):8767–75. sive Alport syndrome. Kidney Int. 2002;61
23. Rheault MN, Kren SM, Hartich LA, Wall M, (6):1947–56.
Thomas W, Mesa HA, et al. X-inactivation modifies 36. Jais JP, Knebelmann B, Giatras I, De Marchi M,
disease severity in female carriers of murine X-linked Rizzoni G, Renieri A, et al. X-linked Alport syn-
Alport syndrome. Nephrol Dial Transplant. 2010;25 drome: natural history and genotype-phenotype cor-
(3):764–9. relations in girls and women belonging to
24. Storey H, Savige J, Sivakumar V, Abbs S, Flinter 195 families: a “European Community Alport Syn-
FA. COL4A3/COL4A4 mutations and features in drome Concerted Action” study. J Am Soc Nephrol.
individuals with autosomal recessive Alport syn- 2003;14:2603–10.
drome. J Am Soc Nephrol. 2013;24:1945–54. 37. Pochet JM, Bobrie G, Landais P, Goldfarb B,
25. Pescucci C, Mari F, Longo I, Vogiatzi P, Caselli R, Grunfeld JP. Renal prognosis in Alport’s and related
Scala E, et al. Autosomal-dominant Alport syndrome: syndromes: influence of the mode of inheritance.
natural history of a disease due to COL4A3 or Nephrol Dial Transplant. 1989;4(12):1016–21.
COL4A4 gene. Kidney Int. 2004;65(5):1598–603. 38. Colville DJ, Savige J. Alport syndrome. A review of
26. Fallerini C, Dosa L, Tita R, Del Prete D, Feriozzi S, the ocular manifestations. Ophthalmic Genet.
Gai G, et al. Unbiased next generation sequencing 1997;18(4):161–73.
analysis confirms the existence of autosomal domi- 39. Gubler M, Levy M, Broyer M, Naizot C, Gonzales G,
nant Alport syndrome in a relevant fraction of cases. Perrin D, et al. Alport’s syndrome: a report of 58 cases
Clin Genet. 2014;86:252–7. and a review of the literature. Am J Med.
27. Moriniere V, Dahan K, Hilbert P, Lison M, Lebbah S, 1981;70:493–505.
Topa A, et al. Improving mutation screening in famil- 40. Kashtan CE, Ding J, Gregory M, Gross O, Heidet L,
ial hematuric nephropathies through next generation Knebelmann B, et al. Clinical practice recommenda-
sequencing. J Am Soc Nephrol. 2014;25:2740–51. tions for the treatment of Alport syndrome: a
26 Inherited Glomerular Diseases 797

statement of the Alport Syndrome Research Collabo- 55. Kashtan CE, Gubler MC, Sisson-Ross S, Mauer
rative. Pediatr Nephrol. 2013;28(1):5–11. M. Chronology of renal scarring in males with Alport
41. Kim KH, Kim Y, Gubler MC, Steffes MW, Lane PH, syndrome. Pediatr Nephrol. 1998;12(4):269–74.
Kashtan CE, et al. Structural-functional relationships 56. Rumpelt HJ. Hereditary nephropathy (Alport syn-
in Alport syndrome. J Am Soc Nephrol. 1995;5 drome): correlation of clinical data with glomerular
(9):1659–68. basement membrane alterations. Clin Nephrol.
42. Rheault MN. Women and Alport syndrome. Pediatr 1980;13(5):203–7.
Nephrol. 2012;27(1):41–6. 57. Kashtan CE, Kleppel MM, Gubler
43. Grunfeld J-P, Noel LH, Hafez S, Droz D. Renal prog- MC. Immunohistologic findings in Alport syndrome.
nosis in women with hereditary nephritis. Clin Contrib Nephrol. 1996;117:142–53.
Nephrol. 1985;23:267–71. 58. Gubler MC, Knebelmann B, Beziau A, Broyer M,
44. Savige J, Liu J, DeBuc DC, Handa JT, Hageman GS, Pirson Y, Haddoum F, et al. Autosomal recessive
Wang YY, et al. Retinal basement membrane abnor- Alport syndrome: immunohistochemical study of
malities and the retinopathy of Alport syndrome. type IV collagen chain distribution. Kidney Int.
Invest Ophthalmol Vis Sci. 2010;51(3):1621–7. 1995;47(4):1142–7.
45. Rhys C, Snyers B, Pirson Y. Recurrent corneal ero- 59. van der Loop FT, Monnens LA, Schroder CH,
sion associated with Alport’s syndrome. Kidney Int. Lemmink HH, Breuning MH, Timmer ED,
1997;52:208–11. et al. Identification of COL4A5 defects in Alport’s
46. Burke JP, Clearkin LG, Talbot JF. Recurrent corneal syndrome by immunohistochemistry of skin. Kidney
epithelial erosions in Alport’s syndrome. Acta Int. 1999;55(4):1217–24.
Ophthalmol. 1991;69:555–7. 60. Massella L, Onetti Muda A, Faraggiana T, Bette C,
47. Teekhasaenee C, Nimmanit S, Wutthiphan S, Renieri A, Rizzoni G. Epidermal basement membrane
Vareesangthip K, Laohapand T, Malasitr P, alpha 5(IV) expression in females with Alport syn-
et al. Posterior polymorphous dystrophy and Alport drome and severity of renal disease. Kidney Int.
syndrome. Ophthalmology. 1991;98:1207–15. 2003;64(5):1787–91.
48. Tan R, Colville D, Wang YY, Rigby L, Savige 61. Wester DC, Atkin CL, Gregory MC. Alport syn-
J. Alport retinopathy results from “severe” COL4A5 drome: clinical update. J Am Acad Audiol.
mutations and predicts early renal failure. Clin J Am 1995;6:73–9.
Soc Nephrol. 2010;5(1):34–8. 62. Cosgrove D, Samuelson G, Meehan DT, Miller C,
49. Zhou J, Mochizuki T, Smeets H, Antignac C, McGee J, Walsh EJ, et al. Ultrastructural, physiolog-
Laurila P, de Paepe A, et al. Deletion of the paired ical, and molecular defects in the inner ear of a gene-
a5(IV) and a6(IV) collagen genes in inherited smooth knockout mouse model of autosomal Alport syn-
muscle tumors. Science. 1993;261:1167–9. drome. Hear Res. 1998;121:84–98.
50. Antignac C, Heidet L. Mutations in Alport syndrome 63. Harvey SJ, Mount R, Sado Y, Naito I, Ninomiya Y,
associated with diffuse esophageal leiomyomatosis. Harrison R, et al. The inner ear of dogs with X-linked
Contrib Nephrol. 1996;117:172–82. nephritis provides clues to the pathogenesis of hearing
51. Heidet L, Cai Y, Sado Y, Ninomiya Y, Thorner P, loss in X-linked Alport syndrome. Am J Pathol.
Guicharnaud L, et al. Diffuse leiomyomatosis 2001;159(3):1097–104.
associated with X-linked Alport syndrome: extracel- 64. Kleppel MM, Santi PA, Cameron JD, Wieslander J,
lular matrix study using immunohistochemistry Michael AF. Human tissue distribution of novel base-
and in situ hybridization. Lab Invest. 1997;76 ment membrane collagen. Am J Pathol.
(2):233–43. 1989;134:813–25.
52. Jonsson JJ, Renieri A, Gallagher PG, Kashtan CE, 65. Zehnder AF, Adams JC, Santi PA, Kristiansen AG,
Cherniske EM, Bruttini M, et al. Alport syndrome, Wacharasindhu C, Mann S, et al. Distribution of
mental retardation, midface hypoplasia, and type IV collagen in the cochlea in Alport syndrome.
elliptocytosis: a new X linked contiguous Arch Otolaryngol Head Neck Surg.
gene deletion syndrome? J Med Genet. 1998;35 2005;131:1007–13.
(4):273–8. 66. Merchant SN, Burgess BJ, Adams JC, Kashtan CE,
53. Vitelli F, Piccini M, Caroli F, Franco B, Malandrini A, Gregory MC, Santi PA, et al. Temporal bone histopa-
Pober B, et al. Identification and characterization of a thology in Alport syndrome. Laryngoscope. 2004;114
highly conserved protein absent in the Alport syn- (9):1609–18.
drome (A), mental retardation (M), midface hypopla- 67. Cheong HI, Kashtan CE, Kim Y, Kleppel MM,
sia (M), and elliptocytosis (E) contiguous gene Michael AF. Immunohistologic studies of type IV
deletion syndrome (AMME). Genomics. 1999;55 collagen in anterior lens capsules of patients with
(3):335–40. Alport syndrome. Lab Invest. 1994;70:553–7.
54. Kashtan CE, Segal Y, Flinter F, Makanjuola D, Gan 68. Ohkubo S, Takeda H, Higashide T, Ito M, Sakurai M,
JS, Watnick T. Aortic abnormalities in males with Shirao Y, et al. Immunohistochemical and molecular
Alport syndrome. Nephrol Dial Transplant. 2010;25 genetic evidence for type IV collagen alpha 5 chain
(11):3554–60. abnormality in the anterior lenticonus associated with
798 M.N. Rheault and C.E. Kashtan

Alport syndrome. Arch Ophthalmol. 2003;121 protein-7 inhibits progression of chronic renal fibrosis
(6):846–50. associated with two genetic mouse models. Am J
69. Chen L, Miyamura N, Ninomiya Y, Handa Physiol Renal Physiol. 2003;285(6):F1060–7.
JT. Distribution of the collagen IV isoforms in 82. Zeisberg M, Khurana M, Rao VH, Cosgrove D,
human Bruch’s membrane. Br J Ophthalmol. Rougier JP, Werner MC, et al. Stage-specific action
2003;87(2):212–5. of matrix metalloproteinases influences progressive
70. Gyoneva L, Segal Y, Dorfman KD, Barocas hereditary kidney disease. PLoS Med. 2006;3(4):
VH. Mechanical response of wild-type and Alport e100.
murine lens capsules during osmotic swelling. Exp 83. Sugimoto H, Mundel TM, Sund M, Xie L,
Eye Res. 2013;113:87–91. Cosgrove D, Kalluri R. Bone-marrow-derived stem
71. Sonarkhan S, Ramappa M, Chaurasia S, Mulay cells repair basement membrane collagen defects and
K. Bilateral anterior lenticonus in a case of Alport reverse genetic kidney disease. Proc Natl Acad Sci U
syndrome: a clinical and histopathological correlation S A. 2006;103(19):7321–6.
after successful clear lens extraction. BMJ Case Rep. 84. Gross O, Borza DB, Anders HJ, Licht C, Weber M,
2014 doi:10.1136/bcr-2013-202036. Segerer S, et al. Stem cell therapy for Alport syn-
72. Kato T, Watanabe Y, Nakayasu K, Kanai A, Yajima drome: the hope beyond the hype. Nephrol Dial
Y. The ultrastructure of the lens capsule abnormalities Transplant. 2009;24(3):731–4.
in Alport’s syndrome. Jpn J Ophthalmol. 85. Chen D, Jefferson B, Harvey SJ, Zheng K, Gartley
1998;42:401–5. CJ, Jacobs RM, et al. Cyclosporine a slows the pro-
73. Kiryluk K, Novak J. The genetics and gressive renal disease of Alport syndrome (X-linked
immunobiology of IgA nephropathy. J Clin Invest. hereditary nephritis): results from a canine model.
2014;124(6):2325–32. J Am Soc Nephrol. 2003;14(3):690–8.
74. Redahan L, Doyle R, O’Shaughnessy M, Dorman A, 86. Callis L, Vila A, Carrera M, Nieto J. Long-term
Little M, Conlon P. Familial MPGN – a case series: a effects of cyclosporine A in Alport’s syndrome. Kid-
clinical description of familial membranoproliferative ney Int. 1999;55(3):1051–6.
glomerulonephritis amongst three Irish families. Ren 87. Charbit M, Gubler MC, Dechaux M, Gagnadoux MF,
Fail. 2014;36(8):1333–6. Grunfeld JP, Niaudet P. Cyclosporin therapy in
75. Martin P, Heiskari N, Zhou J, Leinonen A, patients with Alport syndrome. Pediatr Nephrol.
Tumelius T, Hertz JM, et al. High mutation detection 2007;22(1):57–63.
rate in the COL4A5 collagen gene in suspected Alport 88. Massella L, Muda AO, Legato A, Di Zazzo G,
syndrome using PCR and direct DNA sequencing. J Giannakakis K, Emma F. Cyclosporine A treatment
Am Soc Nephrol. 1998;9:2291–301. in patients with Alport syndrome: a single-center
76. Gross O, Schulze-Lohoff E, Koepke ML, experience. Pediatr Nephrol. 2010;25(7):1269–75.
Beirowski B, Addicks K, Bloch W, 89. Cohen EP, Lemann J. In hereditary nephritis
et al. Antifibrotic, nephroprotective potential of ACE angiotensin-converting enzyme inhibition decreases
inhibitor vs AT1 antagonist in a murine model of renal proteinuria and may slow the rate of progression. Am
fibrosis. Nephrol Dial Transplant. 2004;19 J Kidney Dis. 1996;27:199–203.
(7):1716–23. 90. Proesmans W, Van Dyck M. Enalapril in children with
77. Gross O, Beirowski B, Koepke ML, Kuck J, Alport syndrome. Pediatr Nephrol. 2004;19
Reiner M, Addicks K, et al. Preemptive ramipril ther- (3):271–5.
apy delays renal failure and reduces renal fibrosis in 91. Webb NJ, Lam C, Shahinfar S, Strehlau J, Wells TG,
COL4A3-knockout mice with Alport syndrome. Kid- Gleim GW, et al. Efficacy and safety of losartan in
ney Int. 2003;63(2):438–46. children with Alport syndrome – results from a
78. Gross O, Koepke ML, Beirowski B, Schulze-Lohoff- subgroup analysis of a prospective, randomized,
E, Segerer S, Weber M. Nephroprotection by placebo- or amlodipine-controlled trial. Nephrol
antifibrotic and anti-inflammatory effects of the Dial Transplant. 2011;26(8):2521–6.
vasopeptidase inhibitor AVE7688. Kidney Int. 92. Webb NJ, Shahinfar S, Wells TG, Massaad R, Gleim
2005;68:456–63. GW, McCrary Sisk C, et al. Losartan and enalapril are
79. Sayers R, Kalluri R, Rodgers KD, Shield CF, Meehan comparable in reducing proteinuria in children with
DT, Cosgrove D. Role for transforming growth Alport syndrome. Pediatr Nephrol. 2013;28
factor-beta 1 in Alport renal disease progression. Kid- (5):737–43.
ney Int. 1999;56:1662–73. 93. Gross O, Licht C, Anders HJ, Hoppe B, Beck B,
80. Ninichuk V, Gross O, Reichel C, Kandoga A, Pawar Tonshoff B, et al. Early angiotensin-converting
RD, Ciubar R, et al. Delayed chemokine receptor enzyme inhibition in Alport syndrome delays renal
1 blockade prolongs survival in collagen failure and improves life expectancy. Kidney Int.
4A3-deficient mice with Alport disease. J Am Soc 2012;81(5):494–501.
Nephrol. 2005;16:977–85. 94. Gross O, Friede T, Hilgers R, Gorlitz A, Gavenis K,
81. Zeisberg M, Bottiglio C, Kumar N, Maeshima Y, Ahmed R, et al. Safety and efficacy of the
Strutz F, Muller GA, et al. Bone morphogenic ACE-inhibitor ramipril in Alport syndrome: the
26 Inherited Glomerular Diseases 799

double-blind, randomized, placebo-controlled, multi- 109. Gregory MC. The clinical features of thin basement
center phase III EARLY PRO-TECT Alport trial in membrane nephropathy. Semin Nephrol. 2005;25
pediatric patients. ISRN Pediatr. 2012;2012:436046. (3):140–5.
95. Temme J, Kramer A, Jager KJ, Lange K, Peters F, 110. Auwardt R, Savige J, Wilson D. A comparison of the
Muller GA, et al. Outcomes of male patients with clinical and laboratory features of thin basement
Alport syndrome undergoing renal replacement ther- membrane disease (TBMD) and IgA glomerulone-
apy. Clin J Am Soc Nephrol. 2012;7(12):1969–76. phritis (IgA GN). Clin Nephrol. 1999;52(1):1–4.
96. Gross O, Weber M, Fries JW, Muller GA. Living 111. Nieuwhof CM, de Heer F, de Leeuw P, van Breda
donor kidney transplantation from relatives with Vriesman PJ. Thin GBM nephropathy: premature glo-
mild urinary abnormalities in Alport syndrome: merular obsolescence is associated with hypertension
long-term risk, benefit and outcome. Nephrol Dial and late onset renal failure. Kidney Int. 1997;51
Transplant. 2009;24(5):1626–30. (5):1596–601.
97. Kashtan CE. Renal transplantation in patients with 112. van Paassen P, van Breda Vriesman PJ, van Rie H,
Alport syndrome. Pediatr Transplant. 2006;10 Tervaert JW. Signs and symptoms of thin
(6):651–7. basement membrane nephropathy: a prospective
98. Brainwood D, Kashtan C, Gubler MC, Turner regional study on primary glomerular disease-The
AN. Targets of alloantibodies in Alport anti- Limburg Renal Registry. Kidney Int. 2004;66
glomerular basement membrane disease after renal (3):909–13.
transplantation. Kidney Int. 1998;53:762–6. 113. Voskarides K, Damianou L, Neocleous V, Zouvani I,
99. Dehan P, Van Den Heuvel LPWJ, Smeets HJM, Christodoulidou S, Hadjiconstantinou V,
Tryggvason K, Foidart J-M. Identification of post- et al. COL4A3/COL4A4 mutations producing focal
transplant anti-a5(IV) collagen alloantibodies in segmental glomerulosclerosis and renal failure in thin
X-linked Alport syndrome. Nephrol Dial Transplant. basement membrane nephropathy. J Am Soc Nephrol.
1996;11:1983–8. 2007;18(11):3004–16.
100. Kalluri R, van den Heuvel LP, Smeets HJM, Schroder 114. Vogler C, McAdams AJ, Homan SM. Glomerular
CH, Lemmink HH, Boutaud A, et al. A COL4A3 basement membrane and lamina densa in infants and
gene mutation and post-transplant anti-a3 children: an ultrastructural evaluation. Pediatr Pathol.
(IV) collagen alloantibodies in Alport syndrome. Kid- 1987;7:527–34.
ney Int. 1995;47:1199–204. 115. Steffes MW, Barbosa J, Basgen JM, Sutherland DER,
101. Wang XP, Fogo AB, Colon S, Giannico G, Abul-Ezz Najarian JS, Mauer SM. Quantitative glomerular mor-
SR, Miner JH, et al. Distinct epitopes for anti- phology of the normal human kidney. Kidney Int.
glomerular basement membrane Alport alloanti- 1983;49:82–6.
bodies and goodpasture autoantibodies within the 116. Lang S, Stevenson B, Risdon RA. Thin basement
noncollagenous domain of {alpha}3(IV) collagen: a membrane nephropathy as a cause of recurrent
janus-faced antigen. J Am Soc Nephrol. haematuria in childhood. Histopathology.
2005;16:3563–71. 1990;16:331–7.
102. Marks MI, Drummond KN. Benign familial hematu- 117. Milanesi C, Rizzoni G, Braggion F, Galdiolo
ria. Pediatrics. 1969;44:590–3. D. Electron microscopy for measurement of glomer-
103. McConville JM, West CD, McAdams AJ. Familial ular basement membrane width in children with
and nonfamilial benign hematuria. J Pediatr. benign familial hematuria. Appl Pathol.
1966;69:207–14. 1984;2:199–204.
104. Pardo V, Berian MG, Levi DF, Strauss J. Benign 118. Dische FE. Measurement of glomerular basement
primary hematuria: clinicopathologic study of membrane thickness and its application to the diag-
65 patients. Am J Med. 1979;67:817–22. nosis of thin-membrane nephropathy. Arch Pathol
105. Aarons I, Smith PS, Davies RA, Woodroffe AJ, Lab Med. 1992;116:43–9.
Clarkson AR. Thin membrane nephropathy: a 119. Gubler MC, Beaufils H, Noel LH, Habib
clinico-pathological study. Clin Nephrol. 1989;32 R. Significance of thin glomerular basement mem-
(4):151–8. branes in hematuric children. Contrib Nephrol.
106. Tiebosch ATMG, Frederik PM, van Breda Vriesman 1990;80:147–56.
PJC, Mooy JMV, van Rie H, van de Wiel TWM, 120. Pettersson E, Tornroth T, Wieslander J. Abnormally
et al. Thin-basement-membrane nephropathy in thin glomerular basement membrane and the
adults with persistent hematuria. N Engl J Med. Goodpasture epitope. Clin Nephrol. 1990;33:105–9.
1989;320:14–8. 121. Blumenthal SS, Fritsche C, Lemann J. Establishing
107. Haas M. Thin glomerular basement membrane the diagnosis of benign familial hematuria: the impor-
nephropathy: incidence in 3471 consecutive renal tance of examining the urine sediment of family mem-
biopsies examined by electron microscopy. Arch bers. JAMA. 1988;259:2263–6.
Pathol Lab Med. 2006;130(5):699–706. 122. Jefferson JA, Lemmink HH, Hughes AE, Hill CM,
108. Tryggvason K, Patrakka J. Thin basement membrane Smeets HJ, Doherty CC, et al. Autosomal dominant
nephropathy. J Am Soc Nephrol. 2006;17(3):813–22. Alport syndrome linked to the type IV collage alpha 3
800 M.N. Rheault and C.E. Kashtan

and alpha 4 genes (COL4A3 and COL4A4). Nephrol 136. Sweeney E, Fryer A, Mountford R, Green A, McIn-
Dial Transplant. 1997;12(8):1595–9. tosh I. Nail patella syndrome: a review of the pheno-
123. Plaisier E, Chen Z, Gekeler F, Benhassine S, type aided by developmental biology. J Med Genet.
Dahan K, Marro B, et al. Novel COL4A1 mutations 2003;40(3):153–62.
associated with HANAC syndrome: a role for the 137. Bongers EM, Huysmans FT, Levtchenko E, de Rooy
triple helical CB3[IV] domain. Am J Med Genet JW, Blickman JG, Admiraal RJ, et al. Genotype-
A. 2010;152A(10):2550–5. phenotype studies in nail-patella syndrome show
124. Weng YC, Sonni A, Labelle-Dumais C, de Leau M, that LMX1B mutation location is involved in the
Kauffman WB, Jeanne M, et al. COL4A1 mutations risk of developing nephropathy. Eur J Hum Genet.
in patients with sporadic late-onset intracerebral hem- 2005;13(8):935–46.
orrhage. Ann Neurol. 2012;71(4):470–7. 138. Chen H, Lun Y, Ovchinnikov D, Kokubo H, Oberg
125. Zhang KW, Tonna S, Wang YY, Rana K, Padavarat S, KC, Pepicelli CV, et al. Limb and kidney defects in
Savige J. Do mutations in COL4A1 or COL4A2 Lmx1b mutant mice suggest an involvement of
cause thin basement membrane nephropathy LMX1B in human nail patella syndrome. Nat Genet.
(TBMN)? Pediatr Nephrol. 2007;22(5):645–51. 1998;19(1):51–5.
126. Pierson M, Cordier J, Hervouuet F, Rauber 139. Dreyer SD, Zhou G, Baldini A, Winterpacht A,
G. [An unusual congenital and familial congenital Zabel B, Cole W, et al. Mutations in LMX1B cause
malformative combination involving the eye and kid- abnormal skeletal patterning and renal dysplasia in
ney]. J Genet Hum. 1963;12:184–213. nail patella syndrome. Nat Genet. 1998;19(1):47–50.
127. Zenker M, Aigner T, Wendler O, Tralau T, 140. Vollrath D, Jaramillo-Babb VL, Clough MV,
Muntefering H, Fenski R, et al. Human laminin McIntosh I, Scott KM, Lichter PR, et al. Loss-of-
beta2 deficiency causes congenital nephrosis with function mutations in the LIM-homeodomain gene,
mesangial sclerosis and distinct eye abnormalities. LMX1B, in nail-patella syndrome. Hum Mol Genet.
Hum Mol Genet. 2004;13(21):2625–32. 1998;7(7):1091–8.
128. Hofstaetter C, Neumann I, Lennert T, Dudenhausen 141. Hamlington JD, Jones C, McIntosh I. Twenty-two
JW. Prenatal diagnosis of diffuse mesangial novel LMX1B mutations identified in nail patella
glomerulosclerosis by ultrasonography: a longitudi- syndrome (NPS) patients. Hum Mutat. 2001;18
nal study of a case in an affected family. Fetal Diagn (5):458.
Ther. 1996;11(2):126–31. 142. Knoers NV, Bongers EM, van Beersum SE, Lommen
129. Glastre C, Cochat P, Bouvier R, Colon S, Cottin X, EJ, van Bokhoven H, Hol FA. Nail-patella syndrome:
Giffon D, et al. Familial infantile nephrotic syndrome identification of mutations in the LMX1B gene in
with ocular abnormalities. Pediatr Nephrol. 1990;4 Dutch families. J Am Soc Nephrol. 2000;11
(4):340–2. (9):1762–6.
130. Swietlinski J, Maruniak-Chudek I, Niemir ZI, 143. McIntosh I, Dreyer SD, Clough MV, Dunston JA,
Wozniak A, Wilinska M, Zacharzewska J. A case of Eyaid W, Roig CM, et al. Mutation analysis of
atypical congenital nephrotic syndrome. Pediatr LMX1B gene in nail-patella syndrome patients. Am
Nephrol. 2004;19(3):349–52. J Hum Genet. 1998;63(6):1651–8.
131. Mohney BG, Pulido JS, Lindor NM, Hogan MC, 144. Dreyer SD, Morello R, German MS, Zabel B,
Consugar MB, Peters J, et al. A novel mutation of Winterpacht A, Lunstrum GP, et al. LMX1B
LAMB2 in a multigenerational mennonite family transactivation and expression in nail-patella syn-
reveals a new phenotypic variant of Pierson syn- drome. Hum Mol Genet. 2000;9(7):1067–74.
drome. Ophthalmology. 2011;118(6):1137–44. 145. Morello R, Zhou G, Dreyer SD, Harvey SJ,
132. Matejas V, Hinkes B, Alkandari F, Al-Gazali L, Ninomiya Y, Thorner PS, et al. Regulation of glomer-
Annexstad E, Aytac MB, et al. Mutations in the ular basement membrane collagen expression by
human laminin beta2 (LAMB2) gene and the associ- LMX1B contributes to renal disease in nail patella
ated phenotypic spectrum. Hum Mutat. 2010;31 syndrome. Nat Genet. 2001;27(2):205–8.
(9):992–1002. 146. Miner JH, Morello R, Andrews KL, Li C, Antignac C,
133. Jarad G, Cunningham J, Shaw AS, Miner JH. Shaw AS, et al. Transcriptional induction of slit
Proteinuria precedes podocyte abnormalities diaphragm genes by Lmx1b is required in
inLamb2/ mice, implicating the glomerular base- podocyte differentiation. J Clin Invest. 2002;109
ment membrane as an albumin barrier. J Clin Invest. (8):1065–72.
2006;116(8):2272–9. 147. Rohr C, Prestel J, Heidet L, Hosser H, Kriz W,
134. Mino RA, Mino VH, Livingstone RG. Osseous dys- Johnson RL, et al. The LIM-homeodomain transcrip-
plasia and dystrophy of the nails; review of the liter- tion factor Lmx1b plays a crucial role in podocytes.
ature and report of a case. Am J Roentgenol Radium J Clin Invest. 2002;109(8):1073–82.
Ther. 1948;60(5):633–41. 148. Heidet L, Bongers EM, Sich M, Zhang SY, Loirat C,
135. Hawkins CF, Smith OE. Renal dysplasia in a family Meyrier A, et al. In vivo expression of putative
with multiple hereditary abnormalities including iliac LMX1B targets in nail-patella syndrome kidneys.
horns. Lancet. 1950;1(6609):803–8. Am J Pathol. 2003;163(1):145–55.
26 Inherited Glomerular Diseases 801

149. Burghardt T, Kastner J, Suleiman H, Rivera-Milla E, syndrome) to chromosome 22q11-13. Am J Hum


Stepanova N, Lottaz C, et al. LMX1B is essential for Genet. 1999;65(6):1711–7.
the maintenance of differentiated podocytes in adult 164. Toren A, Rozenfeld-Granot G, Rocca B, Epstein CJ,
kidneys. J Am Soc Nephrol. 2013;24(11):1830–48. Amariglio N, Laghi F, et al. Autosomal-dominant
150. Boyer O, Woerner S, Yang F, Oakeley EJ, Linghu B, giant platelet syndromes: a hint of the same genetic
Gribouval O, et al. LMX1B mutations cause heredi- defect as in Fechtner syndrome owing to a similar
tary FSGS without extrarenal involvement. J Am Soc genetic linkage to chromosome 22q11-13. Blood.
Nephrol. 2013;24(8):1216–22. 2000;96(10):3447–51.
151. Sweeney E, Fryer AE, Mountford RC, Green AJ, 165. Lalwani AK, Goldstein JA, Kelley MJ, Luxford W,
McIntosh I. Nail patella syndrome: a study of Castelein CM, Mhatre AN. Human nonsyndromic
123 patients from 43 British families and the detection hereditary deafness DFNA17 is due to a mutation in
of 16 novel mutations of LMX1B. Am J Hum Genet. nonmuscle myosin MYH9. Am J Hum Genet.
2001;69(4):189. 2000;67(5):1121–8.
152. Bongers EM, Gubler MC, Knoers NV. Nail-patella 166. Seri M, Cusano R, Gangarossa S, Caridi G, Bordo D,
syndrome. Overview on clinical and molecular find- Lo Nigro C, et al. Mutations in MYH9 result in the
ings. Pediatr Nephrol. 2002;17(9):703–12. May-Hegglin anomaly, and Fechtner and Sebastian
153. Ben-Bassat M, Cohen L, Rosenfeld J. The glomerular syndromes. The May-Hegglin/Fechtner Syndrome
basement membrane in the nail-patella syndrome. Consortium. Nat Genet. 2000;26(1):103–5.
Arch Pathol. 1971;92(5):350–5. 167. Seri M, Pecci A, Di Bari F, Cusano R, Savino M,
154. Hoyer JR, Michael AF, Vernier RL. Renal disease in Panza E, et al. MYH9-related disease: May-Hegglin
nail-patella syndrome: clinical and morphologic stud- anomaly, Sebastian syndrome, Fechtner syndrome,
ies. Kidney Int. 1972;2(4):231–8. and Epstein syndrome are not distinct entities but
155. Ikeda K, Yokoyama H, Tomosugi N, Kida H, represent a variable expression of a single illness.
Ooshima A, Kobayashi K. Primary glomerular fibro- Medicine. 2003;82(3):203–15.
sis: a new nephropathy caused by diffuse intra- 168. Seri M, Savino M, Bordo D, Cusano R, Rocca B,
glomerular increase in atypical type III collagen Meloni I, et al. Epstein syndrome: another renal dis-
fibers. Clin Nephrol. 1990;33(4):155–9. order with mutations in the nonmuscle myosin heavy
156. Gubler MC, Dommergues JP, Foulard M, chain 9 gene. Hum Genet. 2002;110(2):182–6.
Bensman A, Leroy JP, Broyer M, et al. Collagen 169. Arrondel C, Vodovar N, Knebelmann B, Grunfeld JP,
type III glomerulopathy: a new type of hereditary Gubler MC, Antignac C, et al. Expression of the
nephropathy. Pediatr Nephrol. 1993;7(4):354–60. nonmuscle myosin heavy chain IIA in the human
157. Salcedo JR. An autosomal recessive disorder with kidney and screening for MYH9 mutations in Epstein
glomerular basement membrane abnormalities similar and Fechtner syndromes. J Am Soc Nephrol. 2002;13
to those seen in the nail patella syndrome: report of a (1):65–74.
kindred. Am J Med Genet. 1984;19(3):579–84. 170. Najafian B, Mauer M, Hopkin RJ, Svarstad E. Renal
158. Imbasciati E, Gherardi G, Morozumi K, Gudat F, complications of Fabry disease in children. Pediatr
Epper R, Basler V, et al. Collagen type III Nephrol. 2013;28(5):679–87.
glomerulopathy: a new idiopathic glomerular disease. 171. Weidemann F, Kramer J, Duning T, Lenders M,
Am J Nephrol. 1991;11(5):422–9. Canaan-Kuhl S, Krebs A, et al. Patients with Fabry
159. Epstein CJ, Sahud MA, Piel CF, Goodman JR, disease after enzyme replacement therapy dose reduc-
Bernfield MR, Kushner JH, et al. Hereditary tion versus treatment switch. J Am Soc Nephrol.
macrothrombocytopathia, nephritis and deafness. 2014;25(4):837–49.
Am J Med. 1972;52(3):299–310. 172. Desnick RJ, Banikazemi M, Wasserstein M. Enzyme
160. Peterson LC, Rao KV, Crosson JT, White replacement therapy for Fabry disease, an inherited
JG. Fechtner syndrome – a variant of Alport’s syn- nephropathy. Clin Nephrol. 2002;57(1):1–8.
drome with leukocyte inclusions and macrothrombo- 173. Germain DP, Waldek S, Banikazemi M, Bushinsky
cytopenia. Blood. 1985;65(2):397–406. DA, Charrow J, Desnick RJ, et al. Sustained, long-
161. Clare NM, Montiel MM, Lifschitz MD, Bannayan term renal stabilization after 54 months of agalsidase
GA. Alport’s syndrome associated with macrothrom- beta therapy in patients with Fabry disease. J Am Soc
bopathic thrombocytopenia. Am J Clin Pathol. Nephrol. 2007;18(5):1547–57.
1979;72(1):111–7. 174. Maroteaux P, Humbel R, Strecker G, Michalski JC,
162. Naito I, Nomura S, Inoue S, Kagawa M, Kawai S, Mande R. A new type of sialidosis with kidney dis-
Gunshin Y, et al. Normal distribution of collagen IV in ease: nephrosialidosis. I. Clinical, radiological and
renal basement membranes in Epstein’s syndrome. J nosological study. Arch Fr Pediatr. 1978;35
Clin Pathol. 1997;50(11):919–22. (8):819–29.
163. Toren A, Amariglio N, Rozenfeld-Granot G, Simon 175. Chen W, Yang S, Shi H, Guan W, Dong Y, Wang Y,
AJ, Brok-Simoni F, Pras E, et al. Genetic linkage of et al. Histological studies of renal biopsy in a boy with
autosomal-dominant Alport syndrome with leukocyte nephrosialidosis. Ultrastruct Pathol. 2011;35
inclusions and macrothrombocytopenia (Fechtner (4):168–71.
802 M.N. Rheault and C.E. Kashtan

176. Kashtan CE, Nevins TE, Posalaky Z, Vernier RL, Fish involvement. Adv Nephrol Necker Hosp.
AJ. Proteinuria in a child with sialidosis: case report 1981;10:167–85.
and histological studies. Pediatr Nephrol. 1989;3 191. Hirashio S, Ueno T, Naito T, Masaki T. Characteristic
(2):166–74. kidney pathology, gene abnormality and treatments in
177. Faraggiana T, Churg J. Renal lipidoses: a review. LCAT deficiency. Clin Exp Nephrol. 2014;18
Hum Pathol. 1987;18(7):661–79. (2):189–93.
178. Varga J, Wohlgethan JR. The clinical and biochemical 192. Rajpal JS, Mapel-Lentz J, Mancera AD, Reed RC,
spectrum of hereditary amyloidosis. Semin Arthritis Kim Y, Chavers BM. Familial LCAT deficiency in a
Rheum. 1988;18(1):14–28. child with nephrotic syndrome. Clin Nephrol.
179. Portincasa P, Scaccianoce G, Palasciano G. Familial 2014;82:211–4.
Mediterranean fever: a fascinating model of inherited 193. Saito T, Sato H, Kudo K, Oikawa S, Shibata T,
autoinflammatory disorder. Eur J Clin Invest. 2013;43 Hara Y, et al. Lipoprotein glomerulopathy: glomerular
(12):1314–27. lipoprotein thrombi in a patient with hyperlipopro-
180. Dode C, Cuisset L, Delpech M, Grateau G. teinemia. Am J Kidney Dis. 1989;13(2):148–53.
TNFRSF1A-associated periodic syndrome 194. Liao MT, Tsai IJ, Cheng HT, Lin WC, Chang YW, Lin
(TRAPS), Muckle-Wells syndrome (MWS) and YH, et al. A rare cause of childhood-onset nephrotic
renal amyloidosis. J Nephrol. 2003;16(3):435–7. syndrome: lipoprotein glomerulopathy. Clin Nephrol.
181. Levy M, Gubler MC, Hadchouel M, Niaudet P, 2012;78(3):237–40.
Habib R, Odievre M. [Alpha-1-antitrypsin deficiency 195. Saito T, Matsunaga A, Ito K, Nakashima H. Topics in
and renal involvement]. Nephrologie. 1985;6 lipoprotein glomerulopathy: an overview. Clin Exp
(2):65–70. Nephrol. 2014;18(2):214–7.
182. Davis ID, Burke B, Freese D, Sharp HL, Kim Y. The 196. Miyata T, Sugiyama S, Nangaku M, Suzuki D,
pathologic spectrum of the nephropathy associated Uragami K, Inagi R, et al. Apolipoprotein E2/E5
with alpha 1-antitrypsin deficiency. Hum Pathol. variants in lipoprotein glomerulopathy recurred in
1992;23(1):57–62. transplanted kidney. J Am Soc Nephrol. 1999;10
183. Montanelli A, Mainardi E, Pini L, Corda L, Grassi (7):1590–5.
V. Alpha-1-antitrypsin deficiency and nephropathy. 197. Oikawa S, Matsunaga A, Saito T, Sato H, Seki T,
Nephron. 2002;90(1):114–5. Hoshi K, et al. Apolipoprotein E Sendai (arginine
184. Morris H, Morgan MD, Wood AM, Smith SW, 145!proline): a new variant associated with lipopro-
Ekeowa UI, Herrmann K, et al. ANCA-associated tein glomerulopathy. J Am Soc Nephrol. 1997;8
vasculitis is linked to carriage of the Z allele of (5):820–3.
alpha(1) antitrypsin and its polymers. Ann Rheum 198. Storm T, Zeitz C, Cases O, Amsellem S, Verroust PJ,
Dis. 2011;70(10):1851–6. Madsen M, et al. Detailed investigations of proximal
185. Elzouki AN, Lindgren S, Nilsson S, Veress B, tubular function in Imerslund-Grasbeck syndrome.
Eriksson S. Severe alpha1-antitrypsin deficiency BMC Med Genet. 2013;14:111.
(PiZ homozygosity) with membranoproliferative glo- 199. Grasbeck R. Imerslund-Grasbeck syndrome (selec-
merulonephritis and nephrotic syndrome, reversible tive vitamin B(12) malabsorption with proteinuria).
after orthotopic liver transplantation. J Hepatol. Orphanet J Rare Dis. 2006;1:17.
1997;26(6):1403–7. 200. Lupski JR, Reid JG, Gonzaga-Jauregui C, Rio
186. Li L, Krantz ID, Deng Y, Genin A, Banta AB, Collins Deiros D, Chen DC, Nazareth L, et al. Whole-genome
CC, et al. Alagille syndrome is caused by mutations in sequencing in a patient with Charcot-Marie-Tooth
human Jagged1, which encodes a ligand for Notch1. neuropathy. N Engl J Med. 2010;362(13):1181–91.
Nat Genet. 1997;16(3):243–51. 201. Gherardi R, Belghiti-Deprez D, Hirbec G, Bouche P,
187. McDaniell R, Warthen DM, Sanchez-Lara PA, Pai A, Weil B, Lagrue G. Focal glomerulosclerosis associ-
Krantz ID, Piccoli DA, et al. NOTCH2 mutations ated with Charcot-Marie-Tooth disease. Nephron.
cause Alagille syndrome, a heterogeneous disorder 1985;40(3):357–61.
of the notch signaling pathway. Am J Hum Genet. 202. Paul MD, Fernandez D, Pryse-Phillips W, Gault
2006;79(1):169–73. MH. Charcot-Marie-Tooth disease and nephropathy
188. Kamath BM, Podkameni G, Hutchinson AL, Leonard in a mother and daughter with a review of the litera-
LD, Gerfen J, Krantz ID, et al. Renal anomalies in ture. Nephron. 1990;54(1):80–5.
Alagille syndrome: a disease-defining feature. Am J 203. Boyer O, Nevo F, Plaisier E, Funalot B, Gribouval O,
Med Genet A. 2012;158A(1):85–9. Benoit G, et al. INF2 mutations in Charcot-Marie-
189. Habib R, Dommergues JP, Gubler MC, Hadchouel M, Tooth disease with glomerulopathy. N Engl J Med.
Gautier M, Odievre M, et al. Glomerular 2011;365(25):2377–88.
mesangiolipidosis in Alagille syndrome 204. Kraemer KH, Patronas NJ, Schiffmann R, Brooks BP,
(arteriohepatic dysplasia). Pediatr Nephrol. 1987;1 Tamura D, DiGiovanna JJ. Xeroderma pigmentosum,
(3):455–64. trichothiodystrophy and Cockayne syndrome: a com-
190. Gjone E. Familial lecithin:cholesterol acyltransferase plex genotype-phenotype relationship. Neuroscience.
deficiency – a new metabolic disease with renal 2007;145(4):1388–96.
26 Inherited Glomerular Diseases 803

205. Hirooka M, Hirota M, Kamada M. Renal lesions in predominant fibronectin deposits: a newly recognized
Cockayne syndrome. Pediatr Nephrol. 1988;2 hereditary disease. Kidney Int. 1995;48(1):163–70.
(2):239–43. 214. Assmann KJ, Koene RA, Wetzels JF. Familial
206. Shinohara O, Kubota C, Kimura M, Nishimura G, glomerulonephritis characterized by massive
Takahashi S. Essential osteolysis associated with deposits of fibronectin. Am J Kidney Dis. 1995;25
nephropathy, corneal opacity, and pulmonary steno- (5):781–91.
sis. Am J Med Genet. 1991;41(4):482–6. 215. Gemperle O, Neuweiler J, Reutter FW, Hildebrandt F,
207. Green JS, Parfrey PS, Harnett JD, Farid NR, Cramer Krapf R. Familial glomerulopathy with giant fibrillar
BC, Johnson G, et al. The cardinal manifestations of (fibronectin-positive) deposits: 15-year follow-up in a
Bardet-Biedl syndrome, a form of Laurence-Moon- large kindred. Am J Kidney Dis. 1996;28(5):668–75.
Biedl syndrome. N Engl J Med. 1989;321 216. Castelletti F, Donadelli R, Banterla F, Hildebrandt F,
(15):1002–9. Zipfel PF, Bresin E, et al. Mutations in FN1 cause
208. Harnett JD, Green JS, Cramer BC, Johnson G, glomerulopathy with fibronectin deposits. Proc Natl
Chafe L, McManamon P, et al. The spectrum of Acad Sci USA. 2008;105(7):2538–43.
renal disease in Laurence-Moon-Biedl syndrome. N 217. Vollmer M, Jung M, Ruschendorf F, Ruf R,
Engl J Med. 1988;319(10):615–8. Wienker T, Reis A, et al. The gene for human fibro-
209. M’Hamdi O, Ouertani I, Chaabouni-Bouhamed nectin glomerulopathy maps to 1q32, in the region of
H. Update on the genetics of Bardet-Biedl syndrome. the regulation of complement activation gene cluster.
Mol Syndromol. 2014;5(2):51–6. Am J Hum Genet. 1998;63(6):1724–31.
210. Marshall JD, Maffei P, Collin GB, Naggert 218. Otsuka Y, Takeda A, Horike K, Inaguma D, Goto N,
JK. Alstrom syndrome: genetics and clinical over- Watarai Y, et al. A recurrent fibronectin
view. Curr Genomics. 2011;12(3):225–35. glomerulopathy in a renal transplant patient: a case
211. Goldstein JL, Fialkow PJ. The Alstrom syndrome report. Clin Transplant. 2012;26 Suppl 24:58–63.
Report of three cases with further delineation of the 219. Emma F, Bertini E, Salviati L, Montini G. Renal
clinical, pathophysiological, and genetic aspects of involvement in mitochondrial cytopathies. Pediatr
the disorder. Medicine. 1973;52(1):53–71. Nephrol. 2012;27(4):539–50.
212. Elkayam L, Matalon A, Tseng CH, Axelrod 220. Seidowsky A, Hoffmann M, Glowacki F, Dhaenens
F. Prevalence and severity of renal disease in familial CM, Devaux JP, de Sainte Foy CL, et al. Renal
dysautonomia. Am J Kidney Dis. 2006;48(5):780–6. involvement in MELAS syndrome – a series of
213. Strom EH, Banfi G, Krapf R, Abt AB, Mazzucco G, 5 cases and review of the literature. Clin Nephrol.
Monga G, et al. Glomerulopathy associated with 2013;80(6):456–63.
Idiopathic Nephrotic Syndrome
in Children: Genetic Aspects 27
Olivia Boyer, Kálmán Tory, Eduardo Machuca, and
Corinne Antignac

Contents Autosomal Dominant Forms of Isolated


SRNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 814
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 806 Mutations in ACTN4 Encoding α-Actinin-4 . . . . . . . . 814
Autosomal Recessive Forms of Isolated Mutations in TRPC6 Encoding the Transient
SRNS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 810 Receptor Potential Cation Channel 6 . . . . . . . . . . . . . . . . 815
Mutations in NPHS2 Encoding Podocin . . . . . . . . . . . . 810 Mutations in INF2 Encoding the Inverted
Mutations in NPHS1 Encoding Nephrin . . . . . . . . . . . . 812 Formin 2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 815
Mutations in PLCE1 Encoding Phospholipase C Less Commonly Mutated Genes . . . . . . . . . . . . . . . . . . . . 816
Epsilon 1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 812 NGS Findings and Redefinition of Known
Less Commonly Mutated Genes . . . . . . . . . . . . . . . . . . . . 813 Gene-Associated Phenotypes . . . . . . . . . . . . . . . . . . . . . . . . 816
NGS Findings and Redefinition of Known Syndromic Forms of SRNS . . . . . . . . . . . . . . . . . . . . . . . . 818
Gene-Associated Phenotypes . . . . . . . . . . . . . . . . . . . . . . . . 814 SRNS with Genitourinary Features:
Denys–Drash and Frasier Syndromes . . . . . . . . . . . . . . . 818
SRNS with Ocular Features: Pierson
Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 818
O. Boyer
SRNS with Osseous Dysplasia: Nail-Patella
Service de Néphrologie Pédiatrique, Hôpital
and Schimke Syndromes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 819
Necker-Enfants Malades, Université Paris-Descartes,
SRNS with Central Nervous System
Paris, France
Involvement: Metabolic Diseases and
e-mail: olivia.boyer@nck.aphp.fr
Galloway–Mowat Syndrome . . . . . . . . . . . . . . . . . . . . . . . . 820
K. Tory SRNS with Peripheral Neurological
Laboratory of Hereditary Kidney Diseases, Inserm UMR Involvement: Charcot–Marie–Tooth Disease
1163, Paris, France and FSGS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 823
SRNS with Cutaneous Features: Epidermolysis
Department of Pediatrics, Semmelweis University,
Bullosa and FSGS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 824
Budapest, Hungary
e-mail: kalman.tory@inserm.fr; tory.kalman@med. Familial Forms of Steroid-Sensitive
semmelweis-univ.hu Nephrotic Syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 825
E. Machuca Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 825
Department of Nephrology, Medical School, Pontificia
Universidad Católica de Chile, Santiago, Chile References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 826
C. Antignac (*)
Laboratory of Hereditary Kidney Diseases, Inserm UMR
1163, Paris, France
Paris Descartes, Sorbonne Paris Cité University, Imagine
Institute, Paris, France
Department of Genetics, MARHEA reference center,
Necker – Enfants Malades Hospital, Paris, France
e-mail: corinne.antignac@inserm.fr

# Springer-Verlag Berlin Heidelberg 2016 805


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_23
806 O. Boyer et al.

Introduction responsible for most of the cases with congenital


nephrotic syndrome (CNS) and might be found in
Over the last decades, screening of large cohorts infantile forms of SRNS [14, 15]. NPHS2 muta-
of pediatric patients presenting with steroid- tions are the most frequent causes of early-onset
resistant nephrotic syndrome (SRNS) for gene autosomal recessive SRNS [16] and account for
mutations has revealed the importance of genetic 37.5 % of the cases with NS presenting in the first
disorders in the pathogenesis of proteinuric year of life among European populations [1].
glomerulopathies. Genetic forms of nephrotic Autosomal dominant (AD) forms of SRNS are
syndrome have been considered as infrequent dis- infrequent and generally observed in adults, except
orders; however, at least 66 % of the cases for WT1-related SRNS. Variable degrees of pro-
presenting with SRNS during the first year of teinuria are detected, and progression to end-stage
life [1] and up to 30 % of SRNS cases with an kidney disease (ESKD) is usually slow [17, 18].
onset below 25 years of age have an underlying Since most patients do not exhibit full-blown
monogenic defect [2]. From a clinical perspective, nephrotic syndrome, these forms are usually
theoretically all patients with hereditary SRNS referred to as AD focal segmental glomerulo-
will be resistant to immunosuppressive agents sclerosis (FSGS).
and will not experience relapse after transplanta- The advent of next-generation sequencing
tion [3–5]. Whereas partial remission may be (NGS) techniques in the past few years has tre-
achieved under cyclosporin A, shown to stabilize mendously facilitated the identification of new
the podocyte actin cytoskeleton in vitro through gene mutations in genetic diseases, most of them
the synaptopodin pathway [6], such treatment being rare mutations involving few families.
cannot induce complete remission in SRNS Moreover, it has revealed mutations in unex-
related to podocyte gene mutations [4, 7–10]. pected genes and has widened the phenotypes
Gene discovery efforts aimed at unraveling the associated with podocyte gene mutations.
causes of Mendelian forms of nephrotic syndrome Recently, mutations in genes involved in
have resulted in the identification of mutations in tubulointerstitial and developmental nephropa-
novel genes that encode proteins crucial for the thies have been associated with isolated SRNS
establishment and maintenance of the glomerular such as the homozygous p.P209L TTC21B muta-
filtration barrier. These discoveries have helped tion known to also lead to nephronophthisis [19]
decipher the pathophysiologic mechanisms of or PAX2 mutations known as a cause of congenital
the glomerular filtration process by revealing anomalies of the kidney and urinary tract
that most of the defective proteins are essential (CAKUT) and papillorenal syndrome [20]. More
for glomerular podocyte function. Podocytes are importantly, NGS approaches have wholly modi-
highly differentiated epithelial cells with an fied the diagnosis screening algorithms, and
octopus-like shape maintained by a finely regu- podocyte gene panels are increasingly used as
lated cytoskeleton. Their multiple cell extensions the first step of genetic analyses in selected
named “foot processes” are interdigitated and patients such as children or adults with familial
connected by slit diaphragms, final glomerular FSGS [2, 21, 22].
filtration barriers. The list of genes mutated in This chapter will review the available epidemi-
syndromic and non-syndromic forms of SRNS is ologic data, genotype–phenotype correlations,
constantly evolving and consists to date of more and the mechanisms by which mutations in
than 30 genes inherited in an autosomal recessive, genes implicated in hereditary forms of isolated
autosomal dominant, or mitochondrial fashion SRNS lead to proteinuric glomerular disease.
(Table 1) [11–13]. Among these genes, NPHS1 A genetic overview on recently discovered genes
and NPHS2 encoding nephrin and podocin, responsible for rarer cases of hereditary
respectively, are by far the two main genes impli- syndromic SRNS and advances in the study of
cated in SRNS [2, 9]. Mutations in NPHS1 are familial SSNS are presented as well.
27

Table 1 Genes implicated in hereditary SRNS and FSGS and associated phenotypes
Typical age
Number of families at dg of Recurrent extrarenal
Gene Desc. Trans. Encoded protein (pts) publishedc PU/NS Age at ESKD Typical histology involvement
Autosomal recessive isolated SRNS
NPHS1 [14] 1998 AR Nephrin >270(>300) 0–10 yrs 7 ms–15 yrs MGC, FSGS –
NPHS2 [16] 2000 AR Podocin >600(>850) 0–40 yrs 2 yrs–50 yrs MGC, FSGS –
PLCE1 [76] 2006 AR Phospholipase C epsilon 50(71) 0–8 yrs 5 ms–12 yrs DMS, FSGS –
1
CD2AP [101] 2007 AR CD-2 associated protein 1(1) 10 mo 3 yrs FSGS –
PTPRO [92] 2011 AR Receptor-type tyrosine- 2(5) 5–14 yrs 18 yrs (n = 1) MGC, FSGS –
protein phosphatase O
MYO1E [85] 2011 AR Myosin 1E 10(14) 2 ms–9 yrs 6 yrs to 15 yrs FSGS –
ADCK4 [238] 2013 AR aarF domain containing 8(15) <1–21 yrs 7–23 yrs FSGS Goiter
kinase 4
TTC21B [19] 2014 AR Intraflagellar transport 7(13) 9–30 yrs 1–35 yrs FSGS –
protein IFT139
CRB2 [93] 2015 AR Crumbs homolog 5(4) 9 mo to 6 yrs NA FSGS Cerebral ventriculomegaly
2 protein with congenital NS
Autosomal dominant isolated SRNS
Idiopathic Nephrotic Syndrome in Children: Genetic Aspects

ACTN4 [105] 2000 AD α-Actinin-4 12(>75) 3–54 yrs 6–59 yrs FSGS –
TRPC6 [43] 2005 AD Transient receptor >28(>62) 2–75 yrs 0–20 yrs FSGS –
potential 6 after onset
INF2d[122] 2010 AD Inverted formin 2 >60(>221) 5–72 yrs 13–70 yrs FSGS –d
LMX1Bd[129] 2013 AD LIM homeodomain 8(>28) 5–70 yrs 28–70 yrs FSGS –d
transcription factor 1B
ARHGAP24 2011 AD Rho-GTPase-activating 1(3) NA 12–29 yrs FSGS –
[126] protein 24
ANLN [127] 2014 AD Anillin 2(12) 9–69 yrs 35–75 yrs FSGS –
COL4A3/ 2014 AD Collagen IV alpha-3 and 16(57) 2–70 yrs 12–82 yrs FSGS Hearing impairment
COL4A4 alpha-4 chains
[131]
WT1[145] 1992 AD Wilms’ tumor protein 7(31) 3–59 yrs 10–69 yrs FSGS Ambiguous genitalia
PAX2d[20] 2014 AD Paired box protein 2 7(24) 7–68 yrs 30–58 yrs FSGS –d
(continued)
807
808

Table 1 (continued)
Typical age
Number of families at dg of Recurrent extrarenal
Gene Desc. Trans. Encoded protein (pts) publishedc PU/NS Age at ESKD Typical histology involvement
Syndromic forms
WT1 [154] 1991 AD Wilms’ tumor protein Denys–Drash >170 0–10 yrs 0–15 yrs DMS Wilms’ tumor in both genders,
Sd genital anomalies in 46,XY
patients
Frasier Sd >60 7 mo to 5 yrs to 34 yrs FSGS Pseudohermaphroditism,
34 yrs gonadoblastoma in 46,XY
patients
LAMB2 [168] 2004 AR Laminin β2 subunit >60 (>70) 0–6 yrs 0–21 yrs DMS Microcoria, abnormal lens, VI,
hypotonia, motor delay
LMX1B [128] 1998 AD LIM homeodomain >35 (>54) 9–76 yrs 22–52 yrs FSGS, Hypoplastic nails/patellae,
transcription factor 1B NPS-nephropathy iliac horns
SMARCAL1 2002 AR SWI/SNF-related, (>90) 2–12 yrs 3.8–22 yrs FSGS Spondyloepiphyseal
[204] matrix-associated, actin- dysplasia, T-cell deficiency,
dependent regulator of cerebral ischemia, broad nasal
chromatin, subfamily tip, hyperpigmented macules
a-like 1
MTTL1 [208] 1990 Maternal tRNALeu(UUR) >30 (>45) 5–47 yrs 13–51 yrs FSGS MELAS: myopathy,
encephalopathy, lactic
acidosis, and stroke-like
episode, diabetes, and
deafness
COQ2 [227] 2006 AR Para-hydroxybenzoate- 10(13) 0–30 mo 0 to 5 yrsa FSGS Encephalomyopathy,
polyprenyltransferase hypotonia, seizures, lactate
acidosis
PDSS2 [233] 2006 AR Decaprenyl-diphosphate 3(3) 0–23 mo ND No data Encephalomyopathy,
synthase subunit 2 hypotonia, seizures, lactate
acidosis
COQ6 [237] 2011 AR Coenzyme Q10 8(16) 2 mo to 6.4 5 ms to 9.3 yrs FSGS SND, seizure
monooxygenase 6 yrs
O. Boyer et al.
27

SCARB2 2008 AR Lysosome membrane 18(24) 9 to >59 yrs 9 to >59 yrs FSGS Progressive myoclonic
[241, 242] protein 2 (LIMP-2) epilepsy, cerebral and
cerebellar atrophy, peripheral
neuropathy, hearing
impairment
WDR73 [261] 2014 AR WD40-repeat-containing 2(3) 5 to >13 yrs 5 yrs FSGS Secondary microcephaly,
protein intellectual deficiency, cortical
and cerebellar atrophy, facial
dysmorphy, optic atrophy
ARHGDIA 2013 AR Rho-GDP dissociation 3(6) 0–2.4 yrs 6 wks to 3 yrs DMS Intellectual disability
[262, 263] inhibitor α
INF2 [124] 2011 AD Inverted formin 2 27(44) 10–46 yrs 12–47 yrs FSGS Charcot–Marie–Tooth
neuropathy, deafness
ITGB4 [278] 1998 AR Integrin β4 subunit 2(2) 2 mo to NA FSGS Junctional epidermolysis
10 yrs bullosa, nail dystrophy,
recurrent hemorrhagic cystitis,
desquamation of laryngeal
mucosa
LAMB3 [279] 2005 AR Laminin β3 subunit 1(1) 4 mo NA (b5 mo) DMS Epidermolysis bullosa, nail
dystrophy
ITGA3 2012 AR Integrin α3 subunit 6(6) 0–4 yrs 2 wks to 4 yrs FSGS Interstitial lung disease,
[280, 281] epidermolysis bullosa
Idiopathic Nephrotic Syndrome in Children: Genetic Aspects

CD151 [283] 2004 AR Tetraspanin CD151 2y NA NA Thickening of the Pretibial skin lesions, SND,
GBM (n = 1) lacrimal duct stenosis, nail
dystrophy
a
Besides CoQ supplementation
b
Died (at the age of)
c
Only patients with renal phenotype are considered
d
Only patients published as isolated FSGS are considered
AD autosomal dominant, AR autosomal recessive, desc. description, DMS diffuse mesangial sclerosis, ESKD end-stage kidney disease, FSGS focal segmental glomerulosclerosis, MGC
minimal glomerular changes, mo months, NA not available, NPS nail-patella syndrome, NS nephrotic syndrome, pts patients, PU proteinuria, SND sensorineural deafness, Sd syndrome, trans.
transmission, VI severe visual impairment, wks weeks, yrs years
809
810 O. Boyer et al.

Autosomal Recessive Forms region, is required for podocin to bind cholesterol


of Isolated SRNS and regulate the activity of associated proteins
[39]. Podocin accumulates in oligomeric form in
Mutations in NPHS2 Encoding Podocin lipid-raft microdomains of the slit-diaphragm
plasma membrane where it recruits nephrin
Gene Identification and Protein thereby augmenting the nephrin-induced activa-
Characterization tion of the AP-1 transcription factor and also
Using a positional cloning approach, Boute interacts with CD2AP and TRPC6 [40–43]. The
et al. identified mutations in NPHS2 (MIM resulting protein complex represents an important
#604766) encoding podocin in pedigrees of link between the slit diaphragm and the podocyte
patients from Europe and Northern Africa who cytoskeleton and is crucial for structural organi-
presented with childhood-onset autosomal reces- zation and regulation of filtration function of the
sive non-syndromic SRNS [16]. Subsequent stud- slit diaphragm, mechanosensory signaling,
ies further defined the phenotype associated with podocyte survival, cell polarity, and cytoskeletal
mutations in the NPHS2 gene, revealing that organization [37, 39, 42–45].
patients usually develop NS from birth to 6 years
of age, do not respond to immunosuppressive Allelic Variants and Genotype/Phenotype
agents, and reach ESKD before the end of the Correlations
first decade of life [1, 3, 4, 23]. Histological find- Mutations in the NPHS2 gene include a full spec-
ings range from minimal glomerular changes trum of missense changes, protein-truncating non-
(MCD), in patients biopsied early, to FSGS at sense and frameshift mutations, and splice-site
later stages [16]. NPHS2 mutations are approxi- variants and involve all eight coding exons [4, 16,
mately responsible for 30–40 % of familial and for 24]. To date, more than 110 pathogenic mutations
10–30 % of sporadic cases of SRNS [3, 4, 24–27]. and 40 variants of unknown significance have been
Interestingly, NPHS2 mutations were also reported [27]. Mutations are frequently found in
found to be a frequent cause of congenital NS pediatric patients with SRNS originating from Cen-
(CNS) manifesting in utero or within the first tral Europe, North America, Turkey, the Middle
3 months of life (15–39 %), infantile NS with East, North Africa, and South America. Contrari-
onset between 4 months and 1 year of age wise, NPHS2 mutations are rarely detected in cases
(29–35.2 %) [1, 23, 27, 28], and also rarely from Japan, China, Korea, and sub-Saharan Africa.
adult-onset SRNS [29–36], thereby further broad- Several founder mutations have been identified
ening the spectrum of phenotypes attributable to including p.R138Q in Europe [16, 24], p.R138X
podocin mutations [27]. in the Israeli-Arab population [46], p.V260E in the
The NPHS2 gene spans a 25-kb region, con- Comoros Islands, and p.A284V in South America
sists of eight exons, and encodes the 42-kDa pro- [27]. The p.R138Q mutation in exon 3 is the most
tein podocin with 383 amino acid residues. prevalent mutation in European series [47]. It was
Podocin is almost exclusively expressed in the found, respectively, in 32 % and 44 % of all
kidney at the slit diaphragm of podocytes, from affected NPHS2 alleles in two large European
the capillary-loop stage [16, 37, 38]. It is a lipid series [3, 26]. The resulting protein is retained in
raft-associated protein belonging to the stomatin the endoplasmic reticulum (ER) and loses its ability
family. Podocin is predicted to be an integral to recruit nephrin in lipid rafts [40]. Interestingly,
membrane protein inserted in the membrane missense mutations leading to retention of the
through a short hydrophobic region resulting in a mutant protein in the ER (such as the p.R138Q)
hairpin-like topology with intracellular NH2- and result in an earlier onset of disease than mutations
COOH-terminal ends [16]. It also contains a that do not disrupt the mutant protein traffic to the
prohibitin homology (PHB) domain that, in addi- plasma membrane (20.8  4 vs. 128.7  9
tion to palmitoylation sites in the hydrophobic months) [48]. A potential therapeutic strategy that
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 811

might delay the onset and reduce the severity of the asymptomatic. This has direct implications in
disease in patients with gene mutations causing genetic counseling, as individuals carrying p.
ER-retention of the mutant plasma membrane pro- R229Q are only at risk of having an affected
tein (e.g., slit-diaphragm proteins) relies on chem- child if their spouse carries a dominant negative
ical chaperones. Several of these molecules have NPHS2 mutation, when the rate of transmission
been used in in vitro systems, allowing for targeting may be dramatically different from those expected
of the mutant protein to the plasma membrane [49]. in autosomal recessive disorders [55].
The mean age at onset of NS in patients carry- Finally, tri-allelic inheritance of NPHS1 and
ing two pathogenic mutations ranges between 2.6 NPHS2 mutations has been occasionally reported
and 3.4 years of age [3, 26]. Weber et al. also [25, 56, 57], but additional studies are needed to
found that patients with frameshift or nonsense better understand the complex genetics of renal
mutations in the homozygous or compound het- disease progression in the setting of nephrotic
erozygous states have an earlier onset of nephrotic syndrome.
syndrome than those carrying missense mutations
[3]. In addition, individuals homozygous for the p.
R138Q mutation present with early-onset disease Clinical Relevance
[3, 26]. At least two mutations, p.V180M and The identification of NPHS2 mutations in children
p.R238S, are associated with a milder phenotype, presenting with nephrotic syndrome may have
including later age at onset of NS and age at important clinical implications. Patients with
ESKD [3]. The p.R138X mutation has been asso- mutations in the NPHS2 gene do not respond to
ciated with a high incidence of cardiac abnormal- steroid or immunosuppressive therapy, although a
ities in children [50], although this finding has not partial reduction of proteinuria has been reported
been confirmed in patients carrying other with cyclosporine A in a small number of cases
mutations [51]. [1, 4]. Avoidance or withdrawal of immunosup-
The p.R229Q variant is the most frequently pressive therapies in these patients would spare
reported non-synonymous NPHS2 variant in Cau- them from the potential risks and side effects
casians [52], in whom the observed frequency of associated with these drugs. Moreover, patients
heterozygotes ranges from 0.03 to 0.13 [3, 4, 34, with two pathogenic NPHS2 mutations have a
52–54]. In African-Americans and sub-Saharan significantly lower risk of relapse after transplan-
populations, the p.R229Q allele is infrequent tation than cases in which mutations were not
[52]. In vitro studies demonstrated decreased identified (8 % vs. 30 %) [3, 4, 58, 59]. Patients
binding of the p.R229Q mutant protein to nephrin, bearing mutations in the heterozygous state have a
suggesting that this variant may be pathogenic risk comparable to those without mutations
[34]. Patients carrying the p.R229Q non-neutral [60]. In contrast with the mechanism of relapse
polymorphism associated with a pathogenic in cases with NPHS1 mutations, there is no evi-
mutation have a significantly later onset of disease dence to support a role for anti-podocin antibodies
[3, 26] and represent almost all adult-onset cases [3, 61, 62]. The recent discovery of the nonpatho-
with NPHS2 mutations [30, 34, 36]. Indeed, genic association of the p.R229Q variant with
patients with such mutations develop ESKD at a C-terminal NPHS2 mutations has given a possible
mean age of 26.1  18.9 years [36]. Our group clue to this unclear phenomenon [55]. Indeed, a
demonstrated that the p.R229Q variant is only subset of patients with podocin mutation and
pathogenic when trans-associated to specific posttransplant recurrence may actually have an
C-terminal mutations, which exert a dominant immune form of SRNS and fortuitously bear the
negative effect on podocinR229Q through an p.R229Q variant and a C-terminal NPHS2 muta-
altered dimerization and mislocalization [55]. tion in trans. Other possible mechanisms of recur-
On the contrary, patients bearing the p.R229Q rence of proteinuria in these patients may involve
variant at the homozygous state are de novo glomerulopathy or drug toxicity.
812 O. Boyer et al.

Animal Models demonstrated that in vitro treatment with a chem-


In the two mouse models of constitutive Nphs2 ical chaperone may allow for trafficking of
inactivation [63] or knock-in of the murine equiv- ER-retained mutants to the plasma membrane
alent of the human mutation p.R138Q [64], mice [70, 71].
develop congenital nephrotic syndrome with mas- However, later findings have broadened the
sive foot process effacement, as well as unex- spectrum of renal disease related to nephrin muta-
pected lesions of mesangiolysis and mesangial tions since patients with childhood-onset and even
sclerosis, and succumb to ESKD within 5 weeks adult-onset SRNS may rarely carry NPHS1 muta-
of life. These effects are modulated not only by tions [15, 72–74]. Indeed, the p.R1160X mutation
genetic background but also, in Nphs2 null mice, may result in a milder phenotype in about 50 % of
by the maternal environment [65]. Conversely, the cases with histological findings consistent with
conditional model of podocin inactivation specif- CNF and either mild proteinuria or complete
ically in podocytes in the mature kidney [66] remission up to 19 years of age [56]. Thereafter,
results in progressive NS and renal insufficiency NPHS1 mutations were identified in 10/160
with FSGS lesions rather than diffuse mesangial patients (142 families) presenting with SRNS at
sclerosis (DMS) lesions, hence recapitulating the a mean age of 3 years (range: 6 months to 8 years)
phenotype of patients bearing NPHS2 mutations. and MCD or FSGS on biopsy [15]. Affected cases
The later model is therefore an invaluable tool to were compound heterozygous for at least one
test novel therapeutic strategies in SRNS. The “mild” missense mutation, which exhibited nor-
crucial role of podocin in glomerular physiology mal trafficking to the plasma membrane and
was further confirmed by the absence of slit dia- maintained the abilities to form nephrin
phragm and an altered plasma filtration in homodimers and to heterodimerize with NEPH1.
zebrafish using podocin morpholino [67]. After- These findings may explain the lesser severity of
wards, it has also been shown that the Drosophila disease observed in these cases.
ortholog of podocin Mec2 is required for the func-
tion of the nephrocyte, a podocyte-like cell with a
filtration slit diaphragm in flies [68]. Mutations in PLCE1 Encoding
Phospholipase C Epsilon 1

Mutations in NPHS1 Encoding Nephrin A positional cloning approach coupled to gene


expression profiling in rat glomeruli identified
NPHS1 (MIM #602716) has been identified as the the PLCE1 gene (MIM #608414), encoding phos-
major gene involved in congenital nephrotic syn- pholipase C epsilon 1, as a good candidate in
drome of the Finnish type (CNF) [14]. The clini- 7 families with SRNS. PLCε1 catalyzes the
cal aspects of CNF and the identification of the hydrolysis of phosphoinositides to generate two
NPHS1 gene and characterization of nephrin are second messengers – inositol 1,4,5-triphosphate
extensively described in the corresponding chap- (IP3) and diacylglycerol (DAG) [75] – that initiate
ter. Briefly, nephrin is a transmembrane protein intracellular pathways of cell growth and differ-
with eight immunoglobulin-like modules, a fibro- entiation. PLCε1 is highly expressed in podocytes
nectin domain, a single transmembrane domain, and interacts with the cell junction-associated pro-
and a cytosolic C-terminal end [69]. It localizes to tein IQGAP1 [76], a nephrin-binding partner
the slit diaphragm and is critical for its architec- involved in cell morphology and adhesion
ture. The spectrum of NPHS1 mutations includes [77]. Mutational analysis subsequently revealed
protein-truncating nonsense and frameshift inser- truncating and missense mutations in several of its
tion/deletion mutations, splice-site changes, and 34 exons [76]. In the initial cohort reported by
missense variants. Most of these mutants are Hinkes et al., all patients with homozygous trun-
retained in the ER and lead to a severe CNF cating PLCE1 mutations had DMS as opposed to
phenotype. Interestingly, Liu et al. have two patients with missense mutations who
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 813

exhibited FSGS with a later onset of NS [76]. Sub- feature in NS [84]. MYO1E encodes myosin 1E,
sequently these investigators showed that PLCE1 a member of the ubiquitously expressed class I
truncating mutations accounted for 28.6 % of myosins, implicated in podocyte actin cytoskele-
cases of isolated DMS in a cohort of 40 patients ton organization and motility. Its knocking down
from 35 families mostly of Turkish origin [78]. In in both zebrafish and mouse models causes
contrast, our group observed either truncating or nephrotic syndrome [87–89], and myo1e/
missense mutations in both FSGS and DMS mice recapitulate the GBM lesions observed in
patients and no clear phenotype–genotype corre- humans [84, 88].
lation in a series of 168 patients [79]. A more
recent study reported an earlier age of onset in
patients with PLCE1 splice-site mutations com- PTPRO
pared with C-terminal truncating mutations or The protein tyrosine phosphatase receptor type O
missense mutations [2]. Interestingly in the origi- (PTPRO, also called GLEPP1) was first identified
nal description, two patients bearing truncating as a podocyte-specific transmembrane protein
mutations achieved complete remission when localized to the apical side of foot processes in
treated early and remained free of proteinuria rabbit glomeruli [90]. Ptpro/ mice do not
after several years of follow-up [76, 80]. Later, develop albuminuria but have reduced glomerular
four families with transiently affected or even filtration rate and hypertension after nephrectomy
asymptomatic family members – although homozy- [91] and exhibit altered, amoeboid-shaped
gous for truncating mutations – have been podocytes with broad and short major processes.
published, suggesting an incomplete penetrance of PTPRO (MIM #600579) mutations were identified
PLCE1-associated nephrotic syndrome [79, 81], in two Turkish consanguineous families
an uncommon phenomenon in autosomal recessive by homozygosity mapping [92]. Three of the five
diseases [82, 83]. A second-locus modifier, yet to affected siblings had developed SRNS between the
be identified, could explain this phenomenon, ages of 5 and 14 years with FSGS or MCD on
although variations in 19 candidate genes including histology and severe foot process effacement, and
16 phospholipases were ruled out [79]. The reported one progressed to ESKD at the age of 18 years.
incomplete penetrance is consistent with the lack
of renal phenotype observed in Plce1/ mice [76].
CRB2
Homozygous or compound heterozygous CRB2
mutations (MIM #609720) have been identified
Less Commonly Mutated Genes by exome sequencing in four families with
childhood-onset isolated SRNS with FSGS diag-
MYO1E nosed between 9 months and 6 years of age
By homozygosity mapping and high-throughput [93]. CRB2 encodes the putative transmembrane
sequencing, MYO1E mutations (MIM #601479) Crumbs homolog 2 protein. The knockout of
have been identified in two Italian families with zebrafish Crb2b results in foot process efface-
isolated SRNS [84, 85]. Among the ten families ment, and podocyte arborization and polarity
with MYO1E mutations published thus far, most defects [93]. In accordance with the embryonic
affected children had developed ESKD between lethality of Crb2 knockout mice, all affected chil-
6 and 13 years of age, although a normal renal dren carried a missense mutation on at least one
function could be observed in a 14-year-old child allele [94]. Of note, CRB2 mutations were also
[84–86]. Patients typically develop FSGS [84–86] detected in five fetuses and one infant with cere-
with disorganization of the glomerular basement bral ventriculomegaly and echogenic kidneys
membrane (GBM) and tubular alterations. Con- with histopathological findings of congenital
sistently, three of the five patients with available nephrosis [95] (▶ Chap. 25, “Congenital
data had microscopic hematuria, an uncommon Nephrotic Syndrome”).
814 O. Boyer et al.

CD2AP identified by exome sequencing combined to link-


CD2-associated protein was originally cloned as an age analyses in seven families with FSGS. TTC21B
interaction partner of CD2, a T lymphocyte signal- is therefore the first ciliary gene involved in a glo-
ing protein expressed on the cell surface, implicated merular disorder. Patients presented late-onset pro-
in T-cell adhesion to antigen-presenting cells teinuria at ages 9–23 years, high blood pressure and
[96]. In the kidney, CD2AP is expressed in proxi- ESKD at ages 15–32 years. Moreover, characteris-
mal tubules, collecting ducts and podocytes where tic morphological alterations of nephronophthisis
it interacts with nephrin [97], podocin [37], and such as tubular basement membrane thickening
F-actin [98] suggesting its role in anchoring the were also present in these patients, indicating the
SD to the actin cytoskeleton. Cd2ap/ mice dem- coexistence of primary glomerular and tubuloin-
onstrate not only an impaired T-cell function in vitro terstitial alterations. Besides, various hepatic fea-
but also congenital nephrotic syndrome leading to tures have been detected in one family and several
ESKD by 6–7 weeks of age [97], the latter being cases found mutated since the original publication
prevented by podocyte-specific expression of (unpublished data). In accordance to data in rat
CD2AP in Cd2ap/ mice [99]. Intriguingly, tissues, a primary cilium was found to be present
Cd2ap+/ mice are susceptible to develop protein- in human podocytes in fetal kidney biopsies and
uria secondary to nephrotoxic antibodies, showing undifferentiated cultured podocytes, but not in adult
that CD2AP haploinsufficiency may contribute to kidney sections and differentiated cells [19]. It was
multifactorial glomerular injury [100]. Though sev- further shown that IFT139 was expressed at the
eral studies screened for CD2AP mutations (MIM cilium base in fetal and undifferentiated podocytes,
#604241) in SRNS, only a single patient has thus whereas in mature and differentiated podocytes,
far been identified with two mutated alleles IFT139 localized along the expanding and complex
[101]. He carried a homozygous last-exon nonsense microtubule network. In undifferentiated cells,
mutation (p.R612*) that truncated the protein for IFT139 knockdown induced ciliary defects that
the last 28 amino acids and markedly diminished its were partially rescued by overexpression of
interaction with F-actin [101]. Patient lymphocytes IFT139P209L, suggesting a hypomorphic effect of
displayed no CD2AP expression, consistent with a this allele. However, this hypomorphic effect was
complete loss of function. He presented with failure not sufficient to rescue the cytoskeletal disorgani-
to thrive secondary to nephrotic-range proteinuria zation yielded by IFT139 knockdown in differenti-
and microscopic hematuria at the age of 10 months, ated cells. The modest consequences of the
with FSGS on histology, progressed to ESKD by hypomorphic variant p.P209L on ciliogenesis with
the age of 3 years, and underwent transplantation more deleterious effects on cytoskeletal regulation
two years later with no recurrence. Both parents are in accordance with the late phenotype observed
were heterozygous for the mutation and had normal in mutated patients [19]. Other ciliary proteins
glomerular filtration rate and no proteinuria, ruling expressed in mature podocytes and sharing addi-
out a haploinsufficient phenotype in this family. tional roles in microtubule maintenance may be
involved in SRNS.

NGS Findings and Redefinition


of Known Gene-Associated Phenotypes Autosomal Dominant Forms
of Isolated SRNS
TTC21B
Mutations of the ciliary gene TTC21B (MIM Mutations in ACTN4 Encoding
#612014), encoding the retrograde intraflagellar a-Actinin-4
transport-A protein IFT139, were initially identified
as a cause of nephronophthisis [102]. Unexpectedly, A genome-wide scan performed in a 100-member
its most common mutation (p.P209L), a founder kindred allowed Mathis et al. to map the first locus
mutation in Portugal and North Africa, was recently of AD FSGS on chromosome 19q13 [103, 104].
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 815

Linkage analysis including additional families channel, subfamily C, member 6 [115]. Subse-
helped to reduce the size of the region and led to quently, TRPC6 mutations were evaluated to be
the identification of three nonconservative mis- responsible for approximately 2–5 % of AD FSGS
sense mutations in the ACTN4 gene (MIM [43, 113, 116, 117]. Notably, TRPC6 mutations
#604638) [105]. ACTN4 encodes the ubiquitous have been described in six children, one of whom
actin-binding protein α-actinin-4, highly was a 2-year-old boy with collapsing FSGS and
expressed in podocytes [105]. α-actinin-4 inter- rapid progression to ESKD due to a de novo
connects actin filaments in podocyte foot pro- mutation [116–119].
cesses [106]. Most reported mutations are TRP channels are involved in mechanosensation,
missense mutations involving residues within or ion homeostasis, cell growth, and PLC-dependent
next to the actin-binding site of α-actinin-4 [105, calcium entry into cells [120, 121]. TRPC6 is
107]. Some of them have been shown to increase expressed in podocytes, specifically at the slit dia-
the affinity of α-actinin-4 to F-actin and/or induce phragm where it interacts with podocin and nephrin
α-actinin-4 mislocalization and the formation of [43]. Some of the mutations were shown to enhance
α-actinin-4 and F-actin aggregates around the TRPC6-mediated calcium signals suggesting a
nucleus with subsequent impairment of podocyte gain-of-function mechanism, whereas others do
spreading and motility [105, 107–110]. Knock-in not [115]. Diverse mechanisms may result in
and knockout mouse models and in vitro data dysregulation of the ion channel or may affect the
suggest both gain-of-function and loss-of-func- interaction with other slit-diaphragm proteins [43].
tion mechanisms [108]. Affected cases typically
present with proteinuria starting in the teenage
years or later, with FSGS lesions on kidney his-
tology, and slowly progress to ESKD in the fifth Mutations in INF2 Encoding
decade of life, although earlier cases have been the Inverted Formin 2
seldom reported [105, 111, 112]. Disease is
incompletely penetrant. Further mutation screen- M. Pollak’s group identified heterozygous muta-
ing of the ACTN4 gene in cases with familial and tions in the INF2 gene (MIM #610982) encoding
sporadic forms of FSGS allowed the identification the inverted formin 2 in 12 % of AD FSGS families
of several additional mutations. Overall, ACTN4 [122]. Subsequent studies confirmed that INF2
mutations seem to account for approximately 3–4 mutations are the first cause of AD FSGS [113,
% of AD FSGS [107, 111, 113]. 123]. Age at proteinuria onset ranges from 5 to
72 years and at ESKD onset from 13 to 70 years.
Incomplete penetrance was also reported by most
authors. Conversely, mutations in this gene seem to
Mutations in TRPC6 Encoding be responsible for <1 % of sporadic cases of FSGS
the Transient Receptor Potential Cation due to de novo mutations [113, 123]. Our group
Channel 6 later demonstrated that INF2 mutations are the
main cause of Charcot–Marie–Tooth disease
Winn et al. identified a second locus for AD FSGS with FSGS (see below) [124]. Formins are ubiqui-
in a large family from New Zealand [114]. tous proteins governing various cellular phenome-
Affected cases presented with nephrotic-range nons such as actin cytoskeleton remodeling, cell
proteinuria in their third or fourth decade and polarity and morphogenesis, and cytokinesis [125].
developed progressive renal insufficiency within INF2 belongs to the diaphanous-related formin
10 years after onset. Using fine-mapping and family, which prototypic member is mDia1. INF2
candidate-gene screening, the same group of contains the formin homology domains FH1 and
investigators subsequently detected a missense FH2 involved in actin elongation and nucleation,
mutation in the TRPC6 gene (MIM #603652), respectively, as well as the diaphanous
encoding the transient receptor potential cation autoregulatory domain (DAD) and diaphanous
816 O. Boyer et al.

inhibitory domain (DID) which play a role in NGS Findings and Redefinition
INF2 auto-inhibition. The vast majority of INF2 of Known Gene-Associated Phenotypes
mutations are localized in the DID region. These
results further confirmed the crucial role of an intact LMX1B Mutations and FSGS Without
actin cytoskeleton in podocyte dynamics and Nail-Patella
function. LMX1B mutations lead to nail-patella syndrome
(NPS) (MIM #161200) associating dysplasia of
the patellae, nails and elbows, iliac horns, glau-
coma, and in some cases FSGS with specific
Less Commonly Mutated Genes
lesions characterized by the presence of type III
collagen fibrils in the GBM by electron micros-
ARHGAP24
copy (cf. below) [128]. By linkage analysis and
Using a candidate-gene approach, ARHGAP24
exome sequencing, a novel LMX1B mutation was
encoding the actin-regulating protein Rho-GAP
unexpectedly identified in a pedigree of five
24 (MIM #610586), highly expressed in
patients with AD FSGS but no glomerular base-
podocytes, was sequenced in 310 patients with
ment membrane anomaly suggestive of nail-
FSGS [126]. A loss-of-function mutation was
patella-like renal disease by electron microscopy
identified in one family with AD FSGS. The
and no extrarenal features [129]. Mutations in two
mother had died of ESKD at 29 years of age,
other families were subsequently identified by
and two offsprings had reached ESKD at 12 and
Sanger sequencing among a cohort of 73 addi-
20 years of age, respectively. This mutation
tional unrelated families with FSGS. The patients
involved the GAP catalytic domain, very con-
presented significant albuminuria between 5 and
served between species. This is another example
70 years of age, and some of them reached ESKD
of actin-regulating genes in which mutations lead
between 26 and 70 years of age. Another novel
to hereditary FSGS.
sequence variant (p.R249Q) involving the same
domain of the protein has been recently described
ANLN in an AD pedigree with similar phenotype, i.e.,
More recently, mutations in the ANLN gene (MIM late-onset isolated FSGS [130]. LMX1B encodes a
#616027) encoding anillin were identified by homeodomain-containing transcription factor that
exome sequencing combined to linkage analyses is essential during development. The two muta-
in two families with AD FSGS [127]. Patients’ tions involve the same residue of LMX1B and are
phenotype was similar to FSGS related to other expected to diminish the interaction between the
AD genes. The age at proteinuria onset varied LMX1B homeodomain and DNA molecule by in
from 9 to 69 years, with FSGS on biopsies and silico analyses. These data demonstrate that muta-
ESKD occurring between 35 and 75 years. Anillin tions in genes involved in syndromic forms of
is an actin-binding protein required for cytokine- SRNS may also be responsible for isolated
sis. It also interacts with the slit-diaphragm pro- FSGS and highlight the need to include these
tein CD2AP and the formin mDia2. In the kidney, genes in next-generation sequencing diagnosis
anillin is mainly expressed in tubular cells but also approaches in FSGS.
in podocytes of control subjects [127]. Con-
versely, anillin is overexpressed on kidney biop- COL4A3/COL4A4 Mutations
sies of patients with idiopathic FSGS. In vitro, and Isolated FSGS
anillin mutants have an impaired binding to Next-generation sequencing brought up several
CD2AP resulting in abnormal podocyte motility. additional surprising findings. Searching for
These results add to the expanding group of mutations in a pedigree of three sisters referred
podocyte cytoskeleton genes involved in FSGS for familial SRNS diagnosed between 8 and
pathophysiology. 13 years of age and FSGS on biopsy,
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 817

R. Gbadegesin’s group identified two compound Frasier syndromes (cf. below) related to exon
heterozygous variants of COL4A3 segregating with 8 and 9 mutations and specific splice-site mutations
the disease [131]. The three children also had in exon 9, respectively [139, 140]. Female patients
microscopic hematuria and two had hearing mostly present with isolated nephropathy (FSGS or
impairment. COL4A3 and COL4A4 encode the α3 DMS) or nephropathy in association with develop-
and four chains of collagen IV, respectively. mental tumors and have normal puberty although
Mutations in these genes are responsible for Alport streak dysgenetic ovaries have been seldom
syndrome of autosomal dominant (MIM #104200) described in 46,XX children [139].
or recessive inheritance (MIM #203780) or benign Very surprisingly, through classical Sanger
familial hematuria (MIM #141200). Unlike the sequencing and more recently NGS, WT1 muta-
classical X-linked and the AR forms of childhood tions were identified in pedigrees with isolated
Alport syndrome, AD forms of the disease (~10 % AD FSGS, including males without genital abnor-
cases) have a late onset and a high phenotypic malities or developmental tumors who transmitted
variability with inconstant gross hematuria and the mutation [141–145]. All patients carried a
hearing loss [132]. Consequently, the authors missense mutation in exon 8 or 9 (R458Q,
searched for COL4A3 and COL4A4 mutations in S461F, or R471K) affecting the second and third
a cohort of 69 unrelated families with FSGS and zinc fingers [141–143]. These results suggest
found a rare heterozygous variant in six of them, including WT1 testing in the genetic work-up not
translating into a mutation rate of 10 % of familial only of females with sporadic FSGS but also in
FSGS [131]. None of the patients had characteristic males with familial AD FSGS.
GBM lesions suggestive of Alport syndrome by
optical or electron microscopy. Most heterozygous PAX2 Mutations and AD FSGS
patients were detected with proteinuria during NGS also allowed the identification of PAX2 muta-
adulthood (2–65 years of age). Nevertheless, all tions (MIM #167409) segregating with the disease
patients with available data had associated hema- in seven families with AD FSGS [20]. The
turia. Subsequent reports confirmed that COL4A3 24 affected patients had various degrees of protein-
and COL4A4 mutations are rather frequent causes uria diagnosed between 7 and 68 years (mostly
of AD FSGS (~10–12 %) [133, 134]. Only one second to fourth decades) and FSGS on biopsy
patient had available electronic microscopy exam- (10/24 patients). Nine had reached ESKD. PAX2
ination results and exhibited only segmental thin- is a transcription factor expressed from the fourth
ning of the GBM [133]. These data emphasize the week of gestation in the kidney as well as otic and
need to perform COL4A3 and COL4A4 gene optic vesicles and the hindbrain. Heterozygous
screening in familial FSGS, especially in case of PAX2 mutations were thus far known as a cause
microscopic hematuria or deafness, or in the of CAKUT and papillorenal syndrome character-
absence of other SRNS gene mutation. ized by renal hypodysplasia, optic nerve coloboma,
and rarely sensorineural hearing loss. In the afore-
WT1 Mutations and AD FSGS mentioned series from Barua et al. [20], kidney
WT1 encodes a transcription factor of the zinc ultrasound in patients with PAX2-related FSGS
finger family that plays a crucial role in kidney either was normal or could reveal echoic or small
and genital tract development [135, 136]. Its kidneys, dilated renal pelvis, or calyceal diverticu-
expression persists in podocytes in the adult lum. Most patients did not exhibit any extrarenal
kidney, and its integrity is required for proper glo- symptom, but papillorenal syndrome was diag-
merular filtration barrier function [137]. Heterozy- nosed retrospectively in one family. PAX2 muta-
gous mutations of WT1 (MIM #607102), mostly de tions could result in FSGS secondary to nephron
novo mutations, are associated to a wide spectrum loss but also in specific podocyte injury through
of syndromes of autosomal dominant inheritance in dysregulation of target genes such as the transcrip-
children [138]. These include Denys–Drash and tion factor WT1 [20].
818 O. Boyer et al.

Syndromic Forms of SRNS with FSGS on biopsy and slow progression to


ESKD toward adolescence or early adulthood
SRNS with Genitourinary Features: [158]. As in DDS, inheritance is autosomal dom-
Denys–Drash and Frasier Syndromes inant, although most cases are sporadic due to de
novo mutations. WT1 encodes numerous
The WT1 gene was first found to be inactivated in isoforms, which are products of alternative trans-
Wilms’ tumor [146]. It encodes a zinc finger tran- lation start sites, alternative splicing, and RNA
scription factor and translational regulator that editing [159]. Of particular interest are WT1
functions both as a tumor suppressor and as a (+KTS) and WT1(KTS) variants, which differ
critical regulator of kidney and gonadal develop- by the presence of the three amino acids KTS
ment [135, 136, 147]. The key role of WT1 in between zinc fingers 3 and 4. The presence of
kidney development has been highlighted by the this insert influences the molecular and biochem-
development of animal and in vitro models show- ical properties of the resulting protein. While
ing complete failure of kidney development in the WT1(KTS) binds DNA efficiently and acts as
absence of WT1 expression [147–149]. Mutations a transcriptional activator, WT1(+KTS) seems to
in the WT1 gene are associated with varied have higher affinity to RNA and splicing factors
syndromic forms of glomerular disease and geni- [160]. Heterozygous mutations in the donor splice
tourinary abnormalities, as well as isolated cases site of intron 9 of WT1 are causative of Frasier
of SRNS. syndrome and lead to reduction of the +KTS
Denys–Drash syndrome (DDS, MIM 194080) isoform of the protein [161]. This results in an
is a rare urogenital disorder comprising nephrop- alteration of the normal ratio of +KTS/-KTS
athy due to DMS, associated with male isoforms in the cell [140].
pseudohermaphroditism and predisposition to De novo deletion of the 11p13 locus leads to
Wilms’ tumors [150, 151]. Nephrotic syndrome WAGR syndrome (MIM 194072), characterized
presents in the first months of life, may be pre- by Wilms’ tumors, aniridia, genitourinary abnor-
ceded by isolated proteinuria, and is always resis- malities, and intellectual disability [162]. Aniridia
tant to steroid therapy. Progression to ESKD is due to the deletion of the PAX6 gene, which
occurs most often before 4 years of age, and no resides in the same locus than WT1.
recurrence is observed after renal transplantation Noteworthily, 46,XX females carrying either
[152]. Wilms’ tumor may be the first presentation missense or splice-site WT1 mutation commonly
of the disease or may be discovered later during do not exhibit genital anomalies, have normal
the course of nephropathy by systematic ultra- puberty, and therefore present with isolated spo-
sound screening. Patients with DDS bear hetero- radic SRNS [8, 155, 163–165]. Genetic counsel-
zygous missense or truncating mutations in exons ing is essential in such patients, since they have a
8 and 9, mostly de novo, affecting the DNA- and theoretical 50 % risk to transmit the mutation
RNA-binding affinity of the second and third zinc [163, 164].
finger domains [153–156]. Isolated cases of DMS
have also been attributed to WT1 mutations,
mostly in 46,XX patients [139, 157]. SRNS with Ocular Features: Pierson
Frasier syndrome (FS, MIM 136680) is char- Syndrome
acterized by male pseudohermaphroditism with
usually normal female external genitalia but Pierson syndrome (OMIM #609049) is character-
streak gonads and increased susceptibility to ized by congenital nephrotic syndrome with DMS
gonadoblastomas in 46,XY patients, whereas 46, and an irregular GBM on kidney biopsy, ocular
XX girls have both normal external and internal anomalies mostly microcoria, and developmental
genitalia. FS is associated with childhood-onset delay [166, 167]. Using both homozygosity map-
proteinuria, usually between 2 and 6 years of age, ping [168] and a candidate-gene approach at the
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 819

locus owing to the CNS phenotype of Lamb2 null nails that may be observed at birth, iliac horns, and
mice [169], LAMB2 truncating mutations were hypoplastic patellae [179]. Nephropathy may
identified in families with Pierson syndrome. occur in 25–50 % of the cases [180–184]. This
Interestingly, the current literature also includes manifests at any age – mostly after the second
reports of milder LAMB2 variants resulting in decade – as microalbuminuria progressing to pro-
apparently isolated congenital nephrotic syn- teinuria that may be intermittent, usually associ-
drome with minor or no ocular defects and normal ated with hematuria. Overt nephrotic syndrome is
psychomotor development [170–172], as well as not a common feature, and progression to ESKD
childhood onset of Pierson syndrome in a occurs in 5–14 % of the cases, usually many years
non-consanguineous family of seven affected after proteinuria onset [184, 185]. Light micros-
individuals, with nephrotic syndrome and ESKD copy of renal tissue usually reveals nonspecific
presenting between 5 and 10 years of age and changes [186], whereas GBM exhibits ultrastruc-
progressive blindness around 2 years of age tural abnormalities that are the most specific his-
[173, 174]. Laminins are heterotrimeric extracel- tological hallmarks of NPS [180, 187–189].
lular matrix proteins that provide the basic scaf- Typically, there is irregular thickening and split-
fold for assembly of the other components of the ting of the GBM, with electron-lucent areas, and
GBM, including type IV collagen, nidogen/ the presence of clusters of fibrillar type III colla-
entactin, and sulfated proteoglycans [175]. gen within the GBM and the mesangial matrix.
Lamb2 null mice recapitulate the clinical features Finally, primary open angle glaucoma and senso-
observed in patients with Pierson syndrome [169]. rineural hearing impairment have been recognized
Interestingly, proteinuria precedes podocyte foot as less frequent features of the disease [184, 190].
process effacement in these mice suggesting that LMX1B is a transcription factor belonging to
the slit-diaphragm integrity is not sufficient to the LIM homeodomain family of proteins that
maintain proper glomerular filtration functions plays an essential role in the normal development
[176]. Indeed, expression and localization of of dorsal limb structures, GBM, anterior segment
podocin and synaptopodin appeared normal at of the eye, and some types of neurons. Approxi-
onset of proteinuria in transgenic mice expressing mately 85 % of families with NPS present muta-
the p.R246Q lamb2 mutation, whereas they were tions in LMX1B. The majority of mutations results
perturbed upon development of heavy proteinuria in protein truncation. Missense mutations gener-
[177]. More importantly, overexpression of lamb1 ally involve substitutions in the homeodomain
in lamb2-null mice could prevent the develop- region critical for DNA binding [182–184, 191,
ment of nephrotic syndrome and extend life span 192]. Recently, entire-gene deletions were
[178], hence raising the prospect of future reported by Bongers et al., confirming that
therapies. haploinsufficiency of the LMX1B transcription
factor underlies this disease [193]. The precise
role of LMX1B in the kidney is only partially
SRNS with Osseous Dysplasia: Nail- elucidated. The targeted disruption of the Lmx1b
Patella and Schimke Syndromes gene (LIM homeobox transcription factor 1 β) in
mice recapitulates the NPS phenotype [194] but
Nail-Patella Syndrome: Mutations only in homozygous mutant mice, whereas het-
in LMX1B erozygous littermates do not exhibit any evident
Nail-patella syndrome (NPS; MIM 161200) is an abnormalities. In Lmx1b-deficient mice, the abun-
autosomal dominant disorder with complete pen- dance of α3 and α4 chains of collagen IV are
etrance and variable phenotypic expression, char- markedly diminished [195], as were the levels of
acterized by pleiotropic developmental defects of podocin [196, 197], CD2AP [196], synaptopodin,
dorsal limb structures. The most characteristic and VEGF [197]. No such anomaly has been
findings include absent, hypoplastic, or dystrophic observed in patients bearing heterozygous
820 O. Boyer et al.

mutations in the LMX1B gene [198]. The latter have a milder course of disease, surviving beyond
findings may be explained by the fact that these 15 years of age. The functional targets of
patients carry one functional allele and one SMARCAL1 remain unidentified.
mutated allele, whereas mice have two mutant
alleles.
SRNS with Central Nervous System
Schimke Immuno-osseous Dysplasia: Involvement: Metabolic Diseases and
Mutations in SMARCAL1 Galloway–Mowat Syndrome
Schimke immuno-osseous dysplasia (SIOD,
OMIM 242900) is a rare incompletely penetrant Mitochondriopathies with SRNS: MELAS
autosomal recessive disorder characterized by and COQ10 Deficiencies
spondyloepiphyseal dysplasia, progressive renal Mitochondriopathies are a diverse group of disor-
dysfunction due to FSGS, and T-cell immunode- ders due to structural, biochemical, or genetic
ficiency [199]. Additional inconstant features derangements of mitochondria [206]. Renal dys-
include cerebral ischemia, migraine-like head- function is a rare event and may result from muta-
aches, deficiency of other blood cell lineages, tions encoded by the mitochondrial or nuclear
hyperpigmented macules, corneal opacities, genomes. The mitochondrial genome codes for
microdontia, intellectual delay, recurrent infec- 13 essential subunits of the mitochondrial respi-
tions, premature atherosclerosis, hypothyroidism, ratory chain, as well as the 22 transfer RNA
cerebellar atrophy, and testicular hypoplasia with (tRNA) and two ribosomal RNA (rRNA)
atrophy and azoospermia [200–203]. Kidney dis- genes [207].
ease manifests typically with proteinuria evolving The c.3243A > G point mutation in the
to overt SRNS, which is diagnosed between the tRNALeu(UUR) mitochondrial gene is associated
first year of life and 14 years of age [200, 201]. with MELAS syndrome (myopathy, encephalop-
Patients who survive infectious complications athy, lactic acidosis, and stroke-like episodes)
progress to ESKD between 5 and 15 years of [208]. Some patients carrying tRNALeu(UUR) gene
age. Numerous cases have been transplanted with- mutations may present with diabetes and deafness
out evidence of relapse in the allograft [201]; [209, 210], cardiomyopathy [211], progressive
however, the evolution of cerebrovascular and external ophthalmoplegia, and FSGS [212–217],
infectious complications does not seem to with or without nephrotic syndrome
improve after transplantation. [215–221]. Although most affected cases are diag-
A genome-wide scan, performed in four fami- nosed in adulthood and have glomerular disease
lies, detected significant linkage at chromosome associated with other manifestations of mitochon-
2q35, and mutations were identified in the drial disease, some patients present with isolated
SMARCAL1 gene [204]. This gene encodes a mem- nephropathy or may have an earlier onset during
ber of an SNF2 subfamily of proteins that mediate adolescence [216, 219, 221, 222]. Likewise, a
DNA-nucleosome restructuring during gene deletion in the mitochondrial DNA has been asso-
regulation and DNA replication, recombination, ciated with FSGS in a cohort of Japanese patients
methylation, and gene repair (SWI/SNF-related and in a Turkish patient [220, 223].
matrix-associated actin-dependent regulator of Nuclear genes, in which mutations lead to
chromatin, subfamily-a-like-1 gene). The majority syndromic or isolated SRNS, encode proteins of
of mutations identified are nonsense and frameshift the coenzyme Q10 pathway. Coenzyme Q10
mutations, likely leading to loss of function [204]. (COQ10, ubiquinone) is a lipid-soluble component
Clewing et al. showed that SMARCAL1 biallelic of all cell membranes, vital for the transport of
mutations accounted for the phenotype in 38 of electrons from complexes I and II to complex III
72 independent cases with SIOD [205], revealing of the mitochondrial respiratory chain, a potent
the genetic heterogeneity of this syndrome. antioxidant, and a cofactor of mitochondrial dehy-
Patients with two missense mutations tended to drogenases and pyrimidine synthesis proteins [224].
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 821

At least ten enzymes are required for COQ10 mouse model in which a spontaneous Pdss2 muta-
synthesis, which, when mutated, lead to heteroge- tion occurred, develop an isolated renal disease
neous multisystemic disorders associating myopa- with dysmorphic mitochondria and later collaps-
thy, seizures, ataxia, deafness, optic atrophy, and ing glomerular lesions [225, 234–236]. Impor-
cardiomyopathy among others. SRNS may develop tantly, the renal phenotype of kd/kd mice can be
in patients with COQ10 deficiency with or without at least partially rescued by CoQ10 supplementa-
extrarenal features, as a result of COQ2, COQ6, tion [225, 226].
PDSS2, or ADCK4 gene mutations. Altogether, COQ6, encoding a monooxygenase, was iden-
mutations in these genes are identified in about tified by homozygosity mapping [237] in five
1 % of SRNS cases [2]. The characteristic histolog- families from Turkey or Lebanon. Four of them
ical finding is FSGS, often of the collapsing are consanguineous and carry one of two homo-
type, with dysmorphic podocyte mitochondria by zygous founder missense mutations (p.G255R
electron microscopy [224]. Podocytes are not con- and p.A353D). The fifth unrelated patient is com-
sidered to have a high energetic requirement, and pound heterozygous for two truncating mutations.
the role of COQ10 in podocyte function remains to The affected children manifested with proteinuria
be clarified [225, 226]. However, the beneficial at the median age of 1.2 years (range: 0.2–6.4) and
effects of early COQ10 supplementation strongly progressed to ESKD at a median age of 1.7 years
support the primary role of COQ10 in podocyte (range: 0.4–9.3) [237]. Renal biopsy revealed
function. FSGS in seven cases and DMS in one. Sensori-
The first COQ10 gene found to be mutated in neural deafness was reported in all nine individ-
SRNS was COQ2 encoding para-hydroxybenzoate- uals examined. Additionally, seizures were
polyprenyl transferase, identified by candidate-gene reported in two of the 11 patients. Two children
approach [227]. Most children present with severe were supplemented with CoQ10 with a beneficial
renal and neurological involvement, whereas only effect in both of them [237].
two unrelated children had isolated SRNS, and two The most recently identified gene implicated in
infants died at the age of 5 and 6 months with no COQ10 biosynthesis is ADCK4, encoding aarF
renal involvement [227–231]. Eight of the nine domain containing kinase 4. Using exome
reported children developed ESKD or died within sequencing and homozygosity mapping, Ashraf
the first 18 months of life. The ninth child, who had et al. identified ADCK4 mutations in 14 patients
an affected brother, was diagnosed early at the age from seven families [238]. The subsequent
of 1 year and could benefit from COQ10 supple- targeted sequencing of ADCK4 in 400 sporadic
mentation since NS onset that allowed preservation cases resulted in the identification of a single case
of renal and neurologic function until last examina- with a homozygous mutation. Affected patients
tion at the age of 5 years [232]. developed SRNS between the ages of 3 and
The first child described with PDSS2 muta- 21 years, typically with FSGS on kidney histology
tions was diagnosed with neonatal Leigh syn- (collapsing variant in three cases), and progressed
drome associated to severe NS at the age of to ESKD at a median age of 15 years (range:
7 months and succumbed to refractory status 7–23). A single patient with infantile nephrotic
epilepticus at 8 months of age [233]. He was syndrome and developmental delay received
found to have COQ10 deficiency in muscle and COQ10 supplementation with subsequent reduc-
fibroblasts and compound heterozygous PDSS2 tion of proteinuria. Intriguingly, apart from goiter
mutations by candidate-gene approach. Two addi- reported in two unrelated adolescents, no other
tional patients presented SRNS at birth and recurrent extrarenal involvement is associated to
2 years of age, associated to cerebral palsy in the NS [238]. A second cohort of 26 patients from
oldest [2]. PDSS2 encodes the second subunit of 12 families confirmed a late-onset and mostly
decaprenyl diphosphate synthase, the first enzyme renal-limited phenotype of ADCK4 mutations
of the COQ10 biosynthetic pathway and one of with a median age at onset of proteinuria of
the rate-limiting enzymes [233]. The kd/kd mice, a 14 years, ESKD during the second decade, and
822 O. Boyer et al.

mild neurological features or retinitis pigmentosa in the SCARB2 gene (encoding LIMP-2). These
in only 3/26 patients [304]. Knockdown of mutations led to a downregulation of SCARB2
ADCK4 orthologs in zebrafish and drosophila mRNA and undetectable protein levels in western
recapitulates the nephrosis phenotype [238]. The blots of cell lysates from lymphoblastoid B cell
encoded aarF domain containing kinase 4 is ubiq- lines from the two affected subjects [242]. LIMP-
uitous, expressed at high levels in podocytes 2 (lysosomal integral membrane protein) is the
where it localizes to foot processes and mitochon- receptor for β-glucocerebrosidase, a lysosomal
dria [238], but its function is unknown. However, enzyme deficient in Gaucher disease [243].
several lines of evidence strongly support the role LIMP-2 is considered to play a role in the biogen-
of ADKC4 in CoQ10 synthesis: (1) its high esis and maintenance of endosomal and lysosomal
sequence similarity with ABC1/COQ8 of Saccha- compartments [244]. The Limp2 knockout mouse
romyces cerevisiae [239], (2) low COQ10 levels model displays some features of AMRF such as
in patient fibroblasts and/or lymphocytes, proteinuria, cochlear deafness, demyelinating
(3) ADCK4 interaction with CoQ6 and CoQ7 in peripheral neuropathy, but also uretericpelvic
cultured podocytes, and (4) the rescue of altered obstruction [242, 245, 246]. The role of LIMP-2
cell migration in ADCK4 knockdown cultured in the podocytes and the pathophysiologic events
podocytes by COQ10 supplementation leading to NS and FSGS in patients with SCARB2
[238]. Given the relatively high prevalence of mutations remain to be elucidated.
ADCK4 mutations in late-onset SRNS/FSGS, the
lack of extrarenal involvement characteristic of Galloway–Mowat Syndrome: Mutations
mitochondriopathies, and the potential benefit in WDR73
from early COQ10 supplementation, ADCK4 Galloway–Mowat syndrome (GMS, OMIM
sequencing should be considered in all adolescent #251300) is a rare autosomal recessive disorder
patients. Moreover, mutated patients should be characterized by SRNS, microcephaly, and severe
repeatedly screened for subclinical extrarenal neurological impairment [247]. Disease frequency
symptoms. is unknown; however, more than 60 cases
have been reported since the original description
Action Myoclonus–Renal Failure in 1968 [247, 248]. Inconstant morphological
Syndrome: Mutations in SCARB2 defects include hiatus hernia, micrognathia,
Action myoclonus–renal failure syndrome arachnodactyly, and floppy ears. The distinctive
(AMRF, MIM 254900) is an autosomal recessive neurological hallmark is microcephaly, which
disease characterized by progressive myoclonic might be congenital (primary microcephaly) or
epilepsy associated with renal failure. It typically may develop after birth (secondary microcephaly).
presents at 15–25 years of age with neurological Structural brain abnormalities may include cerebral
symptoms including tremor, action myoclonus, atrophy and gyration defects and are associated
seizures, and later ataxia, whereas cognitive func- with severe psychomotor impairment, hypotonia,
tion is preserved. Proteinuria is usually diagnosed and seizures in half of all cases. Sensorineural
concomitantly with the onset of neurological blindness and deafness have also been described.
symptoms at a median age of 19 years [240]. Pro- Renal manifestations of GMS range from isolated
gression to ESKD occurs generally within 5 years proteinuria to overt SRNS, which rapidly pro-
after onset of proteinuria [240, 241]. The renal gresses to ESKD after a few months. Proteinuria
pathology is characterized by FSGS, sometimes is usually discovered within the first year of life,
of the collapsing type [240]. By linkage analysis although congenital onset is not unusual as are
and gene expression profiling to prioritize gene cases in which the onset of NS is close to the
sequencing within the region, Berkovic third year of life [247, 249–256]. Kidney histology
et al. identified homozygous truncating mutations may reveal either MC, FSGS that might be of the
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 823

collapsing type, or DMS, the latter more frequent in podocyte physiology has been demonstrated in
early-onset forms [251, 257–260]. The prognosis animal models. The podocyte-specific knockout
of GMS is poor, and most affected children reach of Cdc42 results in podocyte injury [267], and
ESKD between 36 and 72 months and die before inducible transgenic mice expressing either consti-
the age of 6 years, although there are rarer cases tutively active or dominant negative RhoA develop
with preserved renal function after this age albuminuria [266, 268]. All three identified
[256, 260]. ARHGDIA mutations have been shown to abrogate
WDR73 has been very recently reported as the the interaction of Rho-GDIα with its Rho-GTPase
first gene mutated in Galloway–Mowat syndrome partners, resulting in hyperactivation of RAC1 and
(GMS) by homozygosity mapping and whole CDC42 and altered podocyte motility [262,
exome sequencing in an index family and thereaf- 263]. Mouse and zebrafish models recapitulate
ter in a second family by testing a cohort of the NS phenotype [262, 263], which can be par-
30 unrelated individuals [261]. This demonstrates tially rescued by RAC1 inhibitors, hence opening a
the large genetic heterogeneity of GMS. Two of the window of opportunity for therapy of some forms
three affected children developed NS at the age of of hereditary SRNS [264, 269].
5 and 8 years with FSGS, whereas the third had
normal urinary sediment at the age of 7. Facial
dysmorphy, optic atrophy, secondary microcephaly SRNS with Peripheral Neurological
with severe intellectual deficiency, and cortical and Involvement: Charcot–Marie–Tooth
cerebellar atrophy with thin corpus callosum were Disease and FSGS
present in all three children. An abnormal morphol-
ogy of cell nuclei was found both in the patients’ INF2 and Charcot–Marie–Tooth Disease
fibroblasts and in WDR73-depleted cultured with FSGS
podocytes, which could be rescued in the latter Since the 1960s, an increased prevalence of
either by the reexpression of the wild-type protein nephropathies, particularly FSGS, has been
or microtubule depolymerization. WDR73 is there- documented in patients with Charcot–Marie–Tooth
fore suggested to play a role in the organization of disease [270]. Charcot–Marie–Tooth disease refers
the microtubule network [261]. to a heterogeneous group of inherited chronic
peripheral motor and sensory neuropathies, with
ARHGDIA Mutations and Early-Onset an estimated prevalence of 1 in 2,500 live births.
SRNS with Intellectual Disability Affected individuals typically present progressive
Mutations of ARHGDIA, encoding Rho-GDP dis- distal muscle weakness and atrophy, reduced ten-
sociation inhibitor α (Rho-GDIα), have been iden- don reflexes, foot and hand deformities, and rarely
tified by whole exome sequencing in six children mild to moderate sensorineural hearing loss. INF2
from three consanguineous families [262, 263] which mutations account for 12–17 % of AD FSGS
with infantile NS, DMS on histology, and ESKD cases encodes the formin INF2 known to interact
before 3 years of age. Intellectual disability was with the myelin and lymphocyte proteins MAL and
reported in three children, one with sensorineural MAL2 and Cdc42 all implicated in essential steps
hearing loss and another with seizures and cortical of myelination and myelin maintenance. Through a
blindness. Rho family GTPases are key regulators candidate-gene approach, INF2 was sequenced in
of the actin cytoskeleton and thus cell adhesion, 16 families with FSGS and Charcot–Marie–Tooth
migration, and division [126, 264–267]. Rho- disease, and mutations were identified in 75 % of
GDIα inhibits the three canonical Rho-GTPases them, but in none of a series of 50 patients with
RHOA, RAC1, and CDC42, by sequestering Charcot–Marie–Tooth disease and no renal
them in their inactive GDP-bound states in the involvement [124]. INF2 is expressed in Schwann
cytosol [262, 263, 267]. Their pivotal role in cells, where it colocalizes and interacts with MAL,
824 O. Boyer et al.

and INF2 mutations enhance INF2 binding to with a milder phenotype and developed FSGS at
Cdc42, thereby perturbing myelination. Since the the age of 10 years [2]. The mechanisms by which
original publication, 32 patients from 21 families mutations in the ITGB4 gene induce glomerular
have been described in the literature [124, disease remain unknown, and a fortuitous associ-
271–274]. Affected patients developed proteinuria ation cannot be ruled out. Similarly, although
between 7 and 30 years of age with FSGS and LAMB3 mutations are a frequent cause of junc-
Charcot–Marie–Tooth disease of the intermediate tional epidermolysis bullosa, renal involvement
type (i.e., with both demyelinating and axonal has thus far been reported only in a single child
lesions and intermediate median-nerve conduction who developed mixed proteinuria but mostly
velocities) during the second decade, and ESKD nephrotic syndrome with DMS on histology and
occurred in some patients between 12 and 47 years died at the age of 5 months [279]. Immunohisto-
of age. Because of the phenotypic variability, phy- pathologic findings in the patient’s kidney biopsy
sicians should be alert for proteinuria in all patients revealed both altered expression of laminin α
with Charcot–Marie–Tooth disease, and similarly, chains in the GBM and lack of laminin-5 in the
a careful clinical neurological evaluation should be tubular basement membrane that may account for
considered for patients with FSGS. glomerular and tubular involvement, respectively.
Despite possible incidental renal features in the
abovementioned case reports, other integrin and
laminin genes expressed in both skin basement
SRNS with Cutaneous Features: membrane and GBM might be good candidates
Epidermolysis Bullosa and FSGS to explain the dual dermo-renal phenotype.
Indeed, mutations of ITGA3 (MIM #614748)
SRNS with Epidermolysis Bullosa: encoding the integrin α3 subunit have been iden-
Mutations in ITGB4, ITGA3, CD151, or tified using homozygosity mapping and
LAMB3 candidate-gene approach by two different groups
Junctional epidermolysis bullosa is an autosomal in patients with epidermolysis bullosa and NS
recessive skin disorder in which blisters occur at [280, 281]. Most of the six unrelated children
the lamina lucida in the skin basement membrane. reported thus far developed CNS, severe intersti-
The disease is genetically heterogeneous with col- tial lung disease, and epidermolysis bullosa lead-
lagen, integrin, and laminin gene mutations ing to ESKD, respiratory failure, and death during
underlying most cases. Nephrotic syndrome and the first 2 years of life [2, 280, 281]. In a single
renal failure may occur in some patients with patient, SRNS was diagnosed later during child-
epidermolysis bullosa [275]. The most common hood at 4 years of age [2]. The integrin α3 subunit
histological finding is secondary amyloidosis heterodimerizes with the β1 subunit to form
[276, 277]. The association of FSGS and integrin α3β1 expressed at high levels in
epidermolysis bullosa was first reported in a podocytes, where it associates with the tetraspanin
male infant with pyloric atresia, junctional CD151 [281–286] and binds laminins [287].
epidermolysis bullosa, nephrotic-range protein- Consistently, mutations in CD151 (MIM
uria and FSGS diagnosed 6 weeks after birth, #609057) encoding the tetraspanin CD151 have
desquamation of laryngeal mucosa necessitating also been reported in children with SRNS and
tracheostomy, and recurrent hemorrhagic cystitis epidermolysis bullosa from two unrelated families
leading to the obstruction of the vesicoureteral [283]. The three affected children developed
junction within the first year of life [278]. Muta- pretibial epidermolysis bullosa, neurosensory
tional analysis of the β4-integrin gene ITGB4 deafness, bilateral lachrymal duct stenosis, nail
revealed a homozygous missense mutation affect- dystrophy, and thickening and splitting of the
ing the second fibronectin type III domain. tubular basement membrane. CD151 strongly
A second child, compound heterozygous for a interacts with integrin α3β1 and assures its distri-
splice-site and a missense mutation, presented bution along the GBM [283–286]. Cd151/
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 825

mice recapitulate the human phenotype in specific unexpected finding. Moreover, one would expect
strains [288, 289]. In contrast to LAMB3 and the underlying defect in hereditary SSNS to lie in
ITGB4 genes, no patient with ITGA3 or CD151 a gene involved in the immune system, instead of
gene mutations and epidermolysis bullosa without a transmembrane podocyte protein. Therefore, the
an apparent renal phenotype has been reported role of EMP2 in the pathogenesis of NS needs
to date. further clarification.

Familial Forms of Steroid-Sensitive Perspectives


Nephrotic Syndrome
Hereditary forms of NS are far more common than
The incidence of steroid-sensitive NS (SSNS) in previously thought ten years ago. The highest
the pediatric population ranges between 2 and rates of mutation detection are in patients
7/100,000. Although most cases are sporadic, presenting with proteinuria in the first year of
several reports have confirmed the existence of life and subsequently decrease among older
hereditary forms of this disease [290–298]. The patients [2]. Most of the cases with hereditary
exact incidence of familial forms of SSNS is forms of NS have a disease onset within early
unknown, but according to a single survey, it childhood, are resistant to immunosuppressive
may represent up to 3 % of the cases therapy, and do not relapse after kidney transplan-
[291]. Based on the low proportion of the familial tation. As a large proportion of patients with NS
over the sporadic cases, it is unlikely that SSNS do not have an identified underlying genetic
and the immune forms of SRNS were monogenic defect, and given the complexity of the patho-
in majority. Indeed, HLA-DQA1 and PLCG2 mis- physiology of the kidney, numerous further
sense variants have been very recently identified genes are expected to be mutated in NS. The
as risk loci in SSNS by genome-wide association most recently identified SRNS genes are
study [299]. A homozygous nonsense mutation of extremely rarely mutated and concern a very low
EMP2, encoding epithelial membrane protein 2, number of families. It is plausible that more com-
has been detected by homozygosity mapping and plex patterns of inheritance, as described in
exome sequencing in a consanguineous Turkish patients bearing bi- or tri-allelic variants or as
family with two affected children [300]. Both observed with the p.R229Q NPHS2 variant [55],
children had frequently relapsing SSNS at the will be increasingly recognized. Indeed, disease
age of 2 years with sustained remission under predisposing mutations may lead to variable phe-
cyclophosphamide. Subsequent screening of notypic expression and penetrance depending
1,600 individuals with NS revealed EMP2 muta- upon unidentified environmental and genetic fac-
tions in two Turkish siblings who presented with tors [79]. Moreover, common variants in genes
frequently relapsing SSNS, in a third Turkish expressed in podocytes may account for an
infant with SSNS, but also in an African- increased risk of FSGS and ESKD observed in
American patient, who developed steroid- and selected ethnic groups, as has been described with
cyclosporine-resistant NS at the age of 3 years the APOL1 gene, in which several haplotypes con-
[300]. EMP2 is a tetraspan protein that contains fer a major risk effect for FSGS in individuals of
four transmembrane domains and is highly African ancestry [301]. The accessibility to next-
enriched in kidney glomeruli where it localizes generation sequencing techniques has deeply facil-
to both podocytes and non-podocyte glomerular itated the screening of mutations in a broader
cells. The knockdown of emp2 in zebrafish results approach of podocyte-specific genes [2, 302,
in pericardial effusion, a surrogate of 303]. However, these diagnosis approaches face
NS. Nevertheless, as the coexistence of SSNS the difficulty of determining the causal mutation(s)
and SRNS within the same family is exceptional, among a great number of potential pathogenic var-
their common monogenic origin is a much iants. To date, several fascinating disorders, such as
826 O. Boyer et al.

familial forms of SSNS, connecting podocyte phys- steroid-resistant nephrotic syndrome. Clin J Am Soc
iology and the immune system, remain incom- Nephrol. 2010;5(11):2075–84.
8. Ruf RG, Schultheiss M, Lichtenberger A, Karle SM,
pletely solved. The constant advances in the Zalewski I, Mucha B, et al. Prevalence of WT1 muta-
understanding of hereditary kidney diseases and tions in a large cohort of patients with steroid-resistant
renal physiology also open the way to therapeutic and steroid-sensitive nephrotic syndrome. Kidney Int.
innovations. A promising therapy, still explored at a 2004;66(2):564–70.
9. Santin S, Bullich G, Tazon-Vega B, Garcia-Maset R,
basic level, involves protein chaperones. These Gimenez I, Silva I, et al. Clinical utility of genetic
drugs redirect the trafficking of missense mutant testing in children and adults with steroid-resistant
proteins to the plasma membrane when abnormally nephrotic syndrome. Clin J Am Soc Nephrol.
retained in subcellular organelles. Additional 2011;17.
10. Santin S, Tazon-Vega B, Silva I, Cobo MA,
encouraging results have been obtained with drugs Gimenez I, Ruiz P, et al. Clinical value of NPHS2
that stabilize the podocyte actin cytoskeleton. It is analysis in early- and adult-onset steroid-resistant
now clear that genetic diagnosis of cases with nephrotic syndrome. Clin J Am Soc Nephrol.
SRNS or familial NS is necessary to allow for 2011;6(2):344–54.
11. Pollak MR. Familial FSGS. Adv Chronic Kidney Dis.
accurate genetic counseling, to avoid ineffective 2014;21(5):422–5.
therapies and even start early suitable treatment 12. Buscher AK, Weber S. Educational paper: the
such as ubiquinone in COQ10 deficiency, and, podocytopathies. Eur J Pediatr. 2012;171(8):
hopefully in the near future, to offer specific 1151–60.
13. Saleem MA. New developments in steroid-resistant
mutation-based therapies. nephrotic syndrome. Pediatr Nephrol. 2013;28(5):
699–709.
14. Kestila M, Lenkkeri U, Mannikko M, Lamerdin J,
McCready P, Putaala H, et al. Positionally cloned
References gene for a novel glomerular protein–nephrin–is
mutated in congenital nephrotic syndrome. Mol
1. Hinkes BG, Mucha B, Vlangos CN, Gbadegesin R, Cell. 1998;1(4):575–82.
Liu J, Hasselbacher K, et al. Nephrotic syndrome in 15. Philippe A, Nevo F, Esquivel EL, Reklaityte D,
the first year of life: two thirds of cases are caused by Gribouval O, Tete MJ, et al. Nephrin mutations can
mutations in 4 genes (NPHS1, NPHS2, WT1, and cause childhood-onset steroid-resistant nephrotic syn-
LAMB2). Pediatrics. 2007;119(4):e907–19. drome. J Am Soc Nephrol. 2008;19:1871–78.
2. Sadowski CE, Lovric S, Ashraf S, Pabst WL, Gee HY, 16. Boute N, Gribouval O, Roselli S, Benessy F, Lee H,
Kohl S, et al. A single-gene cause in 29.5% of cases of Fuchshuber A, et al. NPHS2, encoding the glomerular
steroid-resistant nephrotic syndrome. J Am Soc protein podocin, is mutated in autosomal recessive
Nephrol. 2015;26(6):1279–89. steroid-resistant nephrotic syndrome. Nat Genet.
3. Weber S, Gribouval O, Esquivel EL, Moriniere V, 2000;24(4):349–54.
Tete MJ, Legendre C, et al. NPHS2 mutation analysis 17. Conlon PJ, Butterly D, Albers F, Rodby R, Gunnells
shows genetic heterogeneity of steroid-resistant JC, Howell DN. Clinical and pathologic features of
nephrotic syndrome and low post-transplant recur- familial focal segmental glomerulosclerosis. Am J
rence. Kidney Int. 2004;66(2):571–9. Kidney Dis. 1995;26(1):34–40.
4. Ruf RG, Lichtenberger A, Karle SM, Haas JP, 18. Rana K, Isbel N, Buzza M, Dagher H, Henning P,
Anacleto FE, Schultheiss M, et al. Patients with muta- Kainer G, et al. Clinical, histopathologic, and genetic
tions in NPHS2 (podocin) do not respond to standard studies in nine families with focal segmental
steroid treatment of nephrotic syndrome. J Am Soc glomerulosclerosis. Am J Kidney Dis. 2003;41(6):
Nephrol. 2004;15(3):722–32. 1170–8.
5. Winn MP. 2007 Young Investigator Award: TRP’ing 19. Cong EH, Bizet AA, Boyer O, Woerner S,
into a new era for glomerular disease. J Am Soc Gribouval O, Filhol E, et al. A homozygous missense
Nephrol. 2008;19(6):1071–5. mutation in the ciliary gene TTC21B causes familial
6. Faul C, Donnelly M, Merscher-Gomez S, Chang YH, FSGS. J Am Soc Nephrol. 2014;25(11):2435–43.
Franz S, Delfgaauw J, et al. The actin cytoskeleton of 20. Barua M, Stellacci E, Stella L, Weins A, Genovese G,
kidney podocytes is a direct target of the Muto V, et al. Mutations in PAX2 associate with
antiproteinuric effect of cyclosporine A. Nat Med. adult-onset FSGS. J Am Soc Nephrol. 2014;25(9):
2008;14(9):931–8. 1942–53.
7. Buscher AK, Kranz B, Buscher R, Hildebrandt F, 21. Bullich G, Trujillano D, Santin S, Ossowski S,
Dworniczak B, Pennekamp P, et al. Immunosuppres- Mendizabal S, Fraga G, et al. Targeted next-
sion and renal outcome in congenital and pediatric generation sequencing in steroid-resistant nephrotic
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 827

syndrome: mutations in multiple glomerular genes 34. Tsukaguchi H, Sudhakar A, Le TC, Nguyen T, Yao J,
may influence disease severity. Eur J Hum Genet. Schwimmer JA, et al. NPHS2 mutations in late-onset
2015;23(9):1192–9. focal segmental glomerulosclerosis: R229Q is a
22. Lovric S, Fang H, Vega-Warner V, Sadowski CE, Gee common disease-associated allele. J Clin Invest.
HY, Halbritter J, et al. Rapid detection of monogenic 2002;110(11):1659–66.
causes of childhood-onset steroid-resistant nephrotic 35. Tsukaguchi H, Yager H, Dawborn J, Jost L,
syndrome. Clin J Am Soc Nephrol. 2014;9(6): Cohlmia J, Abreu PF, et al. A locus for adolescent
1109–16. and adult onset familial focal segmental glomerulo-
23. Hinkes B, Vlangos C, Heeringa S, Mucha B, sclerosis on chromosome 1q25-31. J Am Soc
Gbadegesin R, Liu J, et al. Specific podocin mutations Nephrol. 2000;11(9):1674–80.
correlate with age of onset in steroid-resistant 36. Machuca E, Hummel A, Nevo F, Dantal J, Martinez F,
nephrotic syndrome. J Am Soc Nephrol. 2008;19(2): Al-Sabban E, et al. Clinical and epidemiological
365–71. assessment of steroid-resistant nephrotic syndrome
24. Karle SM, Uetz B, Ronner V, Glaeser L, associated with the NPHS2 R229Q variant. Kidney
Hildebrandt F, Fuchshuber A. Novel mutations in Int. 2009;75(7):727–35.
NPHS2 detected in both familial and sporadic 37. Schwarz K, Simons M, Reiser J, Saleem MA,
steroid-resistant nephrotic syndrome. J Am Soc Faul C, Kriz W, et al. Podocin, a raft-associated
Nephrol. 2002;13(2):388–93. component of the glomerular slit diaphragm,
25. Caridi G, Bertelli R, Di Duca M, Dagnino M, interacts with CD2AP and nephrin. J Clin Invest.
Emma F, Onetti Muda A, et al. Broadening the spec- 2001;108(11):1621–9.
trum of diseases related to podocin mutations. J Am 38. Roselli S, Gribouval O, Boute N, Sich M,
Soc Nephrol. 2003;14(5):1278–86. Benessy F, Attie T, et al. Podocin localizes in the
26. Hinkes B, Vlangos C, Heeringa S, Mucha B, kidney to the slit diaphragm area. Am J Pathol.
Gbadegesin R, Liu J, et al. Specific podocin mutations 2002;160(1):131–9.
correlate with age of onset in steroid-resistant 39. Huber TB, Schermer B, Muller RU, Hohne M,
nephrotic syndrome. J Am Soc Nephrol. 2008;19(2): Bartram M, Calixto A, et al. Podocin and MEC-2
365–71. bind cholesterol to regulate the activity of associated
27. Bouchireb K, Boyer O, Gribouval O, Nevo F, Huynh- ion channels. Proc Natl Acad Sci U S A.
Cong E, Moriniere V, et al. NPHS2 mutations in 2006;103(46):17079–86.
steroid-resistant nephrotic syndrome: a mutation 40. Huber TB, Simons M, Hartleben B, Sernetz L,
update and the associated phenotypic spectrum. Schmidts M, Gundlach E, et al. Molecular basis of
Hum Mutat. 2014;35(2):178–86. the functional podocin-nephrin complex: mutations in
28. Machuca E, Benoit G, Nevo F, Tete MJ, Gribouval O, the NPHS2 gene disrupt nephrin targeting to lipid raft
Pawtowski A, et al. Genotype-phenotype correlations microdomains. Hum Mol Genet. 2003;12(24):
in non-Finnish congenital nephrotic syndrome. J Am 3397–405.
Soc Nephrol. 2010;21(7):1209–17. 41. Huber TB, Kottgen M, Schilling B, Walz G, Benzing
29. He N, Zahirieh A, Mei Y, Lee B, Senthilnathan S, T. Interaction with podocin facilitates nephrin signal-
Wong B, et al. Recessive NPHS2 (Podocin) mutations ing. J Biol Chem. 2001;276(45):41543–6.
are rare in adult-onset idiopathic focal segmental 42. Huber TB, Schermer B, Benzing T. Podocin orga-
glomerulosclerosis. Clin J Am Soc Nephrol. nizes ion channel-lipid supercomplexes: implications
2007;2(1):31–7. for mechanosensation at the slit diaphragm. Nephron
30. McKenzie LM, Hendrickson SL, Briggs WA, Dart Exp Nephrol. 2007;106(2):e27–31.
RA, Korbet SM, Mokrzycki MH, et al. NPHS2 vari- 43. Reiser J, Polu KR, Moller CC, Kenlan P, Altintas
ation in sporadic focal segmental glomerulosclerosis. MM, Wei C, et al. TRPC6 is a glomerular slit
J Am Soc Nephrol. 2007;18(11):2987–95. diaphragm-associated channel required for normal
31. Aucella F, De Bonis P, Gatta G, Muscarella LA, renal function. Nat Genet. 2005;37(7):739–44.
Vigilante M, di Giorgio G, et al. Molecular analysis 44. Sellin L, Huber TB, Gerke P, Quack I, Pavenstadt H,
of NPHS2 and ACTN4 genes in a series of 33 Italian Walz G. NEPH1 defines a novel family of podocin
patients affected by adult-onset nonfamilial focal seg- interacting proteins. FASEB J. 2003;17(1):115–7.
mental glomerulosclerosis. Nephron Clin Pract. 45. Huber TB, Hartleben B, Kim J, Schmidts M,
2005;99(2):c31–6. Schermer B, Keil A, et al. Nephrin and CD2AP asso-
32. Monteiro EJ, Pereira AC, Pereira AB, Krieger JE, ciate with phosphoinositide 3-OH kinase and stimu-
Mastroianni-Kirsztajn G. NPHS2 mutations in adult late AKT-dependent signaling. Mol Cell Biol.
patients with primary focal segmental glomerulo- 2003;23(14):4917–28.
sclerosis. J Nephrol. 2006;19(3):366–71. 46. Frishberg Y, Rinat C, Megged O, Shapira E,
33. Tonna SJ, Needham A, Polu K, Uscinski A, Appel Feinstein S, Raas-Rothschild A. Mutations in
GB, Falk RJ, et al. NPHS2 variation in focal and NPHS2 encoding podocin are a prevalent cause of
segmental glomerulosclerosis. BMC Nephrol. steroid-resistant nephrotic syndrome among Israeli-
2008;9:13. Arab children. J Am Soc Nephrol. 2002;13(2):400–5.
828 O. Boyer et al.

47. Niaudet P. Podocin and nephrotic syndrome: Transplant Cooperative Study. J Am Soc Nephrol.
implications for the clinician. J Am Soc Nephrol. 1992;2 Suppl 12:S258–63.
2004;15(3):832–4. 60. Weber S, Tonshoff B. Recurrence of focal-segmental
48. Roselli S, Moutkine I, Gribouval O, Benmerah A, glomerulosclerosis in children after renal transplanta-
Antignac C. Plasma membrane targeting of podocin tion: clinical and genetic aspects. Transplantation.
through the classical exocytic pathway: effect of 2005;80 Suppl 1:S128–34.
NPHS2 mutations. Traffic (Copenhagen, Denmark). 61. Carraro M, Caridi G, Bruschi M, Artero M, Bertelli R,
2004;5(1):37–44. Zennaro C, et al. Serum glomerular permeability
49. Ohashi T, Uchida K, Uchida S, Sasaki S, Nihei activity in patients with podocin mutations (NPHS2)
H. Intracellular mislocalization of mutant podocin and steroid-resistant nephrotic syndrome. J Am Soc
and correction by chemical chaperones. Histochem Nephrol. 2002;13(7):1946–52.
Cell Biol. 2003;119(3):257–64. 62. Becker-Cohen R, Bruschi M, Rinat C, Feinstein S,
50. Frishberg Y, Feinstein S, Rinat C, Becker-Cohen R, Zennaro C, Ghiggeri GM, et al. Recurrent nephrotic
Lerer I, Raas-Rothschild A, et al. The heart of children syndrome in homozygous truncating NPHS2 muta-
with steroid-resistant nephrotic syndrome: is it all tion is not due to anti-podocin antibodies. Am J
podocin? J Am Soc Nephrol. 2006;17(1):227–31. Transplant. 2007;7(1):256–60.
51. Caridi G, Dagnino M, Carrea A, Massella L, 63. Roselli S, Heidet L, Sich M, Henger A,
Amore A, Emma F, et al. Lack of cardiac anomalies Kretzler M, Gubler MC, et al. Early glomerular
in children with NPHS2 mutations. Nephrol Dial filtration defect and severe renal disease in
Transplant. 2007;22(5):1477–9. podocin-deficient mice. Mol Cell Biol. 2004;24(2):
52. Franceschini N, North KE, Kopp JB, McKenzie L, 550–60.
Winkler C. NPHS2 gene, nephrotic syndrome and 64. Philippe A, Weber S, Esquivel EL, Houbron C,
focal segmental glomerulosclerosis: a HuGE review. Hamard G, Ratelade J, et al. A missense mutation in
Genet Med. 2006;8(2):63–75. podocin leads to early and severe renal disease in
53. Pereira AC, Pereira AB, Mota GF, Cunha RS, mice. Kidney Int. 2008;73(9):1038–47.
Herkenhoff FL, Pollak MR, et al. NPHS2 R229Q 65. Ratelade J, Lavin TA, Muda AO, Morisset L,
functional variant is associated with Mollet G, Boyer O, et al. Maternal environment inter-
microalbuminuria in the general population. Kidney acts with modifier genes to influence progression of
Int. 2004;65(3):1026–30. nephrotic syndrome. J Am Soc Nephrol. 2008;19(8):
54. Kottgen A, Hsu CC, Coresh J, Shuldiner AR, 1491–9.
Berthier-Schaad Y, Gambhir TR, et al. The associa- 66. Mollet G, Ratelade J, Boyer O, Muda AO, Morisset L,
tion of podocin R229Q polymorphism with increased Lavin TA, et al. Podocin inactivation in mature kid-
Albuminuria or reduced estimated GFR in a large neys causes focal segmental glomerulosclerosis and
population-based sample of US adults. Am J Kidney nephrotic syndrome. J Am Soc Nephrol. 2009;20(10):
Dis. 2008;52(5):868–75. 2181–9.
55. Tory K, Menyhard DK, Woerner S, Nevo F, 67. Kramer-Zucker AG, Wiessner S, Jensen AM, Drum-
Gribouval O, Kerti A, et al. Mutation-dependent mond IA. Organization of the pronephric filtration
recessive inheritance of NPHS2-associated steroid- apparatus in zebrafish requires Nephrin, Podocin and
resistant nephrotic syndrome. Nat Genet. 2014;46 the FERM domain protein Mosaic eyes. Dev Biol.
(3):299–304. 2005;285(2):316–29.
56. Koziell A, Grech V, Hussain S, Lee G, Lenkkeri U, 68. Zhang F, Zhao Y, Han Z. An in vivo functional anal-
Tryggvason K, et al. Genotype/phenotype correlations ysis system for renal gene discovery in Drosophila
of NPHS1 and NPHS2 mutations in nephrotic syn- pericardial nephrocytes. J Am Soc Nephrol.
drome advocate a functional inter-relationship in glo- 2013;24(2):191–7.
merular filtration. Hum Mol Genet. 2002;11(4):379–88. 69. Putaala H, Sainio K, Sariola H, Tryggvason K.
57. Schultheiss M, Ruf RG, Mucha BE, Wiggins R, Primary structure of mouse and rat nephrin cDNA
Fuchshuber A, Lichtenberger A, et al. No evidence and structure and expression of the mouse gene.
for genotype/phenotype correlation in NPHS1 and J Am Soc Nephrol. 2000;11(6):991–1001.
NPHS2 mutations. Pediatric Nephrology (Berlin, 70. Liu L, Done SC, Khoshnoodi J, Bertorello A,
Germany). 2004;19(12):1340–8. Wartiovaara J, Berggren PO, et al. Defective nephrin
58. Jungraithmayr TC, Bulla M, Dippell J, Greiner C, trafficking caused by missense mutations in the
Griebel M, Leichter HE, et al. Primary focal segmen- NPHS1 gene: insight into the mechanisms of congen-
tal glomerulosclerosis–long-term outcome after pedi- ital nephrotic syndrome. Hum Mol Genet.
atric renal transplantation. Pediatr Transplant. 2001;10(23):2637–44.
2005;9(2):226–31. 71. Liu XL, Done SC, Yan K, Kilpelainen P, Pikkarainen T,
59. Tejani A, Stablein DH. Recurrence of focal segmental Tryggvason K. Defective trafficking of nephrin mis-
glomerulosclerosis posttransplantation: a special sense mutants rescued by a chemical chaperone. J Am
report of the North American Pediatric Renal Soc Nephrol. 2004;15(7):1731–8.
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 829

72. Kitamura A, Tsukaguchi H, Hiramoto R, Shono A, human autosomal recessive steroid-resistant


Doi T, Kagami S, et al. A familial childhood- nephrotic syndrome. Kidney Int. 2011;80(4):389–96.
onset relapsing nephrotic syndrome. Kidney Int. 86. Al-Hamed MH, Al-Sabban E, Al-Mojalli H,
2007;71(9):946–51. Al-Harbi N, Faqeih E, Al Shaya H, et al. A molecular
73. Philippe A, Nevo F, Esquivel EL, Reklaityte D, genetic analysis of childhood nephrotic syndrome in a
Gribouval O, Tete MJ, et al. Nephrin mutations can cohort of Saudi Arabian families. J Hum Genet.
cause childhood-onset steroid-resistant nephrotic syn- 2013;58(7):480–9.
drome. J Am Soc Nephrol. 2008;19(10):1871–8. 87. Mao J, Wang D, Mataleena P, He B, Niu D,
74. Santin S, Garcia-Maset R, Ruiz P, Gimenez I, Katayama K, et al. Myo1e impairment results in
Zamora I, Pena A, et al. Nephrin mutations cause actin reorganization, podocyte dysfunction, and pro-
childhood- and adult-onset focal segmental teinuria in zebrafish and cultured podocytes. PLoS
glomerulosclerosis. Kidney Int. 2009;76(12): One. 2013;8(8):e72750.
1268–76. 88. Krendel M, Kim SV, Willinger T, Wang T,
75. Wing MR, Bourdon DM, Harden TK. PLC-epsilon: a Kashgarian M, Flavell RA, et al. Disruption of Myo-
shared effector protein in Ras-, Rho-, and G alpha beta sin 1e promotes podocyte injury. J Am Soc Nephrol.
gamma-mediated signaling. Mol Interv. 2003;3(5): 2009;20(1):86–94.
273–80. 89. Chase SE, Encina CV, Stolzenburg LR, Tatum AH,
76. Hinkes B, Wiggins RC, Gbadegesin R, Vlangos CN, Holzman LB, Krendel M. Podocyte-specific
Seelow D, Nurnberg G, et al. Positional cloning knockout of myosin 1e disrupts glomerular filtration.
uncovers mutations in PLCE1 responsible for a Am J Physiol Renal Physiol. 2012;303(7):
nephrotic syndrome variant that may be reversible. F1099–106.
Nat Genet. 2006;38(12):1397–405. 90. Thomas PE, Wharram BL, Goyal M, Wiggins JE,
77. Lehtonen S, Ryan JJ, Kudlicka K, Iino N, Zhou H, Holzman LB, Wiggins RC. GLEPP1, a renal glomer-
Farquhar MG. Cell junction-associated proteins ular epithelial cell (podocyte) membrane protein-
IQGAP1, MAGI-2, CASK, spectrins, and alpha- tyrosine phosphatase. Identification, molecular
actinin are components of the nephrin multiprotein cloning, and characterization in rabbit. J Biol Chem.
complex. Proc Natl Acad Sci U S A. 2005;102 1994;269(31):19953–62.
(28):9814–9. 91. Wharram BL, Goyal M, Gillespie PJ, Wiggins JE,
78. Gbadegesin R, Hinkes BG, Hoskins BE, Vlangos CN, Kershaw DB, Holzman LB, et al. Altered podocyte
Heeringa SF, Liu J, et al. Mutations in PLCE1 are a structure in GLEPP1 (Ptpro)-deficient mice associ-
major cause of isolated diffuse mesangial sclerosis ated with hypertension and low glomerular filtration
(IDMS). Nephrol Dial Transplant. 2008;23(4): rate. J Clin Invest. 2000;106(10):1281–90.
1291–7. 92. Ozaltin F, Ibsirlioglu T, Taskiran EZ, Baydar DE,
79. Boyer O, Benoit G, Gribouval O, Nevo F, Kaymaz F, Buyukcelik M, et al. Disruption of
Pawtowski A, Bilge I, et al. Mutational analysis of PTPRO causes childhood-onset nephrotic syndrome.
the PLCE1 gene in steroid resistant nephrotic syn- Am J Hum Genet. 2011;89(1):139–47.
drome. J Med Genet. 2010;47(7):445–52. 93. Ebarasi L, Ashraf S, Bierzynska A, Gee HY,
80. Quaggin SE. A new piece in the nephrotic puzzle. Nat McCarthy HJ, Lovric S, et al. Defects of CRB2
Genet. 2006;38(12):1360–1. cause steroid-resistant nephrotic syndrome. Am J
81. Gilbert RD, Turner CL, Gibson J, Bass PS, Haq MR, Hum Genet. 2015;96(1):153–61.
Cross E, et al. Mutations in phospholipase C epsilon 94. Xiao Z, Patrakka J, Nukui M, Chi L, Niu D,
1 are not sufficient to cause diffuse mesangial sclero- Betsholtz C, et al. Deficiency in Crumbs homolog
sis. Kidney Int. 2009;75(4):415–9. 2 (Crb2) affects gastrulation and results in embryonic
82. Riazuddin S, Castelein CM, Ahmed ZM, Lalwani lethality in mice. Dev Dyn. 2011;240(12):2646–56.
AK, Mastroianni MA, Naz S, et al. Dominant modi- 95. Slavotinek A, Kaylor J, Pierce H, Cahr M, DeWard
fier DFNM1 suppresses recessive deafness DFNB26. SJ, Schneidman-Duhovny D, et al. CRB2 mutations
Nat Genet. 2000;26(4):431–4. produce a phenotype resembling congenital nephro-
83. Katsanis N, Ansley SJ, Badano JL, Eichers ER, Lewis sis, Finnish type, with cerebral ventriculomegaly and
RA, Hoskins BE, et al. Triallelic inheritance in raised alpha-fetoprotein. Am J Hum Genet. 2015;96
Bardet-Biedl syndrome, a Mendelian recessive disor- (1):162–9.
der. Science. 2001;293(5538):2256–9. 96. Dustin ML, Olszowy MW, Holdorf AD, Li J,
84. Mele C, Iatropoulos P, Donadelli R, Calabria A, Bromley S, Desai N, et al. A novel adaptor protein
Maranta R, Cassis P, et al. MYO1E mutations and orchestrates receptor patterning and cytoskeletal
childhood familial focal segmental glomerulo- polarity in T-cell contacts. Cell. 1998;94(5):667–77.
sclerosis. N Engl J Med. 2011;365(4):295–306. 97. Shih NY, Li J, Karpitskii V, Nguyen A, Dustin ML,
85. Sanna-Cherchi S, Burgess KE, Nees SN, Caridi G, Kanagawa O, et al. Congenital nephrotic syndrome in
Weng PL, Dagnino M, et al. Exome sequencing iden- mice lacking CD2-associated protein. Science.
tified MYO1E and NEIL1 as candidate genes for 1999;286(5438):312–5.
830 O. Boyer et al.

98. Lehtonen S, Zhao F, Lehtonen E. CD2-associated glomerulosclerosis associated with an ACTN4 muta-
protein directly interacts with the actin cytoskeleton. tion and paternal germline mosaicism. Am J Kidney
Am J Physiol Renal Physiol. 2002;283(4):F734–43. Dis. 2008;51(5):834–8.
99. Grunkemeyer JA, Kwoh C, Huber TB, Shaw AS. 113. Barua M, Brown EJ, Charoonratana VT, Genovese G,
CD2-associated protein (CD2AP) expression in Sun H, Pollak MR. Mutations in the INF2 gene
podocytes rescues lethality of CD2AP deficiency. account for a significant proportion of familial but
J Biol Chem. 2005;280(33):29677–81. not sporadic focal and segmental glomerulosclerosis.
100. Kim JM, Wu H, Green G, Winkler CA, Kopp JB, Kidney Int. 2013;83(2):316–22.
Miner JH, et al. CD2-associated protein haploinsuf- 114. Winn MP, Conlon PJ, Lynn KL, Howell DN,
ficiency is linked to glomerular disease susceptibility. Slotterbeck BD, Smith AH, et al. Linkage of a gene
Science. 2003;300(5623):1298–300. causing familial focal segmental glomerulosclerosis
101. Lowik MM, Groenen PJ, Pronk I, Lilien MR, to chromosome 11 and further evidence of genetic
Goldschmeding R, Dijkman HB, et al. Focal segmen- heterogeneity. Genomics. 1999;58(2):113–20.
tal glomerulosclerosis in a patient homozygous 115. Winn MP, Conlon PJ, Lynn KL, Farrington MK,
for a CD2AP mutation. Kidney Int. 2007;72(10): Creazzo T, Hawkins AF, et al. A mutation in the
1198–203. TRPC6 cation channel causes familial focal segmen-
102. Davis EE, Zhang Q, Liu Q, Diplas BH, Davey LM, tal glomerulosclerosis. Science. 2005;308(5729):
Hartley J, et al. TTC21B contributes both causal 1801–4.
and modifying alleles across the ciliopathy spectrum. 116. Heeringa SF, Moller CC, Du J, Yue L, Hinkes B,
Nat Genet. 2011;43(3):189–96. Chernin G, et al. A novel TRPC6 mutation that causes
103. Mathis BJ, Calabrese KE, Slick GL. Familial glomer- childhood FSGS. PLoS One. 2009;4(11):e7771.
ular disease with asymptomatic proteinuria and 117. Santin S, Ars E, Rossetti S, Salido E, Silva I, Garcia-
nephrotic syndrome: a new clinical entity. J Am Oste- Maset R, et al. TRPC6 mutational analysis in a large
opath Assoc. 1992;92(7):875–80. 7. cohort of patients with focal segmental glomerulo-
104. Mathis BJ, Kim SH, Calabrese K, Haas M, Seidman sclerosis. Nephrol Dial Transplant. 2009;24(10):
JG, Seidman CE, et al. A locus for inherited focal 3089–96.
segmental glomerulosclerosis maps to chromosome 118. Gigante M, Caridi G, Montemurno E, Soccio M,
19q13. Kidney Int. 1998;53(2):282–6. D’Apolito M, Cerullo G, et al. TRPC6 mutations in
105. Kaplan JM, Kim SH, North KN, Rennke H, Correia children with steroid-resistant nephrotic syndrome
LA, Tong HQ, et al. Mutations in ACTN4, encoding and atypical phenotype. Clin J Am Soc Nephrol.
alpha-actinin-4, cause familial focal segmental 2011;6(7):1626–34.
glomerulosclerosis. Nat Genet. 2000;24(3):251–6. 119. Lion M, Boyer O, Niaudet P, Charbit M, Eckart P,
106. Ichimura K, Kurihara H, Sakai T. Actin filament Nevo F, et al. Mutation de TRPC6: a rare cause of
organization of foot processes in rat podocytes. steroid-resistant nephrotic syndrome in children.
J Histochem Cytochem. 2003;51(12):1589–600. Pediatr Nephrol. 2012;27:1702.
107. Weins A, Kenlan P, Herbert S, Le TC, Villegas I, 120. Nilius B, Owsianik G, Voets T, Peters JA. Transient
Kaplan BS, et al. Mutational and biological analysis receptor potential cation channels in disease. Physiol
of alpha-actinin-4 in focal segmental glomerulo- Rev. 2007;87(1):165–217.
sclerosis. J Am Soc Nephrol. 2005;16(12):3694–701. 121. Spassova MA, Hewavitharana T, Xu W, Soboloff J,
108. Yao J, Le TC, Kos CH, Henderson JM, Allen PG, Gill DL. A common mechanism underlies
Denker BM, et al. Alpha-actinin-4-mediated FSGS: stretch activation and receptor activation of TRPC6
an inherited kidney disease caused by an aggregated channels. Proc Natl Acad Sci U S A. 2006;103
and rapidly degraded cytoskeletal protein. PLoS Biol. (44):16586–91.
2004;2(6):e167. 122. Brown EJ, Schlondorff JS, Becker DJ, Tsukaguchi H,
109. Michaud JL, Lemieux LI, Dube M, Vanderhyden BC, Tonna SJ, Uscinski AL, et al. Mutations in the formin
Robertson SJ, Kennedy CR. Focal and segmental gene INF2 cause focal segmental glomerulosclerosis.
glomerulosclerosis in mice with podocyte-specific Nat Genet. 2010;42(1):72–6.
expression of mutant alpha-actinin-4. J Am Soc 123. Boyer O, Benoit G, Gribouval O, Nevo F, Tete MJ,
Nephrol. 2003;14(5):1200–11. Dantal J, et al. Mutations in INF2 are a major cause of
110. Michaud JL, Chaisson KM, Parks RJ, Kennedy autosomal dominant focal segmental glomerulo-
CR. FSGS-associated alpha-actinin-4 (K256E) sclerosis. J Am Soc Nephrol. 2011;22(2):239–45.
impairs cytoskeletal dynamics in podocytes. Kidney 124. Boyer O, Nevo F, Plaisier E, Funalot B, Gribouval O,
Int. 2006;70(6):1054–61. Benoit G, et al. INF2 mutations in Charcot-
111. Pollak MR, Alexander MP, Henderson JM. A case of Marie-tooth disease with glomerulopathy. N Engl J
familial kidney disease. Clin J Am Soc Nephrol. Med. 2011;365(25):2377–88.
2007;2(6):1367–74. 125. Chesarone MA, DuPage AG, Goode BL. Unleashing
112. Choi HJ, Lee BH, Cho HY, Moon KC, Ha IS, formins to remodel the actin and microtubule cyto-
Nagata M, et al. Familial focal segmental skeletons. Nat Rev Mol Cell Biol. 2010;11(1):62–74.
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 831

126. Akilesh S, Suleiman H, Yu H, Stander MC, Lavin P, mutation database. Am J Hum Genet. 1998;62(4):
Gbadegesin R, et al. Arhgap24 inactivates Rac1 in 824–33.
mouse podocytes, and a mutant form is associated 140. Klamt B, Koziell A, Poulat F, Wieacker P, Scambler P,
with familial focal segmental glomerulosclerosis. Berta P, et al. Frasier syndrome is caused by defective
J Clin Invest. 2011;121(10):4127–37. alternative splicing of WT1 leading to an altered ratio
127. Gbadegesin RA, Hall G, Adeyemo A, Hanke N, of WT1 +/KTS splice isoforms. Hum Mol Genet.
Tossidou I, Burchette J, et al. Mutations in the gene 1998;7(4):709–14.
that encodes the F-actin binding protein anillin cause 141. Hall G, Gbadegesin RA, Lavin P, Wu G, Liu Y, Oh
FSGS. J Am Soc Nephrol. 2014;25(9):1991–2002. EC, et al. A novel missense mutation of Wilms’
128. Dreyer SD, Zhou G, Baldini A, Winterpacht A, Tumor 1 causes autosomal dominant FSGS. J Am
Zabel B, Cole W, et al. Mutations in LMX1B cause Soc Nephrol. 2014;26(4):831–43.
abnormal skeletal patterning and renal dysplasia in 142. Benetti E, Caridi G, Malaventura C, Dagnino M,
nail patella syndrome. Nat Genet. 1998;19(1):47–50. Leonardi E, Artifoni L, et al. A novel WT1 gene
129. Boyer O, Woerner S, Yang F, Oakeley EJ, Linghu B, mutation in a three-generation family with progres-
Gribouval O, et al. LMX1B mutations cause heredi- sive isolated focal segmental glomerulosclerosis. Clin
tary FSGS without extrarenal involvement. J Am Soc J Am Soc Nephrol. 2010;5(4):698–702.
Nephrol. 2013;24(8):1216–22. 143. Guaragna MS, Lutaif AC, Piveta CS, Belangero VM,
130. Edwards N, Rice SJ, Raman S, Hynes AM, Maciel-Guerra AT, Guerra Jr G, et al. Two distinct
Srivastava S, Moore I, et al. A novel LMX1B muta- WT1 mutations identified in patients and relatives
tion in a family with end-stage renal disease of with isolated nephrotic proteinuria. Biochem Biophys
‘unknown cause’. Clin Kidney J. 2015;8(1):113–9. Res Commun. 2013;441(2):371–6.
131. Malone AF, Phelan PJ, Hall G, Cetincelik U, 144. Zhu C, Zhao F, Zhang W, Wu H, Chen Y, Ding G,
Homstad A, Alonso AS, et al. Rare hereditary et al. A familial WT1 mutation associated with
COL4A3/COL4A4 variants may be mistaken for incomplete Denys-Drash syndrome. Eur J Pediatr.
familial focal segmental glomerulosclerosis. Kidney 2013;172(10):1357–62.
Int. 2014;86(6):1253–9. 145. Coppes MJ, Liefers GJ, Higuchi M, Zinn AB, Balfe
132. Moriniere V, Dahan K, Hilbert P, Lison M, Lebbah S, JW, Williams BR. Inherited WT1 mutation in Denys-
Topa A, et al. Improving mutation screening in famil- Drash syndrome. Cancer Res. 1992;52(21):6125–8.
ial hematuric nephropathies through next generation 146. Gessler M, Poustka A, Cavenee W, Neve RL, Orkin
sequencing. J Am Soc Nephrol. 2014;25(12): SH, Bruns GA. Homozygous deletion in Wilms
2740–51. tumours of a zinc-finger gene identified by chromo-
133. Xie J, Wu X, Ren H, Wang W, Wang Z, Pan X, some jumping. Nature. 1990;343(6260):774–8.
et al. COL4A3 mutations cause focal segmental 147. Kreidberg JA, Sariola H, Loring JM, Maeda M,
glomerulosclerosis. J Mol Cell Biol. 2014;6(6): Pelletier J, Housman D, et al. WT-1 is required for
498–505. early kidney development. Cell. 1993;74(4):679–91.
134. Lin F, Bian F, Zou J, Wu X, Shan J, Lu W, et al. Whole 148. Moore AW, Schedl A, McInnes L, Doyle M, Hecksher-
exome sequencing reveals novel COL4A3 and Sorensen J, Hastie ND. YAC transgenic analysis
COL4A4 mutations and resolves diagnosis in Chi- reveals Wilms’ tumour 1 gene activity in the prolifer-
nese families with kidney disease. BMC Nephrol. ating coelomic epithelium, developing diaphragm and
2014;15:175. limb. Mech Dev. 1998;79(1–2):169–84.
135. Pritchard-Jones K, Fleming S, Davidson D, 149. Davies JA, Ladomery M, Hohenstein P, Michael L,
Bickmore W, Porteous D, Gosden C, et al. The can- Shafe A, Spraggon L, et al. Development of an
didate Wilms’ tumour gene is involved in genitouri- siRNA-based method for repressing specific genes
nary development. Nature. 1990;346(6280):194–7. in renal organ culture and its use to show that the
136. Rivera MN, Haber DA. Wilms’ tumour: connecting Wt1 tumour suppressor is required for nephron dif-
tumorigenesis and organ development in the kidney. ferentiation. Hum Mol Genet. 2004;13(2):235–46.
Nat Rev Cancer. 2005;5(9):699–712. 150. Denys P, Malvaux P, Van Den Berghe H, Tanghe W,
137. Morrison AA, Viney RL, Saleem MA, Ladomery Proesmans W. Association of an anatomo-
MR. New insights into the function of the Wilms pathological syndrome of male pseudohermaph-
tumor suppressor gene WT1 in podocytes. Am J roditism, Wilms’ tumor, parenchymatous nephropa-
Physiol Renal Physiol. 2008;295(1):F12–7. thy and XX/XY mosaicism. Arch Fr Pediatr.
138. Niaudet P, Gubler MC. WT1 and glomerular diseases. 1967;24(7):729–39.
Pediatr Nephrol. 2006;21(11):1653–60. 151. Drash A, Sherman F, Hartmann WH, Blizzard RM. A
139. Jeanpierre C, Denamur E, Henry I, Cabanis MO, syndrome of pseudohermaphroditism, Wilms’ tumor,
Luce S, Cecille A, et al. Identification of constitu- hypertension, and degenerative renal disease.
tional WT1 mutations, in patients with isolated dif- J Pediatr. 1970;76(4):585–93.
fuse mesangial sclerosis, and analysis of genotype/ 152. Habib R, Loirat C, Gubler MC, Niaudet P,
phenotype correlations by use of a computerized Bensman A, Levy M, et al. The nephropathy
832 O. Boyer et al.

associated with male pseudohermaphroditism and syndrome and occur in exons 8 and 9. Pediatr Res.
Wilms’ tumor (Drash syndrome): a distinctive glo- 2006;59(2):325–31.
merular lesion–report of 10 cases. Clin Nephrol. 166. Zenker M, Tralau T, Lennert T, Pitz S, Mark K,
1985;24(6):269–78. Madlon H, et al. Congenital nephrosis, mesangial
153. Hastie ND. Dominant negative mutations in the sclerosis, and distinct eye abnormalities with
Wilms tumour (WT1) gene cause Denys-Drash microcoria: an autosomal recessive syndrome. Am J
syndrome–proof that a tumour-suppressor gene Med Genet. 2004;130(2):138–45.
plays a crucial role in normal genitourinary develop- 167. Pierson M, Cordier J, Hervouuet F, Rauber G.
ment. Hum Mol Genet. 1992;1(5):293–5. [An unusual congenital and familial congenital
154. Pelletier J, Bruening W, Kashtan CE, Mauer SM, malformative combination involving the eye and
Manivel JC, Striegel JE, et al. Germline mutations kidney]. J Genet Hum. 1963;12:184–213.
in the Wilms’ tumor suppressor gene are associated 168. Zenker M, Aigner T, Wendler O, Tralau T,
with abnormal urogenital development in Denys- Muntefering H, Fenski R, et al. Human laminin
Drash syndrome. Cell. 1991;67(2):437–47. beta2 deficiency causes congenital nephrosis with
155. Lipska BS, Ranchin B, Iatropoulos P, Gellermann J, mesangial sclerosis and distinct eye abnormalities.
Melk A, Ozaltin F, et al. Genotype-phenotype Hum Mol Genet. 2004;13(21):2625–32.
associations in WT1 glomerulopathy. Kidney Int. 169. Noakes PG, Miner JH, Gautam M, Cunningham JM,
2014;85(5):1169–78. Sanes JR, Merlie JP. The renal glomerulus of mice
156. Little M, Wells C. A clinical overview of WT1 gene lacking s-laminin/laminin beta 2: nephrosis despite
mutations. Hum Mutat. 1997;9(3):209–25. molecular compensation by laminin beta 1. Nat
157. Ito S, Takata A, Hataya H, Ikeda M, Kikuchi H, Genet. 1995;10(4):400–6.
Hata J, et al. Isolated diffuse mesangial sclerosis 170. Hasselbacher K, Wiggins RC, Matejas V, Hinkes BG,
and Wilms tumor suppressor gene. J Pediatr. Mucha B, Hoskins BE, et al. Recessive missense
2001;138(3):425–7. mutations in LAMB2 expand the clinical spectrum
158. Moorthy AV, Chesney RW, Lubinsky M. Chronic of LAMB2-associated disorders. Kidney Int.
renal failure and XY gonadal dysgenesis: “Frasier” 2006;70(6):1008–12.
syndrome–a commentary on reported cases. Am J 171. Choi HJ, Lee BH, Kang JH, Jeong HJ, Moon KC, Ha
Med Genet Suppl. 1987;3:297–302. IS, et al. Variable phenotype of Pierson syndrome.
159. Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call Pediatr Nephrol (Berlin Germany). 2008;23(6):
KM, Housman DE. Alternative splicing and genomic 995–1000.
structure of the Wilms tumor gene WT1. Proc Natl 172. Kagan M, Cohen AH, Matejas V, Vlangos C, Zenker
Acad Sci U S A. 1991;88(21):9618–22. M. A milder variant of Pierson syndrome. Pediatr
160. Hammes A, Guo JK, Lutsch G, Leheste JR, Nephrol (Berlin Germany). 2008;23(2):323–7.
Landrock D, Ziegler U, et al. Two splice variants of 173. Matejas V, Al-Gazali L, Amirlak I, Zenker M. A
the Wilms’ tumor 1 gene have distinct functions dur- syndrome comprising childhood-onset glomerular
ing sex determination and nephron formation. Cell. kidney disease and ocular abnormalities with progres-
2001;106(3):319–29. sive loss of vision is caused by mutated LAMB2.
161. Barbaux S, Niaudet P, Gubler MC, Grunfeld JP, Nephrol Dial Transplant. 2006;21(11):3283–6.
Jaubert F, Kuttenn F, et al. Donor splice-site mutations 174. Matejas V, Hinkes B, Alkandari F, Al-Gazali L,
in WT1 are responsible for Frasier syndrome. Nat Annexstad E, Aytac MB, et al. Mutations in the
Genet. 1997;17(4):467–70. human laminin beta2 (LAMB2) gene and the associ-
162. Miller RW, Fraumeni Jr JF, Manning MD. Association ated phenotypic spectrum. Hum Mutat. 2010;31(9):
of Wilms’s tumor with Aniridia, hemihypertrophy and 992–1002.
other congenital malformations. N Engl J Med. 175. Sasaki T, Fassler R, Hohenester E. Laminin: the crux
1964;270:922–7. of basement membrane assembly. J Cell Biol.
163. Demmer L, Primack W, Loik V, Brown R, 2004;164(7):959–63.
Therville N, McElreavey K. Frasier syndrome: a 176. Jarad G, Cunningham J, Shaw AS, Miner JH.
cause of focal segmental glomerulosclerosis in a Proteinuria precedes podocyte abnormalities
46 XX female. J Am Soc Nephrol. 1999;10(10): inLamb2/ mice, implicating the glomerular base-
2215–8. ment membrane as an albumin barrier. J Clin Invest.
164. Denamur E, Bocquet N, Mougenot B, Da Silva F, 2006;116(8):2272–9.
Martinat L, Loirat C, et al. Mother-to-child transmit- 177. Chen YM, Kikkawa Y, Miner JH. A missense
ted WT1 splice-site mutation is responsible for LAMB2 mutation causes congenital nephrotic syn-
distinct glomerular diseases. J Am Soc Nephrol. drome by impairing laminin secretion. J Am Soc
1999;10(10):2219–23. Nephrol. 2011;22(5):849–58.
165. Mucha B, Ozaltin F, Hinkes BG, Hasselbacher K, Ruf 178. Suh JH, Jarad G, VanDeVoorde RG, Miner JH.
RG, Schultheiss M, et al. Mutations in the Wilms’ Forced expression of laminin beta1 in podocytes pre-
tumor 1 gene cause isolated steroid resistant nephrotic vents nephrotic syndrome in mice lacking laminin
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 833

beta2, a model for Pierson syndrome. Proc Natl Acad 193. Bongers EM, de Wijs IJ, Marcelis C, Hoefsloot LH,
Sci U S A. 2011;108(37):15348–53. Knoers NV. Identification of entire LMX1B gene
179. Guidera KJ, Satterwhite Y, Ogden JA, Pugh L, deletions in nail patella syndrome: evidence for
Ganey T. Nail patella syndrome: a review of 44 haploinsufficiency as the main pathogenic mecha-
orthopaedic patients. J Pediatr Orthop. 1991;11(6): nism underlying dominant inheritance in man. Eur J
737–42. Hum Genet. 2008;16(10):1240–4.
180. Bennett WM, Musgrave JE, Campbell RA, Elliot D, 194. Chen H, Lun Y, Ovchinnikov D, Kokubo H, Oberg
Cox R, Brooks RE, et al. The nephropathy of the nail- KC, Pepicelli CV, et al. Limb and kidney defects in
patella syndrome. Clinicopathologic analysis of Lmx1b mutant mice suggest an involvement of
11 kindred. Am J Med. 1973;54(3):304–19. LMX1B in human nail patella syndrome. Nat Genet.
181. Looij Jr BJ, te Slaa RL, Hogewind BL, van de Kamp 1998;19(1):51–5.
JJ. Genetic counselling in hereditary osteo- 195. Morello R, Zhou G, Dreyer SD, Harvey SJ,
onychodysplasia (HOOD, nail-patella syndrome) Ninomiya Y, Thorner PS, et al. Regulation of glomer-
with nephropathy. J Med Genet. 1988;25(10):682–6. ular basement membrane collagen expression by
182. McIntosh I, Dreyer SD, Clough MV, Dunston JA, LMX1B contributes to renal disease in nail patella
Eyaid W, Roig CM, et al. Mutation analysis of syndrome. Nat Genet. 2001;27(2):205–8.
LMX1B gene in nail-patella syndrome patients. Am 196. Miner JH, Morello R, Andrews KL, Li C, Antignac C,
J Hum Genet. 1998;63(6):1651–8. Shaw AS, et al. Transcriptional induction of slit
183. Knoers NV, Bongers EM, van Beersum SE, Lommen diaphragm genes by Lmx1b is required in
EJ, van Bokhoven H, Hol FA. Nail-patella syndrome: podocyte differentiation. J Clin Invest. 2002;109(8):
identification of mutations in the LMX1B gene in 1065–72.
Dutch families. J Am Soc Nephrol. 2000;11(9): 197. Rohr C, Prestel J, Heidet L, Hosser H, Kriz W, John-
1762–6. son RL, et al. The LIM-homeodomain transcription
184. Bongers EM, Huysmans FT, Levtchenko E, de Rooy factor Lmx1b plays a crucial role in podocytes. J Clin
JW, Blickman JG, Admiraal RJ, et al. Genotype- Invest. 2002;109(8):1073–82.
phenotype studies in nail-patella syndrome show 198. Heidet L, Bongers EM, Sich M, Zhang SY, Loirat C,
that LMX1B mutation location is involved in the Meyrier A, et al. In vivo expression of putative
risk of developing nephropathy. Eur J Hum Genet. LMX1B targets in nail-patella syndrome kidneys.
2005;13(8):935–46. Am J Pathol. 2003;163(1):145–55.
185. Sweeney E, Fryer A, Mountford R, Green A, 199. Schimke RN, Horton WA, King CR. Chondroitin-
McIntosh I. Nail patella syndrome: a review of the 6-sulphaturia, defective cellular immunity, and
phenotype aided by developmental biology. J Med nephrotic syndrome. Lancet. 1971;2(7733):1088–9.
Genet. 2003;40(3):153–62. 200. Saraiva JM, Dinis A, Resende C, Faria E, Gomes C,
186. Gubler MC. Inherited diseases of the glomerular base- Correia AJ, et al. Schimke immuno-osseous dyspla-
ment membrane. Nat Clin Pract Nephrol. 2008;4(1): sia: case report and review of 25 patients. J Med
24–37. Genet. 1999;36(10):786–9.
187. Ben-Bassat M, Cohen L, Rosenfeld J. The glomerular 201. Boerkoel CF, O’Neill S, Andre JL, Benke PJ,
basement membrane in the nail-patella syndrome. Bogdanovic R, Bulla M, et al. Manifestations and
Arch Pathol. 1971;92(5):350–5. treatment of Schimke immuno-osseous dysplasia:
188. Hoyer JR, Michael AF, Vernier RL. Renal disease in 14 new cases and a review of the literature. Eur J
nail-patella syndrome: clinical and morphologic stud- Pediatr. 2000;159(1–2):1–7.
ies. Kidney Int. 1972;2(4):231–8. 202. Clewing JM, Antalfy BC, Lucke T, Najafian B,
189. Gubler MC, Levy M, Naizot C, Habib R. Marwedel KM, Hori A, et al. Schimke immuno-
Glomerular basement membrane changes in hereditary osseous dysplasia: a clinicopathological correlation.
glomerular diseases. Ren Physiol. 1980;3(1–6):405–13. J Med Genet. 2007;44(2):122–30.
190. Lichter PR, Richards JE, Downs CA, Stringham HM, 203. Lucke T, Clewing JM, Boerkoel CF, Hartmann H,
Boehnke M, Farley FA. Cosegregation of open-angle Das AM, Knauth M, et al. Cerebellar atrophy in
glaucoma and the nail-patella syndrome. Am J Schimke-immuno-osseous dysplasia. Am J Med
Ophthalmol. 1997;124(4):506–15. Genet. 2007;143A(17):2040–5.
191. Vollrath D, Jaramillo-Babb VL, Clough MV, 204. Boerkoel CF, Takashima H, John J, Yan J,
McIntosh I, Scott KM, Lichter PR, et al. Loss-of- Stankiewicz P, Rosenbarker L, et al. Mutant chroma-
function mutations in the LIM-homeodomain gene, tin remodeling protein SMARCAL1 causes Schimke
LMX1B, in nail-patella syndrome. Hum Mol Genet. immuno-osseous dysplasia. Nat Genet. 2002;30(2):
1998;7(7):1091–8. 215–20.
192. Hamlington JD, Jones C, McIntosh I. Twenty-two 205. Clewing JM, Fryssira H, Goodman D, Smithson SF,
novel LMX1B mutations identified in nail patella Sloan EA, Lou S, et al. Schimke immunoosseous
syndrome (NPS) patients. Hum Mutat. 2001;18(5): dysplasia: suggestions of genetic diversity. Hum
458. Mutat. 2007;28(3):273–83.
834 O. Boyer et al.

206. DiMauro S, Moraes CT. Mitochondrial encephalo- 219. Cheong HI, Chae JH, Kim JS, Park HW, Ha IS,
myopathies. Arch Neurol. 1993;50(11):1197–208. Hwang YS, et al. Hereditary glomerulopathy associ-
207. Anderson S, Bankier AT, Barrell BG, de Bruijn MH, ated with a mitochondrial tRNA(Leu) gene mutation.
Coulson AR, Drouin J, et al. Sequence and organiza- Pediatr Nephrol (Berlin, Germany). 1999;13(6):
tion of the human mitochondrial genome. Nature. 477–80.
1981;290(5806):457–65. 220. Yamagata K, Muro K, Usui J, Hagiwara M, Kai H,
208. Goto Y, Nonaka I, Horai S. A mutation in the tRNA Arakawa Y, et al. Mitochondrial DNA mutations in
(Leu)(UUR) gene associated with the MELAS sub- focal segmental glomerulosclerosis lesions. J Am Soc
group of mitochondrial encephalomyopathies. Nephrol. 2002;13(7):1816–23.
Nature. 1990;348(6302):651–3. 221. Guery B, Choukroun G, Noel LH, Clavel P, Rotig A,
209. van den Ouweland JM, Lemkes HH, Ruitenbeek W, Lebon S, et al. The spectrum of systemic involvement
Sandkuijl LA, de Vijlder MF, Struyvenberg PA, et al. in adults presenting with renal lesion and mitochon-
Mutation in mitochondrial tRNA(Leu)(UUR) gene in drial tRNA(Leu) gene mutation. J Am Soc Nephrol.
a large pedigree with maternally transmitted type II 2003;14(8):2099–108.
diabetes mellitus and deafness. Nat Genet. 1992;1 222. Ueda Y, Ando A, Nagata T, Yanagida H, Yagi K,
(5):368–71. Sugimoto K, et al. A boy with mitochondrial disease:
210. Chinnery PF, Turnbull DM. Mitochondrial DNA and asymptomatic proteinuria without neuromyopathy.
disease. Lancet. 1999;354 Suppl 1:SI17–21. Pediatr Nephrol (Berlin, Germany). 2004;19(1):
211. Yoshida R, Ishida Y, Hozumi T, Ueno H, 107–10.
Kishimoto M, Kasuga M, et al. Congestive heart 223. Unal S, Kalkanoglu HS, Kocaefe C, Gucer S, Ozen S,
failure in mitochondrial diabetes mellitus. Lancet. Turanli G, et al. Four-month-old infant with focal
1994;344(8933):1375. segmental glomerulosclerosis and mitochondrial
212. Jansen JJ, Maassen JA, van der Woude FJ, Lemmink DNA deletion. J Child Neurol. 2005;20(1):83–4.
HA, van den Ouweland JM, t’ Hart LM, et al. Muta- 224. Emma F, Bertini E, Salviati L, Montini G. Renal
tion in mitochondrial tRNA(Leu(UUR)) gene associ- involvement in mitochondrial cytopathies. Pediatr
ated with progressive kidney disease. J Am Soc Nephrol. 2012;27(4):539–50.
Nephrol. 1997;8(7):1118–24. 225. Hallman TM, Peng M, Meade R, Hancock WW,
213. Kurogouchi F, Oguchi T, Mawatari E, Yamaura S, Madaio MP, Gasser DL. The mitochondrial and kid-
Hora K, Takei M, et al. A case of mitochondrial ney disease phenotypes of kd/kd mice under germfree
cytopathy with a typical point mutation for MELAS, conditions. J Autoimmun. 2006;26(1):1–6.
presenting with severe focal-segmental glomerulo- 226. Saiki R, Lunceford AL, Shi Y, Marbois B, King R,
sclerosis as main clinical manifestation. Am J Pachuski J, et al. Coenzyme Q10 supplementation
Nephrol. 1998;18(6):551–6. rescues renal disease in Pdss2kd/kd mice with muta-
214. Nakamura S, Yoshinari M, Doi Y, Yoshizumi H, tions in prenyl diphosphate synthase subunit 2. Am J
Katafuchi R, Yokomizo Y, et al. Renal complications Physiol Renal Physiol. 2008;295(5):F1535–44.
in patients with diabetes mellitus associated with an A 227. Quinzii C, Naini A, Salviati L, Trevisson E, Navas P,
to G mutation of mitochondrial DNA at the 3243 Dimauro S, et al. A mutation in para-
position of leucine tRNA. Diabetes Res Clin Pract. hydroxybenzoate-polyprenyl transferase (COQ2)
1999;44(3):183–9. causes primary coenzyme Q10 deficiency. Am J
215. Doleris LM, Hill GS, Chedin P, Nochy D, Bellanne- Hum Genet. 2006;78(2):345–9.
Chantelot C, Hanslik T, et al. Focal segmental 228. Diomedi-Camassei F, Di Giandomenico S, Santorelli
glomerulosclerosis associated with mitochondrial FM, Caridi G, Piemonte F, Montini G, et al. COQ2
cytopathy. Kidney Int. 2000;58(5):1851–8. nephropathy: a newly described inherited mitochon-
216. Hotta O, Inoue CN, Miyabayashi S, Furuta T, driopathy with primary renal involvement. J Am Soc
Takeuchi A, Taguma Y. Clinical and pathologic fea- Nephrol. 2007;18(10):2773–80.
tures of focal segmental glomerulosclerosis with 229. Jakobs BS, van den Heuvel LP, Smeets RJ, de Vries
mitochondrial tRNALeu(UUR) gene mutation. Kid- MC, Hien S, Schaible T, et al. A novel mutation in
ney Int. 2001;59(4):1236–43. COQ2 leading to fatal infantile multisystem disease.
217. Hirano M, Konishi K, Arata N, Iyori M, Saruta T, J Neurol Sci. 2013;326(1–2):24–8.
Kuramochi S, et al. Renal complications in a patient 230. Mollet J, Giurgea I, Schlemmer D, Dallner G,
with A-to-G mutation of mitochondrial DNA at the Chretien D, Delahodde A, et al. Prenyldiphosphate
3243 position of leucine tRNA. Internal Med (Tokyo, synthase, subunit 1 (PDSS1) and OH-benzoate
Japan). 2002;41(2):113–8. polyprenyltransferase (COQ2) mutations in ubiqui-
218. Lowik MM, Hol FA, Steenbergen EJ, Wetzels JF, none deficiency and oxidative phosphorylation disor-
van den Heuvel LP. Mitochondrial tRNALeu(UUR) ders. J Clin Invest. 2007;117(3):765–72.
mutation in a patient with steroid-resistant 231. Scalais E, Chafai R, Van Coster R, Bindl L, Nuttin C,
nephrotic syndrome and focal segmental glomerulo- Panagiotaraki C, et al. Early myoclonic epilepsy,
sclerosis. Nephrol Dial Transplant. 2005;20(2): hypertrophic cardiomyopathy and subsequently a
336–41. nephrotic syndrome in a patient with CoQ10
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 835

deficiency caused by mutations in para- 245. Gamp AC, Tanaka Y, Lullmann-Rauch R, Wittke D,
hydroxybenzoate-polyprenyl transferase (COQ2). D’Hooge R, De Deyn PP, et al. LIMP-2/LGP85 defi-
Eur J Paediatr Neurol. 2013;17(6):625–30. ciency causes ureteric pelvic junction obstruction,
232. Montini G, Malaventura C, Salviati L. Early coenzyme deafness and peripheral neuropathy in mice. Hum
Q10 supplementation in primary coenzyme Q10 defi- Mol Genet. 2003;12(6):631–46.
ciency. N Engl J Med. 2008;358(26):2849–50. 246. Knipper M, Claussen C, Ruttiger L, Zimmermann U,
233. Lopez LC, Schuelke M, Quinzii CM, Kanki T, Lullmann-Rauch R, Eskelinen EL, et al. Deafness
Rodenburg RJ, Naini A, et al. Leigh syndrome with in LIMP2-deficient mice due to early loss of the
nephropathy and CoQ10 deficiency due to decaprenyl potassium channel KCNQ1/KCNE1 in marginal
diphosphate synthase subunit 2 (PDSS2) mutations. cells of the stria vascularis. J Physiol. 2006;576
Am J Hum Genet. 2006;79(6):1125–9. (Pt 1):73–86.
234. Hancock WW, Tsai TL, Madaio MP, Gasser 247. Galloway WH, Mowat AP. Congenital microcephaly
DL. Cutting Edge: multiple autoimmune pathways with hiatus hernia and nephrotic syndrome in two
in kd/kd mice. J Immunol. 2003;171(6):2778–81. sibs. J Med Genet. 1968;5(4):319–21.
235. Barisoni L, Madaio MP, Eraso M, Gasser DL, Nelson 248. Ekstrand JJ, Friedman AL, Stafstrom CE. Galloway-
PJ. The kd/kd mouse is a model of collapsing Mowat syndrome: neurologic features in two sibling
glomerulopathy. J Am Soc Nephrol. 2005;16(10): pairs. Pediatr Neurol. 2012;47(2):129–32.
2847–51. 249. Shapiro LR, Duncan PA, Farnsworth PB, Lefkowitz
236. Peng M, Jarett L, Meade R, Madaio MP, Hancock M. Congenital microcephaly, hiatus hernia and
WW, George Jr AL, et al. Mutant prenyltransferase- nephrotic syndrome: an autosomal recessive syn-
like mitochondrial protein (PLMP) and mitochondrial drome. Birth Defects Orig Artic Ser. 1976;12(5):
abnormalities in kd/kd mice. Kidney Int. 2004;66(1): 275–8.
20–8. 250. Roos RA, Maaswinkel-Mooy PD, vd Loo EM,
237. Heeringa SF, Chernin G, Chaki M, Zhou W, Sloan AJ, Kanhai HH. Congenital microcephaly, infantile
Ji Z, et al. COQ6 mutations in human patients produce spasms, psychomotor retardation, and nephrotic
nephrotic syndrome with sensorineural deafness. syndrome in two sibs. Eur J Pediatr. 1987;146(5):
J Clin Invest. 2011;121(5):2013–24. 532–6.
238. Ashraf S, Gee HY, Woerner S, Xie LX, Vega-Warner- 251. Garty BZ, Eisenstein B, Sandbank J, Kaffe S,
V, Lovric S, et al. ADCK4 mutations promote Dagan R, Gadoth N. Microcephaly and congenital
steroid-resistant nephrotic syndrome through CoQ10 nephrotic syndrome owing to diffuse mesangial scle-
biosynthesis disruption. J Clin Invest. 2013;123(12): rosis: an autosomal recessive syndrome. J Med Genet.
5179–89. 1994;31(2):121–5.
239. Turunen M, Olsson J, Dallner G. Metabolism and 252. Yalcinkaya F, Tumer N, Ekim M, Kuyucu S, Cakar N,
function of coenzyme Q. Biochim Biophys Acta. Ensari C. Congenital microcephaly and infantile
2004;1660(1–2):171–99. nephrotic syndrome–a case report. Pediatr Nephrol.
240. Badhwar A, Berkovic SF, Dowling JP, Gonzales M, 1994;8(1):72–3.
Narayanan S, Brodtmann A, et al. Action myoclonus- 253. Yu CH, Tsai WS, Wang PJ, Tsau YK, Tseng GC,
renal failure syndrome: characterization of a unique Wang TR. Congenital nephrotic syndrome with
cerebro-renal disorder. Brain. 2004;127(Pt 10): microcephaly: report of a case. J Formos Med
2173–82. Assoc. 1994;93(6):528–30.
241. Balreira A, Gaspar P, Caiola D, Chaves J, Beirao I, 254. Hashimoto K, Takeuchi Y, Kida Y, Hasegawa H,
Lima JL, et al. A nonsense mutation in the LIMP-2 Kantake M, Sasaki A, et al. Three siblings of fatal
gene associated with progressive myoclonic epilepsy infantile encephalopathy with olivopontocerebellar
and nephrotic syndrome. Hum Mol Genet. hypoplasia and microcephaly. Brain Dev.
2008;17(14):2238–43. 1998;20(3):169–74.
242. Berkovic SF, Dibbens LM, Oshlack A, Silver JD, 255. de Vries BB, van’t Hoff WG, Surtees RA, Winter
Katerelos M, Vears DF, et al. Array-based gene dis- RM. Diagnostic dilemmas in four infants with
covery with three unrelated subjects shows SCARB2/ nephrotic syndrome, microcephaly and severe devel-
LIMP-2 deficiency causes myoclonus epilepsy and opmental delay. Clin Dysmorphol. 2001;10(2):
glomerulosclerosis. Am J Hum Genet. 2008;82(3): 115–21.
673–84. 256. Steiss JO, Gross S, Neubauer BA, Hahn A. Late-onset
243. Reczek D, Schwake M, Schroder J, Hughes H, nephrotic syndrome and severe cerebellar atrophy in
Blanz J, Jin X, et al. LIMP-2 is a receptor for lyso- Galloway-Mowat syndrome. Neuropediatrics.
somal mannose-6-phosphate-independent targeting 2005;36(5):332–5.
of beta-glucocerebrosidase. Cell. 2007;131(4): 257. Cohen AH, Turner MC. Kidney in Galloway-Mowat
770–83. syndrome: clinical spectrum with description of
244. Eskelinen EL, Tanaka Y, Saftig P. At the acidic edge: pathology. Kidney Int. 1994;45(5):1407–15.
emerging functions for lysosomal membrane pro- 258. Mildenberger E, Lennert T, Kunze J, Jandeck C,
teins. Trends Cell Biol. 2003;13(3):137–45. Waldherr R, Versmold H. Diffuse mesangial sclerosis:
836 O. Boyer et al.

association with unreported congenital anomalies and 272. Caridi G, Lugani F, Dagnino M, Gigante M,
placental enlargement. Acta Paediatr. 1998;87(12): Iolascon A, Falco M, et al. Novel INF2 mutations in
1301–3. an Italian cohort of patients with focal segmental
259. Nakazato H, Hattori S, Karashima S, Kawano T, glomerulosclerosis, renal failure and Charcot-
Seguchi S, Kanahori M, et al. Another autosomal Marie-Tooth neuropathy. Nephrol Dial Transplant.
recessive form of focal glomerulosclerosis with neu- 2014;29 Suppl 4:iv80–6.
rological findings. Pediatr Nephrol. 2002;17(1):16–9. 273. Rodriguez PQ, Lohkamp B, Celsi G, Mache CJ,
260. Shiihara T, Kato M, Kimura T, Matsunaga A, Joh K, Auer-Grumbach M, Wernerson A, et al. Novel INF2
Hayasaka K. Microcephaly, cerebellar atrophy, and mutation p. L77P in a family with glomerulopathy
focal segmental glomerulosclerosis in two brothers: a and Charcot-Marie-Tooth neuropathy. Pediatr
possible mild form of Galloway-Mowat syndrome. Nephrol. 2013;28(2):339–43.
J Child Neurol. 2003;18(2):147–9. 274. Mademan I, Deconinck T, Dinopoulos A, Voit T,
261. Colin E, Huynh Cong E, Mollet G, Guichet A, Schara U, Devriendt K, et al. De novo INF2 mutations
Gribouval O, Arrondel C, et al. Loss-of-function expand the genetic spectrum of hereditary neuropathy
mutations in WDR73 are responsible for Microceph- with glomerulopathy. Neurology. 2013;81(22):1953–8.
aly and steroid-resistant nephrotic syndrome: 275. Fine JD, Johnson LB, Weiner M, Stein A, Cash S,
Galloway-Mowat syndrome. Am J Hum Genet. DeLeoz J, et al. Inherited epidermolysis bullosa and
2014;95(6):637–48. the risk of death from renal disease: experience of the
262. Gee HY, Saisawat P, Ashraf S, Hurd TW, Vega- National Epidermolysis Bullosa Registry. Am J
Warner V, Fang H, et al. ARHGDIA mutations Kidney Dis. 2004;44(4):651–60.
cause nephrotic syndrome via defective RHO GTPase 276. Sams Jr WM, Smith Jr JG. Epidermolysis bullosa
signaling. J Clin Invest. 2013;123(8):3243–53. acquisita, dermal elastosis amyloidosis. Arch
263. Gupta IR, Baldwin C, Auguste D, Ha KC, El Dermatol. 1964;90:137–42.
Andalousi J, Fahiminiya S, et al. ARHGDIA: a 277. Kaneko K, Kakuta M, Ohtomo Y, Shimizu T,
novel gene implicated in nephrotic syndrome. J Med Yamashiro Y, Ogawa H, et al. Renal amyloidosis in
Genet. 2013;50(5):330–8. recessive dystrophic epidermolysis bullosa. Derma-
264. Togawa A, Miyoshi J, Ishizaki H, Tanaka M, tology. 2000;200(3):209–12.
Takakura A, Nishioka H, et al. Progressive impair- 278. Kambham N, Tanji N, Seigle RL, Markowitz GS,
ment of kidneys and reproductive organs in mice Pulkkinen L, Uitto J, et al. Congenital focal segmental
lacking Rho GDIalpha. Oncogene. 1999;18 glomerulosclerosis associated with beta4 integrin
(39):5373–80. mutation and epidermolysis bullosa. Am J Kidney
265. Faul C, Asanuma K, Yanagida-Asanuma E, Kim K, Dis. 2000;36(1):190–6.
Mundel P. Actin up: regulation of podocyte structure 279. Hata D, Miyazaki M, Seto S, Kadota E, Muso E,
and function by components of the actin cytoskeleton. Takasu K, et al. Nephrotic syndrome and aberrant
Trends Cell Biol. 2007;17(9):428–37. expression of laminin isoforms in glomerular basement
266. Zhu L, Jiang R, Aoudjit L, Jones N, Takano T. membranes for an infant with Herlitz junctional
Activation of RhoA in podocytes induces focal seg- epidermolysis bullosa. Pediatrics. 2005;116(4):e601–7.
mental glomerulosclerosis. J Am Soc Nephrol. 280. Has C, Sparta G, Kiritsi D, Weibel L, Moeller A,
2011;22(9):1621–30. Vega-Warner V, et al. Integrin alpha3 mutations with
267. Scott RP, Hawley SP, Ruston J, Du J, Brakebusch C, kidney, lung, and skin disease. N Engl J Med.
Jones N, et al. Podocyte-specific loss of Cdc42 leads 2012;366(16):1508–14.
to congenital nephropathy. J Am Soc Nephrol. 281. Nicolaou N, Margadant C, Kevelam SH,
2012;23(7):1149–54. Lilien MR, Oosterveld MJ, Kreft M, et al. Gain of
268. Wang L, Ellis MJ, Gomez JA, Eisner W, Fennell W, glycosylation in integrin alpha3 causes lung
Howell DN, et al. Mechanisms of the proteinuria disease and nephrotic syndrome. J Clin Invest.
induced by Rho GTPases. Kidney Int. 2012;81(11): 2012;122(12):4375–87.
1075–85. 282. Kreidberg JA, Donovan MJ, Goldstein SL, Rennke H,
269. Shibata S, Nagase M, Yoshida S, Kawarazaki W, Shepherd K, Jones RC, et al. Alpha 3 beta 1 integrin
Kurihara H, Tanaka H, et al. Modification of miner- has a crucial role in kidney and lung organogenesis.
alocorticoid receptor function by Rac1 GTPase: Development. 1996;122(11):3537–47.
implication in proteinuric kidney disease. Nat Med. 283. Karamatic Crew V, Burton N, Kagan A, Green CA,
2008;14(12):1370–6. Levene C, Flinter F, et al. CD151, the first member of
270. Lemieux G, Neemeh JA. Charcot-Marie-Tooth dis- the tetraspanin (TM4) superfamily detected on eryth-
ease and nephritis. Can Med Assoc J. 1967;97(20): rocytes, is essential for the correct assembly of human
1193–8. basement membranes in kidney and skin. Blood.
271. Toyota K, Ogino D, Hayashi M, Taki M, Saito K, 2004;104(8):2217–23.
Abe A, et al. INF2 mutations in Charcot-Marie-Tooth 284. Sterk LM, Geuijen CA, van den Berg JG, Claessen N,
disease complicated with focal segmental glomerulo- Weening JJ, Sonnenberg A. Association of the
sclerosis. J Peripher Nerv Syst. 2013;18(1):97–8. tetraspanin CD151 with the laminin-binding integrins
27 Idiopathic Nephrotic Syndrome in Children: Genetic Aspects 837

alpha3beta1, alpha6beta1, alpha6beta4 and and HLA antigens in Bengali children. Pediatr
alpha7beta1 in cells in culture and in vivo. J Cell Nephrol. 2001;16(4):346–9.
Sci. 2002;115(Pt 6):1161–73. 296. Ruf RG, Fuchshuber A, Karle SM, Lemainque A,
285. Yauch RL, Berditchevski F, Harler MB, Reichner J, Huck K, Wienker T, et al. Identification of the first
Hemler ME. Highly stoichiometric, stable, and spe- gene locus (SSNS1) for steroid-sensitive nephrotic
cific association of integrin alpha3beta1 with CD151 syndrome on chromosome 2p. J Am Soc Nephrol.
provides a major link to phosphatidylinositol 2003;14(7):1897–900.
4-kinase, and may regulate cell migration. Mol Biol 297. Landau D, Oved T, Geiger D, Abizov L, Shalev H,
Cell. 1998;9(10):2751–65. Parvari R. Familial steroid-sensitive nephrotic syn-
286. Yauch RL, Kazarov AR, Desai B, Lee RT, Hemler drome in Southern Israel: clinical and genetic obser-
ME. Direct extracellular contact between integrin vations. Pediatr Nephrol. 2007;22(5):661–9.
alpha(3)beta(1) and TM4SF protein CD151. J Biol 298. Chehade H, Cachat F, Girardin E, Rotman S, Correia
Chem. 2000;275(13):9230–8. AJ, Fellmann F, et al. Two new families with
287. Nishiuchi R, Takagi J, Hayashi M, Ido H, Yagi Y, hereditary minimal change disease. BMC Nephrol.
Sanzen N, et al. Ligand-binding specificities of 2013;14:65.
laminin-binding integrins: a comprehensive survey of 299. Gbadegesin RA, Adeyemo A, Webb NJ, Greenbaum
laminin-integrin interactions using recombinant LA, Abeyagunawardena A, Thalgahagoda S, et al.
alpha3beta1, alpha6beta1, alpha7beta1 and alpha6beta4 HLA-DQA1 and PLCG2 are candidate risk loci for
integrins. Matrix Biol. 2006;25(3):189–97. childhood-onset steroid-sensitive nephrotic syndrome.
288. Baleato RM, Guthrie PL, Gubler MC, Ashman LK, J Am Soc Nephrol. 2015;26(7):1701–10.
Roselli S. Deletion of CD151 results in a strain- 300. Gee HY, Ashraf S, Wan X, Vega-Warner V, Esteve-
dependent glomerular disease due to severe alter- Rudd J, Lovric S, et al. Mutations in EMP2 cause
ations of the glomerular basement membrane. Am J childhood-onset nephrotic syndrome. Am J Hum
Pathol. 2008;173(4):927–37. Genet. 2014;94(6):884–90.
289. Sachs N, Kreft M, van den Bergh Weerman MA, 301. Genovese G, Friedman DJ, Ross MD, Lecordier L,
Beynon AJ, Peters TA, Weening JJ, et al. Kidney Uzureau P, Freedman BI, et al. Association of
failure in mice lacking the tetraspanin CD151. J Cell trypanolytic ApoL1 variants with kidney disease in
Biol. 2006;175(1):33–9. African Americans. Science. 2010;329(5993):841–5.
290. Motoyama O, Sugawara H, Hatano M, Fujisawa T, 302. Giglio S, Provenzano A, Mazzinghi B, Becherucci F,
Iitaka K. Steroid-sensitive nephrotic syndrome in two Giunti L, Sansavini G, et al. Heterogeneous genetic
families. Clin Exp Nephrol. 2009;13(2):170–3. alterations in sporadic nephrotic syndrome associate
291. White RH. The familial nephrotic syndrome I A with resistance to immunosuppression. J Am Soc
European survey. Clin Nephrol. 1973;1(4):215–9. Nephrol. 2014;26(1):230–6.
292. Bensman A, Vasmant D, Mougenot B, Baudon JJ, 303. McCarthy HJ, Bierzynska A, Wherlock M,
Muller JY. [Steroid-responsive nephrotic syndrome Ognjanovic M, Kerecuk L, Hegde S, et al. Simulta-
in infants: 2 familial case reports]. Arch Fr Pediatr. neous sequencing of 24 genes associated with steroid-
1982;39(6):381–3. resistant nephrotic syndrome. Clin J Am Soc Nephrol.
293. Mallmann R. Idiopathic nephrotic syndrome and 2013;8(4):637–48.
hexadactyly in two brothers. Pediatr Nephrol. 304. Korkmaz E, Lipska-Ziętkiewicz BS, Boyer O,
1998;12(5):417–9. Gribouval O, Fourrage C, Tabatabaei M, Schnaidt S,
294. Fuchshuber A, Gribouval O, Ronner V, Kroiss S, Gucer S, Kaymaz F, Arici M, Dinckan A, Mir S,
Karle S, Brandis M, et al. Clinical and genetic evalu- Bayazit AK, Emre S, Balat A, Rees L, Shroff R,
ation of familial steroid-responsive nephrotic syn- Bergmann C, Mourani C, Antignac C, Ozaltin F,
drome in childhood. J Am Soc Nephrol. Schaefer F, PodoNet Consortium. ADCK4-associated
2001;12(2):374–8. glomerulopathy causes adolescence-onset FSGS. J
295. Kari JA, Sinnott P, Khan H, Trompeter RS, Snodgrass Am Soc Nephrol. 2015 May 12. pii:
GJ. Familial steroid-responsive nephrotic syndrome ASN.2014121240. [Epub ahead of print].
Idiopathic Nephrotic Syndrome in
Children: Clinical Aspects 28
Patrick Niaudet and Olivia Boyer

Contents Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 853


Symptomatic Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 853
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 840 Specific Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 854
Associated Disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 841 Initial Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 855
Clinical and Biological Features . . . . . . . . . . . . . . . . . . . 841 Treatment of Relapses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 856
Urinalysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 842 Steroid Side Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 857
Blood Chemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 842 Alternative Treatments (Fig. 9) . . . . . . . . . . . . . . . . . . . . . . 857

Complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 843 Long-Term Outcome of Children


Hypovolemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 843 with Steroid-Sensitive Nephrosis . . . . . . . . . . . . . . . . . . 863
Acute Renal Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 843 Treatment of Steroid-Resistant INS . . . . . . . . . . . . . . . . . 863
Infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 844 Recurrence of Proteinuria After Renal
Thrombosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 844 Transplantation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 867
Growth . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 845 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 868
Chronic Renal Failure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 845
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 869
Renal Biopsy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 846
Minimal Change Nephropathy . . . . . . . . . . . . . . . . . . . . . . 846
Diffuse Mesangial Proliferation . . . . . . . . . . . . . . . . . . . . . 846
Focal and Segmental Glomerular Sclerosis . . . . . . . . . 848
Immunofluorescence Patterns . . . . . . . . . . . . . . . . . . . . . . . 850
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 851
The Anionic Glomerular Basement Membrane . . . . . 851
Immunological Anomalies and Circulating
Permeability Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 851

P. Niaudet (*) • O. Boyer


Service de Néphrologie Pédiatrique, Hôpital
Necker-Enfants Malades, Université Paris-Descartes,
Paris, France
e-mail: pniaudet@gmail.com; olivia.boyer@nck.aphp.fr

# Springer-Verlag Berlin Heidelberg 2016 839


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_24
840 P. Niaudet and O. Boyer

The most common cause of nephrotic syndrome corticosteroids, whereas some steroid-resistant
in children is idiopathic nephrotic syndrome patients have no sclerotic changes on biopsy spec-
(INS), also called nephrosis [1]. INS is defined imens containing adequate tissue [10]. Therefore,
by the combination of a nephrotic syndrome (mas- some authors believe that, although histological
sive proteinuria, hypoalbuminemia, hyperlipid- variants of the INS carry prognostic significance,
emia, and edema) and nonspecific histological they cannot at present be considered as separate
abnormalities of the glomeruli including minimal entities.
changes, focal and segmental glomerular sclerosis The term MCD has become synonymous with
(FSGS), and diffuse mesangial proliferation. On steroid-responsive nephrotic syndrome although
electron microscopy, glomeruli show an efface- renal biopsy is usually not performed in patients
ment of epithelial cell (podocyte) foot processes who respond to steroid therapy. Indeed, in many
and no significant deposits of immunoglobulins or centers, renal biopsy is recommended only for
complement by immunofluorescence. In a major- those patients who fail to respond to steroids.
ity of children, only minimal changes are seen on Consequently, renal biopsy findings in recent
light microscopy. These children are referred to as published series are not representative of the true
having “minimal change disease.” incidence of various histopathological categories
Many authors consider minimal change dis- seen in INS. It is therefore more appropriate to
ease (MCD) and FSGS as separate diseases classify the patients according to their response to
because of differences in response to corticoste- steroid therapy. Response to steroid therapy
roids and subsequent clinical course. Indeed, carries a greater prognostic weight than the histo-
these pathologic features carry prognostic signif- logical features seen on initial renal biopsy. Thus,
icance. Patients with FSGS have more frequently two types of INS can be defined: steroid-
hematuria, are often resistant to corticosteroid responsive nephrotic syndrome, in which protein-
treatment, and progress more often to renal fail- uria rapidly resolves after starting steroid therapy,
ure. Recent data have comforted the belief that and steroid-resistant nephrotic syndrome, in
MCD and FSGS are different entities. MCD is a which steroids do not induce remission.
functional podocyte disease, whereas FSGS
appears to be a structural podocytopathy [2]. The
notion of “podocyte dysregulation” [2, 3], the Epidemiology
different expression cyclin-dependent kinase
inhibitors in MCD and in FSGS, the role of The incidence of INS varies with age, race, and
these cell cycle disturbances leading to podocyte geography. INS is observed in all parts of the
proliferation and maturation [4], and the identifi- world. The annual incidence of steroid-responsive
cation of parvovirus B19 in glomeruli of patients nephrotic syndrome in children in the USA and in
with FSGS [5, 6] are in favor of distinct entities. Europe has been estimated to 1–3 per 100,000
The high circulating levels of the soluble uroki- children below age of 16 [11–13], with a cumula-
nase receptor (suPAR) found in FSGS but not in tive prevalence of 16 per 100,000 children. Similar
MCD [7, 8] support the hypothesis that MCD and figures were reported in New Zealand [16]. Geo-
FSGS as podocytopathies originating from differ- graphical and/or ethnic differences are well known.
ent glomerular permeability factors. In the United Kingdom, for example, the incidence
In the early stages, FSGS and MCD are indis- of INS is sixfold greater in Asian than in European
tinguishable [9]. The best illustration of this is the children [14]; this is also true for Indians [15] and
aspect of a transplant biopsy carried out shortly for Japanese and Southwest Asians. INS is less
after relapse of proteinuria following transplanta- frequent in Africa [16–18]. Such differences under-
tion in a patient whose primary renal disease was line the role of genetic as well as environmental
FSGS. Despite heavy proteinuria, the glomeruli factors in the pathogenesis of the disease.
initially show minimal changes. A significant A male preponderance in children, with a male
number of patients with FSGS respond to to female ratio of up to 3.8:1 [11, 19], is frequently
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 841

reported. Both sexes are similarly affected in Table 1 Conditions associated with idiopathic nephrotic
adolescents. syndrome
The familial occurrence of INS is well known Allergy
in patients with steroid-resistant idiopathic Pollen
nephrotic syndrome (see ▶ Chap. 27, “Idiopathic Fungi
Nephrotic Syndrome in Children: Genetic Cow’s milk
Aspects”). House dust
Bee stings
Cat fur
Poison ivy
Associated Disorders
Drugs
Nonsteroidal anti-inflammatory drugs
INS is, by definition, a primary disease. Neverthe-
Ampicillin
less, in a number of cases, an upper respiratory Gold
tract infection, an allergic reaction, or another Lithium
factor may immediately precede the development Mercury
or relapse of the disease. Trimethadione
Many agents or conditions have been reported Malignancies
to be associated with INS such as infectious dis- Hodgkin disease
eases, drugs, allergy, vaccinations, and malignan- Non-Hodgkin lymphoma
cies (Table 1). The question remains whether Colon carcinoma
these factors are real causes, a simple coincidence, Bronchogenic carcinoma
or precipitating agents. Others
Allergy is associated with up to 30 % of cases Viral infection
[20–22]. Among a list of anecdotal cases, the Kimura’s disease
allergens reported include fungi, poison ivy, tim- Diabetes mellitus
othy grass pollen, house dust, medusa stings, bee Myasthenia gravis
Immunization
stings, and cat fur. A food allergen may be respon-
sible for relapses of steroid-sensitive nephrotic
syndrome, such as cow’s milk and egg. Laurent
et al. evaluated the effect of an oligoantigenic diet A number of drugs may induce a nephrotic
given for 10–15 days to 13 patients. This diet syndrome with the histopathological appearance
coincided with improvement of proteinuria in of minimal change disease. The list comprises
nine, including complete remission in five [23]. drugs such as nonsteroidal anti-inflammatory
The association between minimal change dis- agents, salazopyrin and mesalamine, D-penicilla-
ease and malignancies mainly concerns lympho- mine, lithium, rifampicin, heavy metals (gold,
matous disorders: Hodgkin’s disease and mercury), and trimethadione.
non-Hodgkin’s lymphomas [24, 25]. The
nephrotic syndrome may be the presenting feature
of the disease. It usually disappears after success- Clinical and Biological Features
ful treatment of the malignancy. Eosinophilic
lymphoid granuloma in orientals (Kimura’s dis- The disease may occur during the first year of
ease) has also been reported in association with life, but it usually starts between the ages of 2 and
INS [26, 27]. 7 years. The nephrotic syndrome is rarely dis-
Several cases of minimal change disease have covered during a routine urine analysis. The
been reported in association with the onset of onset is often preceded by an upper respiratory
insulin-dependent diabetes mellitus. The disease tract infection. The disease is characterized by a
is usually responsive to corticosteroids and fol- sudden onset, edema being the major presenting
lows a relapsing course. symptom. It becomes clinically detectable when
842 P. Niaudet and O. Boyer

fluid retention exceeds 3–5 % of body weight. proteins. The selectivity of proteinuria may be
Periorbital edema, frequently misdiagnosed as appreciated by polyacrylamide gel electrophore-
allergy, is often the initial symptom. Edema is sis or by the evaluation of the Cameron index,
gravity dependent, localized to the lower extrem- which is the ratio of IgG (MW 150 kDa) to trans-
ities in the upright position and to the dorsal part ferrin (80 kDa) clearances. A favorable index
of the body in reclining position. This edema is would be below 0.10 or better below 0.05; a
white, soft, and pitting, keeping the marks of poor index is above 0.15 or 0.20. Such poor
clothes or finger pressure. Anasarca may develop index is more often associated with steroid-
with ascites and pleural and pericardial effusions. resistant nephrotic syndrome and FSGS.
Although there may also be abdominal disten- However, there is a considerable overlap in
sion, dyspnea is rare. Periorbital edema may limit results, and the test has limited value. The amount
eye opening, and edema of the scrotum and of protein excreted in the urine does not reflect
penis, or labia, may be seen. A rapid formation the quantity of protein crossing the glomerular
of ascites is often associated with abdominal pain basement membrane since a significant amount
and malaise: these symptoms may also be related is reabsorbed in the proximal tubule. Some chil-
to concomitant hypovolemia. Abdominal pain is dren with severe steroid-resistant nephrotic syn-
occasionally due to a complication such as peri- drome and tubulointerstitial lesions have both
tonitis, thrombosis, or, rarely, pancreatitis. Car- glomerular and tubular proteinuria with an
diovascular shock is not unusual, secondary to increased excretion of β-2 microglobulin,
the sudden fall of plasma albumin, with abdom- retinol-binding protein, and lysozyme due to an
inal pain and symptoms of peripheral circulatory impaired protein reabsorption in the proximal
failure with cold extremities and hypotension. tubule.
Emergency symptomatic treatment is needed. The urine sediment of patients with INS often
Blood pressure is usually normal but sometimes contains fat bodies. Hyaline casts are also usually
elevated. found in patients with massive proteinuria, but
Macroscopic hematuria is observed in a few granular casts are not present unless there is asso-
cases. The disease may also be revealed by a ciated acute renal failure and acute tubular necro-
complication. Peritonitis due to Streptococcus sis. Macroscopic hematuria is rare, occurring in
pneumoniae is a classical mode of onset 3 % of patients. Microscopic hematuria is present
[28]. Deep vein or arterial thromboses and pulmo- in 20 % of cases and has no histopathologic or
nary embolism may also occur during the first prognostic significance.
attack or during a relapse. Urinary sodium excretion is low (<5 mmol/
24 h), associated with sodium retention and
edema. Kaliuresis is usually higher than natriure-
Urinalysis sis, but it may be reduced in oliguric patients.

Proteinuria is detected by dipstick testing 3 or 4+.


Quantitative evaluation gives figures ranging Blood Chemistry
from less than 1 g to more than 10 g/day. The
nephrotic range proteinuria is defined as >50 Serum proteins are markedly reduced below 50 g/l
mg/kg/day or 40 mg/h/m2. In young children it in 80 % of patients and below 40 g/l in 40 %.
may be difficult to perform 24-h urine collection, Albumin concentration usually falls below 20 g/l
and urinary protein/creatinine ratio or U albumin/ and may be less than 10 g/l. Electrophoresis shows
U creatinine ratio in untimed urine specimens is not only low albumin levels but also increased α-2
useful. For these two indices, the nephrotic range globulins and, to a lesser extent, β-globulins, while
is 200–400 mg/mmol. γ-globulins are decreased. IgG is markedly
In most cases, proteinuria is highly selective, decreased, IgA slightly reduced, IgM is increased,
consisting of albumin and lower molecular weight while IgE is normal or increased. Among other
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 843

proteins, fibrinogen and β-lipoproteins are loss of erythropoietin may also contribute to ane-
increased, and antithrombin III is decreased. mia [36]. Thrombocytosis is common and may
Hyperlipidemia is a consequence of (I) an reach 5.108 or 109/l.
increased hepatic synthesis of cholesterol, triglyc-
erides, and lipoproteins; (II) a decreased catabo-
lism of lipoproteins due to a decreased activity of
lipoprotein lipase which normally transforms Complications
VLDL to LDL via IDL; and (III) a decreased
LDL receptor activity and an increased urinary Hypovolemia
loss of HDL [29, 30]. Total cholesterol and LDL
cholesterol are elevated, while HDL cholesterol A few children are severely hypovolemic, and this
remains unchanged or low, particularly HDL2, complication is observed typically at onset or
leading to an increased LDL/HDL cholesterol early during a relapse (Table 2). Sepsis, diarrhea,
ratio [31]. Patients with severe hypoalbuminemia or diuretics may precipitate hypovolemia. These
have increased triglycerides and VLDL. children often complain from abdominal pain and
Apoproteins, apo B, apo CII, and apo CIII, are have low blood pressure and cold extremities.
also elevated. The levels of lipoprotein Lp(a) are Hemoconcentration with a raised hematocrit
elevated in nephrotic patients, and this further accompanies hypovolemia.
contributes to an increased risk of cardiovascular
and thrombotic complications.
Serum electrolytes are usually within the nor- Acute Renal Failure
mal range. A low sodium level may be related to
dilution from inappropriate renal retention of Renal function is usually within normal limits at
water due to hypovolemia and inappropriate presentation. A reduction of the GFR, secondary
antidiuretic hormone secretion. The mild reduc- to hypovolemia, infection, or thrombosis, is fre-
tion of plasma sodium concentration is often an quent [37, 38]. A reduced GFR may be found in
artifact related to hyperlipidemia. Serum potas- patients with normal effective plasma flow.
sium may be high in oliguric patients. Serum Bohman et al. showed a close relationship
calcium is consistently low as a result of between the degree of foot process fusion and
hypoproteinemia. Ionized calcium is usually nor- both GFR and filtration fraction, suggesting that
mal but may be decreased due to urinary loss of fusion of foot processes could lead to a reduction
25-hydroxyvitamin D3 [32] and normal but inap- of glomerular filtering area and/or of permeability
propriate levels of calcitriol [33]. Blood urea to water and small solutes [39]. This reduction is
nitrogen and creatinine concentrations are usually transitory, with a rapid return to normal after
within the normal range or slightly increased in remission. Van de Walle et al. found that changes
relation to a modest reduction in the glomerular in glomerular permeability may also have a major
filtration rate (GFR). role in acute renal failure [37].
A few patients with FSGS and a poor subse-
quent outcome present with a Fanconi syndrome:
glycosuria, aminoaciduria, urinary bicarbonate
Table 2 Nephrotic hypovolemia
loss, and hypokalemia [34]. A defect in urinary
acidification has also been reported [35]. Clinical features Precipitating factors
Abdominal pain Severe relapse
Hematology Hypotension Infection
Hemoglobin levels and hematocrit are increased Sluggish circulation Diuretics
Relative polycythemia Paracentesis
in patients with plasma volume contraction. Ane-
Acute tubular necrosis Diarrhea
mia with microcytosis may be observed, probably
Thrombosis
related to urinary loss of siderophilin. The urinary
844 P. Niaudet and O. Boyer

Table 3 Infections in nephrotic syndrome Table 4 Thrombosis in nephrotic syndrome


Clinical syndrome Risk factors Clinical syndrome Risk factor
Pneumococcal Low IgG Pulmonary emboli Hyperviscosity
peritonitis Pulmonary artery Low antithrombin III
Haemophilus infection Low factor B thrombosis
Gram negative sepsis Edematous tissue Cerebral venous High fibrinogen
Staphylococcus Impaired lymphocyte thrombosis
cellulitis function Renal vein Platelet hyperaggregability
Corticosteroids thrombosis
Immunosuppressive drugs Peripheral venous Underlying genetic
thrombosis thrombophilic tendency
Artery thrombosis Hypovolemia
Central venous catheter
Immobilization
Marked oliguria may occur in children Infection
[40]. Oliguric renal failure may be the presenting
symptom. Renal failure may be secondary to bilat-
eral renal vein thrombosis, which is recognized by
sonography or to interstitial nephritis, which has Viral infections may be observed in patients
been reported, especially with furosemide. Skin receiving corticosteroids or immunosuppressive
rash and eosinophilia are suggestive of this agents. Chickenpox is often observed in these
diagnosis. young children and may be life-threatening if
Acute renal failure is usually reversible, espe- acyclovir is not started rapidly. Interestingly, mea-
cially with intravenous infusion of albumin sles infection may induce long-lasting remissions.
[41]. In some cases, where glomerular structure
is normal on renal histology, renal failure may last
for as long as a year and sometimes be Thrombosis
irreversible [42].
Nephrotic patients are at risk of developing
thromboembolic complications, which may be
Infections life threatening [44]. The majority of thromboem-
bolic complications occur within the first 3 months
Bacterial infections are frequent in nephrotic chil- after the onset of nephrotic syndrome. Children
dren (Table 3). Sepsis may occur at the onset of aged >12 years are at higher risk of this compli-
the disease. The most common infection is peri- cation than younger children. Several factors con-
tonitis, often with Streptococcus pneumoniae tribute the increased risk of thrombosis, including
[43]. Other organisms may be responsible: hypercoagulability state, hypovolemia, immobili-
E. coli, streptococcus B, Haemophilus influenzae, zation, infection, the presence of central venous
and other gram-negative organisms. Apart from catheter, and a possible underlying genetic
peritonitis, children may develop meningitis, thrombophilic tendency [45] (Table 4). A number
pneumonitis, and cellulitis. Several factors may of hemostatic abnormalities have been described
explain the propensity of nephrotic children to in nephrotic patients: increase in platelet count
develop bacterial infections: low IgG levels due and platelet aggregability, increase in
to an impaired synthesis, urinary loss of factor B procoagulant proteins with high molecular weight
and impaired T lymphocyte function. Factor B is a such as fibrinogen and factors V and VIII,
cofactor of C3b of the alternative pathway of decreased level of the anticoagulant protein anti-
complement, which has an important role in thrombin III is due to its urinary loss, and
opsonization of bacteria such as Streptococcus decreased fibrinolytic activity due to the increased
pneumoniae. concentrations of both α2-macroglobulin and
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 845

lipoprotein(a) [46]. The incidence of thromboem- The only “benefit” of the decrease of GFR is the
bolic complications in nephrotic children is close improvement of the nephrotic syndrome due to a
to 3 % [45, 47]. Venous thrombosis account for decrease of proteinuria.
most of the cases of thromboembolic complica- We have retrospectively analyzed in the Enfants
tions, while arterial thrombosis is much less fre- Malades series the outcome of 181 children with
quent. In a recent study including 370 children, steroid-resistant INS who have been followed for at
venous thromboembolism accounted for 97 % of least 5 years. Eighty-five percent were primary non-
the cases [48]. Two reports suggest that the per- responders and 15 % late nonresponders. Initial
centage of subclinical thromboembolic complica- renal biopsy had shown minimal changes in
tions may be far more common [49, 50]. In the 62 cases and FSGS in 119 cases. Renal survival
first study, lung scintigraphic examination in rates were 65 % at 5 years, 50 % at 10 years, and
26 children showed evidence of pulmonary embo- 34 % at 15 years. Interestingly, the rate of progres-
lism in 7 (28 %) and residual changes in another sion to end-stage renal failure was similar in patients
10 (38.5 %). In the second study, 28 % of children with minimal changes or FSGS on initial biopsy.
with nephrotic syndrome without respiratory The data reported in other series are difficult to
symptoms had subclinical pulmonary embolism. compare as most of them deal with patients with
Pulmonary embolism should be suspected in FSGS. The Southwest Pediatric Nephrology Study
cases of pulmonary or cardiovascular symptoms group reported 75 children with FSGS followed for
and may be confirmed by computerized tomogra- periods of 7–217 months [54]. Twenty-one percent
phy pulmonary angiography (CTPA) [51]. Renal had progressed to end-stage renal failure, 23 % had
vein thrombosis should be suspected in cases with decreased glomerular filtration rate, 37 % had a
sudden macroscopic hematuria or acute renal fail- persistent nephrotic syndrome, and 11 % were in
ure. Doppler ultrasonography shows an increase remission. Paik et al. retrospectively analyzed
in kidney size and the absence of blood flow in the 92 children with steroid-resistant FSGS and found
renal vein. Thrombosis may affect the arteries renal survival rates at 5, 10, and 15 years of 84 %,
such as pulmonary arteries or other deep veins. 64 %, and 53 %, respectively [55]. Poor prognostic
factors of chronic renal failure were asymptomatic
proteinuria at presentation, initial renal failure, and
Growth higher proportion of glomeruli with segmental
sclerosis. Gipson et al. reported that the response
Growth may be severely affected in children with to treatment was a good predictor of long-term
persistent nephrotic syndrome. Depletion of hor- renal survival in children with FSGS [56]. Indeed,
mones due to urinary losses is a possible cause of more than 50 % of patients who had not achieve
stunting. Hypothyroidism related to urinary loss complete or partial remission of proteinuria had
of iodinated proteins has been observed and may progressed to ESRF after 3 years, whereas com-
be corrected [52]. A low plasma IgF1 and IgF2 plete remission was associated with a 90 %
level associated with a urinary loss of the carrier decreased risk of ESRF.
proteins has also been reported [53]. Progression to end-stage renal failure has been
reported to be more rapid in patients of African or
Hispanic descent when compared with Cauca-
Chronic Renal Failure sians. Ingulli and Tejani found that among 57 Afri-
can American and Hispanic children, 50 % of
The main difference between steroid responders them had reached end-stage renal failure within
and steroid nonresponders is the tendency of the 3 years, and 95 % had reached this stage after
latter to develop end-stage renal failure, which is 6 years [57]. In addition, among the children
seen in less than 3 % of responders. This compli- with INS, the proportion of those with steroid-
cation occurs in 50 % or more of the steroid- resistant FSGS tends to be more important in
resistant patients after a follow-up of 10 years. African American and Hispanic children.
846 P. Niaudet and O. Boyer

Renal Biopsy Podocyte foot process fusion is generalized and


constant (Figs. 2 and 3); its extent is closely
Renal biopsy is usually not indicated in a child aged related to the degree of proteinuria [59]. Other
1–10 years with typical symptoms and a complete epithelial changes consist of microvillus forma-
remission with corticosteroids. However, it is indi- tion and the presence of numerous protein
cated in children less than 1 year of age, when the reabsorption droplets. The glomerular basement
child has macroscopic hematuria or hypertension or membranes are normal with no parietal deposits.
low C3 levels or persistent renal failure. The main The endothelial cells are often swollen
indication is the failure to respond to corticosteroid [60]. Mesangial alterations include mesangial cell
therapy. A renal biopsy may be indicated in patients hyperactivity, increased mesangial matrix, and
who relapse before considering alternative therapy, occasionally finely granular, osmiophilic deposits
namely, anticalcineurin agents. located along the internal side of the basement
Light microscopy shows three morphological membrane. These ultrastructural alterations are
patterns: minimal changes, diffuse mesangial pro- nonspecific and are probably related to massive
liferation, and FSGS. Minimal changes are found proteinuria. Vascular changes are absent in children.
in the majority of children, whereas FSGS is
found in 5–7 % of them and diffuse mesangial
proliferation in 3–5 % [11, 54]. Diffuse Mesangial Proliferation

Some patients with steroid-resistant INS show a


Minimal Change Nephropathy marked increase in mesangial matrix associated
with hypercellularity (Fig. 4) [58, 61, 62]. How-
On light microscopy, glomeruli may be normal ever, peripheral capillary walls are normal, and
with normal capillary walls and normal cellularity immunofluorescence microscopy is negative.
(Fig. 1). Swelling and vacuolation of epithelial Electron microscopy shows foot process fusion
cells and a slight increase in mesangial matrix similar to the changes observed in minimal
are often observed. A mild mesangial hypercel- change disease. The presence of mesangial
lularity may be noted as well as scattered foci of hypercellularity has been found to have prognos-
tubular lesions and interstitial fibrosis [58]. tic significance with a higher rate of progression to
Ultrastructural changes are always present, renal failure [61], but these findings were not
involving podocytes and mesangial stalks. confirmed by other authors.

Fig. 1 Minimal change


disease. Glomeruli appear
normal by light microscopy
with no tubulointerstitial
lesions
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 847

Fig. 2 Electron microscopy. On the left, normal aspect with the podocyte foot processes attached to the glomerular
basement membrane. On the right, minimal change disease with effacement of foot processes

Fig. 3 Scanning electron microscopy showing the normal aspect of podocytes with their foot processes on the left and
their effacement in minimal change disease on the right

Fig. 4 Diffuse mesangial


proliferation with an
increased number of
mesangial cells and
mesangial matrix
848 P. Niaudet and O. Boyer

Fig. 5 FSGS. In early


stage, there is a segmental
collapse of glomerular
capillaries surrounded by
hypertrophic podocytes
containing intracytoplasmic
vacuoles

Fig. 6 FSGS. Segmental


lesion of the tuft
characterized by the
deposition of hyaline
material at the inner side of
the glomerular basement
membrane with a ring of
podocytes separated from
the glomerular basement
membrane by a clear “halo”

Focal and Segmental Glomerular material is often present within the sclerotic
Sclerosis lesions. A clear “halo” zone is observed at the
periphery of the sclerotic segments (Fig. 6). The
The glomerular lesions affect a variable propor- segmental lesion has a different aspect depending
tion of glomeruli [58, 63]. The focal changes are on whether it affects a group of capillary loops free
limited to a part of the tuft, the other capillary in Bowman’s space or is adherent to Bowman’s
loops showing no modification. The lesions capsule. The “free” sclerotic segments are always
always predominate in the deeper cortex at the surrounded by a “crown” of flat or hypertrophied
corticomedullary junction [64]. The segmental podocytes. The podocytes form a continuous layer
lesion affects a few capillary loops, which stick overlying the damaged areas of the tuft and in close
together either at the hilum or at the periphery of apposition to the clear “halo.” When the sclerotic
the tuft or at both (Fig. 5) [65, 66]. The clinical lesion is adherent to Bowman’s capsule, there is a
course has been found to be more benign when the direct synechia between the collapsed capillary
location of these sclerotic lesions is peripheral (the loops and Bowman’s basement membrane. The
tip lesion), although such findings have not been rest of the tuft and the nonsclerotic glomeruli
confirmed by other authors [64, 67–69]. Hyaline show either “minimal changes” or “diffuse
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 849

mesangial proliferation” both with foot process markers (PCNA and Ki-67) are expressed, indicat-
fusion. Glomerular hypertrophy is common in ing a mitotic activity. This “podocyte
FSGS, and when such hypertrophy is found in dysregulation” is accompanied by podocyte detach-
minimal change disease, it is somewhat predictive ment from the glomerular basement membrane.
of further development to FSGS [70, 71]. A pathologic classification of FSGS, desig-
Tubular atrophy and interstitial fibrosis are often nated the Columbia classification, has been pro-
present and apparently proportional to the glomer- posed with five histological variants [77]:
ular damage [63, 72]. Focal glomerular lesions
should therefore be suspected when focal tubular 1. FSGS not otherwise specified (NOS) or classic
and interstitial changes are found associated with FSGS, the most frequent variant.
minimal glomerular changes. Erkan et al. found 2. The perihilar variant with lesions located at
apoptosis in proximal and distal tubular cells of the vascular pole of the glomerular tuft.
children with idiopathic FSGS [73]. There was a 3. The cellular variant with podocyte hyperplasia
correlation between the degree of proteinuria and and endocapillary hypercellularity, foam cells,
the number of apoptotic cells. An elevated tubule and leukocyte infiltration.
cell apoptosis rate at the time of initial biopsy was 4. The tip variant, where the lesion involves the
found to be an independent predictor of progres- tubular pole, is the most benign form of FSGS
sion to end-stage renal disease. with presenting features and outcome more
On electron microscopy, the lesion is character- close to MCD [78].
ized by the presence of paramesangial and 5. Collapsing variant or collapsing
subendothelial, finely granular, osmiophilic glomerulopathy characterized by segmental
deposits [72, 74, 75] with either disappearance or or global wrinkling and collapse of the glomer-
swelling of endothelial cells and an increase in ular capillary walls with prominent hypertro-
mesangial matrix material. Fatty vacuoles may be phy and hyperplasia of the overlying
seen, either in the middle of the abnormal deposit or podocytes (Fig. 7) [77].
in the cytoplasm of endothelial and mesangial cells.
The peripheral synechia, located between All variants share podocyte alterations. This
podocytes and basement membrane, is formed by classification has clinical implications in terms of
the apposition of acellular material in which thin response to therapy and risk of progression to renal
and irregular layers of newly formed basement failure. For example, glomerular tip lesions have
membranes are visible. Modifications of the been associated with better outcomes and collaps-
podocytes consist of focal cytoplasmic degenera- ing variant with worse outcomes [67, 69]. The
tion, breakdown of cell membranes, and detach- glomerular tip lesion was found to be a predictor
ment of epithelial cells from basement of a favorable response to therapy in a pediatric
membranes, with filling of the resulting space by series [79]. Patients with collapsing
cell debris and new membranes [76]. glomerulopathy have a severe nephrotic syndrome
FSGS is characterized by important changes in and rapidly progress to renal failure. The rate of
the podocytes, with major cell cycle derangement response to steroids is poor. The incidence of
[2, 3]. The normal mature podocyte does not divide collapsing glomerulopathy seems to have
and does not express proliferative markers such as increased in the recent years. The main cause of
PCNA and Ki-67. The podocyte expresses several secondary collapsing glomerulopathy is
cell surface proteins such as WT-1, C3b receptor, HIV-associated nephropathy [80]. Many authors
glomerular epithelial protein-1 (GLEPP-1), consider that “collapsing glomerulopathy” is a
podocalyxin, synaptopodin, and vimentin. The distinct form of FSGS which may be “idiopathic,”
first stages of FSGS are characterized by the loss also observed in patients with recurrent nephrotic
of the cell surface proteins (dedifferentiation) and syndrome after renal transplantation or associated
the expression of macrophage markers and with HIV, parvovirus B19 infection, or CMV infec-
cytokeratin (transdifferentiation). Proliferations tion [81–83]. Idiopathic collapsing glomerulopathy
850 P. Niaudet and O. Boyer

Fig. 7 Collapsing
glomerulopathy

Fig. 8 IgM deposits

predominates in blacks and has a poor infection, Alport’s syndrome, hypertension, pyelone-
prognosis [82]. phritis, and obesity. It has also been reported in renal
FSGS is an irreversible scarring process in the hypoplasia with oligomeganephronia, after partial
glomeruli, as shown by the analysis of repeat biop- nephrectomy and in other conditions with a reduction
sies [74, 75, 84]. Studies in experimental animals in nephron number, including reflux nephropathy or
[85] as well as in nephrotic patients have shown obstructive uropathy.
that proteinuria precedes the development of focal
sclerotic lesions. The same sequence was reported
in patients with recurrence of the disease after Immunofluorescence Patterns
transplantation. Within weeks following recurrence
of proteinuria, podocytes observed by electron IgM-associated nephropathy. Immunofluores-
microscopy appear swollen and vacuolated. The cence microscopy is usually negative [86]. How-
podocytes exhibit strong mitotic activity, with ever, mesangial deposits of IgM, IgG, C3, and
multinucleation and expression of the PCNA and more rarely IgA have been reported. The most
Ki-67 proliferation markers. commonly found is IgM (Fig. 8) and Cohen
FSGS is not a specific histopathological lesion: et al. found that patients with IgM deposits in the
similar alterations may be seen in persistent idio- mesangium had a poor response to corticosteroids
pathic proteinuria and heroin-associated nephropathy [87]. Other studies did not confirm these findings.
and, independently, in association with HIV Habib et al. reported on immunofluorescence studies
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 851

in a series of 222 children with INS [88]. Although for the charge selectivity of filtration. The anionic
IgM was the immunoglobulin most frequently pre- (negative) charges of the glomerular basement
sent in the glomeruli (54 of 222), there was no membrane repulse the negatively charged albu-
correlation between IgM deposits, the initial response min molecules, whose isoelectric point is 4.6.
to steroid therapy or the final outcome. IgM deposits The mechanism of proteinuria in the absence
have also been described in association with diffuse of histological alterations on light microscopy has
mesangial proliferation and with FSGS. IgM is a suggested an electrochemical disorder of the
large molecule that can be nonspecifically trapped GBM. Indeed it was shown that the glomerular
in an injured glomerulus, and mesangial IgM Kf is diminished despite increased permeability to
deposits may represent an epiphenomenon. serum albumin due to a loss of glomerular nega-
IgA deposits and minimal change disease. tive charges [95–98].
A number of patients with INS display mesangial
deposits of IgA. They are classified by some as
IgA nephropathy. Others consider that mesangial Immunological Anomalies
IgA in patients with nephrotic syndrome and min- and Circulating Permeability Factors
imal changes, that is, without cellular proliferation
is coincidental [88–90]. This applies to INS asso- In 1974, Shalhoub postulated that minimal change
ciated with mesangial IgA deposits and explains a disease might be secondary to a disorder of the
rapid response to steroids, which would not be the immune system [99]. He hypothesized that the
case in true IgA nephropathy. production of a lymphokine may increase the
C1q glomerulopathy. C1q glomerulopathy permeability of the glomerular filtration barrier
refers to a disorder in which mesangial prolifera- to proteins. The evidences supporting this hypoth-
tion is associated with mesangial deposits on elec- esis were the response of the disease to corticoste-
tron microscopy and prominent C1q deposits on roids and to alkylating agents; the remission
immunofluorescence microscopy [91–93] and no occurring in association with measles, which
clinical and laboratory evidence of systemic lupus depresses cell-mediated immunity; the suscepti-
erythematosus. bility of patients to pneumococcal infections; and
C1q nephropathy has been thought to be a the occurrence of minimal change nephrotic syn-
subgroup of primary FSGS. However, many drome in patients with Hodgkin’s disease. Since
reports describe different symptoms, histopathol- then, several facts have sustained this hypothesis
ogies, therapeutic responses, and prognoses, [99–101]: in SSNS/MCD, the triggering of
suggesting that C1q glomerulopathy may be a relapses by allergy or infections; the unique obser-
combination of several disease groups rather vation of the remission of proteinuria when a
than a single disease entity [94]. C1q kidney from a donor with INS in relapse was
glomerulopathy may be associated with MCD, transplanted to a recipient without INS [102]; in
FSGS, or proliferative glomerulonephritis. SRNS/FSGS, the rapid recurrence of proteinuria
after renal transplantation in 30 % of patients
[103] that may respond to plasma exchanges or
Pathophysiology immunoadsorption [100, 104]; and the induction
of proteinuria in rat following injection of serum
The Anionic Glomerular Basement from patients with posttransplant recurrence of
Membrane nephrotic syndrome [105]. The Buffalo/Mna
strain of rats spontaneously develops proteinuria
The permeability of the glomerular basement with FSGS at 2 months of age. The nephrotic
membrane (GBM) to proteins is determined not syndrome recurs when these rats receive a kidney
only by the size but also by the charge of the from a healthy LEW.1W rat [106]. Conversely,
protein. It is believed that the anionic charge of proteinuria and renal lesions regress when kid-
the glomerular basement membrane is responsible neys from a Buffalo/Mna rat are transplanted
852 P. Niaudet and O. Boyer

into normal LEW.1W rats. A transient neonatal detected significantly high suPAR levels in FSGS
nephrotic syndrome was observed in several chil- patients, especially those with posttransplant
dren born to an affected mother with SRNS recurrence, but not in patients with MCD or mem-
[107, 108]. branous nephropathy [8]. The role of suPAR-
It has long been postulated that the circulating induced αvβ3 integrin activation on proteinuria
permeability factor was a T-cell secreted cytokine was further studied in three animal models:
[109]. Cytokine profile studies during relapses recombinant suPAR injection to suPAR knockout
suggest the implication of Th2-mediated cyto- mice (Plaur/), LPS injection in wild-type mice
kines [110, 111] more specifically IL-13 transplanted with Plaur/ mouse kidney, and a
[112, 113] or IL-4 [114]. The work of Sahali transgenic mouse model overexpressing suPAR
et al. demonstrating a dysregulation of the through repeated cutaneous plasmid
NFκB/lκB pathway [115] and upregulation of electroporations. The authors also showed an acti-
the Th2-specific factor c-maf during relapses vation of the suPAR-αvβ3 integrin pathway in
[116] is in accordance with these results. Aside cultured podocytes incubated with patient sera.
from cytokines, other factors have been suspected However, these data have not been confirmed by
to be involved in the pathogenesis of INS includ- other groups [122]. Cathelin et al. showed that the
ing cardiotrophin-like cytokine-1, hemopexin, administration of recombinant suPAR to C57BL/
and suPAR. Savin et al., using isolated rat glomer- 6J wild-type mice does not result in albuminuria
uli incubated in vitro with the sera of patients with nor podocyte architecture alterations although
recurrence of proteinuria after renal transplanta- leading to suPAR glomerular deposition
tion, described a glomerular permeability factor [123]. In addition, suPAR levels are elevated in
[100]. Using this assay, the same group identified patients with reduced glomerular filtration rate
the cardiotrophin-like cytokine 1(CLC-1), a pro- and in other non-proteinuric diseases such as can-
tein of the IL-6 family [117]. Its concentration is cer and HIV infection [124, 125] with MCD or
100 times higher in the sera of patients with recur- membranous nephropathy [8]. Conversely, some
rent proteinuria after transplantation compared to patients with normal suPAR plasma levels
normal subjects. The injection of CLC-1 to rats develop posttransplant recurrent FSGS [8]. Last,
induces proteinuria. CLC-1 decreases nephrin suPAR levels are not increased in children with
expression in cultured podocytes. Hemopexin, a MCD suggesting distinct pathophysiological
heme-scavenging protein secreted by the liver dur- mechanisms underlying MCD and FSGS.
ing inflammation, induces reversible proteinuria Although high pre-transplant suPAR levels seem
and foot process effacement when infused into to be associated with an increased recurrence risk
rats [118, 119]. Hemopexin has a serine protease after transplantation, the potential role of suPAR as
activity during relapses in children a valuable marker in INS is debated [122, 126, 127].
[120]. Hemopexin might exist in an altered isoform Three studies including a total of 1,151 patients,
with increased activity in patients during relapses 212 of whom with primary FSGS, concluded
as compared with patients in remission. that levels of suPAR do not discriminate
Hemopexin causes nephrin-dependent podocyte between primary FSGS and other causes of glomer-
cytoskeleton rearrangements and disruption of the ular diseases [128–130]. Altogether these data
glomerular filtration barrier permselectivity, both argue against suPAR being per se the FSGS
of which are reversed by incubating cultured factor [131].
podocytes with normal human sera [121]. How- The type of immune cell producing the circulat-
ever, the mechanisms of hemopexin activation dur- ing factor also remains elusive. T-cells have long
ing relapses and their consequences on podocyte been incriminated. More recent data suggest the
structure and function are not yet fully understood. role of B-cells in INS [132, 133]. Indeed, the effi-
Recent findings have suggested the role of ciency of rituximab in preventing relapses in
soluble urokinase receptor (suPAR) in the patho- patients with steroid dependency strongly advo-
genesis of FSGS [8]. The group of Jochen Reiser cates the role of B-cells in INS [134–136]. B-cells
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 853

could be involved by secreting the permeability the roles of the putative circulating factors and
factor (i.e., that could be an immunoglobulin) or their effects on downstream podocyte signaling
through a T-cell to B-cell communication pathways.
dysregulation [137]. However, Fornoni
et al. showed that rituximab also has a direct effect
on podocyte cytoskeleton stabilization [138]. Treatment
It was also reported that CD34(+) hematopoietic
progenitors from patients injected to NOD/SCID Symptomatic Treatment
immunodeficient mice could induce proteinuria
and foot process effacement, whereas peripheral Diet
CD34() mononucleated cells could not [139]. Dietary therapy should include a protein intake of
Accumulating data sustain the role of one or around 130–140 % of the recommended daily
several permeability factors. However, their allowance according to statural age. Salt restriction
effects on podocyte structure and function and is advised for the prevention and the treatment of
the mechanisms driving foot process effacement edema. A very low salt diet is only necessary in
and proteinuria remain unclear. Aside from cases of massive edema. Fluid restriction is
suPAR-αvβ3 integrin pathway, B7.1 (CD80) and recommended for moderate to severe hyponatremia
c-mip signaling have been incriminated. B7.1 (plasma sodium concentration less than 125 meq/l).
(CD80), a T-cell co-stimulator present at the sur- A reduction of saturated fat is recommended. Car-
face of antigen-presenting cells, is increased in bohydrates should be given preferentially as starch
urine of children with MCD [140]. LPS injection or dextrin-maltose, avoiding sucrose, which
in mice induces B7.1 (CD80) expression on increases lipid abnormalities.
podocytes [141] and thereby podocyte signaling
cascades that could lead to foot process efface- Hypovolemia
ment and proteinuria [142]. Differential cloning Hypovolemia occurs as a consequence of rapid
of patient lymphocytes in remission or relapse loss of protein and is sometimes aggravated by the
suggested the role of c-mip, expressed in use of diuretics. This complication needs emer-
podocytes during relapses, T-lymphocytes, and gency treatment by rapid infusion of plasma
Hodgkin lymphoma cells from patients develop- (20 ml/kg) or albumin 20 % (1 g/kg) administrated
ing nephrotic syndrome [116, 143]. C-mip is also with control of heart rate, respiratory rate, and
activated upon LPS or Adriamycin administration blood pressure.
to mice in experimental models of nephrotic syn-
drome [144]. Transgenic mice overexpressing Diuretics
c-mip in podocytes develop massive proteinuria Diuretics should only be used in cases of severe
and foot process effacement [144]. C-mip silenc- edema, after hypovolemia has been corrected.
ing by RNA interference reduces LPS-induced Furosemide is administered at a dose of 1–2
proteinuria [144]. These results are a first step mg/kg. If not effective, spironolactone (5–10
toward targeted therapies in INS. mg/kg) or amiloride (0.2–0.5 mg/kg) may be pre-
In summary, it is currently unclear whether scribed if the plasma creatinine concentration is
INS is caused by one or multiple factors and normal [146]. Patients with severe edema may be
whether this/these putative factors are identical treated with furosemide or, if necessary, furose-
in MCD and FSGS. Indeed, urinary CD80 is mide plus albumin to increase the rate of diuretic
elevated in MCD patients in relapse compared delivery to the kidney. This approach is immedi-
with FSGS patients. In contrast, serum suPAR ately effective, but not long lasting. Moreover,
is significantly elevated in FSGS patients respiratory distress with congestive heart failure
suggesting that these two conditions might have has been observed in some patients [147].
distinct underlying mechanisms [145]. Further Refractory edema with serious effusions may
studies are needed to confirm and better refine require drainage of ascites and/or pleural
854 P. Niaudet and O. Boyer

effusions. Immersion of the body up to the neck in treatment with corticosteroids. Pneumococcal
a bath may be helpful in these cases [148]. vaccine may be performed. It is effective even in
children receiving high doses of steroids, and it is
Thromboemboli not associated with an increased risk of relapse
Nephrotic patients are at risk for thromboembolic [149, 150]. In cases of peritonitis, antibiotics
complications. Prevention of this complication against both S. pneumoniæ and gram-negative
includes regular ambulation, avoidance of organisms are started after peritoneal liquid sam-
hemoconcentration due to hypovolemia, avoid- pling. Varicella is a serious disease in patients
ance of central venous catheter if possible, and receiving immunosuppressive treatment or daily
early treatment of sepsis or volume depletion. corticosteroids. Varicella immunity status should
There are no controlled trials on the efficacy and be checked in these patients. In cases of exposure,
safety of prophylactic anticoagulation to prevent early preventive treatment by acyclovir must be
thromboembolic complications in children with instituted. Varicella vaccination is safe and effec-
nephrotic syndrome. However, several authors tive if the child is in remission even if he/she is on
propose prophylactic measures in children with low-dose alternate-day steroids [151, 152].
risk factors. For example, prophylactic warfarin
therapy may be given to patients with a plasma Hyperlipidemia
albumin concentration below 20 g/l, a fibrinogen Persistent hyperlipidemia is a risk factor for ath-
level over 6 g/l, or an antithrombin III level below erosclerosis and may play a role in the progression
70 % of normal. Patients at risk may also be of chronic renal failure. Experience with
treated with low-dose aspirin and dipyridamole, hypolipidemic drugs in nephrotic patients is still
although there is no evidence that it is limited, but it seems that statins are effective and
beneficial [45]. able to decrease hypercholesterolemia [153, 154].
Heparin is given initially if thrombi do occur, Although long-term side effects of these drugs are
alone or with thrombolytic agents. Anticoagulation not known, it is reasonable to consider a lipid-
is most often initiated with low molecular weight lowering regimen in children with a persistent
heparin, such as enoxaparin at a starting dose of nephrotic syndrome [155].
1 mg/kgBW every 12 h, with the antifactor Xa
assay as a therapeutic drug monitoring. Its pharma- Miscellaneous
cokinetics is more predictable than unfractionated Calcium metabolism may be altered by the uri-
heparin, and it is given subcutaneously, avoiding nary loss of 25-hydroxycholecalciferol and its
venous catheter. In situations where short half-life carrier protein. Preventive treatment with vitamin
and reversible anticoagulation is necessary, D supplements is therefore useful, but does not
unfractionated heparin is utilized. The heparin completely prevent bone loss [156]. Thyroxine
dose necessary to obtain a therapeutic effect is substitution may be indicated, but only in patients
often greater than normal due to decreased anti- with documented hypothyroidism due to urinary
thrombin III level. loss of iodinated proteins.

Antihypertensive Drugs
Any arterial hypertension has to be carefully con- Specific Treatment
trolled, using preferably a β-blocker or a calcium
channel blocker during acute episodes. In cases of Steroid therapy is applied in all cases of INS
permanent hypertension, an angiotensin- whatever the histopathology, even in patients
converting enzyme inhibitor is preferred. with FSGS. The majority of patients are steroid
responsive (Table 5). Urine protein profile (prote-
Infections and Immunizations ome) may in the future help to predict the response
Prophylaxis of S. pneumoniæ with oral penicillin to steroid therapy [157]. Steroid responders may
is often applied in patients during the initial relapse, but the majority still responds to steroids
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 855

Table 5 Definitions normal complement levels. In some cases, the


Nephrotic syndrome: proteinuria >40 mg/h/m2 or >50 treatment is started after a renal biopsy has been
mg/kg/day or protein/creatinine ratio >0.2 g/mmol performed. Prednisone remains the reference
(>2 g/g) and hypoalbuminemia <25 g/l with or drug. Prednisolone has the advantage of being
without edema
soluble in water, making treatment easier in
Remission: proteinuria <4 mg/h/m2 or 0-trace on
Albustix for 3 consecutive days young children, but it may fail to induce remission
Steroid responsive: complete remission achieved with in some patients who respond quickly to the same
steroid therapy dosage of prednisone. Indeed, the intestinal
Steroid resistant: failure to achieve remission following absorption of prednisolone is less than that of
4-week prednisone 60 mg/m2  followed by three prednisone, and this may explain a lesser efficacy
methylprednisolone pulses
in some children.
Relapse: proteinuria >>40 mg/h/m2 or >50 mg/kg/day
or Albustix +++ for 3 consecutive days after having been The ISKDC regimen consists of prednisone,
in remission 60 mg/m2/day with a maximum of 80 mg/day, in
Frequent relapses: 2 or more relapses within 6 months of divided doses for 4 weeks followed by 40 mg/m2/
initial response or 4 or more relapses within a period of day with a maximum of 60 mg/day in divided
1 year doses, on three consecutive days per week for
Steroid dependence: 2 consecutive relapses during
4 weeks [161]. The Arbeitsgemeinschaft f€ur
corticosteroid therapy or within 14 days after cessation of
therapy pädiatrische Nephrologie showed that an
Early non responder: steroid resistance during the first alternate-day regimen (40 mg/m2 every other
episode day for 4 weeks) resulted in a significantly lower
Late non responder: steroid resistance in a patient who number of patients with relapses and fewer
had previously responded to corticosteroid therapy relapses per patient [162]. It also showed that on
alternate days, prednisone could be given in a
single dose rather than in divided doses.
over the subsequent course. Only 1–3 % of A response occurs in most cases within 10–15
patients initially steroid-sensitive subsequently days (median 11 days). According to the Interna-
become steroid-resistant and are defined as “late tional Study of Kidney Disease in Children,
nonresponders” [158]. approximately 90 % of responders enter in remis-
sion within 4 weeks after starting steroids,
whereas less than 10 % go into remission after
Initial Treatment 2–4 more weeks of a daily regimen. A few more
patients go into remission after 8–12 weeks of
Although steroid therapy is often started immedi- daily steroids [11, 13], but prolongation of daily
ately following the diagnosis of nephrotic syn- steroid treatment beyond 4 or 5 weeks increases
drome, it should be stressed that spontaneous the risk of side effects. An alternative for patients
remission occurs in 5 % of cases within 1 or who are not in remission after 4 weeks is to
2 weeks. Therefore, initiation of steroid therapy administer three to four pulses of methylprednis-
may be delayed for a few days [159]. Some of olone (1 g/1.73 m2). This additional regimen
these early spontaneous remissions are definitive. seems to be associated with fewer side effects
Infection must be treated before starting steroids, than prolongation of daily high-dose steroids and
not only to prevent the risk of overwhelming probably produces remission more rapidly in the
sepsis during treatment but also because occult few patients who would have entered into remis-
infection may be responsible for steroid sion during the second month of daily
resistance [160]. therapy [163].
Steroid therapy is started when the diagnosis of A working committee recently developed the
INS is most likely in a child older than 1 year and following KDIGO guidelines [164]. Initial pred-
younger than 11 years of age, without hyperten- nisone therapy consists of 60 mg/m2 or 2 mg/kg
sion, gross hematuria, or extrarenal symptoms and administered as a single daily dose for 4–6 weeks
856 P. Niaudet and O. Boyer

(maximum dose of 60 mg/day), followed by therapy in terms of time to onset of frequently


alternate-day prednisone of 40 mg/m2 or 1.5 relapsing nephrotic syndrome. Another random-
mg/kg (maximum dose of 40 mg/day) continued ized control trial from India showed that extending
for 2–5 months with tapering of the dose. the initial prednisolone treatment from 3 to
The duration of initial steroid therapy influ- 6 months does not influence the course of the
ences the risk of relapse. The APN compared a disease [172].
standard regimen of 4-week daily prednisone and There are no data showing that treating for
4 weeks of alternate-day prednisone with a longer more than 7 months is beneficial. However, a
initial course of 6 weeks of daily prednisone at a study showed that an alternate-day regimen over
dose of 60 mg/m2/day followed by 6 weeks a year did not reduce the rate of relapse compared
alternate-day prednisone at a dose of 40 mg/m2/ to a 5-month alternate-day regimen [173].
day [165]. The subsequent relapse rate within Although the studies were not designed to analyze
12 months following discontinuation of therapy the side effects of glucocorticoids, the authors did
was lower with the prolonged course of therapy not report increased toxicity with longer duration
compared to the standard course (36 % vs. 61 %). of treatment.
Following an 8-week steroid regimen, 50–70 % A slow tapering phase to avoid adrenal sup-
of children experience relapses. Several controlled pression may maintain long-term remission as a
studies have compared the 8-week regimen with study showed that moderate to severe adrenal
longer duration of steroid regimen (3–7 months) suppression was associated with an increased
including 4–8 weeks of daily prednisone followed risk of relapse [174]. Some authors have
by alternate-day prednisone [165–169]. With a suggested a possible prevention by low-dose
follow-up of 2 years, a significant reduction of maintenance hydrocortisone [174, 175]. Another
25–30 % in the relapse rate was observed with a study also concluded that adrenocortical suppres-
prednisone regimen of 3 months or more sion increases the risk of relapse in children on
[170]. Conversely, Teeninga et al. reported the long-term alternate-day steroid therapy [176].
results of a randomized, double-blind, placebo- Increasing initial immunosuppression by
controlled trial involving 150 children presenting adding cyclosporine to steroid therapy does not
with nephrotic syndrome who received either change the 2-year relapse rate [177].
3 months of prednisolone followed by 3 months
of placebo or 6 months of prednisolone with both
groups receiving equal cumulative doses of pred- Treatment of Relapses
nisolone [141]. This study concluded that
extending initial prednisolone treatment from 3 to About 30 % of children experience only one
6 months without increasing cumulative dose did attack and are definitively cured after a single
not benefit clinical outcome. Therefore, prolonged course of steroids. Persistent remission for
treatment regimens that reduce relapses most likely 18–24 months after stopping treatment is likely
result from increased cumulative dose rather than to reflect definitive cure, and the risk of later
treatment duration. More recently, Yoshikawa relapses is low. Ten to twenty percent of patients
et al. reported the results of an open-label, random- relapse several months after stopping treatment
ized controlled trial comparing a 2-month prednis- and are most often cured after three or four epi-
olone therapy with a 6-month therapy in sodes, which respond to a standard course of
246 children with steroid-sensitive nephrotic syn- steroid therapy. The remaining 50–60 % experi-
drome [171]. Time to frequent relapse and time to ence relapses as soon as steroid therapy is stopped
first relapse were also similar in both groups or when dosage is decreased. In some cases, exac-
allowing the authors to conclude that 2 months of erbation of proteinuria is only transient, and spon-
initial prednisolone therapy for steroid-sensitive taneous remissions are observed [193]. The risk of
nephrotic syndrome, despite less prednisolone relapse is greater in children aged less than 5 years
exposure, is not inferior to 6 months of initial at onset. A recent study found an association
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 857

between the GR-9b haplotype of the glucocorti- Steroid Side Effects


coid receptor gene and the outcome of children
with steroid-responsive nephrotic syndrome. Side effects of prolonged steroid therapy are well
Children with this haplotype showed increased known and are observed in children with a steroid-
incidence of first relapse, frequent relapses, and dependent course. Growth retardation is observed
steroid dependence as compared with noncarriers with prolonged daily steroid therapy, and alternate-
[178]. Steroid-dependent patients often raise day therapy may preserve growth [184]. However,
difficult therapeutic problems. when the dose needed to maintain remission is too
Steroid-dependent patients may be treated with high, growth may be impaired [185, 186]. Osteopo-
repeated courses of prednisone, 60 mg/m2/day, rosis has been reported in adults who had suffered
continued 3 days after the urine has become pro- from nephrotic syndrome during childhood [187].
tein free, followed by alternate-day prednisone, However, a study in children and adolescent with
40 mg/m2, for 4 weeks as proposed by the Inter- steroid-dependent nephrotic syndrome failed to
national Study of Kidney Disease in Children find a deleterious effect of alternate-day steroid
[161]. Another option consists of treating relapses therapy on bone mineral content [188]. Biyikli
with daily prednisone, 40–60 mg/m2, until pro- et al. reported that steroid treatment causes a
teinuria has disappeared for 4–5 days. Thereafter, dose-dependent decrease in bone formation, as
prednisone is switched to alternate days, and the shown by the changes in osteocalcin and alkaline
dosage is tapered to 15–20 mg/m2 every other day, phosphatase levels and low 25-hydroxyvitamin D
according to the steroid threshold, that is, the levels [189]. A randomized controlled trial com-
dosage at which the relapse has occurred. Treat- pared vitamin D and calcium supplements with no
ment is then continued for 12–18 months. The first prophylaxis in children receiving high-dose steroid
approach allows better definition in terms of num- therapy during a relapse. The authors found a
ber of relapses but is associated with more decrease in bone mineral content in both groups but
relapses. The latter regimen is associated with less pronounced in the treated group (4.6  2.1 %
less steroid side effects as the cumulative dosage vs. 13.0  4.0 %, respectively; P < 0.001)
is lower. Prolonged courses of alternate-day ste- [156]. Another controlled study showed that
roid therapy are often well tolerated by young bisphosphonates are effective in preventing steroid-
children, and growth velocity is not affected. induced osteoporosis in children receiving long-term
However, prednisone dosage must be as low as steroid therapy [190]. The other side effects include
possible in order to reduce the side effects. In weight gain, cataracts, behavior disturbances, and
adolescents, steroid therapy is often accompanied hypertension.
by decreased growth velocity.
A controlled trial has shown that deflazacort
reduces the risk of relapse in comparison with Alternative Treatments (Fig. 9)
equivalent doses of prednisone, without addi-
tional side effects [179]. Unfortunately, An alternative treatment is required when
deflazacort is not available in many countries. children develop severe side effects of steroid
The role of upper respiratory tract infections in therapy, in children at risk of toxicity (diabetes
exacerbating nephrotic syndrome has been or during puberty), in children with severe
highlighted in all series: 71 % of relapses were relapses accompanied by thrombotic complica-
preceded by such an event in a prospective study, tions or severe hypovolemia, and in those with
although only 45 % of respiratory infections were poor compliance.
followed by an exacerbation of proteinuria Alternative treatments include levamisole,
[180]. The risk of relapse is decreased during which has a steroid-sparing effect, and alkylating
upper respiratory tract infections when steroid agents such as cyclophosphamide or
therapy is given daily for 5–7 days rather than chlorambucil, mycophenolate mofetil, cyclospor-
on alternate days [181–183]. ine, and, more recently, rituximab (Fig. 9).
858

2
4 weeks prednisone, 60 mg/m /day (1)
Complete remission

No relapse Relapse > 3 months Relapse while on alternate day steroid therapy
(30%) after steroid withdrawal or less than 3 months after steroid withdrawal (60%)
(10%)

Daily steroid therapy and switch


Sart again as (1) to alternate day 3-4 days after remission

Prolonged alternate day steroid therapy


and/or levamisole

Persistent remission More relapses and steroid side effects


without steroid side-effects

Course of alkylating agents or MMF

Continue the treatment for 12 to 18 months


Relapse + steroid side effects

No relapse Relapse
Anti-calcineurin ± low dose alternate day steroid therapy

alternate day steroid therapy


for 12-18 months
Anti-calcineurin toxicity, steroid toxicity and relapses

Rituximab

Fig. 9 Proposed algorithm for the treatment of steroid-dependent idiopathic nephrotic syndrome
P. Niaudet and O. Boyer
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 859

Levamisole given alone [204, 208, 209]. The incidence of


The beneficial effect of levamisole was first relapse after cyclophosphamide is significantly
reported by Tanphaichitr et al. [191]. Levamisole higher in patients with FSGS (73 %) or mesangial
was subsequently reported to reduce the risk of proliferation, compared to children with minimal
relapse in steroid-dependent patients [192–196]. change (22 %) [210].
A significant steroid-sparing effect at a dose of 2.5 The Arbeitsgemeinschaft fur pädiatrische
mg/kg every other day was demonstrated in a Nephrologie reported that a treatment for
prospective controlled trial of the British Associ- 12 weeks at a daily dose of 2 mg/kg was more
ation for Paediatric Nephrology [197, 211]. effective than an 8-week course, with 67 % as
Another controlled study confirmed the efficacy compared to 22 % remaining in remission after
of levamisole for preventing relapses [198, 212]. 2 years [211]. However, a randomized trial
Levamisole given for 6 months was compared showed that prolonging the course of cyclophos-
with cyclophosphamide given for 8–12 weeks phamide from 8 to 12 weeks did not further reduce
in a retrospective study involving 51 children the proportion of children experiencing
with steroid-dependent nephrotic syndrome relapses [212].
[199, 213]. The relapse rate and the cumulative However, more recent data show disappointing
dose of prednisone were reduced to the same results of cyclophosphamide treatment for
extent with both drugs. However, the beneficial steroid-dependent patients. Kemper
effect of levamisole is not sustained after stopping et al. reported that only 6 out of 20 children had
treatment. a sustained remission following a 12-week course
Levamisole is well tolerated in most children. of cyclophosphamide [213]. In a retrospective
Side effects occasionally include neutropenia, study, Vester et al. reported that 24 % of 106 chil-
agranulocytosis, vomiting, cutaneous rash, vascu- dren who had received a course of cyclophospha-
litis, and neurological symptoms including insom- mide were still in remission after 10 years [207].
nia, hyperactivity, and seizures [200–202]. A younger age than 3 years at onset is associated
However, levamisole is not widely available [201]. with lower response rate [214]. A recent series
reported lower remission rates of 44 %, 27 %,
Alkylating Agents and 13 % at 1, 2, and 5 years after cyclophospha-
Alkylating agents, such as cyclophosphamide and mide therapy [215]. In another study of 90 chil-
chlorambucil, have been used for more than dren with a steroid-dependent course, sustained
50 years to achieve long-lasting remission. remissions were observed in 31 % of patients at
The efficacy of alkylating agents is illustrated 5-year follow-up [216]. These variations are prob-
by a meta-analysis that compared alkylating ably due to differences in patient populations as
agents with prednisone in maintaining remission. steroid-dependent patients have a lower response
In three trials including 102 patients, oral cyclo- rate than frequently relapsing patients. The degree
phosphamide reduced the risk of relapse at 6–12 of steroid dependency also affects remission rates
months (RR 0.44; 95 % CI 0.26–0.73). Similarly, [217], and cyclophosphamide is less effective in
chlorambucil compared with prednisone alone patients with steroid dependency compared to
reduced the risk of relapse at 12 months in 32 chil- patients with frequent relapses [218].
dren (RR 0.13; 95 % CI 0.03–0.57) [203]. Several Cyclophosphamide has also been administered
studies involving patients with frequently relaps- as monthly boluses at 500 mg/m2 for 6 months.
ing or steroid-dependent nephrotic syndrome Gulati et al. reported a remission rate of 38 % at
showed that cyclophosphamide resulted in 5 years among the 29 steroid-dependent patients
sustained remission in 57–93 % of patients at [219]. However, Donia et al. reported a remission
1 year, 31–66 % at 5 years, and 25 % at 10 years rate of only 5 % at 2 years in a group of 20 steroid-
[204–207]. The duration of remission is higher dependent patients [220]. Prasad et al. compared
when cyclophosphamide is given in association cyclophosphamide given orally or as boluses in
with steroids compared to cyclophosphamide 47 children [221]. The remission rate 6 months
860 P. Niaudet and O. Boyer

after the end of treatment was higher following the courses of treatment. Gonadal toxicity, as with
cyclophosphamide boluses (73 % vs. 38 %) but cyclophosphamide, essentially affects boys. Azo-
was similar with a follow-up of 2 years. ospermia is total and probably irreversible at
Cyclophosphamide toxicity includes bone cumulative doses above 10–20 mg/kg. No case
marrow depression, hemorrhagic cystitis, gastro- of azoospermia was reported in patients given less
intestinal disturbances, alopecia, and infection than 8 mg/kg.
[222]. Leukopenia is frequently observed, but
weekly hematological monitoring may limit its Cyclosporine
severity and concomitant steroids help blunt mar- Cyclosporine has been shown in a number of
row depression. Hemorrhagic cystitis rarely uncontrolled studies to reduce the incidence of
occurs. Alopecia, which is variably pronounced, relapses in 75–90 % of patients with steroid-
remits a few weeks after stopping treatment. Viral dependent INS [230]. However, most patients
infections can be overwhelming if cyclophospha- experience relapses when the dosage is tapered
mide is not stopped in due time. or when cyclosporine is withdrawn [231]. The
Long-term toxicity includes malignancy, pul- patients thus behave with cyclosporine as they
monary fibrosis, ovarian fibrosis, and sterility. did with steroids; that is, they become cyclospor-
Gonadal toxicity is well established, and the risk ine dependent. The relapse rate usually returns to
of sterility is greater in boys than in girls. The the pretreatment rate. Hulton et al. found that
cumulative threshold dose above which patients in whom cyclosporine had been
oligospermia/azoospermia may be feared lies discontinued and later restarted had more
between 150 and 250 mg/kg [223–225]. In relapses, requiring steroids in addition to cyclo-
females, the cumulative dose associated with ste- sporine in order to maintain remission [232].
rility is greater, but not well defined. Pregnancies The effects of cyclosporine have been evalu-
have been reported after treatments longer than ated in two comparative trials in steroid-sensitive
18 months [226]. patients. Cyclosporine at a dosage of 6 mg/kg/day
Most authors would prescribe a 12-week for 3 months, then tapered over 3 months, was
course of oral cyclophosphamide at a daily dose compared with chlorambucil given for 2 months.
of 2 mg/kg. At 12 months, 30 % of patients who had received
Beneficial results have also been achieved with chlorambucil and only 5 % of those who had
chlorambucil in steroid-responsive nephrosis received cyclosporine were still in remission
[227–229]. The response to chlorambucil is [233]. A multicenter randomized controlled trial
related to the clinical pattern of relapse as for compared cyclosporine for 9 months then tapered
cyclophosphamide. A meta-analysis of 26 con- over 3 months, with oral cyclophosphamide for
trolled trials and cohort studies found that the 2- 2 months [234]. After 2 years, 25 % of the patients
and 5-year relapse rates following treatment with (50 % of adults and 20 % of children) who had
cyclophosphamide or chlorambucil were 72 % received cyclosporine had not relapsed, while
and 36 % in frequently relapsing nephrotic syn- 63 % of those treated with cyclophosphamide
drome compared with 40 % and 24 % in steroid- (40 % of adults and 68 % of children) were still
dependent nephrotic syndrome [222]. in remission. During the year following treatment,
Acute toxic effects are less frequent with the relapse rate (1.8 vs. 0.7) and the steroid dosage
chlorambucil than with cyclophosphamide. Leu- required (109 vs. 23 mg/kg/year) were signifi-
kopenia and thrombocytopenia may occur and are cantly higher in children who had received
reversible within 1–3 weeks. Severe microbial cyclosporine.
and viral infections have been reported, including Tejani et al. performed a randomized con-
malignant hepatitis and measles encephalitis. trolled trial comparing low-dose prednisone and
Long-term toxic effects include the risk of cyclosporine versus high-dose prednisone for
developing cancer or leukemia, which has only 8 weeks as first-line treatment in 28 children
been reported in patients who had prolonged [235]. Thirteen of the 14 children receiving the
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 861

combined treatment went into remission com- cyclosporine is a reduced cosmetic side effects
pared to only 8/14 receiving prednisone alone (hypertrichosis, gum hypertrophy).
( p < 0.05). The duration of remission after ending
treatment was comparable in both groups. Severe Mycophenolate Mofetil
hypercholesterolemia may inhibit cyclosporine During the past 10 years, several reports have
efficacy and require higher dosages for similar shown that mycophenolate mofetil (MMF) treat-
results [236, 237]. ment may have a beneficial effect in children with
A prospective, open multicenter trial from steroid-dependent INS [245–251]. These studies
Japan compared the efficacy and safety of two have shown that MMF allowed decreasing or
cyclosporine regimen, a dose adjusted to maintain stopping steroid therapy in 40–75 % of children.
trough level between 60 and 80 ng/ml (group A) However, relapses were nearly constant after ces-
and a fixed dose of 2.5 mg/kg (group B) in chil- sation of treatment. MMF was shown to have a
dren who had initially received cyclosporine for significant cyclosporine and/or steroid-sparing
6 months with a trough level of 80–100 ng/ml effect in children with cyclosporine-dependent
[238]. After 2 years, the rate of sustained remis- INS with a beneficial effect on renal function
sion was significantly higher in group A. [248, 252].
Considering cyclosporine dependency, this In a small randomized trial comparing MMF to
treatment must be pursued to prevent new relapses. cyclosporine, sustained complete remission was
Indeed, cyclosporine exposes to nephrotoxicity. In achieved in 7 of 12 patients who received MMF
case of decreased renal function, it is advisable to and in 11 of 12 patients treated with cyclosporine
reduce dosage or even stop the treatment. Renal suggesting that cyclosporine is more effective
function improvement of is in favor of functional than MMF [253]. A Bayesian study was
renal insufficiency or drug nephrotoxicity. Never- conducted in 23 children with steroid-dependent
theless, lesions of chronic nephrotoxicity can idiopathic nephrotic syndrome having received
develop without any appreciable decline of the prior alkylating agent treatment and 2/3 of them
glomerular filtration rate [239–241]. As it is often levamisole [254]. They were treated with MMF
necessary to continue treatment for a long time, and prednisone according to a defined schedule
repeat renal biopsies are highly advisable to detect [reduction of alternate-day dose to 50 % of
these lesions. They most often consist of tubuloin- pre-MMF dose at 3 months, 25 % at 6 months].
terstitial injury, characterized by stripes of intersti- Twenty-three children completed the study. Four
tial fibrosis containing clusters of atrophic tubules relapsed during the first 6 months and two at
and by vascular lesions. months 8 and 11.5. In the 19 patients free of
Other side effects are of less concern: hyper- relapse during the first 6 months, median predni-
tension, hyperkalemia, hypertrichosis, gum sone maintenance dose decreased from
hypertrophy, and hypomagnesemia are common 25 (10–44) to 9 (7.5–11.2) mg/m2 and cumulative
but easily manageable. Cyclosporine treatment dose from 459 (382–689) to 264 (196–306)
has no deleterious effect on bone mineral mg/m2/ before and on MMF, respectively. The
content [242]. authors concluded that MMF reduces relapse
rate and steroid dose in children with steroid-
Tacrolimus dependent nephrotic syndrome and should be
In a retrospective study of ten children, it was proposed before cyclosporine.
observed that tacrolimus was not better than cyclo- These studies confirm the efficacy and safety
sporine for the management of severe steroid- of MMF in patients with steroid-dependent
dependent nephrotic syndrome [243]. In a series nephrotic syndrome and support its use for a lon-
of five children treated with tacrolimus, two devel- ger duration than 12 months. Doses of 450–600
oped insulin-dependent diabetes mellitus, which mg/m2/day in two divided doses are usually
resolved after stopping tacrolimus therapy [244]. given. Side effects including gastrointestinal dis-
However, one advantage of tacrolimus over turbances (abdominal pain, diarrhea) and
862 P. Niaudet and O. Boyer

hematologic abnormalities are rare. Many authors in stable remission without drugs after 9 months.
now recommend the use of MMF rather than The authors concluded that rituximab and lower
alkylating agents in children with steroid- doses of prednisone and calcineurin inhibitors are
dependent nephrotic syndrome who suffer from non-inferior to standard therapy in maintaining
side effects of steroid therapy. However, random- short-term remission in children with INS depen-
ized trials should be performed before such dent on both drugs and allow their temporary
recommendations can be made. withdrawal.
Iijima et al. performed a multicentre, double-
Rituximab blind, randomized, placebo-controlled trial
Several retrospective series suggest that rituximab involving 48 patients with complicated frequently
(RTX), a chimeric anti-CD20 monoclonal anti- relapsing or steroid-dependent nephrotic syn-
body that depletes B-cell lymphocytes, is effec- drome who received rituximab (375 mg/m2) or
tive in steroid-dependent or calcineurin inhibitor- placebo once weekly for 4 weeks [258]. All
dependent patients allowing to reduce or stop patients received standard steroid treatment for a
immunosuppression [255–257]. In a case series relapse at screening and stopped taking immuno-
of 22 children with severe steroid or cyclosporine- suppressive agents by 169 days after randomiza-
dependent NS, the administration of RTX allowed tion. The median relapse-free period was
the discontinuation of one or more immunosup- significantly longer in the rituximab group
pressive agents [134]. The response to rituximab (267 days, 95 % CI 223–374) than in the placebo
appears to be better if the patient is in remission at group (101 days, 70–155; hazard ratio: 0.27,
the time of the infusion. Seven patients were 0.14–0.53; p < 0.0001). The rate of relapse by
nephrotic at the time of RTX treatment. Remission day 85 was 42 % in the rituximab group and 83 %
was induced in three of the seven proteinuric in the placebo group, and all patients in the trial
patients. RTX was effective in all patients when had relapsed by 19 months.
administered during a proteinuria-free period in Indeed, a significant proportion of patients
association with other immunosuppressive relapse after rituximab administration. Most
agents. Therefore, rituximab should not be admin- relapses occur simultaneously with the recovery
istered during a relapse but after remission has of B-cell lymphocytes. Maintenance therapy with
been induced by increased doses of steroids. One MMF has been shown to be effective in
or more immunosuppressive treatments could be preventing relapse after treatment with rituximab
withdrawn in 19 patients, with no relapse of pro- [259]. Fujinaga et al. found that cyclosporine was
teinuria and without increasing other drug dosage. even more effective than MMF for maintaining
When relapses occurred, they were associated remission after a single infusion of RTX [260].
with an increase in CD19 cell count. Adverse Although rituximab is well tolerated in most
effects were observed in 45 % of cases, but most patients, it may be associated with adverse effects
of them were mild and transient. including infusion-related reactions (hypotension,
Ravani et al. randomized 54 children (mean fever, skin rash, diarrhea, and bronchospasm).
age 11  4 years) with INS dependent on predni- Patients may develop serious infections second-
sone and calcineurin inhibitors for >12 months ary to leukopenia and/or hypogammaglo-
[135]. RTX with lower doses of prednisone and bulinemia. Patients with minimal change disease
calcineurin inhibitors was compared to current have depressed serum IgG levels. This is more
therapy alone. Three-month proteinuria was pronounced during relapses, but persists during
70 % lower in the RTX arm as compared with remission [261]. It was recently reported that
standard therapy arm (intention to treat); relapse rituximab prolongs this IgG depletion [262]. Sev-
rates were 18.5 % (intervention) and 48.1 % (stan- eral cases of progressive multifocal leukoence-
dard arm) (P = 0.029). Probabilities of being phalopathy caused by JC polyomavirus have
drug-free at 3 months were 62.9 % and 3.7 %, been reported in patients with hematological dis-
respectively (P < 0.001); 50 % of RTX cases were orders or lupus. In addition, there is one published
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 863

case report of death due to lung fibrosis [263] and 26 patients over the age of 20 years, of whom
another one of severe myocarditis requiring heart 5 were still relapsing in adulthood [273].
transplantation [264] in two children with Koskimies et al. reported on the follow-up of
nephrotic syndrome treated with rituximab. Addi- children with INS observed in Finland from 1967
tional severe adverse effects reported in childhood to 1976: 94 of 114 cases had responded to corti-
nephrotic syndrome include Pneumocystis carinii costeroids. Twenty-four percent of steroid
pneumonia [134, 265] and severe immune- responders had no relapse, 22 % had infrequent
mediated ulcerative gastrointestinal disease relapses, and 54 % had frequent relapses. More
[266]. These life-threatening complications may than two-thirds were in remission at time of report
be underestimated in the literature. Moreover, the [274]. None of these patients developed renal
long-term immunological consequences of insufficiency, and none died from the disease.
repeated rituximab infusions in children are not Lahdenkari et al. reported a 30-year follow-up of
known. the patients reported previously by Koskimies
The KDIGO Glomerulonephritis Workgroup et al. [275]. Of 104 patients, 10 % had further
suggested that rituximab be considered only in relapses in adulthood.
children who have continuing relapses despite Fakhouri et al. reported on the outcome in
optimal combinations of prednisone and steroid- adulthood of 102 patients born between 1970
sparing agents or in patients who had developed and 1975 [187]. Forty-two percent presented at
calcineurin inhibitor nephrotoxicity [267]. Well- least one relapse in adulthood. A young age at
defined randomized control studies are mandatory onset and a high number of relapses during child-
to examine which drug has the best risk/benefit hood were associated with a high risk of relapse in
profile and to better define the place of rituximab adulthood. Similarly, Ruth et al. in a study of
in the treatment of childhood steroid-sensitive 42 patients found that 14 (33 %) relapsed in adult-
nephrotic syndrome [268], knowing that hood [276]. The higher relapse rates in these two
rituximab does not cure the disease and repeated reports probably reflect patient selection with
courses may be necessary [269]. more steroid-dependent cases compared to
Koskimies’ series.
This generally good long-term prognosis is
Long-Term Outcome of Children also observed in patients with steroid-responsive
with Steroid-Sensitive Nephrosis focal and segmental glomerulosclerosis: the
19 children reported by Arbus et al. remained
Steroid-dependent patients may have a prolonged responders, and none had renal insufficiency
course. However, if the patient continues to after a mean follow-up of 10 years [277].
respond to steroids, the risk of progression to
chronic renal failure is minimal.
Schärer and Minges found that 22 % of patients Treatment of Steroid-Resistant INS
had only one attack and 35 % of the relapsing
patients continued to relapse after 10 years Resistance to steroid therapy is defined by the
[270]. Trompeter et al. reported the late outcome absence of remission after 1 month of daily pred-
of 152 children steroid-responsive nephrotic syn- nisone therapy at a dose of 60 mg/m2/day
drome after a follow-up of 14–19 years: [161]. Some authors continue the treatment for
127 (83 %) were in remission, 4 had hypertension, 1 or 2 weeks, while others favor a series of three
10 were still relapsing, and 11 had died [271]. The methylprednisolone pulses (1,000 mg/1.73 m2)
duration of the disease was longer in children who every other day [163]. Indeed, the side effects of
had started before the age of 6 years. Wynn this regimen are less than those induced by an
et al. found that 15 % of 132 patients had a increase in the daily prednisone dose. Persistence
persistent relapsing course with a mean follow- of proteinuria 1 week after this treatment defines
up of 27.5 years [272]. Lewis et al. reported on steroid resistance. A recent report found that
864 P. Niaudet and O. Boyer

prednisone, 60 mg/m2/day, 4 weeks


+ 3 methylprednisolone pulses

Persistent nephrotic syndrome

Renal biopsy

Cyclosporine or tacrolimus + prednisone

Complete remission No response

stop cyclosporine or tacrolimus and prednisone


No relapse Relapse
often steroid
responsive
Symptomatic therapy MMF + MP pulses?
(ACEI, statins) IV cyclophosphamide?
Rituximab?
Galactose?

Fig. 10 Proposed algorithm for the treatment of steroid-resistant idiopathic nephrotic syndrome

children with initial steroid resistance show difficult to analyze, as a significant proportion of
decreased glucocorticoid receptor expression in the patients progress to cure gradually, with a slow
peripheral blood mononuclear cells before decrease of proteinuria, and in such cases,
starting therapy compared to steroid-responsive relapses are rare.
children. This low expression may be one of the
pathophysiological mechanisms of steroid resis- Pulse Methylprednisolone
tance in children [278]. Mendoza et al. have proposed methylpredniso-
Overall, the prognosis of steroid-resistant idi- lone pulse therapy. It consists of methylpredniso-
opathic nephrotic syndrome is poor, with a high lone (30 mg/kg intravenously), administered
proportion of children progressing to end-stage every other day for 2 weeks, weekly for
renal failure. This explains that intensive treat- 8 weeks, every other week for 8 weeks, monthly
ment regimens have been tried. The results of for 9 months, and then every other month for
immunosuppressive treatments should take into 6 months in association with oral prednisone
account the fact that children with genetic forms and, if necessary, cyclophosphamide or
of INS most often fail to respond to any therapy. chlorambucil [279]. At an average of over
However, many published trials include patients 6 years of follow-up, 21 of 32 children were in
who had not been tested for mutations in the complete remission, and the 5-year incidence of
different genes involved in steroid-resistant INS. end-stage renal disease was approximately 5 %
Moreover, most studies are nonrandomized and versus 40 % in historical controls [280]. Two pub-
include a small number of patients. lications reported similar results [281, 282].
The interpretation of treatment is also compli- Although these results are better than those seen
cated by the fact that many studies include with any other therapy, other reports described
patients with minimal change disease, diffuse less favorable results [283, 284].
mesangial proliferation, and FSGS, while others
only include patients with FSGS. Calcineurin Inhibitors
The treatment of these patients remains diffi- A combination of calcineurin inhibitor with
cult. Several therapeutic options have been pro- low-dose steroid therapy for at least 6 months is
posed (Fig. 10). The results of these treatments are presently the best option as a first-line therapy.
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 865

Cyclosporine resistant FSGS who received prolonged and inten-


The rate of complete remission is significantly sified treatment with combined cyclosporine and
higher when cyclosporine is given in combination steroids including methylprednisolone pulses.
with steroids [230]. This treatment resulted in sustained remission in
The French Society of Pediatric Nephrology 84 % of children with non-genetic forms of
reported the results of a prospective trial including steroid-resistant INS [289].
65 children treated with cyclosporine, 150–200 Three randomized trials involving 49 children
mg/m2, and prednisone, 30 mg/m2/day for showed that complete remissions and partial
1 month and on alternate days for 5 months there- remissions were observed in 31 % and 38 % of
after [285]. Twenty-seven patients (42 %) went patients which was significantly higher than in the
into complete remission (48 % of those with min- control arms [290–292].
imal changes and 30 % of those with FSGS) and
four (6 %) partial remission, whereas 34 (52 %) Tacrolimus
failed to respond to the combined treatment. The There is evidence from case series that tacrolimus
remission rate was higher in patients who had is effective in children with steroid-resistant INS
initially responded to steroids but had developed [293–295]. Gulati et al. found that tacrolimus and
late steroid resistance (71 %) compared with ini- prednisone are more effective than cyclophospha-
tially steroid nonresponders (33 %). Interestingly, mide pulses in children with steroid-resistant
eight patients who relapsed after cyclosporine nephrotic syndrome, including those with mini-
treatment responded to steroid and experienced a mal change disease [296]. Tacrolimus was found
steroid-dependent course. Progression to chronic to be as effective as cyclosporine in a randomized
or terminal renal failure was observed only in trial involving 41 children [297]. Both groups
patients who had had a partial remission (1 patient) received alternate-day steroids and enalapril. The
or in those who did not respond to the treatment rate of remission at 6 months was 85.7 % with
(12 patients including 5 with minimal change tacrolimus and 80 % with cyclosporine. Interest-
disease and 7 with FSGS). ingly, the proportion of patients with relapses was
Ingulli et al. reported that prolonged cyclospor- significantly higher in the cyclosporine group.
ine treatment in children with steroid-resistant Conversely, Wang et al. found that tacrolimus
FSGS reduces proteinuria and blunts the progres- was associated with higher efficacy and lower
sion to end-stage renal failure [286]. The dose of renal toxicity in comparison to CsA, with no dif-
cyclosporine (4–20 mg/kg/day) was titrated to the ference in the rate of relapse [298].
serum cholesterol level to achieve a remission. In
this study, only 5 of the 21 treated patients (24 %) Alkylating Agents
progressed to ESRF compared to 42 of 54 patients Although alkylating agents have little therapeutic
from an historical group who had not received this effect in steroid-resistant FSGS, for unknown rea-
treatment. Gregory et al. treated 15 children with sons, they are still widely used either alone or in
steroid-resistant INS with an association of mod- combination with corticosteroids. The Interna-
erate doses of cyclosporine and prednisone. They tional Study of Kidney Disease in Children
observed a remission in 13 children after a mean reported on 60 children with steroid-resistant
duration of treatment of 2 months [287]. Singh FSGS who were randomly allocated to receive
et al. reported the effect of cyclosporine in 42 chil- either prednisone 40 mg/m2 on alternate days for
dren with steroid-resistant FSGS [288]. The mean 12 months (control group) or cyclophosphamide,
proteinuria decreased from 7.1 to 1.8 g/day, while 2.5 mg/kgBW for 3 months plus prednisone
the serum albumin increased from 2.1 to 3.5 g/dl. 40 mg/m2 on alternate days for 12 months
The mean serum creatinine increased from [299]. Complete remissions were observed in
0.85 to 1.26 mg/dl. Twenty-five patients achieved 28 % of children in the control group and in
complete remission. Ehrich et al. reported a retro- 25 % of children who received cyclophospha-
spective study including 25 children with steroid- mide. The authors concluded that there was no
866 P. Niaudet and O. Boyer

beneficial effect of cyclophosphamide in these cyclosporine was more effective in reducing pro-
patients. Rennert et al. treated ten children with teinuria. The authors concluded that the small
steroid-resistant FSGS with cyclophosphamide sample size might have prevented detection of a
pulses. Only two of the five initial nonresponders moderate treatment effect.
went into remission, whereas all five late nonre-
sponders achieved complete remission [300]. In a Rituximab
prospective study of 24 patients, Bajpai et al. also At present, there is no evidence that rituximab is
found that therapy with intravenous cyclophos- effective in patients with steroid-resistant
phamide had limited efficacy in patients with ini- nephrotic syndrome, although retrospective case
tial corticosteroid resistance, while sustained series report that treatment with rituximab is effec-
remission was likely to occur in patients with tive in some patients [255, 306–310]. Bagga
late resistance and those with absence of signifi- et al. reported three complete and two partial
cant tubulointerstitial changes on renal histology remissions in five patients receiving rituximab
[301]. Intravenous cyclophosphamide pulses [306]. Kamei et al. treated ten steroid and
were found to be more effective than oral cyclo- calcineurin inhibitor-resistant patients with addi-
phosphamide in children with steroid-resistant tional rituximab. Seven patients achieved com-
minimal change disease. In this trial, patients plete remission, one partial remission, and two
receiving cyclophosphamide pulses had more failed to respond. The seven patients with com-
sustained remissions despite lower cumulative plete remission preserved normal renal function
dose [302]. Another study found that finally, without proteinuria [308]. However, subsequent
Gulati et al. found that tacrolimus and predniso- reports did not confirm such good results [307,
lone were superior to IV cyclophosphamide and 311–313]. Gulati et al. reported that rituximab had
prednisolone with a rate of complete remission of induced a complete remission in 27 % of 33
52.4 % and 14.8 %, respectively [296]. steroid-resistant patients and a partial remission
in 21 % of them. The rate of remission was
Mycophenolate Mofetil higher in patients with minimal changes on renal
There is no convincing data for the beneficial biopsy and in patients who were late nonre-
effect of mycophenolate mofetil in children with sponders [307]. Magnasco et al. reported the
steroid-resistant FSGS. Mendizabal et al. treated results of an open-label randomized trial including
five patients, and only one achieved complete 31 children aged 2–16 years. All received
remission [250]. Mycophenolate mofetil in asso- calcineurin inhibitors and prednisolone and 16 of
ciation with methylprednisolone pulses and them received in addition two rituximab infu-
angiotensin-converting enzyme inhibitors was sions. Proteinuria remained unchanged in these
reported to significantly reduce proteinuria patients, and none entered partial or complete
[303]. Another study involving 52 patients found remission [312].
a rate of complete remission of 23 % and partial
remission of 35.5 % following therapy with MMF Galactose
[304]. A prospective trial of the NIH compared Savin et al. showed that galactose binds to the
cyclosporine with low-dose alternate-day predni- focal segmental glomerulosclerosis permeability
sone to a combination of oral pulse dexametha- factor and that oral administration of galactose
sone and mycophenolate mofetil [305]. Partial or decreased the activity of the plasma permeability
complete remission was achieved in 22 of the factor [314]. Following this observation, De Smet
66 patients in the mycophenolate/dexamethasone et al. reported the remission of proteinuria in a
group and 33 of the 72 cyclosporine-treated patient with a nephrotic syndrome resistant to
patients at 12 months. Thus, this study did not corticosteroids, immunosuppression, and plasma-
find a difference in rates of proteinuria remission pheresis [315]. However, Sgambat et al. did not
following 12 months of cyclosporine compared to observe a decrease of proteinuria in seven chil-
mycophenolate/dexamethasone, although dren with steroid-resistant nephrotic syndrome
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 867

despite a reduction of the plasma permeability Lipid-Lowering Agents


factor activity [316]. Hyperlipidemia has been shown in experimental
animals to accelerate the progression of glomeru-
Nonsteroidal Anti-inflammatory Drugs lar sclerosis. Controlled trials in adult patients
These drugs may decrease proteinuria. Several have shown that lipid-lowering agents can prevent
authors have used indomethacin in the treatment the decline of renal function. No such studies have
of INS with variable results. Donker et al. found a been performed in children.
reduction of proteinuria in patients with FSGS but
with a simultaneous reduction of glomerular fil-
tration rate [317]. Velosa et al. also found a clear Recurrence of Proteinuria After Renal
reduction in proteinuria in patients treated with Transplantation
meclofenamate [318].
The detrimental effect of nonsteroidal anti- The major problem of patients with INS who
inflammatory agents on renal function is well progress to end-stage renal failure is the risk of
known, and patients with renal disease seem more recurrence of proteinuria after renal transplanta-
vulnerable [319, 320]. Positive sodium balance, tion. In children, recurrence of proteinuria occurs
increased edema, and risk of arterial hypertension in most cases within the first hours or the first days
are recognized complications. The decrease of glo- after transplantation. A high proportion of patients
merular filtration rate observed with nonsteroidal with immediate recurrence show delayed graft
anti-inflammatory drugs is usually reversible in function [325, 326]. In some patients, proteinuria
salt-sodium-depleted patients. However, irrevers- recurs several months later. Proteinuria is most
ible renal failure has been reported with a high often associated with a nephrotic syndrome.
incidence in a prospective study in children with Transplant biopsy when performed early shows
steroid-resistant FSGS [321]. minimal glomerular changes with foot process
fusion [327, 328]. Podocyte foot process efface-
Angiotensin-Converting Enzyme ment can be observed within minutes after reper-
Inhibitors fusion and correlates with proteinuria [329].
Numerous studies in adults have demonstrated Lesions of FSGS appear after several days or
that angiotensin-converting enzyme (ACE) inhib- weeks, which is a strong argument to consider
itors and angiotensin II receptor blockers (ARBs) these lesions as secondary rather than the cause
slow the rate of progression of proteinuric chronic of heavy proteinuria [327]. Therefore, it is more
renal disease. Captopril was reported to decrease appropriate to speak of recurrence of nephrotic
dramatically nephrotic range proteinuria due to syndrome rather that recurrence of FSGS.
renovascular hypertension and secondary FSGS The overall risk of recurrence is estimated to be
[322]. A decrease in proteinuria, and even com- between 20 % and 30 % [330]. The risk is higher in
plete remission, has also been observed in patients children compared to adults [331]. Initial steroid
with chronic glomerulopathies with or without sensitivity in children with steroid-resistant
hypertension [323]. A 50 % decrease of protein- nephrotic syndrome is associated with a higher risk
uria without a concomitant decrease in glomerular of recurrence. In a large series of children without
filtration rate was reported in children with genetic or familial steroid-resistant nephrotic syn-
steroid-resistant INS [324]. Marked benefits with drome, 26 of 28 late nonresponders (92.9 %) expe-
an ACE inhibitor and/or ARB therapy, plus rienced recurrence versus 26 of 86 initial
mycophenolate, were observed in nine children nonresponders (30.2 %) [332]. In children, recur-
with steroid-resistant FSGS [303]. At 6 months, rence is more frequent when the disease has started
mycophenolate plus angiotensin blockade after the age of 6 years than before [333, 334].
resulted in a 72 % decrease in proteinuria from The most likely explanation is that the incidence of
baseline values, a benefit that was maintained for a genetic forms of steroid-resistant INS is higher in
minimum period of 24 months. younger children and these patients have a very low
868 P. Niaudet and O. Boyer

risk of recurrence. A rapid progression of the disease disappeared after 20.8  8.4 days (range: 12–40
to end-stage renal failure is a major factor associated days). Persistent remission was observed in
with recurrence: in most series, when the duration of 11 patients with a follow-up of 3.7  3 years.
disease has been shorter than 3 years, the nephrotic Actuarial graft survival was 92 % and 70 % at
syndrome recurs in half of the patients [333, 335]. 1 and 5 years [352].
Recurrence is less frequent in African-Americans Most patients with recurrence are treated with
than in whites and Hispanics [330]. The histopatho- plasma exchanges or immunoabsorption. In a
logical pattern observed on the first biopsy during review of the literature, Ponticelli found that
the course of the disease is also an important predic- 49 of 70 children who had received plasma
tive factor [331, 333, 336, 337]. Recurrence occurs exchanges had entered complete or partial remis-
in 50–80 % of patients in whom initial biopsy sion of proteinuria [355–361]. The best results are
showed diffuse mesangial proliferation but in only observed when plasma exchanges are performed
25 % of patients with minimal changes on first early after recurrence. Complete and sustained
biopsy. Most patients who experience a recurrence remission was observed in nine of ten adult
in a first graft also show recurrence in a second graft patients treated with high dose of oral steroids,
[326, 338, 339]. Conversely, when the graft has been 14 days of intravenous cyclosporine, and plasma
lost due to rejection without recurrence, a second exchanges for 9 months [362]. A few studies of
graft can be performed safely as recurrence is excep- preemptive plasma exchanges have been reported
tional in this setting [343]. Donor type (living that show inconstant efficacy.
or deceased donor) does not change recurrence Nozu et al. first reported the case of a child with
risk [340]. posttransplant lymphoproliferative disease in
Recurrence increases the risk of delayed graft whom rituximab induced a complete remission
function and early and late graft losses. Graft of a recurrent nephrotic syndrome [363]. Pescovitz
failure occurs in about 50 % of patients with et al. reported the case of a child with recurrent
recurrence [330, 341, 342]. However, some FSGS resistant to plasma exchanges and cyclo-
patients may show good renal function for several sporine who developed at month 5 a posttrans-
years despite persistent nephrotic syndrome. plantation lymphoproliferative disease and had a
The beneficial role of cyclosporine in recurrent complete remission of proteinuria following six
steroid-resistant INS is still debated. There is no infusions of rituximab [364]. Other reports con-
evidence that cyclosporine can prevent the recur- firmed the beneficial effect of rituximab given
rence of nephrotic syndrome following transplan- alone or more often with plasma exchanges
tation [343–348]. Following the introduction of [365–368], but failures have also been reported
cyclosporine, the incidence of recurrence did not [369, 370].
change, but graft survival was improved. In
patients who have recurrent disease, high doses
of oral cyclosporine or intravenous cyclosporine Conclusion
may be effective [349–353]. This beneficial effect
may be related to the immunosuppressive effect of Most children with idiopathic nephrotic syndrome
cyclosporine and also to the direct effect of cyclo- respond to corticosteroid therapy. Although about
sporine on podocytes by blocking calcineurin- half of them experience relapses requiring long-
mediated dephosphorylation of synaptopodin term steroid therapy and several corticosteroid-
and the resulting stabilization of actin [354]. In sparing agents, these children maintain normal
our group, 17 children with recurrence were renal function, and the severity of the disease is
treated with intravenous cyclosporine at an initial mainly related to the complications of the treat-
dose of 3 mg/kg/day, which was afterward ments needed to maintain remission.
adapted in order to maintain permanent whole There is evidence to suggest that minimal
blood levels between 250 and 350 ng/ml. In changes and FSGS represent a spectrum of dis-
14 of the 17 cases (82 %), proteinuria completely eases with FSGS at the severe end [367]. Indeed,
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 869

several disease entities are included in the term 11. International Study of Kidney Disease in Children,
“steroid-resistant INS.” Defects in podocyte pro- Nephrotic syndrome in children: prediction of histo-
pathology from clinical and laboratory characteristics
teins have been identified in some cases, and these at time of diagnosis. A report of the International
patients do not respond to any therapy. In other Study of Kidney Disease in Children. Kidney Int.
patients, circulating factor(s) produced by 1978;13(2):159–65.
immune cells increase the glomerular basement 12. McKinney PA, Feltbower RG, Brocklebank JT,
Fitzpatrick MM. Time trends and ethnic patterns of
membrane permeability to proteins. More than childhood nephrotic syndrome in Yorkshire,
50 % of steroid-resistant patients progress to UK. Pediatr Nephrol. 2001;16(12):1040–4.
end-stage renal failure. Recurrence of the 13. Schlesinger ER, Sultz HA, Mosher WE, Feldman JG.
nephrotic syndrome occurs in 30–40 % of them The nephrotic syndrome. Its incidence and implica-
tions for the community. Am J Dis Child.
after renal transplantation. 1968;116(6):623–32.
14. Wong W. Idiopathic nephrotic syndrome in
New Zealand children, demographic, clinical fea-
tures, initial management and outcome after twelve-
References month follow-up: results of a three-year national sur-
veillance study. J Paediatr Child Health. 2007;43(5):
1. Eddy AA, Symons JM. Nephrotic syndrome in child- 337–41.
hood. Lancet. 2003;362(9384):629–39. 15. Srivastava RN, Mayekar G, Anand R, Choudhry VP,
2. Bariety J, Bruneval P, Hill G, Irinopoulou T, Ghai OP, Tandon HD. Nephrotic syndrome in Indian
Mandet C, Meyrier A. Posttransplantation relapse of children. Arch Dis Child. 1975;50(8):626–30.
FSGS is characterized by glomerular epithelial cell 16. Anochie I, Eke F, Okpere A. Childhood nephrotic
transdifferentiation. J Am Soc Nephrol. 2001;12(2): syndrome: change in pattern and response to steroids.
261–74. J Natl Med Assoc. 2006;98(12):1977–81.
3. Barisoni L, Mokrzycki M, Sablay L, Nagata M, 17. Coovadia HM, Adhikari M, Morel-Maroger L.
Yamase H, Mundel P. Podocyte cell cycle regulation Clinico-pathological features of the nephrotic
and proliferation in collapsing glomerulopathies. Kid- syndrome in South African children. Q J Med.
ney Int. 2000;58(1):137–43. 1979;48(189):77–91.
4. Shankland SJ, Eitner F, Hudkins KL, Goodpaster T, 18. Doe JY, Funk M, Mengel M, Doehring E, Ehrich
D’Agati V, Alpers CE. Differential expression of JH. Nephrotic syndrome in African children: lack of
cyclin-dependent kinase inhibitors in human glomer- evidence for ‘tropical nephrotic syndrome’? Nephrol
ular disease: role in podocyte proliferation and matu- Dial Transplant. 2006;21(3):672–6.
ration. Kidney Int. 2000;58(2):674–83. 19. Hayslett JP, Kashgarian M, Bensch KG, Spargo BH,
5. Moudgil A, Nast CC, Bagga A, Wei L, Nurmamet A, Freedman LR, Epstein FH. Clinicopathological cor-
Cohen AH, et al. Association of parvovirus B19 relations in the nephrotic syndrome due to primary
infection with idiopathic collapsing glomerulopathy. renal disease. Medicine (Baltimore). 1973;52(2):
Kidney Int. 2001;59(6):2126–33. 93–120.
6. Tanawattanacharoen S, Falk RJ, Jennette JC, Kopp 20. Meadow SR, Sarsfield JK, Scott DG, Rajah SM.
JB. Parvovirus B19 DNA in kidney tissue of patients Steroid-responsive nephrotic syndrome and allergy:
with focal segmental glomerulosclerosis. Am J Kid- immunological studies. Arch Dis Child. 1981;56(7):
ney Dis. 2000;35(6):1166–74. 517–24.
7. Jefferson JA, Alpers CE. Glomerular disease: 21. Salsano ME, Graziano L, Luongo I, Pilla P, Giordano
‘suPAR’-exciting times for FSGS. Nat Rev Nephrol. M, Lama G. Atopy in childhood idiopathic nephrotic
2013;9(3):127–8. syndrome. Acta Paediatr. 2007;96(4):561–6.
8. Wei C, El Hindi S, Li J, Fornoni A, Goes N, 22. Thomson PD, Stokes CR, Barratt TM, Turner MW,
Sageshima J, et al. Circulating urokinase receptor as Soothill JF. HLA antigens and atopic features in
a cause of focal segmental glomerulosclerosis. Nat steroid-responsive nephrotic syndrome of childhood.
Med. 2011;17(8):952–60. Lancet. 1976;2(7989):765–8.
9. Kashgarian M, Hayslett JP, Siegel NJ. Lipoid nephro- 23. Laurent J, Rostoker G, Robeva R, Bruneau C, Lagrue
sis and focal sclerosis distinct entities or spectrum of G. Is adult idiopathic nephrotic syndrome food
disease. Nephron. 1974;13(2):105–8. allergy? Value of oligoantigenic diets. Nephron.
10. McAdams AJ, Valentini RP, Welch TR. The 1987;47(1):7–11.
nonspecificity of focal segmental glomerulosclerosis. 24. Audard V, Larousserie F, Grimbert P, Abtahi M, Sotto
The defining characteristics of primary focal JJ, Delmer A, et al. Minimal change nephrotic syn-
glomerulosclerosis, mesangial proliferation, and min- drome and classical Hodgkin’s lymphoma: report of
imal change. Medicine (Baltimore). 1997;76(1): 21 cases and review of the literature. Kidney Int.
42–52. 2006;69(12):2251–60.
870 P. Niaudet and O. Boyer

25. Eagen JW. Glomerulopathies of neoplasia. Kidney 41. Fliser D, Zurbruggen I, Mutschler E, Bischoff I,
Int. 1977;11(5):297–303. Nussberger J, Franek E, et al. Coadministration of
26. Nakahara C, Wada T, Kusakari J, Kanemoto K, albumin and furosemide in patients with the nephrotic
Kinugasa H, Sibasaki M, et al. Steroid-sensitive syndrome. Kidney Int. 1999;55(2):629–34.
nephrotic syndrome associated with Kimura disease. 42. Raij L, Keane WF, Leonard A, Shapiro FL. Irrevers-
Pediatr Nephrol. 2000;14(6):482–5. ible acute renal failure in idiopathic nephrotic
27. Rajpoot DK, Pahl M, Clark J. Nephrotic syndrome syndrome. Am J Med. 1976;61(2):207–14.
associated with Kimura disease. Pediatr Nephrol. 43. Krensky AM, Ingelfinger JR, Grupe WE. Peritonitis
2000;14(6):486–8. in childhood nephrotic syndrome: 1970–1980. Am J
28. Alwadhi RK, Mathew JL, Rath B. Clinical profile of Dis Child. 1982;136(8):732–6.
children with nephrotic syndrome not on glucocorti- 44. Kerlin BA, Ayoob R, Smoyer WE. Epidemiology and
coid therapy, but presenting with infection. J Paediatr pathophysiology of nephrotic syndrome-associated
Child Health. 2004;40(1–2):28–32. thromboembolic disease. Clin J Am Soc Nephrol.
29. Joven J, Villabona C, Vilella E, Masana L, Alberti R, 2012;7(3):513–20.
Valles M. Abnormalities of lipoprotein metabolism in 45. Kerlin BA, Haworth K, Smoyer WE. Venous throm-
patients with the nephrotic syndrome. N Engl J Med. boembolism in pediatric nephrotic syndrome. Pediatr
1990;323(9):579–84. Nephrol. 2014;29(6):989–97.
30. Thabet MA, Salcedo JR, Chan JC. Hyperlipidemia in 46. Schlegel N. Thromboembolic risks and complications
childhood nephrotic syndrome. Pediatr Nephrol. in nephrotic children. Semin Thromb Hemost.
1993;7(5):559–66. 1997;23(3):271–80.
31. Vaziri ND. Molecular mechanisms of lipid disorders 47. Lilova MI, Velkovski IG, Topalov IB. Thromboem-
in nephrotic syndrome. Kidney Int. 2003;63(5): bolic complications in children with nephrotic syn-
1964–76. drome in Bulgaria (1974–1996). Pediatr Nephrol.
32. Sato KA, Gray RW, Lemann Jr J. Urinary excretion 2000;15(1–2):74–8.
of 25-hydroxyvitamin D in health and the 48. Kerlin BA, Blatt NB, Fuh B, Zhao S, Lehman A,
nephrotic syndrome. J Lab Clin Med. 1982;99 Blanchong C, et al. Epidemiology and risk factors
(3):325–30. for thromboembolic complications of childhood
33. Freundlich M, Bourgoignie JJ, Zilleruelo G, nephrotic syndrome: a Midwest Pediatric Nephrology
Abitbol C, Canterbury JM, Strauss J. Calcium and Consortium (MWPNC) study. J Pediatr. 2009;155(1):
vitamin D metabolism in children with nephrotic syn- 105–10, 10 e1.
drome. J Pediatr. 1986;108(3):383–7. 49. Hoyer PF, Gonda S, Barthels M, Krohn HP, Brodehl
34. McVicar M, Exeni R, Susin M. Nephrotic syndrome J. Thromboembolic complications in children with
and multiple tubular defects in children: an early sign nephrotic syndrome. Risk and incidence. Acta
of focal segmental glomerulosclerosis. J Pediatr. Paediatr Scand. 1986;75(5):804–10.
1980;97(6):918–22. 50. Zhang LJ, Wang ZJ, Zhou CS, Lu L, Luo S, Lu GM.
35. Rodriguez-Soriano J, Vallo A, Castillo G, Oliveros Evaluation of pulmonary embolism in pediatric
R. Defect in urinary acidification in nephrotic syn- patients with nephrotic syndrome with dual
drome and its correction by furosemide. Nephron. energy CT pulmonary angiography. Acad Radiol.
1982;32(4):308–13. 2012;19(3):341–8.
36. Vaziri ND, Kaupke CJ, Barton CH, Gonzales 51. Cueto SM, Cavanaugh SH, Benenson RS, Redclift
E. Plasma concentration and urinary excretion of MS. Computed tomography scan versus ventilation-
erythropoietin in adult nephrotic syndrome. Am J perfusion lung scan in the detection of pulmonary
Med. 1992;92(1):35–40. embolism. J Emerg Med. 2001;21(2):155–64.
37. Vande Walle J, Mauel R, Raes A, Vandekerckhove K, 52. Feinstein EI, Kaptein EM, Nicoloff JT, Massry
Donckerwolcke R. ARF in children with minimal SG. Thyroid function in patients with nephrotic syn-
change nephrotic syndrome may be related to func- drome and normal renal function. Am J Nephrol.
tional changes of the glomerular basal membrane. Am 1982;2(2):70–6.
J Kidney Dis. 2004;43(3):399–404. 53. Garin EH, Grant MB, Silverstein JH. Insulinlike
38. Cavagnaro F, Lagomarsino E. Peritonitis as a risk growth factors in patients with active nephrotic syn-
factor of acute renal failure in nephrotic children. drome. Am J Dis Child. 1989;143(7):865–7.
Pediatr Nephrol. 2000;15(3–4):248–51. 54. Southwest Pediatric Nephrology Study Group, Focal
39. Bohman SO, Jaremko G, Bohlin AB, Berg U. Foot segmental glomerulosclerosis in children with idio-
process fusion and glomerular filtration rate in pathic nephrotic syndrome. A report of the Southwest
minimal change nephrotic syndrome. Kidney Int. Pediatric Nephrology Study Group. Kidney Int.
1984;25(4):696–700. 1985;27(2):442–9.
40. Sakarcan A, Timmons C, Seikaly MG. Reversible 55. Paik KH, Lee BH, Cho HY, Kang HG, Ha IS, Cheong
idiopathic acute renal failure in children with primary HI, et al. Primary focal segmental glomerular sclero-
nephrotic syndrome. J Pediatr. 1994;125(5 Pt 1): sis in children: clinical course and prognosis. Pediatr
723–7. Nephrol. 2007;22(3):389–95.
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 871

56. Gipson DS, Chin H, Presler TP, Jennette C, Ferris 71. Muda AO, Feriozzi S, Cinotti GA, Faraggiana T.
ME, Massengill S, et al. Differential risk of remission Glomerular hypertrophy and chronic renal failure in
and ESRD in childhood FSGS. Pediatr Nephrol. focal segmental glomerulosclerosis. Am J Kidney
2006;21(3):344–9. Dis. 1994;23(2):237–41.
57. Ingulli E, Tejani A. Racial differences in the incidence 72. Jenis EH, Teichman S, Briggs WA, Sandler P,
and renal outcome of idiopathic focal segmental Hollerman CE, Calcagno PL, et al. Focal segmental
glomerulosclerosis in children. Pediatr Nephrol. glomerulosclerosis. Am J Med. 1974;57(5):695–705.
1991;5(4):393–7. 73. Erkan E, Garcia CD, Patterson LT, Mishra J,
58. Churg J, Habib R, White RH. Pathology of the Mitsnefes MM, Kaskel FJ, et al. Induction of renal
nephrotic syndrome in children: a report for the Inter- tubular cell apoptosis in focal segmental glomerulo-
national Study of Kidney Disease in Children. Lancet. sclerosis: roles of proteinuria and Fas-dependent path-
1970;760(1):1299–302. ways. J Am Soc Nephrol. 2005;16(2):398–407.
59. Powell HR. Relationship between proteinuria and 74. Hyman LR, Burkholder PM. Focal sclerosing
epithelial cell changes in minimal lesion glomerulonephropathy with segmental hyalinosis. A
glomerulopathy. Nephron. 1976;16(4):310–7. clinicopathologic analysis. Lab Invest. 1973;28(5):
60. Kawano K, Wenzl J, McCoy J, Porch J, Kimmelstiel 533–44.
P. Lipoid nephrosis. A multifold blind study, includ- 75. Nagi AH, Alexander F, Lannigan R. Light and elec-
ing quantitation. Lab Invest. 1971;24(6):499–503. tron microscopical studies of focal glomerular sclero-
61. Waldherr R, Gubler MC, Levy M, Broyer M, Habib sis. J Clin Pathol. 1971;24(9):846–50.
R. The significance of pure diffuse mesangial prolif- 76. Grishman E, Churg J. Focal glomerular sclerosis in
eration in idiopathic nephrotic syndrome. Clin nephrotic patients: an electron microscopic study of
Nephrol. 1978;10(5):171–9. glomerular podocytes. Kidney Int. 1975;7(2):111–22.
62. White RH, Glasgow EF, Mills RJ. Clinicopathologi- 77. D’Agati VD, Fogo AB, Bruijn JA, Jennette JC.
cal study of nephrotic syndrome in childhood. Lancet. Pathologic classification of focal segmental
1970;1(7661):1353–9. glomerulosclerosis: a working proposal. Am J Kidney
63. Habib R, Gubler MC. Focal sclerosing glomerulone- Dis. 2004;43(2):368–82.
phritis. Perspect Nephrol Hypertens. 1973;1(Pt 1): 78. Stokes MB, Markowitz GS, Lin J, Valeri AM,
263–78. D’Agati VD. Glomerular tip lesion: a distinct entity
64. Morita M, White RH, Coad NA, Raafat F. The clinical within the minimal change disease/focal segmental
significance of the glomerular location of segmental glomerulosclerosis spectrum. Kidney Int. 2004;65
lesions in focal segmental glomerulosclerosis. Clin (5):1690–702.
Nephrol. 1990;33(5):211–9. 79. Abeyagunawardena AS, Sebire NJ, Risdon RA, Dil-
65. Howie AJ, Brewer DB. The glomerular tip lesion: a lon MJ, Rees L, Van’t Hoff W, et al. Predictors of
previously undescribed type of segmental glomerular long-term outcome of children with idiopathic focal
abnormality. J Pathol. 1984;142(3):205–20. segmental glomerulosclerosis. Pediatr Nephrol.
66. Ito H, Yoshikawa N, Aozai F, Hazikano H, 2007;22(2):215–21.
Sakaguchi H, Akamatsu R, et al. Twenty-seven chil- 80. Detwiler RK, Falk RJ, Hogan SL, Jennette JC.
dren with focal segmental glomerulosclerosis: corre- Collapsing glomerulopathy: a clinically and
lation between the segmental location of the pathologically distinct variant of focal
glomerular lesions and prognosis. Clin Nephrol. segmental glomerulosclerosis. Kidney Int. 1994;45
1984;22(1):9–14. (5):1416–24.
67. Deegens JK, Steenbergen EJ, Borm GF, Wetzels JF. 81. Toth CM, Pascual M, Williams Jr WW, Delmonico
Pathological variants of focal segmental glomerulo- FL, Cosimi AB, Colvin RB, et al. Recurrent collaps-
sclerosis in an adult Dutch population–epidemiology ing glomerulopathy. Transplantation. 1998;65(7):
and outcome. Nephrol Dial Transplant. 2008;23(1): 1009–10.
186–92. 82. Valeri A, Barisoni L, Appel GB, Seigle R, D’Agati
68. Huppes W, Hene RJ, Kooiker CJ. The glomerular tip V. Idiopathic collapsing focal segmental glomerulo-
lesion: a distinct entity or not? J Pathol. 1988;154(2): sclerosis: a clinicopathologic study. Kidney Int.
187–90. 1996;50(5):1734–46.
69. Thomas DB, Franceschini N, Hogan SL, Ten 83. Barisoni L, Kriz W, Mundel P, D’Agati V. The
Holder S, Jennette CE, Falk RJ, et al. Clinical and dysregulated podocyte phenotype: a novel
pathologic characteristics of focal segmental concept in the pathogenesis of collapsing idiopathic
glomerulosclerosis pathologic variants. Kidney Int. focal segmental glomerulosclerosis and
2006;69(5):920–6. HIV-associated nephropathy. J Am Soc Nephrol.
70. Fogo A, Hawkins EP, Berry PL, Glick AD, Chiang 1999;10(1):51–61.
ML, MacDonell Jr RC, et al. Glomerular hypertrophy 84. Nash MA, Greifer I, Olbing H, Bernstein J,
in minimal change disease predicts subsequent pro- Bennett B, Spitzer A. The significance of focal scle-
gression to focal glomerular sclerosis. Kidney Int. rotic lesions of glomeruli in children. J Pediatr.
1990;38(1):115–23. 1976;88(5):806–13.
872 P. Niaudet and O. Boyer

85. Couser WG, Stilmant MM. Mesangial lesions and recurrent focal segmental glomerulosclerosis. N
focal glomerular sclerosis in the aging rat. Lab Invest. Engl J Med. 1996;334(14):878–83.
1975;33(5):491–501. 101. Ritz E. Pathogenesis of “idiopathic” nephrotic syn-
86. Michael AF, McLean RH, Roy LP, Westberg NG, drome. N Engl J Med. 1994;330(1):61–2.
Hoyer JR, Fish AJ, et al. Immunologic aspects of 102. Ali AA, Wilson E, Moorhead JF, Amlot P, Abdulla A,
the nephrotic syndrome. Kidney Int. 1973;3(2): Fernando ON, et al. Minimal-change glomerular
105–15. nephritis. Normal kidneys in an abnormal environ-
87. Cohen AH, Border WA, Glassock RJ. Nehprotic syn- ment? Transplantation. 1994;58(7):849–52.
drome with glomerular mesangial IgM deposits. Lab 103. Hoyer JR, Vernier RL, Najarian JS, Raij L, Simmons
Invest. 1978;38(5):610–9. RL, Michael AF. Recurrence of idiopathic nephrotic
88. Habib R, Girardin E, Gagnadoux MF, Hinglais N, syndrome after renal transplantation. Lancet.
Levy M, Broyer M. Immunopathological findings in 1972;2(7773):343–8.
idiopathic nephrosis: clinical significance of glomer- 104. Dantal J, Godfrin Y, Koll R, Perretto S, Naulet J,
ular “immune deposits”. Pediatr Nephrol. 1988;2(4): Bouhours JF, et al. Antihuman immunoglobulin affin-
402–8. ity immunoadsorption strongly decreases proteinuria
89. Southwest Pediatric Nephrology Study Group, Asso- in patients with relapsing nephrotic syndrome. J Am
ciation of IgA nephropathy with steroid-responsive Soc Nephrol. 1998;9(9):1709–15.
nephrotic syndrome. A report of the Southwest Pedi- 105. Zimmerman SW. Increased urinary protein excretion
atric Nephrology Study Group. Am J Kidney Dis. in the rat produced by serum from a patient with
1985;5(3):157–64. recurrent focal glomerular sclerosis after renal trans-
90. Lai KN, Lai FM, Chan KW, Ho CP, Leung AC, plantation. Clin Nephrol. 1984;22(1):32–8.
Vallance-Owen J. An overlapping syndrome of IgA 106. Le Berre L, Godfrin Y, Gunther E, Buzelin F,
nephropathy and lipoid nephrosis. Am J Clin Pathol. Perretto S, Smit H, et al. Extrarenal effects on the
1986;86(6):716–23. pathogenesis and relapse of idiopathic nephrotic
91. Jennette JC, Hipp CG. C1q nephropathy: a distinct syndrome in Buffalo/Mna rats. J Clin Invest.
pathologic entity usually causing nephrotic syn- 2002;109(4):491–8.
drome. Am J Kidney Dis. 1985;6(2):103–10. 107. Lagrue G, Niaudet P, Guillot F, Lang P. Pregnancy
92. Kersnik Levart T, Kenda RB, Avgustin Cavic M, and glomerulonephritis. Lancet. 1989;2(8670):1037.
Ferluga D, Hvala A, Vizjak A. C1Q nephropathy in 108. Kemper MJ, Wolf G, Muller-Wiefel DE. Transmis-
children. Pediatr Nephrol. 2005;20(12):1756–61. sion of glomerular permeability factor from a mother
93. Markowitz GS, Schwimmer JA, Stokes MB, Nasr S, to her child. N Engl J Med. 2001;344(5):386–7.
Seigle RL, Valeri AM, et al. C1q nephropathy: a 109. Lagrue G, Branellec A, Blanc C, Xheneumont S,
variant of focal segmental glomerulosclerosis. Kid- Beaudoux F, Sobel A, et al. A vascular permeability
ney Int. 2003;64(4):1232–40. factor in lymphocyte culture supernants from patients
94. Mii A, Shimizu A, Masuda Y, Fujita E, Aki K, with nephrotic syndrome. II. Pharmacological and
Ishizaki M, et al. Current status and issues of C1q physicochemical properties. Biomedicine/[publiee
nephropathy. Clin Exp Nephrol. 2009;13(4):263–74. pour l’AAICIG]. 1975;23(2):73–5.
95. Carrie BJ, Salyer WR, Myers BD. Minimal change 110. Grimbert P, Valanciute A, Audard V, Pawlak A, Le
nephropathy: an electrochemical disorder of the glo- gouvelo S, Lang P, et al. Truncation of C-mip
merular membrane. Am J Med. 1981;70(2):262–8. (Tc-mip), a new proximal signaling protein, induces
96. Kitano Y, Yoshikawa N, Nakamura H. Glomerular c-maf Th2 transcription factor and cytoskeleton reor-
anionic sites in minimal change nephrotic syndrome ganization. J Exp Med. 2003;198(5):797–807.
and focal segmental glomerulosclerosis. Clin 111. Mansour H, Cheval L, Elalouf JM, Aude JC,
Nephrol. 1993;40(4):199–204. Alyanakian MA, Mougenot B, et al. T-cell
97. van den Born J, van den Heuvel LP, Bakker MA, transcriptome analysis points up a thymic disorder
Veerkamp JH, Assmann KJ, Berden JH. A monoclo- in idiopathic nephrotic syndrome. Kidney Int.
nal antibody against GBM heparan sulfate induces an 2005;67(6):2168–77.
acute selective proteinuria in rats. Kidney Int. 112. Van Den Berg JG, Aten J, Chand MA, Claessen N,
1992;41(1):115–23. Dijkink L, Wijdenes J, et al. Interleukin-4 and
98. Guasch A, Deen WM, Myers BD. Charge selectivity interleukin-13 act on glomerular visceral epithelial
of the glomerular filtration barrier in healthy and cells. J Am Soc Nephrol. 2000;11(3):413–22.
nephrotic humans. J Clin Invest. 1993;92(5): 113. Yap HK, Cheung W, Murugasu B, Sim SK, Seah CC,
2274–82. Jordan SC. Th1 and Th2 cytokine mRNA profiles in
99. Shalhoub RJ. Pathogenesis of lipoid nephrosis: a dis- childhood nephrotic syndrome: evidence for
order of T-cell function. Lancet. 1974;2(7880): increased IL-13 mRNA expression in relapse. J Am
556–60. Soc Nephrol. 1999;10(3):529–37.
100. Savin VJ, Sharma R, Sharma M, McCarthy ET, Swan 114. Valanciute A, le Gouvello S, Solhonne B, Pawlak A,
SK, Ellis E, et al. Circulating factor associated with Grimbert P, Lyonnet L, et al. NF-kappa B p65 antag-
increased glomerular permeability to albumin in onizes IL-4 induction by c-maf in minimal change
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 873

nephrotic syndrome. J Immunol. 2004;172(1): 128. Meijers B, Maas RJ, Sprangers B, Claes K, Poesen R,
688–98. Bammens B, et al. The soluble urokinase receptor is
115. Sahali D, Pawlak A, Le Gouvello S, Lang P, not a clinical marker for focal segmental glomerulo-
Valanciute A, Remy P, et al. Transcriptional and sclerosis. Kidney Int. 2014;85(3):636–40.
post-transcriptional alterations of IkappaBalpha in 129. Sinha A, Bajpai J, Saini S, Bhatia D, Gupta A,
active minimal-change nephrotic syndrome. J Am Puraswani M, et al. Serum-soluble urokinase receptor
Soc Nephrol. 2001;12(8):1648–58. levels do not distinguish focal segmental glomerulo-
116. Sahali D, Pawlak A, Valanciute A, Grimbert P, sclerosis from other causes of nephrotic syndrome in
Lang P, Remy P, et al. A novel approach to investiga- children. Kidney Int. 2014;85(3):649–58.
tion of the pathogenesis of active minimal-change 130. Wada T, Nangaku M, Maruyama S, Imai E, Shoji K,
nephrotic syndrome using subtracted cDNA library Kato S, et al. A multicenter cross-sectional study of
screening. J Am Soc Nephrol. 2002;13(5):1238–47. circulating soluble urokinase receptor in Japanese
117. McCarthy ET, Sharma M, Savin VJ. Circulating per- patients with glomerular disease. Kidney Int.
meability factors in idiopathic nephrotic syndrome 2014;85(3):641–8.
and focal segmental glomerulosclerosis. Clin J Am 131. Deegens JK, Wetzels JF. Glomerular disease: the
Soc Nephrol. 2010;5(11):2115–21. search goes on: suPAR is not the elusive FSGS factor.
118. Cheung PK, Klok PA, Baller JF, Bakker WW. Induc- Nat Rev Nephrol. 2014;10(8):431–2.
tion of experimental proteinuria in vivo following 132. Grimbert P, Audard V, Remy P, Lang P, Sahali
infusion of human plasma hemopexin. Kidney Int. D. Recent approaches to the pathogenesis of
2000;57(4):1512–20. minimal-change nephrotic syndrome. Nephrol Dial
119. Bakker WW, Borghuis T, Harmsen MC, van den Transplant. 2003;18(2):245–8.
Berg A, Kema IP, Niezen KE, et al. Protease activity 133. Kemper MJ, Meyer-Jark T, Lilova M, Muller-Wiefel
of plasma hemopexin. Kidney Int. 2005;68(2): DE. Combined T- and B-cell activation in childhood
603–10. steroid-sensitive nephrotic syndrome. Clin Nephrol.
120. Bakker WW, van Dael CM, Pierik LJ, van Wijk JA, 2003;60(4):242–7.
Nauta J, Borghuis T, et al. Altered activity of plasma 134. Guigonis V, Dallocchio A, Baudouin V, Dehennault
hemopexin in patients with minimal change disease in M, Hachon-Le Camus C, Afanetti M, et al. Rituximab
relapse. Pediatr Nephrol. 2005;20(10):1410–5. treatment for severe steroid- or cyclosporine-
121. Lennon R, Singh A, Welsh GI, Coward RJ, Satchell S, dependent nephrotic syndrome: a multicentric series
Ni L, et al. Hemopexin induces nephrin-dependent of 22 cases. Pediatr Nephrol. 2008;23(8):1269–79.
reorganization of the actin cytoskeleton in podocytes. 135. Ravani P, Magnasco A, Edefonti A, Murer L, Rossi R,
J Am Soc Nephrol. 2008;19(11):2140–9. Ghio L, et al. Short-term effects of rituximab in chil-
122. Bock ME, Price HE, Gallon L, Langman CB. Serum dren with steroid- and calcineurin-dependent
soluble urokinase-type plasminogen activator recep- nephrotic syndrome: a randomized controlled trial.
tor levels and idiopathic FSGS in children: a single- Clin J Am Soc Nephrol. 2011;6:1308–15.
center report. Clin J Am Soc Nephrol. 2013;8(8): 136. Sellier-Leclerc AL, Macher MA, Loirat C, Guerin V,
1304–11. Watier H, Peuchmaur M, et al. Rituximab efficiency
123. Cathelin D, Placier S, Ploug M, Verpont MC, in children with steroid-dependent nephrotic syn-
Vandermeersch S, Luque Y, et al. Administration of drome. Pediatr Nephrol. 2010;25(6):1109–15.
recombinant soluble urokinase receptor per se is not 137. Haffner D, Fischer DC. Nephrotic syndrome and
sufficient to induce podocyte alterations and protein- rituximab: facts and perspectives. Pediatr Nephrol.
uria in mice. J Am Soc Nephrol. 2014;25:1662–8. 2009;24(8):1433–8.
124. Sier CF, Stephens R, Bizik J, Mariani A, Bassan M, 138. Fornoni A, Sageshima J, Wei C, Merscher-Gomez S,
Pedersen N, et al. The level of urokinase-type plas- Aguillon-Prada R, Jauregui AN, et al. Rituximab tar-
minogen activator receptor is increased in serum of gets podocytes in recurrent focal segmental
ovarian cancer patients. Cancer Res. 1998;58(9): glomerulosclerosis. Sci Transl Med. 2011;3(85):
1843–9. 85ra46.
125. Sidenius N, Sier CF, Ullum H, Pedersen BK, Lepri 139. Sellier-Leclerc AL, Duval A, Riveron S, Macher MA,
AC, Blasi F, et al. Serum level of soluble urokinase- Deschenes G, Loirat C, et al. A humanized mouse
type plasminogen activator receptor is a strong and model of idiopathic nephrotic syndrome suggests a
independent predictor of survival in human immuno- pathogenic role for immature cells. J Am Soc
deficiency virus infection. Blood. 2000;96(13): Nephrol. 2007;18(10):2732–9.
4091–5. 140. Garin EH, Diaz LN, Mu W, Wasserfall C, Araya C,
126. Maas RJ, Deegens JK, Wetzels JF. Serum suPAR in Segal M, et al. Urinary CD80 excretion increases in
patients with FSGS: trash or treasure? Pediatr idiopathic minimal-change disease. J Am Soc
Nephrol. 2013;28:1041–8. Nephrol. 2009;20(2):260–6.
127. Sever S, Trachtman H, Wei C, Reiser J. Is there 141. Teeninga N, Kist-van Holthe JE, van Rijswijk N, de
clinical value in measuring suPAR levels in FSGS? Mos NI, Hop WC, Wetzels JF, et al. Extending pred-
Clin J Am Soc Nephrol. 2013;8(8):1273–5. nisolone treatment does not reduce relapses in
874 P. Niaudet and O. Boyer

childhood nephrotic syndrome. J Am Soc Nephrol. pediatric nephrotic syndrome. Ann Pharmacother.
2013;24(1):149–59. 2004;38(12):2105–14.
142. Reiser J, von Gersdorff G, Loos M, Oh J, Asanuma K, 156. Bak M, Serdaroglu E, Guclu R. Prophylactic calcium
Giardino L, et al. Induction of B7-1 in podocytes is and vitamin D treatments in steroid-treated children
associated with nephrotic syndrome. J Clin Invest. with nephrotic syndrome. Pediatr Nephrol.
2004;113(10):1390–7. 2006;21(3):350–4.
143. Audard V, Zhang SY, Copie-Bergman C, Rucker- 157. Woroniecki RP, Orlova TN, Mendelev N, Shatat IF,
Martin C, Ory V, Candelier M, et al. Occurrence of Hailpern SM, Kaskel FJ, et al. Urinary proteome of
minimal change nephrotic syndrome in classical steroid-sensitive and steroid-resistant idiopathic
Hodgkin lymphoma is closely related to the induction nephrotic syndrome of childhood. Am J Nephrol.
of c-mip in Hodgkin-Reed Sternberg cells and 2006;26(3):258–67.
podocytes. Blood. 2010;115(18):3756–62. 158. Trainin EB, Boichis H, Spitzer A, Edelmann Jr CM,
144. Zhang SY, Kamal M, Dahan K, Pawlak A, Ory V, Greifer I. Late nonresponsiveness to steroids in
Desvaux D, et al. c-mip impairs podocyte proximal children with the nephrotic syndrome. J Pediatr.
signaling and induces heavy proteinuria. Sci Signal. 1975;87(4):519–23.
2010;3(122):ra39. 159. Tune BM, Mendoza SA. Treatment of the idiopathic
145. Cara-Fuentes G, Johnson RJ, Reiser J, Garin nephrotic syndrome: regimens and outcomes in chil-
EH. CD80 and suPAR in patients with minimal dren and adults. J Am Soc Nephrol. 1997;8(5):
change disease and focal segmental glomerulo- 824–32.
sclerosis: diagnostic and pathogenic significance: 160. McEnery PT, Strife CF. Nephrotic syndrome in child-
response. Pediatr Nephrol. 2014;29(8):1467–8. hood. Management and treatment in patients with
146. Doucet A, Favre G, Deschenes G. Molecular mecha- minimal change disease, mesangial proliferation, or
nism of edema formation in nephrotic syndrome: focal glomerulosclerosis. Pediatr Clin North
therapeutic implications. Pediatr Nephrol. Am. 1982;29(4):875–94.
2007;22(12):1983–90. 161. International Study of Kidney Disease in Children,
147. Haws RM, Baum M. Efficacy of albumin and diuretic The primary nephrotic syndrome in children. Identi-
therapy in children with nephrotic syndrome. Pediat- fication of patients with minimal change nephrotic
rics. 1993;91(6):1142–6. syndrome from initial response to prednisone. A
148. Rascher W, Tulassay T, Seyberth HW, Himbert U, report of the International Study of Kidney Disease
Lang U, Scharer K. Diuretic and hormonal responses in Children. J Pediatr. 1981;98(4):561–4.
to head-out water immersion in nephrotic syndrome. 162. Arbetsgemeinschaft fur Padiatrische Nephrologie,
J Pediatr. 1986;109(4):609–14. Alternate-day versus intermittent prednisone in fre-
149. Taylor B, Andrews N, Stowe J, Hamidi-Manesh L, quently relapsing nephrotic syndrome. A report of
Miller E. No increased risk of relapse after meningo- “Arbetsgemeinschaft fur Padiatrische Nephrologie”.
coccal C conjugate vaccine in nephrotic syndrome. Lancet. 1979;1(8113):401–3.
Arch Dis Child. 2007;92(10):887–9. 163. Murnaghan K, Vasmant D, Bensman A. Pulse
150. Ulinski T, Leroy S, Dubrel M, Danon S, Bensman methylprednisolone therapy in severe idiopathic
A. High serological response to pneumococcal childhood nephrotic syndrome. Acta Paediatr Scand.
vaccine in nephrotic children at disease onset on 1984;73(6):733–9.
high-dose prednisone. Pediatr Nephrol. 2008;23(7): 164. Lombel RM, Gipson DS, Hodson EM. Treatment of
1107–13. steroid-sensitive nephrotic syndrome: new guidelines
151. Alpay H, Yildiz N, Onar A, Temizer H, Ozcay S. from KDIGO. Pediatr Nephrol 2013;28:415–26.
Varicella vaccination in children with steroid- 165. Ehrich JH, Brodehl J. Long versus standard predni-
sensitive nephrotic syndrome. Pediatr Nephrol. sone therapy for initial treatment of idiopathic
2002;17(3):181–3. nephrotic syndrome in children. Arbeitsgemeinschaft
152. Furth SL, Arbus GS, Hogg R, Tarver J, Chan C, fur Padiatrische Nephrologie. Eur J Pediatr.
Fivush BA. Varicella vaccination in children with 1993;152(4):357–61.
nephrotic syndrome: a report of the Southwest 166. Bagga A, Hari P, Srivastava RN. Prolonged versus
Pediatric Nephrology Study Group. J Pediatr. standard prednisolone therapy for initial episode of
2003;142(2):145–8. nephrotic syndrome. Pediatr Nephrol. 1999;13(9):
153. Elisaf M, Dardamanis M, Pappas M, Sferopoulos G, 824–7.
Siamopoulos KC. Treatment of nephrotic 167. Ksiazek J, Wyszynska T. Short versus long initial
hyperlipidemia with lovastatin. Clin Nephrol. prednisone treatment in steroid-sensitive nephrotic
1991;36(1):50–2. syndrome in children. Acta Paediatr. 1995;84(8):
154. Wheeler DC, Bernard DB. Lipid abnormalities in the 889–93.
nephrotic syndrome: causes, consequences, and treat- 168. Norero C, Delucchi A, Lagos E, Rosati P. Initial ther-
ment. Am J Kidney Dis. 1994;23(3):331–46. apy of primary nephrotic syndrome in children: eval-
155. Prescott Jr WA, Streetman DA, Streetman DS. The uation in a period of 18 months of two prednisone
potential role of HMG-CoA reductase inhibitors in treatment schedules. Chilean Co-operative Group of
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 875

Study of Nephrotic Syndrome in Children. Rev Med 182. Gulati A, Sinha A, Sreenivas V, Math A, Hari P,
Chil. 1996;124(5):567–72. Bagga A. Daily corticosteroids reduce infection-
169. Ueda N, Chihara M, Kawaguchi S, Niinomi Y, associated relapses in frequently relapsing nephrotic
Nonoda T, Matsumoto J, et al. Intermittent versus syndrome: a randomized controlled trial. Clin J Am
long-term tapering prednisolone for initial therapy in Soc Nephrol. 2011;6(1):63–9.
children with idiopathic nephrotic syndrome. J 183. Mattoo TK, Mahmoud MA. Increased maintenance
Pediatr. 1988;112(1):122–6. corticosteroids during upper respiratory infection
170. Hodson EM, Knight JF, Willis NS, Craig JC. Corti- decrease the risk of relapse in nephrotic syndrome.
costeroid therapy for nephrotic syndrome in children. Nephron. 2000;85(4):343–5.
Cochrane Database Syst Rev. 2005;1:CD001533. 184. Polito C, Oporto MR, Totino SF, La Manna A, Di
171. Yoshikawa N, Nakanishi K, Sako M, Oba MS, Toro R. Normal growth of nephrotic children during
Mori R, Ota E, et al. A multicenter randomized trial long-term alternate-day prednisone therapy. Acta
indicates initial prednisolone treatment for childhood Paediatr Scand. 1986;75(2):245–50.
nephrotic syndrome for two months is not inferior to 185. Donatti TL, Koch VH, Fujimura MD, Okay
six-month treatment. Kidney Int. 2014; doi:10.1038/ Y. Growth in steroid-responsive nephrotic syndrome:
ki.2014.260. a study of 85 pediatric patients. Pediatr Nephrol.
172. Sinha A, Saha A, Kumar M, Sharma S, Afzal K, 2003;18(8):789–95.
Mehta A, et al. Extending initial prednisolone treat- 186. Emma F, Sesto A, Rizzoni G. Long-term linear
ment in a randomized control trial from 3 to 6 months growth of children with severe steroid-responsive
did not significantly influence the course of illness in nephrotic syndrome. Pediatr Nephrol. 2003;18(8):
children with steroid-sensitive nephrotic syndrome. 783–8.
Kidney Int. 2014; doi:10.1038/ki.2014.240. 187. Fakhouri F, Bocquet N, Taupin P, Presne C,
173. Hodson EM, Willis NS, Craig JC. Corticosteroid ther- Gagnadoux MF, Landais P, et al. Steroid-sensitive
apy for nephrotic syndrome in children. Cochrane nephrotic syndrome: from childhood to adulthood.
Database Syst Rev. 2007;4:CD001533. Am J Kidney Dis. 2003;41(3):550–7.
174. Leisti S, Koskimies O. Risk of relapse in steroid- 188. Leonard MB, Feldman HI, Shults J, Zemel BS, Foster
sensitive nephrotic syndrome: effect of stage of BJ, Stallings VA. Long-term, high-dose glucocorti-
post-prednisone adrenocortical suppression. J Pediatr. coids and bone mineral content in childhood
1983;103(4):553–7. glucocorticoid-sensitive nephrotic syndrome. N Engl
175. Schoeneman MJ. Minimal change nephrotic syn- J Med. 2004;351(9):868–75.
drome: treatment with low doses of hydrocortisone. 189. Biyikli NK, Emre S, Sirin A, Bilge I. Biochemical
J Pediatr. 1983;102(5):791–3. bone markers in nephrotic children. Pediatr Nephrol.
176. Abeyagunawardena AS, Hindmarsh P, Trompeter 2004;19(8):869–73.
RS. Adrenocortical suppression increases the risk of 190. Kim SD, Cho BS. Pamidronate therapy for preventing
relapse in nephrotic syndrome. Arch Dis Child. steroid-induced osteoporosis in children with
2007;92(7):585–8. nephropathy. Nephron Clin Pract. 2006;102(3–4):
177. Hoyer PF, Brodeh J. Initial treatment of idiopathic c81–7.
nephrotic syndrome in children: prednisone versus 191. Tanphaichitr P, Tanphaichitr D, Sureeratanan J,
prednisone plus cyclosporine A: a prospective, ran- Chatasingh S. Treatment of nephrotic syndrome
domized trial. J Am Soc Nephrol. 2006;17(4): with levamisole. J Pediatr. 1980;96(3 Pt 1):490–3.
1151–7. 192. Boyer O, Moulder JK, Grandin L, Somers MJ. Short-
178. Teeninga N, Kist-van Holthe JE, van den Akker EL, and long-term efficacy of levamisole as adjunctive
Kersten MC, Boersma E, Krabbe HG, et al. Genetic therapy in childhood nephrotic syndrome. Pediatr
and in vivo determinants of glucocorticoid sensitivity Nephrol. 2008;23(4):575–80.
in relation to clinical outcome of childhood nephrotic 193. Drachman R, Schlesinger M, Alon U, Mor J,
syndrome. Kidney Int. 2014;85:1444–53. Etzioni A, Shapira H, et al. Immunoregulation with
179. Broyer M, Terzi F, Lehnert A, Gagnadoux MF, levamisole in children with frequently relapsing ste-
Guest G, Niaudet P. A controlled study of deflazacort roid responsive nephrotic syndrome. Acta Paediatr
in the treatment of idiopathic nephrotic syndrome. Scand. 1988;77(5):721–6.
Pediatr Nephrol. 1997;11(4):418–22. 194. Mehta KP, Ali U, Kutty M, Kolhatkar U. Immuno-
180. MacDonald NE, Wolfish N, McLaine P, Phipps P, regulatory treatment for minimal change nephrotic
Rossier E. Role of respiratory viruses in exacerbations syndrome. Arch Dis Child. 1986;61(2):153–8.
of primary nephrotic syndrome. J Pediatr. 1986;108 195. Mongeau JG, Robitaille PO, Roy F. Clinical efficacy
(3):378–82. of levamisole in the treatment of primary nephrosis in
181. Abeyagunawardena AS, Trompeter RS. Increasing children. Pediatr Nephrol. 1988;2(4):398–401.
the dose of prednisolone during viral infections 196. Niaudet P, Drachman R, Gagnadoux MF,
reduces the risk of relapse in nephrotic syndrome: Broyer M. Treatment of idiopathic nephrotic syn-
a randomised controlled trial. Arch Dis Child. drome with levamisole. Acta Paediatr Scand.
2008;93(3):226–8. 1984;73(5):637–41.
876 P. Niaudet and O. Boyer

197. British Association for Paediatric Nephrology. Padiatrische Nephrologie. Arch Dis Child. 1987;
Levamisole for corticosteroid-dependent nephrotic 62(11):1102–6.
syndrome in childhood. Lancet. 1991;337(8757): 212. Ueda N, Kuno K, Ito S. Eight and 12 week courses of
1555–7. cyclophosphamide in nephrotic syndrome. Arch Dis
198. Dayal U, Dayal AK, Shastry JC, Raghupathy P. Use Child. 1990;65(10):1147–50.
of levamisole in maintaining remission in steroid- 213. Kemper MJ, Altrogge H, Ludwig K, Timmermann K,
sensitive nephrotic syndrome in children. Nephron. Muller-Wiefel DE. Unfavorable response to cyclo-
1994;66(4):408–12. phosphamide in steroid-dependent nephrotic syn-
199. Al-Saran K, Mirza K, Al-Ghanam G, Abdelkarim drome. Pediatr Nephrol. 2000;14(8–9):772–5.
M. Experience with levamisole in frequently relaps- 214. Kyrieleis HA, Levtchenko EN, Wetzels JF. Long-
ing, steroid-dependent nephrotic syndrome. Pediatr term outcome after cyclophosphamide treatment in
Nephrol. 2006;21(2):201–5. children with steroid-dependent and frequently
200. Barbano G, Ginevri F, Ghiggeri GM, Gusmano relapsing minimal change nephrotic syndrome. Am J
R. Disseminated autoimmune disease during levami- Kidney Dis. 2007;49(5):592–7.
sole treatment of nephrotic syndrome. Pediatr 215. Cammas B, Harambat J, Bertholet-Thomas A, Bouissou
Nephrol. 1999;13(7):602–3. F, Morin D, Guigonis V, et al. Long-term effects of
201. Davin JC, Merkus MP. Levamisole in steroid- cyclophosphamide therapy in steroid-dependent or fre-
sensitive nephrotic syndrome of childhood: the lost quently relapsing idiopathic nephrotic syndrome.
paradise? Pediatr Nephrol. 2005;20(1):10–4. Nephrol Dial Transplant. 2011;26(1):178–84.
202. Palcoux JB, Niaudet P, Goumy P. Side effects of 216. Azib S, Macher MA, Kwon T, Dechartres A,
levamisole in children with nephrosis. Pediatr Alberti C, Loirat C, et al. Cyclophosphamide in
Nephrol. 1994;8(2):263–4. steroid-dependent nephrotic syndrome. Pediatr
203. Hodson EM, Willis NS, Craig JC. Non-corticosteroid Nephrol. 2011;26(6):927–32.
treatment for nephrotic syndrome in children. 217. Zagury A, de Oliveira AL, de Moraes CA, de Araujo
Cochrane Database Syst Rev. 2008;1:CD002290. Montalvao JA, Novaes RH, de Sa VM, et al. Long-
204. Cameron JS, Chantler C, Ogg CS, White RH. Long- term follow-up after cyclophosphamide therapy in
term stability of remission in nephrotic syndrome steroid-dependent nephrotic syndrome. Pediatr
after treatment with cyclophosphamide. Br Med Nephrol. 2011;26(6):915–20.
J. 1974;4(5935):7–11. 218. Arbeitsgemeinschaft fur Padiatrische Nephrologie,
205. Chiu J, Drummond KN. Long-term follow-up Effect of cytotoxic drugs in frequently relapsing
of cyclophosphamide therapy in frequent relapsing nephrotic syndrome with and without steroid depen-
minimal lesion nephrotic syndrome. J Pediatr. dence. N Engl J Med. 1982;306(8):451–4.
1974;84(6):825–30. 219. Gulati S, Pokhariyal S, Sharma RK, Elhence R,
206. McDonald J, Murphy AV, Arneil GC. Long-term Kher V, Pandey CM, et al. Pulse cyclophosphamide
assessment of cyclophosphamide therapy for nephro- therapy in frequently relapsing nephrotic syndrome.
sis in children. Lancet. 1974;2(7887):980–2. Nephrol Dial Transplant. 2001;16(10):2013–7.
207. Vester U, Kranz B, Zimmermann S, Buscher R, Hoyer 220. Donia AF, Gazareen SH, Ahmed HA, Moustafa FE,
PF. The response to cyclophosphamide in steroid- Shoeib AA, Ismail AM, et al. Pulse cyclophospha-
sensitive nephrotic syndrome is influenced by poly- mide inadequately suppresses reoccurrence of mini-
morphic expression of glutathion-S-transferases-M1 mal change nephrotic syndrome in corticoid-
and -P1. Pediatr Nephrol. 2005;20(4):478–81. dependent children. Nephrol Dial Transplant.
208. International study of Kidney Disease in Children, 2003;18(10):2054–8.
Prospective, controlled trial of cyclophosphamide 221. Prasad N, Gulati S, Sharma RK, Singh U, Ahmed M.
therapy in children with nephrotic syndrome. Report Pulse cyclophosphamide therapy in steroid-
of the International study of Kidney Disease in Chil- dependent nephrotic syndrome. Pediatr Nephrol.
dren. Lancet. 1974;2(7878):423–7. 2004;19(5):494–8.
209. Dundon S, O’Callaghan U, Raftery J. Stability of 222. Latta K, von Schnakenburg C, Ehrich JH. A meta-
remission in minimal lesion nephrotic syndrome analysis of cytotoxic treatment for frequently relaps-
after treatment with prednisolone and cyclophospha- ing nephrotic syndrome in children. Pediatr Nephrol.
mide. Int J Pediatr Nephrol. 1980;1(1):22–5. 2001;16(3):271–82.
210. Siegel NJ, Gaudio KM, Krassner LS, McDonald BM, 223. Hsu AC, Folami AO, Bain J, Rance CP. Gonadal
Anderson FP, Kashgarian M. Steroid-dependent function in males treated with cyclophosphamide for
nephrotic syndrome in children: histopathology and nephrotic syndrome. Fertil Steril. 1979;31(2):173–7.
relapses after cyclophosphamide treatment. Kidney 224. Penso J, Lippe B, Ehrlich R, Smith Jr FG. Testicular
Int. 1981;19(3):454–9. function in prepubertal and pubertal male patients
211. Arbeitsgemeinschaft fur Padiatrische Nephrologie, treated with cyclophosphamide for nephrotic syn-
Cyclophosphamide treatment of steroid dependent drome. J Pediatr. 1974;84(6):831–6.
nephrotic syndrome: comparison of eight week with 225. Trompeter RS, Evans PR, Barratt TM. Gonadal func-
12 week course. Report of Arbeitsgemeinschaft fur tion in boys with steroid-responsive nephrotic
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 877

syndrome treated with cyclophosphamide for short 239. Habib R, Niaudet P. Comparison between pre- and
periods. Lancet. 1981;1(8231):1177–9. posttreatment renal biopsies in children receiving
226. Watson AR, Taylor J, Rance CP, Bain J. Gonadal ciclosporine for idiopathic nephrosis. Clin Nephrol.
function in women treated with cyclophosphamide 1994;42(3):141–6.
for childhood nephrotic syndrome: a long-term fol- 240. Iijima K, Hamahira K, Tanaka R, Kobayashi A,
low-up study. Fertil Steril. 1986;46(2):331–3. Nozu K, Nakamura H, et al. Risk factors for
227. Baluarte HJ, Hiner L, Gruskin AB. Chlorambucil cyclosporine-induced tubulointerstitial lesions in
dosage in frequently relapsing nephrotic children with minimal change nephrotic syndrome.
syndrome: a controlled clinical trial. J Pediatr. Kidney Int. 2002;61(5):1801–5.
1978;92(2):295–8. 241. Meyrier A, Noel LH, Auriche P, Callard P. Long-term
228. Grupe WE, Makker SP, Ingelfinger JR. Chlorambucil renal tolerance of cyclosporin A treatment in adult
treatment of frequently relapsing nephrotic syndrome. idiopathic nephrotic syndrome. Collaborative Group
N Engl J Med. 1976;295(14):746–9. of the Societe de Nephrologie. Kidney Int. 1994;
229. Williams SA, Makker SP, Ingelfinger JR, Grupe WE. 45(5):1446–56.
Long-term evaluation of chlorambucil plus predni- 242. Freundlich M. Bone mineral content and mineral
sone in the idiopathic nephrotic syndrome of child- metabolism during cyclosporine treatment of
hood. N Engl J Med. 1980;302(17):929–33. nephrotic syndrome. J Pediatr. 2006;149(3):383–9.
230. Niaudet P, Habib R. Cyclosporine in the treatment of 243. Sinha MD, MacLeod R, Rigby E, Clark
idiopathic nephrosis. J Am Soc Nephrol. 1994;5(4): AG. Treatment of severe steroid-dependent nephrotic
1049–56. syndrome (SDNS) in children with tacrolimus.
231. Ishikura K, Yoshikawa N, Nakazato H, Sasaki S, Nephrol Dial Transplant. 2006;21(7):1848–54.
Iijima K, Nakanishi K, et al. Two-year follow-up of 244. Dittrich K, Knerr I, Rascher W, Dotsch J. Transient
a prospective clinical trial of cyclosporine for fre- insulin-dependent diabetes mellitus in children with
quently relapsing nephrotic syndrome in children. steroid-dependent idiopathic nephrotic syndrome dur-
Clin J Am Soc Nephrol. 2012;7(10):1576–83. ing tacrolimus treatment. Pediatr Nephrol. 2006;
232. Hulton SA, Neuhaus TJ, Dillon MJ, Barratt TM. 21(7):958–61.
Long-term cyclosporin A treatment of minimal- 245. Afzal K, Bagga A, Menon S, Hari P, Jordan SC.
change nephrotic syndrome of childhood. Pediatr Treatment with mycophenolate mofetil and predniso-
Nephrol. 1994;8(4):401–3. lone for steroid-dependent nephrotic syndrome.
233. Niaudet P. Comparison of cyclosporin and Pediatr Nephrol. 2007;22(12):2059–65.
chlorambucil in the treatment of steroid-dependent 246. Bagga A, Hari P, Moudgil A, Jordan SC.
idiopathic nephrotic syndrome: a multicentre random- Mycophenolate mofetil and prednisolone therapy in
ized controlled trial. The French Society of Paediatric children with steroid-dependent nephrotic syndrome.
Nephrology. Pediatr Nephrol. 1992;6(1):1–3. Am J Kidney Dis. 2003;42(6):1114–20.
234. Ponticelli C, Edefonti A, Ghio L, Rizzoni G, 247. Fujinaga S, Ohtomo Y, Umino D, Takemoto M,
Rinaldi S, Gusmano R, et al. Cyclosporin versus Shimizu T, Yamashiro Y, et al. A prospective study
cyclophosphamide for patients with steroid- on the use of mycophenolate mofetil in children with
dependent and frequently relapsing idiopathic cyclosporine-dependent nephrotic syndrome. Pediatr
nephrotic syndrome: a multicentre randomized con- Nephrol. 2007;22(1):71–6.
trolled trial. Nephrol Dial Transplant. 1993;8(12): 248. Gellermann J, Querfeld U. Frequently relapsing
1326–32. nephrotic syndrome: treatment with mycophenolate
235. Tejani A, Suthanthiran M, Pomrantz A. A randomized mofetil. Pediatr Nephrol. 2004;19(1):101–4.
controlled trial of low-dose prednisone and 249. Hogg RJ, Fitzgibbons L, Bruick J, Bunke M, Ault B,
ciclosporin versus high-dose prednisone in nephrotic Baqi N, et al. Mycophenolate mofetil in children with
syndrome of children. Nephron. 1991;59(1):96–9. frequently relapsing nephrotic syndrome: a report
236. Hirano T, Kawamura T, Fukuda S, Kohsaka S, from the Southwest Pediatric Nephrology Study
Yoshikawa N, Yoshida M, et al. Implication of cho- Group. Clin J Am Soc Nephrol. 2006;1(6):1173–8.
lesterol in cyclosporine pharmacodynamics in mini- 250. Mendizabal S, Zamora I, Berbel O, Sanahuja MJ,
mal change nephrotic syndrome. Clin Pharmacol Fuentes J, Simon J. Mycophenolate mofetil in
Ther. 2003;74(6):581–90. steroid/cyclosporine-dependent/resistant nephrotic
237. Ingulli E, Tejani A. Severe hypercholesterolemia syndrome. Pediatr Nephrol. 2005;20(7):914–9.
inhibits cyclosporin A efficacy in a dose-dependent 251. Novak I, Frank R, Vento S, Vergara M, Gauthier B,
manner in children with nephrotic syndrome. J Am Trachtman H. Efficacy of mycophenolate mofetil in
Soc Nephrol. 1992;3(2):254–9. pediatric patients with steroid-dependent nephrotic
238. Ishikura K, Ikeda M, Hattori S, Yoshikawa N, syndrome. Pediatr Nephrol. 2005;20(9):1265–8.
Sasaki S, Iijima K, et al. Effective and safe treatment 252. Barletta GM, Smoyer WE, Bunchman TE, Flynn JT,
with cyclosporine in nephrotic children: a prospec- Kershaw DB. Use of mycophenolate mofetil in
tive, randomized multicenter trial. Kidney Int. steroid-dependent and -resistant nephrotic syndrome.
2008;73(10):1167–73. Pediatr Nephrol. 2003;18(8):833–7.
878 P. Niaudet and O. Boyer

253. Dorresteijn EM, Kist-van Holthe JE, Levtchenko EN, multiple nodular granulomas after rituximab for
Nauta J, Hop WC, van der Heijden AJ. refractory nephrotic syndrome. Pediatr Nephrol.
Mycophenolate mofetil versus cyclosporine for 2013;28(1):145–9.
remission maintenance in nephrotic syndrome. 266. Ardelean DS, Gonska T, Wires S, Cutz E, Griffiths A,
Pediatr Nephrol. 2008;23(11):2013–20. Harvey E, et al. Severe ulcerative colitis after
254. Baudouin V, Alberti C, Lapeyraque AL, Bensman A, rituximab therapy. Pediatrics. 2010;126(1):e243–6.
Andre JL, Broux F, et al. Mycophenolate mofetil for 267. Lombel RM, Gipson DS, Hodson EM. Treatment of
steroid-dependent nephrotic syndrome: a phase II steroid-sensitive nephrotic syndrome: new guidelines
Bayesian trial. Pediatr Nephrol. 2012;27(3):389–96. from KDIGO. Pediatr Nephrol. 2013;28(3):415–26.
255. Ito S, Kamei K, Ogura M, Udagawa T, Fujinaga S, 268. Boyer O, Niaudet P. Nephrotic syndrome: rituximab
Saito M, et al. Survey of rituximab treatment for in childhood steroid-dependent nephrotic syndrome.
childhood-onset refractory nephrotic syndrome. Nat Rev Nephrol. 2013;9(10):562–3.
Pediatr Nephrol. 2013;28(2):257–64. 269. Razzak M. Nephrotic syndrome: rituximab is safe and
256. Ruggenenti P, Ruggiero B, Cravedi P, Vivarelli M, effective in FRNS and SDNS-but where to go from
Massella L, Marasa M, et al. Rituximab in steroid- here? Nat Rev Nephrol. 2014;10(8):421.
dependent or frequently relapsing idiopathic 270. Scharer K, Minges U. Long term prognosis of the
nephrotic syndrome. J Am Soc Nephrol. nephrotic syndrome in childhood. Clin Nephrol.
2014;25:850–63. 1973;1(3):182–7.
257. Sellier-Leclerc AL, Baudouin V, Kwon T, Macher 271. Trompeter RS, Lloyd BW, Hicks J, White RH,
MA, Guerin V, Lapillonne H, et al. Rituximab in Cameron JS. Long-term outcome for children with
steroid-dependent idiopathic nephrotic syndrome in minimal-change nephrotic syndrome. Lancet.
childhood–follow-up after CD19 recovery. Nephrol 1985;1(8425):368–70.
Dial Transplant. 2012;27(3):1083–9. 272. Wynn SR, Stickler GB, Burke EC. Long-term prog-
258. Iijima K, Sako M, Nozu K, Mori R, Tuchida N, nosis for children with nephrotic syndrome. Clin
Kamei K, et al. Rituximab for childhood-onset, com- Pediatr (Phila). 1988;27(2):63–8.
plicated, frequently relapsing nephrotic syndrome or 273. Lewis MA, Baildom EM, Davis N, Houston IB,
steroid-dependent nephrotic syndrome: a multicentre, Postlethwaite RJ. Nephrotic syndrome: from toddlers
double-blind, randomised, placebo-controlled trial. to twenties. Lancet. 1989;1(8632):255–9.
Lancet. 2014;384:1273–81. 274. Koskimies O, Vilska J, Rapola J, Hallman N. Long-
259. Ito S, Kamei K, Ogura M, Sato M, Fujimaru T, term outcome of primary nephrotic syndrome. Arch
Ishikawa T, et al. Maintenance therapy with Dis Child. 1982;57(7):544–8.
mycophenolate mofetil after rituximab in pediatric 275. Lahdenkari AT, Suvanto M, Kajantie E, Koskimies O,
patients with steroid-dependent nephrotic syndrome. Kestila M, Jalanko H. Clinical features and outcome
Pediatr Nephrol. 2011;26(10):1823–8. of childhood minimal change nephrotic syndrome: is
260. Fujinaga S, Someya T, Watanabe T, Ito A, Ohtomo Y, genetics involved? Pediatr Nephrol. 2005;20(8):
Shimizu T, et al. Cyclosporine versus mycophenolate 1073–80.
mofetil for maintenance of remission of steroid- 276. Ruth EM, Kemper MJ, Leumann EP, Laube GF,
dependent nephrotic syndrome after a single infusion Neuhaus TJ. Children with steroid-sensitive nephrotic
of rituximab. Eur J Pediatr. 2013;172(4):513–8. syndrome come of age: long-term outcome. J Pediatr.
261. Giangiacomo J, Cleary TG, Cole BR, Hoffsten P, 2005;147(2):202–7.
Robson AM. Serum immunoglobulins in the 277. Arbus GS, Poucell S, Bacheyie GS, Baumal R. Focal
nephrotic syndrome. A possible cause of minimal- segmental glomerulosclerosis with idiopathic
change nephrotic syndrome. N Engl J Med. nephrotic syndrome: three types of clinical response.
1975;293(1):8–12. J Pediatr. 1982;101(1):40–5.
262. Delbe-Bertin L, Aoun B, Tudorache E, Lapillone H, 278. Hammad A, Yahia S, Gouida MS, Bakr A,
Ulinski T. Does rituximab induce hypogammaglo- El-Farahaty RM. Low expression of glucocorticoid
bulinemia in patients with pediatric idiopathic receptors in children with steroid-resistant nephrotic
nephrotic syndrome? Pediatr Nephrol. 2013;28(3): syndrome. Pediatr Nephrol. 2013;28(5):759–63.
447–51. 279. Mendoza SA, Reznik VM, Griswold WR, Krensky
263. Chaumais MC, Garnier A, Chalard F, Peuchmaur M, AM, Yorgin PD, Tune BM. Treatment of steroid-
Dauger S, Jacqz-Agrain E, et al. Fatal pulmonary resistant focal segmental glomerulosclerosis with
fibrosis after rituximab administration. Pediatr pulse methylprednisolone and alkylating agents.
Nephrol. 2009;24(9):1753–5. Pediatr Nephrol. 1990;4(4):303–7.
264. Sellier-Leclerc AL, Belli E, Guerin V, Dorfmuller P, 280. Tune BM, Kirpekar R, Sibley RK, Reznik VM,
Deschenes G. Fulminant viral myocarditis after Griswold WR, Mendoza SA. Intravenous methyl-
rituximab therapy in pediatric nephrotic syndrome. prednisolone and oral alkylating agent therapy of
Pediatr Nephrol. 2013;28:1875–9. prednisone-resistant pediatric focal segmental
265. Sato M, Ito S, Ogura M, Kamei K, Miyairi I, Miyata I, glomerulosclerosis: a long-term follow-up. Clin
et al. Atypical Pneumocystis jiroveci pneumonia with Nephrol. 1995;43(2):84–8.
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 879

281. Pena A, Bravo J, Melgosa M, Fernandez C, 295. Loeffler K, Gowrishankar M, Yiu V. Tacrolimus ther-
Meseguer C, Espinosa L, et al. Steroid-resistant apy in pediatric patients with treatment-resistant
nephrotic syndrome: long-term evolution after nephrotic syndrome. Pediatr Nephrol. 2004;19(3):
sequential therapy. Pediatr Nephrol. 2007;22(11): 281–7.
1875–80. 296. Gulati A, Sinha A, Gupta A, Kanitkar M, Sreenivas V,
282. Yorgin PD, Krasher J, Al-Uzri AY. Pulse methylpred- Sharma J, et al. Treatment with tacrolimus and pred-
nisolone treatment of idiopathic steroid-resistant nisolone is preferable to intravenous cyclophospha-
nephrotic syndrome. Pediatr Nephrol. 2001;16(3): mide as the initial therapy for children with
245–50. steroid-resistant nephrotic syndrome. Kidney Int.
283. Hari P, Bagga A, Jindal N, Srivastava RN. Treatment 2012;82(10):1130–5.
of focal glomerulosclerosis with pulse steroids 297. Choudhry S, Bagga A, Hari P, Sharma S,
and oral cyclophosphamide. Pediatr Nephrol. Kalaivani M, Dinda A. Efficacy and safety of
2001;16(11):901–5. tacrolimus versus cyclosporine in children with
284. Waldo FB, Benfield MR, Kohaut EC. Methylprednis- steroid-resistant nephrotic syndrome: a randomized
olone treatment of patients with steroid-resistant controlled trial. Am J Kidney Dis. 2009;53(5):760–9.
nephrotic syndrome. Pediatr Nephrol. 1992;6(6): 298. Wang W, Xia Y, Mao J, Chen Y, Wang D, Shen H,
503–5. et al. Treatment of tacrolimus or cyclosporine A in
285. Niaudet P. Treatment of childhood steroid-resistant children with idiopathic nephrotic syndrome. Pediatr
idiopathic nephrosis with a combination of cyclospor- Nephrol. 2012;27(11):2073–9.
ine and prednisone. French Society of Pediatric 299. Tarshish P, Tobin JN, Bernstein J, Edelmann Jr
Nephrology. J Pediatr. 1994;125(6 Pt 1):981–6. CM. Cyclophosphamide does not benefit patients
286. Ingulli E, Singh A, Baqi N, Ahmad H, Moazami S, with focal segmental glomerulosclerosis. A report of
Tejani A. Aggressive, long-term cyclosporine therapy the International Study of Kidney Disease in Chil-
for steroid-resistant focal segmental glomerulo- dren. Pediatr Nephrol. 1996;10(5):590–3.
sclerosis. J Am Soc Nephrol. 1995;5(10):1820–5. 300. Rennert WP, Kala UK, Jacobs D, Goetsch S, Verhaart
287. Gregory MJ, Smoyer WE, Sedman A, Kershaw DB, S. Pulse cyclophosphamide for steroid-resistant focal
Valentini RP, Johnson K, et al. Long-term cyclospor- segmental glomerulosclerosis. Pediatr Nephrol.
ine therapy for pediatric nephrotic syndrome: a clini- 1999;13(2):113–6.
cal and histologic analysis. J Am Soc Nephrol. 1996; 301. Bajpai A, Bagga A, Hari P, Dinda A, Srivastava RN.
7(4):543–9. Intravenous cyclophosphamide in steroid-resistant
288. Singh A, Tejani C, Tejani A. One-center experience nephrotic syndrome. Pediatr Nephrol. 2003;18(4):
with cyclosporine in refractory nephrotic syndrome in 351–6.
children. Pediatr Nephrol. 1999;13(1):26–32. 302. Elhence R, Gulati S, Kher V, Gupta A, Sharma RK.
289. Ehrich JH, Geerlings C, Zivicnjak M, Franke D, Intravenous pulse cyclophosphamide–a new regime
Geerlings H, Gellermann J. Steroid-resistant idio- for steroid-resistant minimal change nephrotic syn-
pathic childhood nephrosis: overdiagnosed and drome. Pediatr Nephrol. 1994;8(1):1–3.
undertreated. Nephrol Dial Transplant. 2007;22(8): 303. Montane B, Abitbol C, Chandar J, Strauss J,
2183–93. Zilleruelo G. Novel therapy of focal glomerulo-
290. Garin EH, Orak JK, Hiott KL, Sutherland SE. Cyclo- sclerosis with mycophenolate and angiotensin block-
sporine therapy for steroid-resistant nephrotic syn- ade. Pediatr Nephrol. 2003;18(8):772–7.
drome. A controlled study. Am J Dis Child. 304. de Mello VR, Rodrigues MT, Mastrocinque TH, Mar-
1988;142(9):985–8. tins SP, de Andrade OV, Guidoni EB, et al.
291. Lieberman KV, Tejani A. A randomized double-blind Mycophenolate mofetil in children with steroid/
placebo-controlled trial of cyclosporine in steroid- cyclophosphamide-resistant nephrotic syndrome.
resistant idiopathic focal segmental glomerulo- Pediatr Nephrol. 2010;25(3):453–60.
sclerosis in children. J Am Soc Nephrol. 1996;7(1): 305. Gipson DS, Trachtman H, Kaskel FJ, Greene TH,
56–63. Radeva MK, Gassman JJ, et al. Clinical trial of focal
292. Ponticelli C, Rizzoni G, Edefonti A, Altieri P, segmental glomerulosclerosis in children and young
Rivolta E, Rinaldi S, et al. A randomized trial of adults. Kidney Int. 2011;80(8):868–78.
cyclosporine in steroid-resistant idiopathic nephrotic 306. Bagga A, Sinha A, Moudgil A. Rituximab in patients
syndrome. Kidney Int. 1993;43(6):1377–84. with the steroid-resistant nephrotic syndrome. N Engl
293. Bhimma R, Adhikari M, Asharam K, Connolly J Med. 2007;356(26):2751–2.
C. Management of steroid-resistant focal segmental 307. Gulati A, Sinha A, Jordan SC, Hari P, Dinda AK,
glomerulosclerosis in children using tacrolimus. Am J Sharma S, et al. Efficacy and safety of treatment
Nephrol. 2006;26(6):544–51. with rituximab for difficult steroid-resistant and
294. Gulati S, Prasad N, Sharma RK, Kumar A, Gupta A, -dependent nephrotic syndrome: multicentric report.
Baburaj VP. Tacrolimus: a new therapy for steroid- Clin J Am Soc Nephrol. 2010;5(12):2207–12.
resistant nephrotic syndrome in children. Nephrol 308. Kamei K, Okada M, Sato M, Fujimaru T, Ogura M,
Dial Transplant. 2008;23(3):910–3. Nakayama M, et al. Rituximab treatment combined
880 P. Niaudet and O. Boyer

with methylprednisolone pulse therapy and 324. Milliner DS, Morgenstern BZ. Angiotensin
immunosuppressants for childhood steroid-resistant converting enzyme inhibitors for reduction of
nephrotic syndrome. Pediatr Nephrol. 2014; proteinuria in children with steroid-resistant
29:1181–7. nephrotic syndrome. Pediatr Nephrol. 1991;5
309. Prytula A, Iijima K, Kamei K, Geary D, Gottlich E, (5):587–90.
Majeed A, et al. Rituximab in refractory nephrotic 325. Kim EM, Striegel J, Kim Y, Matas AJ, Najarian JS,
syndrome. Pediatr Nephrol. 2010;25(3):461–8. Mauer SM. Recurrence of steroid-resistant nephrotic
310. Sinha A, Bagga A. Rituximab therapy in nephrotic syndrome in kidney transplants is associated with
syndrome: implications for patients’ management. increased acute renal failure and acute rejection. Kid-
Nat Rev Nephrol. 2013;9(3):154–69. ney Int. 1994;45(5):1440–5.
311. Kari JA, El-Morshedy SM, El-Desoky S, Alshaya 326. Striegel JE, Sibley RK, Fryd DS, Mauer SM.
HO, Rahim KA, Edrees BM. Rituximab for refractory Recurrence of focal segmental sclerosis in children
cases of childhood nephrotic syndrome. Pediatr following renal transplantation. Kidney Int Suppl.
Nephrol. 2011;26(5):733–7. 1986;19:S44–50.
312. Magnasco A, Ravani P, Edefonti A, Murer L, Ghio L, 327. Morales JM, Andres A, Prieto C, Martinez MA,
Belingheri M, et al. Rituximab in children with resis- Praga M, Ruilope LM, et al. Clinical and histological
tant idiopathic nephrotic syndrome. J Am Soc sequence of recurrent focal segmental glomerulo-
Nephrol. 2012;23(6):1117–24. sclerosis. Nephron. 1988;48(3):241–2.
313. Zachwieja J, Silska M, Ostalska-Nowicka D, 328. Verani RR, Hawkins EP. Recurrent focal segmental
Soltysiak J, Lipkowska K, Blumczynski A, et al. Effi- glomerulosclerosis. A pathological study of the early
cacy and safety of rituximab treatment in children lesion. Am J Nephrol. 1986;6(4):263–70.
with primary glomerulonephritis. J Nephrol. 329. Chang JW, Pardo V, Sageshima J, Chen L, Tsai HL,
2012;25(6):1060–6. Reiser J, et al. Podocyte foot process effacement in
314. Savin VJ, McCarthy ET, Sharma R, Charba D, postreperfusion allograft biopsies correlates with
Sharma M. Galactose binds to focal segmental early recurrence of proteinuria in focal segmental
glomerulosclerosis permeability factor and inhibits glomerulosclerosis. Transplantation. 2012;93(12):
its activity. Transl Res. 2008;151(6):288–92. 1238–44.
315. De Smet E, Rioux JP, Ammann H, Deziel C, Querin S. 330. Fine RN. Recurrence of nephrotic syndrome/focal
FSGS permeability factor-associated nephrotic syn- segmental glomerulosclerosis following renal trans-
drome: remission after oral galactose therapy. plantation in children. Pediatr Nephrol. 2007;22(4):
Nephrol Dial Transplant. 2009;24(9):2938–40. 496–502.
316. Sgambat K, Banks M, Moudgil A. Effect of galactose 331. Senggutuvan P, Cameron JS, Hartley RB, Rigden S,
on glomerular permeability and proteinuria in steroid- Chantler C, Haycock G, et al. Recurrence of focal
resistant nephrotic syndrome. Pediatr Nephrol. segmental glomerulosclerosis in transplanted kid-
2013;28(11):2131–5. neys: analysis of incidence and risk factors in 59 allo-
317. Donker AJ, Brentjens JR, van der Hem GK, Arisz L. grafts. Pediatr Nephrol. 1990;4(1):21–8.
Treatment of the nephrotic syndrome with indometh- 332. Ding WY, Koziell A, McCarthy HJ, Bierzynska A,
acin. Nephron. 1978;22(4–6):374–81. Bhagavatula MK, Dudley JA, et al. Initial steroid
318. Velosa JA, Torres VE. Benefits and risks of nonste- sensitivity in children with steroid-resistant nephrotic
roidal antiinflammatory drugs in steroid-resistant syndrome predicts post-transplant recurrence. J Am
nephrotic syndrome. Am J Kidney Dis. 1986;8(5): Soc Nephrol. 2014;25(6):1342–8.
345–50. 333. Habib R, Hebert D, Gagnadoux MF, Broyer
319. Bennett WM. The adverse renal effects of nonsteroi- M. Transplantation in idiopathic nephrosis. Trans-
dal anti-inflammatory drugs: increasing problems or plant Proc. 1982;14(3):489–95.
overrated risk. Am J Kidney Dis. 1983;2(4):477. 334. Rizzoni G, Ehrich JH, Brunner FP, Geerlings W,
320. Clive DM, Stoff JS. Renal syndromes associated with Fassbinder W, Landais P, et al. Combined report on
nonsteroidal antiinflammatory drugs. N Engl J Med. regular dialysis and transplantation of children in
1984;310(9):563–72. Europe, 1990. Nephrol Dial Transplant. 1991;
321. Kleinknecht C, Broyer M, Gubler MC, Palcoux JB. 6 Suppl 4:31–42.
Irreversible renal failure after indomethacin 335. Leumann EP, Briner J, Donckerwolcke RA,
in steroid-resistant nephrosis. N Engl J Med. Kuijten R, Largiader F. Recurrence of focal segmental
1980;302(12):691. glomerulosclerosis in the transplanted kidney. Neph-
322. Martinez Vea A, Garcia Ruiz C, Carrera M, Oliver JA, ron. 1980;25(2):65–71.
Richart C. Effect of captopril in nephrotic-range pro- 336. Maizel SE, Sibley RK, Horstman JP, Kjellstrand CM,
teinuria due to renovascular hypertension. Nephron. Simmons RL. Incidence and significance of recurrent
1987;45(2):162–3. focal segmental glomerulosclerosis in renal allograft
323. Ferder LF, Inserra F, Daccordi H, Smith RD. Enalapril recipients. Transplantation. 1981;32(6):512–6.
improved renal function and proteinuria in chronic 337. Pinto J, Lacerda G, Cameron JS, Turner DR,
glomerulopathies. Nephron. 1990;55 Suppl 1:90–5. Bewick M, Ogg CS. Recurrence of focal segmental
28 Idiopathic Nephrotic Syndrome in Children: Clinical Aspects 881

glomerulosclerosis in renal allografts. Transplanta- 352. Salomon R, Gagnadoux MF, Niaudet P. Intravenous
tion. 1981;32(2):83–9. cyclosporine therapy in recurrent nephrotic syndrome
338. Cameron JS. Recurrent primary disease and de novo after renal transplantation in children. Transplanta-
nephritis following renal transplantation. Pediatr tion. 2003;75(6):810–4.
Nephrol. 1991;5(4):412–21. 353. Srivastava RN, Kalia A, Travis LB, Diven SC,
339. Ramos EL, Tisher CC. Recurrent diseases in the Gugliuzza KK, Rajaraman S. Prompt remission of
kidney transplant. Am J Kidney Dis. 1994;24 post-renal transplant nephrotic syndrome with high-
(1):142–54. dose cyclosporine. Pediatr Nephrol. 1994;8(1):94–5.
340. Nehus EJ, Goebel JW, Succop PS, Abraham 354. Faul C, Donnelly M, Merscher-Gomez S, Chang YH,
EC. Focal segmental glomerulosclerosis in children: Franz S, Delfgaauw J, et al. The actin cytoskeleton of
multivariate analysis indicates that donor type does kidney podocytes is a direct target of the
not alter recurrence risk. Transplantation. 2013;96(6): antiproteinuric effect of cyclosporine A. Nat Med.
550–4. 2008;14(9):931–8.
341. Cochat P, Fargue S, Mestrallet G, Jungraithmayr T, 355. Dall’Amico R, Ghiggeri G, Carraro M, Artero M,
Koch-Nogueira P, Ranchin B, et al. Disease recur- Ghio L, Zamorani E, et al. Prediction and treatment
rence in paediatric renal transplantation. Pediatr of recurrent focal segmental glomerulosclerosis after
Nephrol. 2009;24(11):2097–108. renal transplantation in children. Am J Kidney Dis.
342. Seikaly MG. Recurrence of primary disease in chil- 1999;34(6):1048–55.
dren after renal transplantation: an evidence-based 356. Fencl F, Simkova E, Vondrak K, Janda J,
update. Pediatr Transplant. 2004;8(2):113–9. Chadimova M, Stejskal J, et al. Recurrence of
343. Artero M, Biava C, Amend W, Tomlanovich S, nephrotic proteinuria in children with focal segmental
Vincenti F. Recurrent focal glomerulosclerosis: natu- glomerulosclerosis after renal transplantation treated
ral history and response to therapy. Am J Med. with plasmapheresis and immunoadsorption: case
1992;92(4):375–83. reports. Transplant Proc. 2007;39(10):3488–90.
344. Banfi G, Colturi C, Montagnino G, Ponticelli C. The 357. Kawaguchi H, Hattori M, Ito K, Takahashi K, Ota K.
recurrence of focal segmental glomerulosclerosis in Recurrence of focal glomerulosclerosis of allografts
kidney transplant patients treated with cyclosporine. in children: the efficacy of intensive plasma exchange
Transplantation. 1990;50(4):594–6. therapy before and after renal transplantation. Trans-
345. Pirson Y, Squifflet JP, Marbaix E, Alexandre GP, van plant Proc. 1994;26(1):7–8.
Ypersele de Strihou C. Recurrence of focal 358. Laufer J, Ettenger RB, Ho WG, Cohen AH, Marik JL,
glomerulosclerosis despite cyclosporin treatment Fine RN. Plasma exchange for recurrent nephrotic
after renal transplantation. Br Med J (Clin Res Ed). syndrome following renal transplantation. Transplan-
1986;292(6531):1336. tation. 1988;46(4):540–2.
346. Schwarz A, Krause PH, Offermann G, Keller F. 359. Ponticelli C. Recurrence of focal segmental glomeru-
Recurrent and de novo renal disease after kidney lar sclerosis (FSGS) after renal transplantation.
transplantation with or without cyclosporine A. Am Nephrol Dial Transplant. 2010;25(1):25–31.
J Kidney Dis. 1991;17(5):524–31. 360. Pradhan M, Petro J, Palmer J, Meyers K, Baluarte
347. Vincenti F, Biava C, Tomlanovitch S, Amend Jr WJ, HJ. Early use of plasmapheresis for recurrent post-
Garovoy M, Melzer J, et al. Inability of cyclosporine transplant FSGS. Pediatr Nephrol. 2003;18(9):934–8.
to completely prevent the recurrence of focal 361. Saleem MA, Ramanan AV, Rees L. Recurrent focal
glomerulosclerosis after kidney transplantation. segmental glomerulosclerosis in grafts treated with
Transplantation. 1989;47(4):595–8. plasma exchange and increased immunosuppression.
348. Voets AJ, Hoitsma AJ, Koene RA. Recurrence of Pediatr Nephrol. 2000;14(5):361–4.
nephrotic syndrome during cyclosporin treatment 362. Canaud G, Martinez F, Noel LH, Mamzer MF,
after renal transplantation. Lancet. 1986;1(8475): Niaudet P, Legendre C. Therapeutic approach to
266–7. focal and segmental glomerulosclerosis recurrence
349. Ingulli E, Tejani A, Butt KM, Rajpoot D, Gonzalez R, in kidney transplant recipients. Transplant Rev
Pomrantz A, et al. High-dose cyclosporine therapy in (Orlando). 2010;24(3):121–8.
recurrent nephrotic syndrome following renal trans- 363. Nozu K, Iijima K, Fujisawa M, Nakagawa A,
plantation. Transplantation. 1990;49(1):219–21. Yoshikawa N, Matsuo M. Rituximab treatment for
350. Mowry J, Marik J, Cohen A, Hogg R, Sahney S, posttransplant lymphoproliferative disorder (PTLD)
Ettenger R. Treatment of recurrent focal segmental induces complete remission of recurrent nephrotic
glomerulosclerosis with high-dose cyclosporine syndrome. Pediatr Nephrol. 2005;20(11):1660–3.
A and plasmapheresis. Transplant Proc. 1993; 364. Pescovitz MD, Book BK, Sidner RA. Resolution of
25(1 Pt 2):1345–6. recurrent focal segmental glomerulosclerosis protein-
351. Raafat RH, Kalia A, Travis LB, Diven SC. High-dose uria after rituximab treatment. N Engl J Med.
oral cyclosporin therapy for recurrent focal segmental 2006;354(18):1961–3.
glomerulosclerosis in children. Am J Kidney Dis. 365. Apeland T, Hartmann A. Rituximab therapy in early
2004;44(1):50–6. recurrent focal segmental sclerosis after renal
882 P. Niaudet and O. Boyer

transplantation. Nephrol Dial Transplant. 2008;23(6): nephropathy) after renal transplantation successfully
2091–4. treated with combinations of plasma exchanges,
366. Hristea D, Hadaya K, Marangon N, Buhler L, immunoglobulin, and rituximab. Transplant Proc.
Villard J, Morel P, et al. Successful treatment of recur- 2006;38(8):2659–60.
rent focal segmental glomerulosclerosis after kidney 369. El-Firjani A, Hoar S, Karpinski J, Bell R, Deschenes
transplantation by plasmapheresis and rituximab. MJ, Knoll GA. Post-transplant focal segmental
Transpl Int Off J Eur Soc Organ Transplant. glomerulosclerosis refractory to plasmapheresis
2007;20(1):102–5. and rituximab therapy. Nephrol Dial Transplant.
367. Meyer TN, Thaiss F, Stahl RA. Immediate versus 2008;23(1):425.
long-term effect of rituximab in recurrent focal seg- 370. Yabu JM, Ho B, Scandling JD, Vincenti F. Rituximab
mental glomerulosclerosis. Transpl Int Off J Eur Soc failed to improve nephrotic syndrome in renal trans-
Organ Transplant. 2008;21(11):1102. plant patients with recurrent focal segmental
368. Westphal S, Hansson S, Mjornstedt L, Molne J, glomerulosclerosis. Am J Transplant Off J Am Soc
Swerkersson S, Friman S. Early recurrence of Transplant Am Soc Transplant Surg. 2008;8(1):
nephrotic syndrome (immunoglobulin m 222–7.
Immune-Mediated Glomerular
Injury in Children 29
Michio Nagata

Contents Growth Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 914


Platelet-Derived Growth Factor (PDGF) . . . . . . . . . . . . 914
Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 884 Transforming Growth Factor (TGF) . . . . . . . . . . . . . . . . . 914
Glomerular Components . . . . . . . . . . . . . . . . . . . . . . . . . . . 885 Fibroblast Growth Factor (FGF) . . . . . . . . . . . . . . . . . . . . . 915
The Glomerular Basement Membrane (GBM) . . . . . . 887 Vascular Endothelial Growth Factor (VEGF) . . . . . . . 915
Mesangial Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 887 Connective Tissue Growth Factor (CTGF) . . . . . . . . . . 915
Podocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 889 Epidermal Growth Factor (EGF) . . . . . . . . . . . . . . . . . . . . 916
Endothelial Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 890 Platelets and the Coagulation Cascade . . . . . . . . . . . 916
Parietal Epithelial Cells (PECs) . . . . . . . . . . . . . . . . . . . . . 891 Platelets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 916
Immune System and Animal Models . . . . . . . . . . . . . 893 Fibrin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 917

Antibody-Mediated Immunity . . . . . . . . . . . . . . . . . . . . . 894 Tissue Factor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 918


Circulating Immune Complexes . . . . . . . . . . . . . . . . . . . . . 894 The Enzymatic Fibrinolysis System . . . . . . . . . . . . . . . . . 918
In Situ Immune Complexes . . . . . . . . . . . . . . . . . . . . . . . . . . 896 Eicosanoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 919
Cell-Mediated Immunity . . . . . . . . . . . . . . . . . . . . . . . . . . . 896 Reactive Oxygen Species (ROS) . . . . . . . . . . . . . . . . . . . 921
T-Cell Component of Adaptive Nitric Oxide (NO) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 922
Immune Response . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 897
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 922
Inflammatory Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 899
Leukocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 899
Polymorphonuclear Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . 900
Monocytes/Macrophages . . . . . . . . . . . . . . . . . . . . . . . . . . . . 901
Mast Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 904
Toll-Like Receptors (TLRs) . . . . . . . . . . . . . . . . . . . . . . . . . 904
Complement Cascade . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 905
Soluble, Secreted Peptides . . . . . . . . . . . . . . . . . . . . . . . . . 909
Interleukins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 909
Interferons . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 911
Tumor Necrosis Factor (TNF) . . . . . . . . . . . . . . . . . . . . . . . 911
Chemokines and Their Receptors . . . . . . . . . . . . . . . . . . . 911

M. Nagata (*)
Graduate School of Comprehensive Human Sciences,
University of Tsukuba, Tsukuba, Japan
e-mail: nagatam@md.tsukuba.ac.jp

# Springer-Verlag Berlin Heidelberg 2016 883


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_25
884 M. Nagata

Overview components and the subsequent activation of sec-


ondary inflammatory mediators. The glomerular
Glomerular injury is the central basis of renal deposition of antibodies is promoted by the
insufficiency, and immunologic mechanisms humoral immune process of Th2 activation, which
play critical roles for initiation and progression leads to antibody production via B-lymphocyte
of glomerular diseases. activation. This deposition may be composed of
Two axes of adaptive immune system involve preformed immune complexes in the circulation,
the pathogenesis of glomerular injury: antibody- which are trapped in the glomerulus, or antibodies
mediated and cell-mediated immunity. In children, (autoantibodies) that bind to intraglomerular anti-
popular immune-mediated glomerular diseases gens or interact with intrinsic glomerular cell-
include poststreptococcal acute glomerulonephri- surface antigens, as a result of changes to the cell
tis, IgA glomerulonephritis, membranoproli- surface (in situ immune complex).
ferative glomerulonephritis (type 1), membranous Antibody deposition on glomerular capillary
glomerulonephritis, ANCA-related glomerulone- walls or the mesangium activates the complement
phritis, and lupus nephritis, all of which reveal cascade, and intrinsic cell-derived chemotactic
various autoimmune features (Table 1). Although factors result in inflammatory cell influx, which is
ANCA-related glomerulonephritis is caused by a crucial event in the glomerular injury by produc-
Th1-predominant T-cell-mediated immunity in a tion/activation of proinflammatory cytokines,
pauci-immune fashion, remainders are thought to chemokines, growth factors, reactive oxygen species
be processed by antibody-mediated immunity. (ROS), eicosanoids, and nitric oxides (NO). These
The major steps of antibody-mediated glomeru- secondary mediators not only stimulate intrinsic
lar injury are immune deposition on glomerular cells to proliferate and/or produce matrix materials,
but they also induce the synthesis of similar mole-
cules by intrinsic glomerular cells. The secondary
Table 1 Autoimmune features of various renal mediators involve severe endothelial injury which
diseases [1] potentially results in dysregulation of local coagula-
Disease Autoimmune features tion system, accompanied by exudation and the
Poststreptococcal GN Anti-C1q, IgG, AECA, anti- formation of cross-linking fibrins. These end
DNA, ANCA, PDI, C3Nef products of the extrinsic coagulation cascade also
IgA nephropathy Anti-glycan, endothelial cells, damage glomeruli, particularly tuft necrosis, the
mesangial cells, IgG
common forerunner of crescentic formation.
Anti-GBM nephritis Anti-GBM, ANCA (20 %)
ANCA-positive Anti-MPO, PR3, cPR3, NET,
An alternative immune mechanism is cell-
nephritis DNA, endothelial cells, mediated immunity, with Th1 activation promot-
LAMP2 ing cytotoxic glomerular injury by influx of
Lupus nephritis Anti-dsDNA, annexin, MPO, delayed-type hypersensitivity (DTH) effectors,
PR3, nucleosome, IgG, C1q, i.e., macrophages, T cells, and fibrin. Proliferative
cardiolipin, MBL, NET
glomerulonephritis in the absence of immune
MPGN I Anti-C3 Nef, C4Nef, C1q
complexes or antibodies suggests a role for cell-
MCD/FSGS None identified
Membranous Anti-PLA2R, DNA, NEP,
mediated immune injury. This is supported by the
nephropathy aldose reductase, SOD2 experiment showing that transfer of sensitized T
Dense deposit disease C3Nef, C4Nef, anti-CHF, cells successfully induced glomerular injury in the
factor B, C1q host. Cytokines secreted by DTH effectors are
C3 nephropathy C3Nef, anti-CHF considered to injure the glomerular cells. It is
AECA anti-endothelial cell antibody, PDI protein disulfide clear that the antibody- and cell-mediated immune
isomerase, NET neutrophil extracellular trap, LAMP2 systems are not independent and often interact in
lysosomal membrane protein 2, MBL mannose binding
lectin pathway, NEP anti-neutral endopeptidase, SOD2 glomerular injury. The complex mechanism of
anti-manganese superoxide dismutase, CHF complement immune-mediated glomerular injury is integrated
H factor (Modified by Ref. [1]) by a recent review by Couser (Fig. 1) [1].
29 Immune-Mediated Glomerular Injury in Children 885

Etiologic events

PAMPS/DAMPS Antigen-
presenting cell

Complement TLRs, NLRs CD4 T cells

B cells
Auto antibodies, TREGs
C5b-9 C5a antigen-antibody
complexes
TH1
Resident Circulating
TH2
glomerular cells inflammatory cells
TH17

Proteases Chemokines
Oxidants Growth factors Glomerular tissue injury
Cytokines Eicosanoids

Fig. 1 Schematic overview of both the innate and adap- differentiation of CD4 helper cells, B cell activation, and
tive immune mechanisms that mediate tissue injury in antibody production. Antibodies lead to circulating com-
GN. Etiologic events expose immunostimulatory PAMPs plex trapping or in situ formation of immune complexes
or DAMPs that activate both the innate (red) and the that can activate both the TLR and complement compo-
adaptive (antigen specific, blue) immune systems, which nents of the innate immune system. Complement activa-
also interact with each other. Activation of the innate tion generates the chemotactic factor C5a that attracts
immune system occurs immediately and involves TLRs circulating inflammatory cells (including neutrophils, mac-
or NLRs on both circulating inflammatory cells and resi- rophages, basophils, and natural killer cells), which release
dent glomerular cells. TLR activation results in release of mediators and damage glomeruli and C5b-9 that activates
inflammatory mediators that cause glomerular injury. resident glomerular cells to do the same. CD4 Th1 and Th2
Some PAMPs and DAMPs can activate complement cells cause tissue injury primarily thorough macrophages
directly through the innate immune system. TLRs are and basophils, respectively, whereas Th17 cells can medi-
also required to activate the adaptive immune system ate glomerular damage directly. CD4 regulatory cells
through antigen-presenting cells that promote (Tregs) downregulate the adaptive immune response [1]

This chapter provides a review of our basic immune-mediated glomerulonephritis may help
understanding of and updated information regard- to explain some aspects of the mechanism(s)
ing immune-mediated glomerular injury, with underlying chronic glomerulonephritis in
emphasis on the pathogenic mechanism(s). humans.
Although numerous scientific advances have
come out of animal experiments, it should be
kept in mind that experimental models of Glomerular Components
immune-mediated glomerular injury are largely
relevant to the acute phases of anti-GBM antibody Glomerulonephritis arises from the responses of
nephritis, nephrotoxic serum nephritis, or anti- intrinsic glomerular cells to inflammatory reac-
Thy1 nephritis of mesangial proliferation. Fur- tions. Glomerular deposition, hypercellularity
thermore, the molecular profiles of the immune (intrinsic and inflammatory cells), and capillary
systems of mice and humans are significantly destruction are typical features of glomerular
different. This review of our basic knowledge of injury (Fig. 2). The biologic characteristics of
886 M. Nagata

Fig. 2 Typical features of immune-mediated glomerular stain), (c) subendothelial immune deposition in lupus
injury seen in human biopsy samples by LM and IF. (a) nephritis (PAM stain), (d) granular and peripheral pattern
Mesangial proliferation in IgA glomerulonephritis (PAS of IgG in membranous glomerulonephritis, (e) fringe pat-
stain), (b) crescentic formation with tuft necrosis and fibrin tern of C3 deposition in MPGN, (f) subendothelial IgG
deposition in ANCA-related glomerulonephritis (PAM deposits in lupus nephritis

the intrinsic glomerular cells are implicated in this involved in glomerular pathology, particularly in
process and described here. Although the cellular cellular crescent formation and adhesion (the
components of the glomerulus are anatomically renal corpuscle comprises these four cell types).
defined by three cell types, mesangial cells, endo- In addition, the biochemical profiles of the extra-
thelial cells, and podocytes, parietal cells are also cellular matrices, glomerular basement
29 Immune-Mediated Glomerular Injury in Children 887

membrane, and mesangial matrices are unique on the GBM, as seen in membranous glomerulo-
and may be targets for antibody deposition, lead- nephritis and lupus nephritis, can also alter the
ing to effects on the corresponding intrinsic cells. GBM by changing its barrier of charge and size
Ultrastructural analyses provide a good opportu- properties.
nity to define the involvement of extracellular
matrices in glomerular injury.
Mesangial Cells

The Glomerular Basement Membrane The mesangium is the axis of the glomerulus,
(GBM) holding the capillaries in a developed dense net-
work of matrix, which includes microfibrils and
The GBM, which is a central component of the fibronectin. Mesangial cells are derived from the
filtration barrier based on charge and size, is com- metanephric mesenchyme, and there is one or two
posed of extracellular matrix materials, including cells within each mesangial area. The biologic
type IV collagen, nidogen (entactin), heparan sul- properties of mesangial cells have some similari-
fate proteoglycans (HSPG), laminin, and perlican. ties with vascular smooth muscle cells. Both cell
GBM is synthesized by podocytes and endothelial types show contraction and cell proliferation in
cells; however, the details of the formation of the response to vasoactive substances and cytokines
membrane from the individually synthesized pro- in vitro.
teins remain unknown. Nevertheless, this unique The lack of intact GBM between endothelial
permeability system traps macromolecules and cells and the mesangium enables bone marrow-
antibodies and is responsible for their glomerular derived inflammatory cells and macromolecules
deposition. to access the mesangium (Fig. 5). Early studies
The GBM also contains potentially harmful showed that resident cells in the mesangium
antigens. Pathogenic autoantibodies directed expressed the phenotype of bone marrow cells
against the C-terminal globular noncollagenous (Ia positive) in the normal state and that their
domain 1 (NC1) of the α3-chain of type IV colla- numbers were increased in glomerulonephritis.
gen, which has a distinct GBM molecular compo- GFP-tagged bone marrow cell transplantation in
sition, promote severe glomerulonephritis in rat anti-Thy1 nephritis, a model of mesangial pro-
patients with Goodpasture syndrome by epitope- liferative glomerulonephritis, has provided evi-
specific autoantibody binding and effector T-cell dence that bone marrow-derived cells are
reaction (Fig. 3; [3]). Similar epitopes are targeted responsible for mesangial hypercellularity.
by alloantibodies in a subset of patients with The mesangial fluid pathway was discovered
Alport nephritis who have received allografts. in an experiment involving the ligation of lymph
GBM is also the target of bioactive substances vessels around the renal artery. This leaky barrier
released from inflammatory cells and intrinsic and fluid pathway allow circulatory macromole-
cells during inflammation. GBM changes may cules, including antibodies, to be trapped in the
directly influence the physiologic stability of fil- mesangium as mesangial deposits. The pheno-
tration, thereby reducing the glomerular filtration types and behaviors of cells in the mesangium
rate and causing proteinuria. Electron microscopy are also modulated by the glomerular scaffold of
has revealed GBM alterations in association with self-produced extracellular matrix materials.
immune deposits, and inflammatory cell influx In glomerular injury, deposition, cell prolifera-
can directly alter the morphology of the GBM tion, matrix production, and cell death occur in the
(Fig. 4). Proteolytic enzymes derived from neu- mesangium. Mesangial proliferation is a typical
trophils may directly alter the GBM, resulting in feature of glomerular injury. Activation of cell
membranolysis, lamination, gaps, and edema cycle molecules, as revealed by immunohisto-
(typical features of GBM injury), which may chemical detection of proliferating cell nuclear
lead to urinary abnormalities. Immune deposition antigen or Ki-67, is detected in the mesangial
888 M. Nagata

Type IV collagen chains


Protomers
1 1
2
1
2 α1.α2.α1
3
3 4
5
α3.α4.α5
4
5
6
5
α5.α6.α5 5

6 7S NC1 Trimer

Auto-antibody

GPA

T A

α3 α5 I
P
S
C P V P
E L
GPA G Y
T
region S

α3.(IV) NC1

α3.α4.α5(IV)
Protomer

Fig. 3 Structure of Goodpasture antigen epitopes in col- were found critical for the epitope of the immunodominant
lagen type IV. (a, c) GP epitope is localized in the NC1 GPA autoantibodies (inset), which appears to be seques-
domain of α3(IV) chain of type IV collagen. In the GBM, tered by an interaction with an α5(IV) chain in the same
the α3(IV) chain is associated with the α4(IV) and α5 protomer (b). Linear staining pattern of IgG in the glomer-
(IV) chains, forming protomers and networks. The GP ulus from a patient with GP syndrome (GP Goodpasture,
epitopes are cryptic within the α3 α4 α5(IV) NC1 hexamer GBM glomerular basement membrane) (Modified figure
and inaccessible for autoantibody binding unless the NC1 from Borza and Hudson [2] with permission)
hexamer dissociates. Several hydrophobic amino acids

cells in glomerulonephritis, indicating mesangial those produced by inflammatory cells, as well as


cell proliferation in situ. various vasoactive substances, such as angioten-
Mesangial activation occurs though receptor- sin, endothelin, and vasopressin. As soluble cyto-
mediated mechanism. Mesangial cell proliferation kines and growth factors act in autocrine,
is stimulated by various cytokines and growth paracrine, and juxtacrine manners, mesangial
factors, including interleukins (ILs), PDGF, and inflammatory cells effectively activate resident
fibroblast growth factor (FGF). In addition, Fc cells, and both cell types accelerate cell prolifera-
fragments of IgG and IgA bind the receptor in tion and matrix production in situ. During active
mesangium and stimulate the cells. For example, glomerulonephritis with mesangial proliferation,
PDGF binds to a tyrosine kinase receptor, which mesangial cells frequently express α-smooth mus-
activates a number of downstream effectors, cle actin (α-SMA), desmin, calponin, and matrix-
including the Ras/MAPK pathways, to activate binding integrin receptors. This may not only be
various transcription factors (c-fos, c-myc, c-jun) an example of phenotypic changes in mesangial
and proto-oncogenes. Activated mesangial cells cells reflecting cell activation, but may also repre-
synthesize cytokines and growth factors similar to sent involvement in further glomerular injury.
29 Immune-Mediated Glomerular Injury in Children 889

Fig. 4 Changes of filtration barrier in glomerulonephritis thickening and irregularity in GBM and foot process
by EM. (a) Lytic change in GBM is accompanied with effacement. (c) Thickening of GBM due to membranolysis
endothelial swelling with loss of fenestra on the back- containing cellular debris, deposits, and cellular infoldings.
ground of inflammatory cells in the capillary. Note foot (d) Endothelial cell swelling and podocyte detachment
process effacement in podocytes with actin accumulation from GBM
in podocyte. (b) Subepithelial dense deposits with

Mesangial proliferation is recognized as a fore- substances. Podocytes are an important compo-


runner of glomerulosclerosis, and several in vitro nent of the filtration barrier, in that they have slit
experiments have revealed that mesangial cells membranes and synthesize GBM components.
synthesize matrix components in response to var- Podocytes are highly specialized postmitotic
ious inflammatory cytokines and growth factors. cells that are derived from the metanephric mes-
Hyperplastic mesangial cells reduce cell numbers enchyme. Indeed, podocytes have a negligible
by apoptosis mediated via a variety of signaling capacity to proliferate, in that they undergo
mechanisms. nuclear division, but cytokinesis is not evident
in vivo. Actin dynamics in podocytes has been
intensively investigated, and its role for cell integ-
Podocytes rity and function emerged. Podocytes are rich in
actin filaments, and the cytoskeletons of their
Podocytes have many intrinsic properties intermediate filaments are linked to foot process
sustained by actin cytoskeleton and some unique maintenance and slit membrane function. Several
molecules that participate in the glomerular path- podocyte markers, such as WT1, CALLA, CD10,
ophysiology. Podocytes are located outside of synaptopodin, podocalyxin, podoplanin, and
glomerular capillaries and do not affect directly nestin, have been identified. The slit membrane-
the bloodstream; instead, they face the primary related molecules such as nephrin and podocin are
ultrafiltrate, which contains various biochemical also sometimes used as specific markers.
890 M. Nagata

m
m

p
m

e
pa
Mf

e
m
p

Fig. 5 Structure of the glomerulus and monocyte influx. Note lack of GBM between endothelial cell and
(a) Schematic presentation of the glomerular profile. The mesangium. Original illustration is courtesy from
glomerulus is composed of four cell types: mesangium (m), Dr. W. Kriz with permission, (b) invasion of macrophages
endothelial cell (e), podocyte ( p), and parietal cell (pa). (Mφ) into mesangium in the biopsy of in MPGN

In addition, the podocyte is a major source of podocyte-membrane protein megalin. Similarly,


growth factors, such as VEGF, TGF-β, and the recently identified neutral endopeptidase
CTGF. Podocyte also expresses angiotensin II (NEP) and M-type phospholipase A2 receptor,
type 1 receptor which mediates local renin- which are located in podocytes, are potentially
angiotensin system for podocyte function. intrinsic antigens that form immune complexes
Podocyte injury is the base of proteinuria, and in situ that cause membranous glomerulonephritis
podocyte loss from GBM (podocytopenia) may in humans [4, 5]. Podocytes express B7-1, which
lead to glomerulosclerosis. is a costimulatory molecule for T cells that is
Immunologic mechanisms including GBM upregulated in injured podocytes and causes
deposition, complement activation, and inflam- dysregulation of the actin cytoskeleton, leading
matory mediators involve podocyte damage, and to proteinuria [6].
alteration of podocyte molecules results in pro-
teinuria and sclerosis. Podocyte injury is visible
by morphology. Electron microscopy reveals Endothelial Cells
cytoplasmic vacuolation, pseudocyst formation,
and foot process effacement with actin accumula- The glomerular endothelial cell is the only intrin-
tion and detachment. In addition, phenotypic sic glomerular cell type that directly faces the
changes, namely, expression of desmin, also circulation. These endothelial cells are different
represent podocyte injury in the rodents. These from those in other organs because of their
changes are induced not only by hemodynamic extraordinary flatness and highly fenestrated fea-
effects, but also by immune-mediated properties. tures, allowing effective filtration. The fenestra-
Membranous glomerulonephritis is an example of tion (70–100 nm in size) is maintained by vascular
immune-mediated podocyte injury, which results endothelial growth factor A (VEGF-A), which is
from complement activation. The antigen derived from the podocytes. The endothelial cell-
responsible for experimental membranous surface layer provides a negatively charged mesh
glomerulonephritis (Heymann nephritis) is the of polysaccharide moieties of glycoproteins,
29 Immune-Mediated Glomerular Injury in Children 891

Table 2 Neutrophil endothelial cells express various adhesion molecules for interaction
Endothelial molecules Leukocyte molecules Major function
P-selectin Sialyl-Lewis X-modified Rolling (neutrophils, monocytes, lymphocytes)
protein
E-selectin Sialyl-Lewis X-modified Rolling and adhesion PMNs, monocytes, T cells)
protein
GlyCam-1, CD34 L-selectin Rolling (neutrophils, monocytes)
ICAM-1 (immunoglobulin CD11/CD18 integrins Adhesion, arrest, transmigration (neutrophils,
family) (LFA-1, Mac-1) monocytes, lymphocytes)
VCAM-1 VLS-4 integrin Adhesion (eosinophils, monocytes, lymphocytes)
(immunoglobulin family)
CD31 CD31 Transmigration (all leukocytes)

glycosaminoglycans (heparan sulfate, sialic acid, double contour of the GBM, as typically seen in
chondroitin sulfate, hyaluronan) which determine transplant glomerulopathy. The mechanism of this
vascular rheology and are probably involved in humoral-mediated rejection is unclear, but MHC
permselectivity. The charge and various adhesion class II antigens on the endothelial surface interact
molecules on the cell surface permit interactions with anti-donor antigens and form an in situ
between endothelial cells and blood-borne fac- immune complex that persistently activates or
tors, including leukocytes, platelets, the third injures endothelial cells, resulting in matrix pro-
component of complement, antigens, and immune duction and subendothelial widening. These fea-
complexes (Table 2). Endothelial cells physiolog- tures have also been noted in MPGN, in which
ically synthesize several coagulation proteins, persistent complement activation is the trigger for
growth factors, extracellular matrix components, endothelial cell injury.
and various substances, including nitric oxide,
prostaglandins, and endothelin, to maintain cell
integrity and the vascular environment. Parietal Epithelial Cells (PECs)
Angiotensin-converting enzyme (ACE) and
endothelin-converting enzyme participate in the Parietal cells, also known as Bowman’s epithe-
local formation of bioactive angiotensin II and lium, are located in the continuous epithelial lin-
mature endothelin, respectively. ing between the podocytes and proximal tubular
Endothelial cells are involved in immune- cells. PECs and podocytes share a common phe-
mediated glomerular injury in many aspects, notype during glomerular differentiation until the
including characteristic cell surface which is a S-shaped body stage. Although PECs seem to
feasible target of inflammatory reaction, expres- play a role in glomerular injury, little is known
sion of MHC proteins, and changes of variety of about their biologic and physiologic properties.
bioactive/physiologic substances. PECs express receptors for PDGF and FGF, and
Endothelial cell proliferation, typically their ligands derived from inflammatory or intrin-
observed in endocapillary proliferative glomerulo- sic cells can stimulate cell proliferation and matrix
nephritis and preeclampsia (also known as synthesis. PECs are the major constituent of the
endotheliosis), is considered as post-injury regen- cellular crescent, which expresses connective tis-
erative responses. Since endothelial cells actively sue growth factor (CTGF) mRNA, suggesting a
regenerate and cell loss is replaced either by neigh- growth factor-mediated mechanism for glomeru-
boring cells or bone marrow-derived cells, endo- lar scar formation. An in vitro study revealed that
thelial cell injury may be usually reversible. FGF-2 stimulated PECs proliferation and PDGF
Endothelial cell injury is also evidenced by fibrin stimulated matrix production in PECs in culture.
deposition, as seen in thrombotic microangiopathy. In addition, tissue factor or cross-linked fibrin
An additional feature of endothelial injury is the stimulates proliferation in PECs in vitro.
892 M. Nagata

a IL-4 IL-12 TGF-β


IL-6
IL-21

GATA3 T-bet RORγT Foxp3


STAT-6 STAT-4 STAT-3

IL-23

Th2 Th1 Th17 Treg


IL-17A, IFN-γ Tregs acquiring
producing affector function
IL-4 IFN-γ Th cell IL-17A IL-10
IL-5 TNF IL-17F TGF-β
IL-13 LT-α IL-21 LT-35
IL-22

lgE, neutralising lgG Macrophage activation Neutrophil activation Regulation of


Eosinophil activation Complement fixing lgG Direct tissue interactions Th subsets
High FcR affinity lgG

Parasitic infection Intracellular infection Extracellular infection Limiting tissue injury


Allergy Classical DTH Tissue injury Peripheral tolerance
Autoimmunity Autoimmunity Graft tolerance
Rejection Rejection

b APC
Th17

IL-23
IL-22

IL-17A, IL-17F

Neutrophil Direct tissue effects,


activation chemokine production
Macrophage
TNF, IL-1β

Fig. 6 (a) Simplified diagram of the Th1, Th2, Th17, and There is evidence that Th17 cells can acquire the capacity
Treg subsets, showing key cytokines produced by these to produce IFN-, and Tregs can, at sites of inflammation,
cells and the cytokines and transcription factors critical to lose their Foxp3 expression and acquire effector function.
the development and expansion of these subsets. The bio- (b) Summary of potential mechanisms by which Th17
logic functions of Th cell subsets are briefly summarized. cytokines could mediate immune renal injury [9]
29 Immune-Mediated Glomerular Injury in Children 893

Table 3 Characteristics of T-helper subsets


Characteristics Th1 cells Th2 cells
Inducing stimuli IL-12, INF-γ, IL-18, IL-27, PRR signaling IL-4
Transcription factors STAT4, STAT 1, T-bet, NFκB STAT6, GATA3
Cytokine produced INF-γ, IL-2, TNF, lymphotoxin-β IL-4, IL-5, IL-13
Chemokine receptor CXCR3, CCR5, CCR1 CCR3, CCR4, CCR8
expression
Antibody isotypes Human: IgG1, IgG2, IgG3 IgG4, IgE
Mouse: IgG2a, IgG2b, IgG3 IgG1, IgE
Effector response Cell-mediated immunity, macrophage activation, antibody- Eosinophil
mediated cellular cytotoxicity activation, allergy

The experimental model of crescentic glomerulo- the glomerulus. This initiation induces the sec-
nephritis has shown that PECs undergo an epithe- ondary inflammatory mediators, including cyto-
lial mesenchymal transition, which is involved in kines and chemical mediators, derived from
scar formation in glomerular crescents [7]. PEC activated leukocytes. T-cell-mediated glomerular
proliferation has also been demonstrated in injury may include rather wide-ranged processes,
nonimmune-mediated glomerulonephritis, partic- since T cell regulates B-cell system. However
ularly in focal segmental glomerulosclerosis and when we concern about T-cell-dependent glomer-
other immune-/nonimmune-mediated glomerulo- ular injury as a narrow meaning, it is less com-
sclerosis conditions. Nitch-1-mediated PEC mon. One example of this mechanism is cytotoxic
migration has shown to be a glomerular wound action of T cells which may damage the intrinsic
healing process in response to severe injury of glomerular cells or alter glomerular filtration bar-
filtration barrier [8]. Cell proliferation, matrix pro- rier, causing proteinuria. This complex immune
duction, and distortion of functional architecture system profiled by T-cell subsets (Th1/Th2/Th17/
in PECs participate in the glomerulosclerosis by Treg) may determine the type of glomerular injury
immune or nonimmune mechanisms. (Table 3).
There are varieties of experimental models of
immune-mediated glomerulonephritis that have
Immune System and Animal Models been used to analyze the nephritogenic mecha-
nisms. Nephrotoxic serum nephritis, a classical
Innate and adaptive immune systems are the path- model initially established by Masugi in 1933, is
ogenic background of immune-mediated glomer- induced by injection of duck anti-rabbit kidney
ular injury. Although molecular and cellular antisera to rabbits resulting in crescentic glomer-
mechanism of immune system is substantially ulonephritis. Anti-GBM antibody nephritis is
complex, it may be convenient to follow the induced by administration of anti-GBM IgG
mechanism of glomerular injury by two steps, showing proliferative (usually crescentic) glomer-
initiation and promotion. Lymphocytes mainly ulonephritis. Both are the basic and widely exam-
drive an initiation, and neutrophils and mono- ined models of glomerulonephritis. There are two
cyte/macrophages largely participate in promo- phases of immune reaction in these models, het-
tion as secondary mediators. erologous and autologous phase. The former is
Lymphocyte-mediated injury is processed by provoked by the binding of injected alloantibody
antibody-mediated (B cell driven) or cell- to GBM, whereas the latter is caused by a host-
mediated (T cell driven) mechanisms, and they produced autoantibody that binds to the GBM.
usually interact and are synergistically involved Heterologous phase alone causes mild and self-
in the tissue damage. B-cell system promotes pro- limiting nephritis, but prolongation or accelera-
duction and in situ deposition of antibodies tion of autologous phase by pre-immunization
(majority forms antigen-antibody complexes) in with IgG of immunized animals provokes severe
894 M. Nagata

glomerulonephritis. Autoantibody may be impor- glomerular diseases, with various histologic pat-
tant to induce severer nephritis. Several mice terns, and the clinical outcomes may be related to
models of lupus, including (NZBXNZW) F1, the nature of the antigen(s) and their glomerular
BXSB, and MRL/lpr mice, reveal spontaneous binding site(s).
and proliferative glomerulonephritis with immune
complex deposition, similar to the human lupus
nephritis. Heymann nephritis is a model of mem- Circulating Immune Complexes
branous glomerulonephritis, and a responsible
antigen is identified as megalin. In this model, If the immune system produces autoantibodies,
complement activation plays significant roles for immune complexes can be formed. Circulating
proteinuria, and proliferative nephritis is not usu- immune complex-mediated glomerulonephritis is
ally seen. All these models are immune complex- primarily seen as a type III hypersensitivity reac-
mediated glomerulonephritis as revealed by tion (Fig. 7). As known in the serum sickness
immunoglobulin and complement deposition, disease model, high amount of foreign serum
regardless of whether circulating or in situ injection results in the appearance of immune
immune complex formation. On the contrary, complex in the circulation which accumulates in
widely used model of anti-Thy1 nephritis reveals the glomerulus and induces acute glomerulone-
mesangial proliferative glomerulonephritis but phritis and vasculitis. Regardless of whether they
pauci-immune. In this model, antibody against are endogenous or exogenous, the antigens are not
thymus immediately binds to receptor on glomerular in origin. Exogenous antigens can be
mesangial cells resulting in cell lysis bacterial (Streptococcus and Staphylococcus
(mesangiolysis) associated with transient activa- spp.), viral (hepatitis B), parasitic (Plasmodium
tion of complements; however, immune complex spp.), or spirochete (Treponema spp.) in origin.
and complements are lost immediately. Mesangial In contrast, DNA and histones are endogenous
proliferation in this model may reflect repair pro- antigens that are typically seen in lupus nephritis.
cess from mesangiolysis to reconstruct glomerular The highly permeabilized glomerular filter is
architecture mimicking glomerulogenesis, rather susceptible to passive trapping of preformed
than the result of mitogenic stimuli of inflamma- immune complexes from the circulation. The
tory mediators (unlike immune-mediated glomer- highly negative charge of the capillary wall and
ulonephritis). Since majority of experimental endothelial glycocalyx surface also attracts circu-
works using these models investigated an acute lating macromolecules.
phase of the diseases, it is necessary to understand The biologic activities of circulating immune
the pathogenetic difference in each model and complexes depend largely on the biochemical and
recognize the limitation of the results to translate quantitative features of the circulating antigens.
human chronic glomerulonephritis. The size of an immune complex is determined by
the ratio of the antigen to antibody. When the
antibody is present in excess, cross-linking of
Antibody-Mediated Immunity immune complexes occurs, and the complex
becomes large. The primary site of such a large
Antibody (humoral)-mediated immune glomeru- immune deposition is either the subendothelium
lar injury is initiated by antigen-antibody complex or the mesangium and efficiently activates the
formation within the glomerulus. B-cell activation complement cascade by binding to Fc receptors
by interleukins (IL-4, IL-5), which are synthe- (the Arthus reaction), thereby further stimulating
sized by antigen-specific CD4+ T cells, is a pre- the inflammatory processes. In contrast, when an
requisite for antigen-specific antibody production. antigen bears few epitopes or when there is an
Many glomerular lesions result from the glomer- excess of antigen relative to antibody, the immune
ular binding of antibodies. Immune-mediated glo- complexes formed tend to be smaller in size and
merular injury is seen in a wide spectrum of soluble. These small immune complexes may be
29
Immune-Mediated Glomerular Injury in Children

Fig. 7 Mechanism of immune deposition and inflammatory reaction in the glomerulus. accumulate at subepithelial area (d). Antigen-antibody complex in the circulation
(a) Circulating immune complex interaction with endothelium results in deposition in deposits on the endothelial surface is an initiation of immune-mediated glomerular
GBM. (b) In case of anti-GBM antibody glomerulonephritis (Goodpasture syndrome), injury. Local activation of complement system chemoattracts inflammatory cells
fixed antigen is a component of GBM (NC1 domain of α3 (IV) chain of type IV followed by secretion of secondary messengers which injure glomerular component
collagen). Thus antibody binds GBM and shows linear distribution. (c) In case of (The figure was published in Kumar et al. Robbins basic pathology, 8th edn. 2007,
membranous glomerulonephritis (Heymann nephritis), antigen is a podocyte compo- p. 545 and 127, Copyright Elsevier)
nent (megalin). Antigen-antibody complex is formed in situ and complex shift to
895
896 M. Nagata

deposited within the GBM or subepithelial areas, Another example of an in situ immune complex
but they often fail to induce inflammation, disease is Heymann nephritis, which serves as a
because the deposition and complements are iso- model of membranous glomerulonephritis in
lated from circulation. When the antigen to anti- humans. Combine this model with isolated per-
body ratio nears equivalence, the immune fused kidney system provided first evidence for
complexes formed tend to be of maximal size in situ immune complex-induced glomerulonephri-
and insoluble. The net charge of the complex is tis [10]. Autoantigenic target in this model is
an additional determinant of immune complex 516 kD protein megalin that binds receptor-
deposition, based on the interactions with the neg- associated protein (RAP), a podocyte-membrane
atively charged GBM and endothelial surface. glycoprotein [11]. In humans, megalin is not
The levels of circulating immune complexes expressed, but phospholipase A2 receptor has
have been shown to correlate with glomerular been identified as the antigen in human membra-
injury in lupus glomerulonephritis. The size and nous glomerulonephritis [5].
biochemical characteristics of immune complexes In clinical practice, various antibodies are
may change with disease activity or with thera- detected in the blood, while circulating immune
peutic modulation. Indeed, immune complex complexes are generally undetectable. It seems
deposition in subendothelial or mesangial cells that in situ immune complex formation is the
occasionally shifts to the subepithelial cells in distinct process for immune deposition in various
lupus nephritis, such as Class IV to Class V trans- glomerular diseases in humans.
formation. Acceleration of glomerular capillary
permeability in the process of glomerulonephritis
may be an alternative factor to change the site of Cell-Mediated Immunity
immune deposition. Although experimental
models suggest induction of glomerulonephritis Cell-mediated immunity can be summarized as
by glomerular immune complex deposition, it the T-cell-dependent interaction of antigen-
remains unclear as to whether passive trapping presenting cells and CD4+ helper T cells through
of preformed immune complexes alone is suffi- the T-cell receptor (TCR), which promotes cyto-
cient to induce glomerulonephritis in humans. kine release and stimulates the proliferation and
activation of effector cells, macrophages, and
CD8+ cytotoxic T cells [12].
In Situ Immune Complexes T-cell-mediated glomerular injury includes sev-
eral mechanisms, such as T-cell-mediated B-cell
In situ immune complex formation is basically the activation, cytotoxic effects, and the direct actions
result of the combination of unbound circulating of lymphokines on the glomerular filtration barrier.
soluble antibody and antigen within the tissue. In The first process is T-cell-driven antibody-mediated
situ tissue antigens are either fixed (components immunity as described above, and the latter two
of the glomerulus) or planted (exogenous) processes are narrowly defined, basically because
(Fig. 7). The best example of an in situ immune of the paucity of antibody deposition in situ.
complex disease is anti-GBM antibody-induced Inappropriate T-cell responses may cause hyper-
glomerulonephritis (anti-GBM antibody nephritis). sensitive immune reactions and provoke glomeru-
In this condition, antibodies are directed against lonephritis. In humans, T-cell infiltration is
a fixed antigen of a component of the GBM, observed in the glomeruli in pauci-immune glo-
i.e., the noncollagenous domain of the α3 chain of merular diseases, such as ANCA-related glomeru-
collagen type IV. These antibodies occasionally lonephritis, and therapeutic interventions suppress
cross-react with the lung alveolar basement mem- glomerular inflammation and T-cell infiltration.
brane, and the resulting simultaneous inflammation Anti-GBM antibody nephritis in Wistar Kyoto
of the kidneys and lungs represents a severe clinical rats revealed infiltration of CD8+ T cells in the
phenotype, known as Goodpasture syndrome. glomeruli, and suppression of circulating CD8+ T
29 Immune-Mediated Glomerular Injury in Children 897

cells resulted in decreased glomerular inflamma- in promoting Th2 responses. Differences in che-
tion. However in mice model of anti-GBM anti- mokine receptor expression between Th1 and Th2
body nephritis, glomerular injury is CD8+ T cell cells also influence the activation patterns of these
independent. These observations provide circum- cells by different chemokines. CXCR3, the recep-
stantial evidence to establish the basis of a T-cell- tor for INF-γ-inducible chemokines, is expressed
dependent glomerular injury mechanism. Transfer at high levels on INF-γ-producing Th1 cells and at
of rat CD4+ cell line specific for Col4α3NC1 suc- low levels on Th2 cells. The CCR3 and CCR4
cessfully induced severe nephritis without glomer- chemokine receptors are expressed by Th2 cells
ular IgG and C3 deposition, which may be the that produce IL-4. Th17 cells are differentiated
direct evidence for T-cell-mediated glomerular from naïve CD4+ T cells by TGF-β and IL-6 and
injury [13]. Conversely, T-cell-mediated injury produce IL-17 which promotes autoimmune dis-
cannot be excluded, even when antibodies ease, such as multiple sclerosis and rheumatoid
(reflecting a B-cell response) are present. arthritis. Th17 cells also secrete IL-21, IL-22, and
IL-9.
The roles of Th1 and Th2 cells in glomerular
T-Cell Component of Adaptive injury have been studied using different strains for
Immune Response Th1 (mice: C57BL/6, rats: Lewis) and Th2 (mice:
BALB/c, rats: Brown Norway) dominance.
The complex immunologic network-driven Induction of nephrotoxic serum nephritis in
immune-mediated glomerular injury involves the Th1-predominant C57BL/6 mice results in severe
primed CD4+ helper T-cell-derived subsets, glomerular injury. Anti-GBM antibody nephritis
including Th1, Th2, Th17, and Treg subsets. in Lewis (Th1) and Brown Norway (Th2) rats
Th1 and Th2 cells can be distinguished based on resulted in more severe glomerulonephritis, with
their cytokine (Th1; INF-γ, Th2; IL-4), chemo- monocytic and T-cell influx and Th1-profile cyto-
kine, and antibody isotype profiles and on which kine production, in Lewis rats than in Brown
generate different immune effector responses. Norway rats. Mice deficient in Th2 cytokines
Th17 was the newly identified subset by expres- show more pronounced crescentic glomerulone-
sion of IL-17A and IL-17F. These cell types dif- phritis, while the administration of Th2 cytokines
ferentiate from naïve precursors, “Th0” cells, ameliorates this disease. Mercuric chloride-
following specific antigen stimulation by pleiotro- induced autoimmune glomerulonephritis in
pic cytokines via their α/β T-cell receptors congenic strains of Brown Norway rats resulted
(Fig. 8). Antigen-presenting dendritic cells in in a membranous-like form of glomerulonephritis.
humans are derived from monocytes or These experiments indicate that Th1 predomi-
plasmacytoid cells, with a bias towards the stim- nance is related to crescentic glomerulonephritis
ulation of either INF-γ or IL-4, IL-5, and IL-10 and that Th2 predominance tends to be involved
production by naïve T cells, as demonstrated in membranous glomerulonephritis.
in vitro (Table 2, Fig. 6). Although every pathway In human anti-GBM antibody nephritis, there
may interact, the predominance of a particular is an INF-γ-predominant, antigen-specific effector
subset may explain the different patterns of glo- cell response in the active stage and IL-10 pre-
merular histology and disease activity, which are dominance in remission. In humans, CD4+ cells
related to disease outcome [15]. and macrophages are present in the cellular cres-
Th1 development is promoted primarily by cents of crescentic glomerulonephritis. In ANCA-
INF-γ and IL-12, and partially by IL-18, IL-27, related glomerulonephritis, a T-cell response with
and IL-23, indicating complex and overlapping Th1 predominance is involved. In the renal tissues
mechanisms for Th1 immunologic activities. of ANCA-related glomerulonephritis, the level of
Th2 responses are driven by IL-4 and IL-10, and INF-γ mRNA is high, and the level of IL-4 mRNA
they inhibit Th1 responses. IL-10 appears to be is low. Peripheral blood T cells show a high INF-γ/
more active in inhibiting Th1 differentiation than IL-4 ratio in patients with ANCA-associated
898 M. Nagata

NaÏve CD4+ T cell

TGF-β

IFN-γ, IL-12 IL-4

IL-6, IL-23 Retinoic acid

STAT1+4 STAT6 STAT3 FoxP3


T-bet GATA3 RORγ t

Th1 Th2 Th17 Treg


Intracellular bacteria Helminth infections Extracellular Downregulation of
Cell-mediated Allergic disease bacteria, fungi the immune response
autoimmunity Cell-mediated in autoimmunity and
IL-4, IL-5, IL-13 autoimmunity infection
IFN-γ, TNF-α
TGF-β, IL-10, IL-35
IL-17, IL-17F, IL-21
IL-22, TNF-α

Fig. 8 Differentiation of CD4t T-cell subsets. After the subtypes show differential activation of transcriptional
encounter of antigen and costimulatory molecules programs and are characterized by the secretion of certain
presented by antigen-presenting cells, naive CD4t T cells effector cytokines that enable them to provide protection
can differentiate into different subpopulations. The lineage against different classes of pathogens and to mediate auto-
commitment of CD4t T cells depends on the cytokine immunity. IFN-γ, interferon-γ; IL, interleukin; TGF-β,
milieu accompanying the T-cell activation process in sec- transforming growth factor-β; Th1, T-helper 1 cell;
ondary lymphoid organs. The diverse T-helper cell TNF-α, tumor necrosis factor-α [14]

glomerulonephritis, as compared with patients and Th2 polarity. Protection of anti-GBM nephri-
with nonproliferative glomerulonephritis. Corti- tis in CD28 deficient mice is due to inhibition of
costeroids diminish this ratio, which suggests autologous antibody production [16]. Selective
that Th1 predominance is linked to ANCA- blockade of B7-1 prevented the development of
associated glomerulonephritis in humans. crescentic glomerulonephritis in anti-GBM
For full activation of CD4+ cells, engagement nephritis in Wistar Kyoto (WKY) rats [17]. In
of T-cell receptor with the antigenic peptide-MHC the same nephritis in mice, administration of
molecule complex in antigen-presenting cells either CD80 or CD86 revealed no effect on
needs secondary costimulatory signals for full glomerulonephritis; however, administration of
activation. T-cell costimulatory molecules include anti-CD80/CD86 antibody attenuated glomerulo-
CD28-B7 family/CD80 (B7-1) + CD86 (B7-2) nephritis [18]. Anti-GBM antibody nephritis in
and CD40 (TNF receptor on B cells)/CD154 CD80 (B7-1)-deficient mice reveals attenuation
(ligand of CD40 on T cells) that determines Th1 of crescentic glomerulonephritis in concert with
29 Immune-Mediated Glomerular Injury in Children 899

reduction of glomerular CD4+ cell accumulation. thus, failure of Treg to control T-cell activity can
By contrast, same nephritis in CD86 (B7-2)-defi- provoke autoimmunity. Transfer of CD4+CD25
cient mice shows disease exacerbation [19]. Thus Treg inhibited murine anti-GBM antibody nephri-
CD80 is pathogenic in crescentic glomerulone- tis via T-cell and macrophage suppression, with
phritis by enhancing survival and proliferation of significant reductions in the levels of INF-γ,
CD4+ cells, whereas CD86 is protective by TNF-α, and TGF-β mRNA. This may be the
enhancing Th2 and attenuating Th1 responses. mechanism of the protective roles of Treg in
Inducible costimulatory molecule (ICOS) and autoimmune-mediated glomerulonephritis. How-
ICOS ligand (ICOSL) are a receptor-ligand pair ever, Treg transfer to lupus-prone mice
that belongs to the CD28/B7 family. Administra- suppressed autoantibody production but not glo-
tion of antibody against ICOSL in mice with merulonephritis [24]. Treg suppresses Th1 and
anti-GBM nephritis increased glomerular accu- Th17 cells by secreting IL-10, and Treg deletion
mulation of T cells and macrophages without aggravated T-cell-mediated glomerulonephritis
affecting systemic immune response [20]. through INF-γ induction and promotion of
This suggests that ICOSL plays protective roles delayed-type hypersensitivity [25, 26]. The
during induction of anti-GBM nephritis by locally T-cell-mediated immune mechanisms of glomer-
reducing accumulation of T cells and macro- ulonephritis have been analyzed in an acute
phages. Blockade of CD154-CD40 costimulatory nephritis model, and the role of T cells in chronic
signals in anti-GBM antibody nephritis in WKY glomerulonephritis, particularly in humans,
rats prevented glomerulonephritis [21]. In addi- remains largely unknown.
tion, anti-GBM nephritis in CD40 chimeric mice
(CD40 is absent in glomeruli but intact in bone
marrow) revealed suppression of glomerulone- Inflammatory Cells
phritis accompanied by reduced level of MCP-1,
IL-10, mRNA, and T-cell and macrophage influx. Leukocytes
This indicates that CD40 expression in intrinsic
glomerular cells promotes Th1 effector cell Leukocytes are bone marrow-derived cells that
response in glomerulonephritis [16]. include granulocytes, lymphocytes, and mono-
Comparing to Th1 and Th2 subsets, the role of cytes/macrophages. Granulocytes include neutro-
Th17 cells on the glomerulonephritis is under phils, eosinophils, and basophils. Neutrophils
investigation. Th17-derived IL-21, IL-22, and (polymorphonuclear cells, PMNs) and macro-
IL-9 induce proinflammatory cytokines and phages play central roles in the acute inflamma-
chemokines in resident cells resulting in recruit- tion process. Neutrophils and macrophages have
ment and activation of leukocytes and macro- common effects in terms of tissue injury, but mac-
phages in situ [14]. Although presence of Th17 rophages are more actively involved in glomeru-
cells in the interstitial nephritis and induction of lar injury by their immunologic functions.
cytokines and chemokines by tubular epithelial Usually, PMNs predominate in the acute phase
cells in vitro has been noted earlier, its importance of glomerulonephritis, e.g., in acute poststrep-
in glomerulonephritis was firstly shown by atten- tococcal glomerulonephritis and ANCA-related
uation of nephrotoxic nephritis in IL-17 knockout glomerulonephritis. Macrophages appear in
mice. Transfer of Th-17-polarized ovalbumin- acute and chronic glomerulonephritis, and both
specific CD4+ effector cells induced proliferative cell types are sometimes copresented in the two
glomerulonephritis [22]. In addition, cross- phases of glomerulonephritis, particularly acute
regulation between Th17 cells and Th1 cells was phase on chronic glomerulonephritis.
suggested by nephrotoxic nephritis in mice In the variety of pathogeneses of glomerulone-
lacking Th1 response [23]. phritis, leukocytes are involved in the loss of
Regulatory T cells (Treg) suppress effector T immune tolerance, T cell-directed adaptive
cells through receptor-mediated mechanism, and immune responses, cellular effectors inducing
900 M. Nagata

injury in delayed-type hypersensitivity-like reac- glomerular permeability, with consequent break-


tions, and macrophage/neutrophil recruitment via down of the ECM, which links mesangiolysis and
the deposition of circulating or in situ immune GBM changes, and injuring the intrinsic cells.
complexes through receptor-mediated fashion Activation of antioxidant mechanisms that
[27]. Leukocytes express Fc-γ receptor on cell involve catalase, superoxide dismutase, and glu-
surface, and it binds the Fc portion of IgG, and tathione peroxidase ordinarily protects tissues
lack of Fc-γ R protects immune complex- from oxidant stress. The role of ROS in glomeru-
mediated glomerulonephritis [27]. In addition, lar injury is separately discussed.
ANCA have been postulated to activate neutro- A good example of neutrophil-mediated glo-
phils, leading to glomerular capillaritis. merular injury is ANCA-related glomerulonephri-
tis [28]. ANCA activates cytokine-primed
neutrophils and monocytes through Fab’2 binding
Polymorphonuclear Cells and Fc receptor engagement in vitro. These PMNs
adhere to endothelial cells and release ROS, pro-
PMNs are implicated in glomerular injury owing teolytic enzymes, and inflammatory cytokines,
to their numerous bioactive/toxic products. PMNs which promote cytotoxic tissue damage. In addi-
are chemoattracted by anaphylatoxin (C5a and tion to these danger enzymes and ROS, comple-
C3a), CXC chemokines, the immune adherence ment- and cytokine-induced neutrophils
of complement receptors, and Fc receptor- occasionally form extracellular fibrillar networks
dependent binding to antibodies. The called neutrophil extracellular traps (NETs). The
corresponding receptors include FcR for IgG, trap is composed of nuclear chromatin with
CR1, CR2, and CR3 for complement fragments embedded granule proteins which are antimicro-
and C1q for the collectins. Locally activated bial peptides and enzymes. Anti-MPO CD4 cells
PMNs express sialyl-Lewis X-modified proteins recognize MPO as a planted glomerular antigen
and interact with endothelial cells, through and act with macrophages to amplify severe glo-
selectin or integrin ligands on the endothelial sur- merular injury [29]. It has been suggested that
face. Various cytokines stimulate endothelial cells NETs involve tuft necrosis in ANCA-related
to express these adhesion molecules (Table 2). glomerulonephritis [30].
Occasionally, PMNs on endothelial cells migrate Intravenous injection of mouse antibodies or
to the mesangium and stimulate cells there, splenocytes specific for mouse MPO alone
through bioactive or toxic products. Among the induces necrotizing crescentic glomerulonephritis
PMN-derived substances, lysosomal enzymes and in Rag2 mice (lack of functioning B cells and T
reactive oxygen species (ROS) are the most harm- cells). In this model, it appeared that PMNs’ infil-
ful for the glomerulus. Lysosomal enzymes tration was conspicuously associated with glo-
include metalloproteases and other proteases, merular necrosis and crescent formation with
which along with cathepsin G exert proteolytic macrophages, whereas lymphocytes were not
activities and damage endothelial cells or promote prominent. Mice depleted of circulating PMNs
lysis of the GBM and mesangium. Microbes, with rat monoclonal antineutrophil antibody
cytokines, immune complexes, and various (NIMP-R14) were completely protected from
inflammatory stimuli promote ROS synthesis in anti-MPO IgG-induced necrotizing crescentic
PMNs through the NADPH oxidase pathway. glomerulonephritis [31]. Local pretreatment of
ROS ordinarily function to destroy phagocytosed cremaster muscle with TNF-α followed by
microbes and bring about cell death. Low-level systemic administration of anti-MPO IgG in
secretion of ROS accelerates the inflammatory wild-type mice leads to enhancement of
cascade by upregulating the expression of cyto- leukocyte-endothelial cell interaction in the
kines, chemokines, and adhesion molecules. At cremasteric microvessels, whereas this effect
high levels, ROS damage endothelial cells, acti- was not seen in Fc receptor γ chain-deficient
vating the coagulation cascade and increasing mice. In addition, coadministration of anti-MPO
29 Immune-Mediated Glomerular Injury in Children 901

Fig. 9 Macrophage infiltration in IgA glomerulonephritis. mesangial proliferation and less in the capillary (PAS
(a) Immunohistochemical detection of macrophages by stain), (b) EM reveals monocytes/macrophages in the
CD68 in IgA glomerulonephritis. Note many mesangium and in the capillary (arrowhead)
immunolabeled cells (brown) locate predominantly in the

antibody with anti-CD18 antibody (CD18; INF-γ. Examination of anti-Thy1 nephritis reveals
integrin β2) has no effect on the local leukocyte- that infiltration per se is insufficient to promote
endothelial cell interaction [32]. This indicates macrophage programming, but this is completed
Fc-γ receptor and integrin β2 are important for shortly after localization to the appropriate local
processing MPO-induced small vessel vasculitis. microenvironment.
Immunization of MPO in mice with bone marrow Human renal biopsies and experimental
transplantation of MPO-positive cells to models of glomerulonephritis reveal glomerular
MPO-deficient recipient (chimera) developed macrophage accumulations, which may be indic-
crescentic glomerulonephritis, whereas chimeric ative of disease activity or severity. IgA glomeru-
mice of MPO-negative bone marrow cells into lonephritis in children represents relatively acute
wild-type mice did not [33], indicating that bone inflammation, and the mesangial influx of macro-
marrow cells are the necessary target to mediate phages leads to mesangial proliferation, as dem-
MPO antibody-induced crescentic glomerulone- onstrated by electron microscopy and
phritis. The local inflammation induced by cyto- immunohistochemistry (Fig. 9). Local activation
kine (e.g., TNF-α)-primed PMNs and anti-MPO of macrophages also occurs in various glomerular
antibody via FcR-γ effectively promotes cyto- diseases, including nonimmune-mediated condi-
toxic endothelial cell damage and activates coag- tions, such as diabetic glomerulosclerosis.
ulation cascade. This occasionally results in Macrophages produce a wide range of
fibrinoid necrosis followed by the breakdown of potentially cytotoxic products, including
GBM and subsequent crescentic formation. proinflammatory cytokines and chemokines, pro-
teolytic enzymes, ROS, eicosanoids, and growth
factors (Table 4, Fig. 10). Experiments on the sup-
Monocytes/Macrophages pression or depletion of macrophages in acute and
chronic glomerulonephritis models have demon-
Macrophages are derived from circulating blood strated reduced glomerular injury [34, 35]. In cres-
monocytes. In normal rat mesangial areas, tissue- centic glomerulonephritis in rats, liposome-MDP
resident macrophages are occasionally present, (dichloromethylene diphosphonate) attenuated
but they are uncommitted. When inflammation glomerular macrophage influx accompanied by
occurs, monocyte-derived macrophages are suppression of CD8 and expression of adhesion
recruited, programmed, and activated, largely by molecule. These findings circumferentially support
902 M. Nagata

Table 4 Macrophage activation status


Status Alteration Product
Classically activated (M1) Upregulation iNOS, IL-1β, TNF-α, IL-6, HMGB1, IL-8, MCP-1, MHC class I
Alternatively activated (M2) Upregulation Mannose receptor, MHC class II, arginase, FIZZ1/Ym1, IL-ra
Downregulation iNOS, TNF-α, IL-6
IL10 activated Upregulation Soluble TNF receptors, IL1ra
Downregulation IL-b, TNF-α, IL-12, IL-6, IL-8, iNOS, MHC class I
Type II activated Upregulation IL-10, TNF-α, IL-6, MHC class I
Downregulation IL-12
Uptake of apoptotic cells Upregulation TGF-b, PGE2
Downregulation TNF-α, IL-8, MCP1
Steroid treated Upregulation IL-10, uptake of apoptotic cells
Downregulation IL-12, TNF-α, IL1-β, IL-6

Fig. 10 Activation of Circulating Adherent Emigrating


macrophages and monocyte
production of various
cytokines. Circulation
monocytes transmigrate to
the site of inflammation,
and T-cell-derived
cytokines (INF-γ) stimulate
activation in monocytes/
macrophage and produce
many soluble factors for
tissue cell injury/tissue
fibrosis. In the glomerulus, Tissue macrophage
macrophages are able to
migrate into mesangium,
subendothelial space, or Immune
urinary space (The figure response:
was published in Kumar activated T cell
et al. Robbins Basic
Pathology, 8th edn. 2007, Activation Cytokine
p. 55, Copyright Elsevier) by microbes, (IFN-γ)
dead cells,
etc.
Activated macrophage

Tissue injury and Fibrosis


inflammation • Growth factors
• Reactive oxygen and (PDGF, FGF, TGFβ)
nitrogen species • Fibrogenic cytokines
• Proteases • Angiogenesis factors
• Cytokines, including (FGF)
chemokines
• Coagulation factors
• AA metabolites
29 Immune-Mediated Glomerular Injury in Children 903

Monocyte

Circulation

Injured tissue
Innate activation by TRL ligands
Increased production of
proinflammatory cytokines,
Macrophage iNOS and ROS

M2 polarization M1 polarization

Alternative activation by IL-4 or IL-13 Classical activation by IFN-γ TNF-α


Increased endocytic activity; increased and LPS
expression of mannose receptor Increase production of
dection-1 and arginase; increased cell proinflamamtory cytokines, iNOS and
growth; increased tissue repair; ROS: increased expression of MHC
increased parasite killing class II molecules and CD86;
increased antigen presentation;
increased microbicidalactivity

Fig. 11 Macrophage heterogeneity during inflammation. activity and produce ROS and iNOS for proinflammatory
Monocytes attracted to the injured tissue are thought to be action. By contrast M2 polarization is promoted by IL-4,
able to acquire distinct phenotypes and physiological func- IL-10, IL-13, or TGF-β, and it acted on the tissue repair and
tions. Largely there are two major phenotypes in macro- parasite killing. However, this hypothesis has been drawn
phages, M1 and M2. In M1 polarization, macrophages are by the in vitro experiments and still remains undetermined
stimulated with INF-γ and reveal high microbicidal in vivo (Modified figures from Gordon et al. [37])

the role of macrophages in the glomerulonephritis. proinflammatory cytokines, NO, and ROS. INF-γ
In this context adoptive transfer of macrophages in TNF-α and/or LPS primes macrophages to convert
leukopenic rats with anti-GBM nephritis revealed classical activation (M1 polarization) and leads to
proteinuria and glomerular cell proliferation and increased production of proinflammatory cyto-
indicates direct evidence for macrophage to induce kines, iNOS, and ROS, as well as the upregulation
glomerulonephritis [36]. Given these results, it has of MHC class II molecule expression and
long been suggested that the scale of macrophage costimulatory CD86 molecules, thereby promoting
influx promotes glomerular injury. However, it has antigen presentation. In contrast, IL-4- or IL-13-
recently become clear that it is the functional prop- primed macrophages undergo alternative activa-
erties, rather than the total number, of macrophages tion (M2 polarization), which is associated with
that determine the severity of inflammation. In increases in endocytosis, arginase, anti-
vitro studies have led to the hypothesis of func- inflammatory cytokines, and the synthesis of extra-
tional heterogeneity in macrophages. Polarized cellular matrix, thereby promoting angiogenesis.
macrophages can be subdivided into several phe- M2 polarization also leads to decreased iNOS and
notypes, particularly classically activated (M1) and NO production and is involved in anti-
alternatively activated (M2) macrophages inflammatory effects, cell growth, and tissue repair.
[37]. The difference in activators determines the Recent findings suggest that M2 macrophages are
heterogeneous phenotype (Fig. 11). Innate activa- induced by IL25 resulting protection in renal dis-
tion is promoted by Toll-like receptor (TLR) eases [38]. This phenotypic heterogeneity of mac-
ligands, which induce the production of rophages may be switched during the course of
904 M. Nagata

chronic glomerulonephritis, depending on the mortality rate and enhanced disease activity, as
background pathophysiology. compared with wild-type mice [41]. This condi-
Macrophage migration inhibition factor (MIF) tion can be rescued by mast cell reconstitution,
is a kind of proinflammatory cytokine to induce suggesting a protective role for mast cells in
cytokines, chemokines, and adhesion molecules immune complex-mediated glomerulonephritis.
in the inflammatory environment. Antagonist for
MIF resulted in the prevention of lupus nephritis
[39]. Activator protein (AP-1) transcription factor Toll-Like Receptors (TLRs)
JUND is a major determinant of macrophage
activity and associated with susceptibility to glo- The recently identified TLR families provide
merulonephritis [40]. During the process of glo- receptor-mediated pathogen recognition and acti-
merular injury, macrophages take up apoptotic vation of the immune system. Ten human and nine
cells, most of which are infiltrated inflammatory murine TLR proteins related to the original Dro-
cells. This uptake causes the macrophages to exert sophila Toll gene have been identified to date.
anti-inflammatory effects, through increased Inflammatory cytokines and chemokines are
expression of TGF-β and prostaglandin E2 induced through the activation of TLRs by a vari-
(PGE2) and decreased production of IL-8, ety of ligands. The biologic activities that result
TNF-α, and IL-1β. Suppression of T-cell prolifer- from TLR binding include activation of antigen-
ative responses also results from the anti- presenting cells, dendritic cell maturation, B-cell
inflammatory effects of macrophages that have activation, and different cytokine production pro-
taken up apoptotic cells. files. The responsibility of TRL in the innate and
adaptive immune response may be a likely back-
ground in the immune-mediated glomerular
Mast Cells injury. Small nuclear RNA and autoantigen for
lupus activate B cells and dendritic cells via
Mast cells are bone marrow-derived cells that play TLR-7. TLR-7 overexpression is associated with
significant roles in innate and adaptive immune antinuclear autoantibody production and lupus-
responses. Mast cells secrete various inflamma- like disease in mice, and backcrossing TLR-7-
tory mediators, including chemical vasomotor deficient mice with MRL lpr/lpr mice attenuated
mediators, cytokines, growth factors, and eicosa- kidney and lung disease. Administration of syn-
noids. These substances attract inflammatory cells thetic oligodeoxynucleotides with immunoregula-
and are involved in mediating tissue remodeling. tory sequences (IRS) which specifically blocks
Mast cell detection in tissues is typically TLR-7 suppressed autoimmune kidney and lung
performed by staining with toluidine blue, Alcian diseases. Endogenous or exogenous CpG-DNA
blue, and safranin, all of which enable the visual- activates TLR-9 and administration of CpG
ization of cell granules. Immunohistochemistry to DNA to MRL-Fas(lpr) mice resulted in disease
detect enzymes specific for mast cells, such as progression accompanied by MCP-1 and
tryptase and chymase, can be used to subdivide RANTES expression and leukocyte recruitment
mast cells into MCT (containing tryptase) and [42]. In ddY mice of spontaneous IgA glomeru-
MCTC (containing tryptase plus chymase) cells. lonephritis, genome-wide scan revealed myeloid
This enzyme visualization method reveals many differentiation factor 88 (MyD88) as a candidate
more mast cells than classic toluidine blue gene for disease progression, and TRL-9, the
staining. Mast cells are present in human glomer- receptor for MyD88, was increased in a conven-
ulonephritis, primarily in the interstitium, and are tionally housed condition. Nasal challenge with
probably involved in fibrosis. The role of mast CpG oligonucleotide (a ligand for TRL-9) aggra-
cells in glomerulonephritis has been examined in vated renal injury with strong Th1 polarization
anti-GBM antibody nephritis in congenitally mast and increased mesangial IgA. TRL-9 polymor-
cell-deficient mice (W/Wv) showing a higher phism in the TLR-9 gene associated with the
29 Immune-Mediated Glomerular Injury in Children 905

disease progression of IgA in humans. TLR-9 activation of complement system that is involved
pathway may be involved in the pathogenesis in the pathogenesis of glomerular injury. Classi-
and determination of severity of IgA glomerulo- cally, inhibition of the complement cascade by
nephritis [43]. HSPN and IgA glomerulonephritis cobra venom factor attenuated various glomeru-
in children showed upregulation of TLRs and lonephritis models. Complement depletion, by
Treg depression in peripheral blood cells, aggregated human IgG, protected against
suggesting autoimmune proteasome switch in complement/PMN-dependent glomerular injury
these diseases [44]. in anti-GBM antibody nephritis, with marked
TLR-3 recognizes dsDNA of viral origin and is attenuation of leukocyte influx. In addition, defec-
expressed in mesangial cells. Cytokines stimulate tive complement regulation system, as seen in
TLR-3 mRNA in mesangial cells in vitro. human and animal models with specific comple-
TLR-3mRNA is significantly upregulated in glo- ment component deficiencies, has provided evi-
meruli associated with enhanced mRNA of dence of a complement-mediated/complement-
RANTES/CCL5 and MCP-1/CCL2 in hepatitis dependent pathway in glomerular injury. This
C-associated glomerulonephritis in humans. This includes certain forms of hemolytic uremic syn-
suggests that immune complex containing viral drome and MPGN type II [46].
RNA activates mesangial cells via TLR-3-medi- The complement system consists of three dis-
ated chemokine/cytokine effects [45]. tinct pathways: the classical, mannan-lectin, and
alternative pathways (Fig. 12). These pathways
sometimes are synergistically involved in the glo-
Complement Cascade merulonephritis, such as simultaneous activation
of lectin pathway and alternative pathway in IgA
The complement system, which is a cascade of glomerulonephritis and purpura nephritis [47].
plasma proteins, involves approximately Of the four classical types of hypersensitivity
30 plasma and membrane-bound proteins that reactions, complement is thought to be involved
play a pivotal function in antimicrobial defense in types II and III. Type II hypersensitivity and
and immune complex clearance. Complement antibody-mediated hypersensitivity target intrin-
components are largely liver derived and are pre- sic glomerular cells or extracellular matrix, pro-
sent in inactive forms (numbered C1 through C9) voking tissue damage by a complement-
in the plasma, and local complement production dependent mechanism. This mechanism typically
occurs in the inflammatory milieu involved in includes Goodpasture syndrome and Heymann
tissue damage, including leukocyte recruitment, nephritis. In type III hypersensitivity, antigen-
cell necrosis, and apoptosis. Immune complex antibody complexes within the glomerulus (glo-
clearance and proinflammatory functions of com- merular capillary endothelium or mesangium) and
plement system provide its complex participation complement bind the C3b receptor or the Fc
in immune-mediated glomerulonephritis. receptor of immune-competent cells and activate
The biologic actions of complement ultimately the production of chemical mediators or cytokines
generate a pore-like membrane attack complex that lead to tissue injury. Lupus nephritis and
(C5-9, MAC), which punches holes in invading poststreptococcal acute glomerulonephritis are
microorganisms. Among the several important promoted by this mechanism.
complement fragments produced in the The complement activation-involved glomeru-
MAC-generating processes are the cleavage prod- lar injury is thought to be mediated by leukocyte
ucts of C3 and C5 (C3a, C5a), collectively termed recruitment via C5a, which accelerates the inflam-
anaphylatoxin, which promote vascular perme- matory process (leukocyte dependent) or through
ability (C3a, C5a) and leukocyte chemotaxis and direct attack by complement fragments on intrinsic
activation (C5a). glomerular components (leukocyte independent).
The presence of complement components in Leukocyte-dependent tissue injury mecha-
the tissue of glomerular diseases represents local nisms are involved in injury to the glomerular
906 M. Nagata

Fig. 12 Schematic depiction of the three pathways of damaged cells, and IgA. Cleavage of C3 produces C3b,
complement activation as they relate to GN. In the adaptive which combines with factor B and properdin to form the
immune system, complement-fixing antibodies (IgG1, alternative pathway C3 convertase that is regulated by
IgG3, IgM) in immune complexes and CRP initiate classic factors H and I to prevent excess C3 activation. Both
pathway activation through C1q, C4, and C2 to form classic and alternative pathway C3 convertases cleave C3
C4b2a, the classic pathway C3 convertase. In the innate leading to release of C3b, which allows C5 convertase
immune system, PAMPs and DAMPs activate complement formation. Cleavage of C5 produces the chemotactic factor
through the MBL or alternative pathways. Some infectious C5a and C5b, which combines with C6, C7, C8, and
PAMPs and DAMPs bind MBL leading to activation of multiple C9 molecules to form the lipophilic C5b-9 mem-
MBL-associated serine proteases (MASPs), which activate brane attack complex that can activate resident glomerular
C3 via C4, C2, and the classic pathway C3 convertase. In cells to become effector cells. C5b-9 formation is regulated
the alternative pathway, direct activation of C3 occurs by cell-bound CRPs such as CD59 [1]
spontaneously (“C3 tickover”) and by foreign surfaces,

capillary. C5a affects the chemotaxis of neutro- and C5L2, and most of the functional effects of
phils, monocytes/macrophages, and eosinophils C5a are mediated by C5aR. Transplantation of
and accelerates the release of inflammatory medi- C5aR-deficient marrow suppressed glomerulone-
ators. C5a also increases the adhesion of leuko- phritis in MPO-deficient mice immunized with
cytes to endothelium, by activating the leukocytes MPO, indicating that C5a involves leukocyte acti-
and increasing the avidity of the surface integrin vation via C5aR [49]. This inflammatory process
for the ligands C3b and C3bi deposited on endo- involves GBM breakdown and activation of the
thelial surface. C5-deficient mice show reduction coagulation system, due to endothelial injury;
of glomerular PMNs infiltration in cryoglobulin- leads to proliferative and matrix changes in the
induced immune complex glomerulonephritis mesangium and parietal cells; and accelerates
[48]. Complement anaphylatoxins bind several podocyte damage. MAC stimulates NF-κB,
receptors including G protein-coupled receptors which is linked to the activation of the
(C3aR and C5aR). C5a binds two receptors, C5aR proinflammatory cytokines IL-8 and MCP-1.
29 Immune-Mediated Glomerular Injury in Children 907

In humans, C3aR is upregulated in the glomerular the MAC of complement, inhibits at the level of
endothelial region in lupus nephritis, but not other C8 activation, preventing MAC formation.
glomerular diseases [50]. Glomerular-resident cells normally express MCP
Leukocyte-independent mechanism of glomer- and CD59, and these molecules are upregulated in
ular injury is seen in Heymann nephritis. Like glomerular disease. Nephrotoxic serum nephritis
type II mechanisms, antigen (megalin) on in mice deficient for either DAF1 or CD59a or
podocyte surface is a target of complement cyto- both DAF1 and CD59a genes reveals that mice
toxicity and results in direct injury for filtration lacking DAF1 gene developed severer proteinuria
barrier and complement depletion-attenuated and histology compared to wild-type or CD59a-
Heymann nephritis. In addition, sublyric levels deficient mice. Severe disease in DAF1 gene defi-
of MAC damage the DNA in podocytes [51]. ciency is associated with glomerular C3 and C9
Glomerulonephritis in complement deficiency deposition, whereas immunoglobulin deposition
provides a clue to understand the roles for com- is comparative. Crry is a rodent homologue with
plement in glomerular injury. C6-deficient/C6- activities similar to DAF and MCP. Blocking
depleted rats with anti-Thy1 nephritis or CD59 or Crry accelerates complement-dependent
Heymann nephritis show attenuated disease activ- glomerulonephritis, and the soluble forms of these
ity. By contrast occurrence of IgA glomerulone- molecules effectively inhibit complement and
phritis even in the hereditary C6 or C9 deficiency attenuate glomerular damage.
in humans indicated that MAC per se is not essen- Alternative mechanism of complement-
tial in the IgA glomerulonephritis. dependent glomerular injury is defective comple-
Interestingly, C1q or C4 deficiency develops ment regulatory proteins. Protective proteins
lupus-like disease in mice, and C1q, C2, and C4 against the complement cascade include
deficiency in humans increased risk of developing factor H, factor H-like protein, factor I, and C4
SLE. Although the phenomenon may contradict binding protein, and functional defects in these
to the many literatures suggesting complement proteins are the background. Factor H is a soluble
system injures the glomerulus, it may be glycoprotein that regulates complement, both in
explained by the scenario that complement defi- the fluid phase and on the cellular surface. It acts
ciency may lead to autoimmune phenomena due in the amplification loop of the alternative path-
to defective clearance of apoptotic cells which are way to regulate C3bBb through dissociation of
a rich source of autoimmunity in SLE. C3bBb into inactive factor Bb and C3bfH,
Glomerular cells are normally protected from followed by irreversible inactivation by factor
complement-mediated injury by cell-surface I. Factor H has affinity for polyanions on the cell
defense mechanisms, and complement regulatory surface and protects the cell from alternative
proteins are expressed in intrinsic glomerular pathway-mediated injury.
cells. CR1 (CD35) preferentially binds to C3b Dense deposit disease (DDD, synonym
and C4b, which are normally expressed in as MPGN type II) is a peculiar disease with
podocytes and protect against complement- persistent complement activation without immu-
dependent cellular damage. CR2 is also expressed noglobulin deposition, as revealed by immunoflu-
in intrinsic glomerular cells, including podocytes orescence [53]. Electron microscopy shows a
and mesangial and endothelial cells, and is characteristic continuous, electron-dense deposi-
upregulated in membranous nephropathy. In addi- tion along the lamina densa of the GBM. The
tion, ER (endoplasmic reticulum) stress protein disease has a poor prognosis, with chronic renal
may have protective role for MAC-mediated cell failure and frequent relapses after renal transplan-
injury as shown in podocytes [52]. tation, which suggests that genetic or humoral
In human glomeruli, decay accelerating factor factors cause the disease. Systemic and persistent
(DAF or CD55) and membrane cofactor protein alternative pathway activation in DDD is gener-
(MCP or CD 46) regulate C3 and C5 activation. ally caused by either the presence of autoanti-
CD59, which is a membrane protein inhibitor of bodies to C3 convertase (C3bBb), C3NeF, which
908 M. Nagata

stabilizes it, or by inherited mutations in or the factor H-deficient mice is blocked by administra-
presence of antibodies against factor H, resulting tion of anti-C5 antibody [58]. Notably, the site of
in the inhibition of convertase inactivation [54]. pathologic lesions is limited to the glomerulus,
C3NeF-mediated convertase stabilization may be suggesting a unique requirement in the GBM for
dependent on the factor H-mediated inactivation factor H-mediated protection against complement
of the convertase. Similar to DDD, glomerulone- attack.
phritis with isolated C3 deposition (C3 glom- Another type of glomerular injury with persis-
erulonephritis) has been recently categorized tent complement activation is atypical hemolytic
and needs differential diagnosis with DDD uremic syndrome (aHUS), which is typically seen
and poststreptococcal acute glomerulonephritis. as a familial condition. A factor H mutation has
C3 glomerulonephritis is a heterogeneous disease been implicated in aHUS, and the HUS database
which includes abnormalities of complement reg- (http://www.fh-hus.org) lists more than 100 muta-
ulatory protein and others [55]. tions related to factor H: 163 factor H mutations,
The genes for the factor H family of proteins 57 factor I mutations, and 49 MCP mutations
localize to the “regulators of complement activa- linked to aHUS, as well as 6 factor H mutations
tion” (RCA) region at 1q32. Few reports have linked to MPGN type II, as of July 2014. Factor H
addressed the potential association between mutations apparently generate different glomerular
human DDD and factor H mutations, although injury phenotypes in MPGN type II and aHUS.
such mutations are important in the pathogenesis In contrast to the factor H mutations in patients
of the disease. A 13-month-old Native American with MPGN type II, aHUS patients with factor H
boy with MPGN type II, who had a C518R muta- mutations are usually heterozygous (few cases
tion in the factor H short consensus repeat (SCR) with homozygous mutation are reported) for the
9 in trans with a C941Y mutation in factor H mutation, and these patients have normal levels of
SCR16, showed retention of the defective factor circulating factor H protein [59]. In addition, factor
H in the endoplasmic reticulum. A relationship H mutations in aHUS patients are typically located
between MPGN type II and factor H has also in the C-terminal region, which is important for
been demonstrated in animal models. Norwegian binding to the cell surface. Transgenic mice that
Yorkshire piglets with the I1166R mutation in lack the exons encoding the C-terminal region of
SCR20 developed MPGN type II; this mutation factor H develop aHUS, closely resembling the
prevented extracellular release of factor H, human disease. These mice show preservation of
resulting in intracellular accumulation. Factor fluid-phase complement activation, associated
H-deficient mice show significant reductions in with defects in endothelial protection against com-
C3 and reproducible MPGN type II, with C3 plement attack, leading to persistent endothelial
deposition in the GBM, whereas combined factor injury. This corresponds to the pathogenic
H and factor I deficiency or factor I deficiency background of aHUS, unlike MPGN type II. Sim-
alone did not show MPGN II [56]. These findings ilarly, heterozygous mutations in MCP have been
in humans and animals suggest that mutated factor reported in cases of familial aHUS. These muta-
H is not delivered to the cell surface and/or is tions resulted in three amino acid changes (posi-
unable to bind to surface-bound C3b, resulting in tions 233–235) and the insertion of a premature
incomplete convertase inactivation. Cases with stop codon, resulting in loss of the transmembrane
DDD with C3 mutation revealed defect of C3 domain of the protein and severely diminishing
structure which is critical for recognition of the the cell-surface expression of MCP [60]. Recently
substrate C3 by the alternative pathway developed anti-C5 antibody (eculizumab) has
convertase and for the regulatory activities of been shown to be effective for children
factor H, DAF, and MCP [57]. Spontaneous with not only aHUS, but also Shiga toxin-
MPGN in factor H-deficient mice was attenuated producing Escherichia coli (STEC-HUS), who
in additional deficient with C5, but not those with are negative for complement regulatory protein
C6. Severe nephritis of anti-GBM nephritis in abnormalities [61].
29 Immune-Mediated Glomerular Injury in Children 909

Soluble, Secreted Peptides similar but occasionally different mechanisms,


and their roles are sometimes disease specific.
Following the initiation of immune-mediated glo- For example, IL-10, IL-12, and IL-18 are
merular injury, regardless of whether it is cell pathogenesis-specific ILs in lupus nephritis and
mediated, antibody mediated, or both, soluble are involved in glomerular injury by mediating
and secreted peptides are significantly involved leukocyte infiltration.
in the promotion of glomerular injury as second IL-1 is produced by macrophages, neutrophils,
messengers. Not only interactions between these and dendritic cells and accelerates helper T-cell
molecules occur among inflammatory cells, but costimulation, B-cell maturation, and macrophage
their direct actions on intrinsic cells and cellular and endothelial cell activation, via the specific
responses are also important in the pathogenic receptors Cd121a/IL1R1 and CD121b/IL1R2.
background of glomerular injury. Various cyto- IL-1β is involved in macrophage-mediated glo-
kines, chemokines, and growth factors are solu- merular injury, as is TNF-α. IL-1 is transiently
ble, secreted peptides that promote the responses expressed during acute glomerular injury and cor-
of intrinsic cells, including cell proliferation, cell relates with the degree of glomerular inflamma-
death, and matrix production/degradation. The tion; it is not expressed during chronic glomerular
cellular responses of intrinsic cells and the fea- damage. IL-1 accelerates leukocyte adherence to
tures of inflammatory cells lead to the glomerular endothelial cell surface, resulting in mesangial
pathology that defines the stage and grade of proliferation, activation of procoagulant activity,
glomerulonephritis. and the synthesis of prostaglandins. Indeed, IL-1
mRNA is expressed in acute glomerular injury,
and the administration of a soluble IL-1 receptor
Interleukins antagonist attenuates the damage. In addition,
IL-1 stimulates mesangial VEGF synthesis via
Interleukins (ILs), initially identified as cytokines the PI3-K/mTOR pathway and stimulates matrix
secreted by leukocytes, are important signaling production by epithelial cells [62].
molecules. To date, more than 35 subtypes have IL-6 is produced by several types of blood-
been identified. ILs are known to mediate cellular borne cells, glomerular endothelial cells, and
immune/inflammatory reactions, and they play mesangial cells. IL-6 is a multifunctional cytokine
key roles in immune-mediated glomerular injury. that is important for B-cell differentiation and
Many cells synthesize ILs that act locally, maturation, immunoglobulin synthesis, acute-
resulting in the proliferation or maturation of T phase protein production, bone marrow progeni-
cells, T-cell interactions with B cells, antibody tor stimulation, mesangial proliferation, and the
production by B cells, and changes in the Th1 activation of monocytes/macrophages. IL-6 is
and Th2 subset balance. detected in the serum and urine in both animals
In the kidneys, ILs stimulate mesangial cells and humans with glomerulonephritis. IL-6-trans-
and endothelial cell proliferation and stimulate the genic mice demonstrate glomerulonephritis, and
production of oxygen radicals, collagenase, cyto- blockade of IL-6 receptors using a neutralizing
kines, chemokines, adhesion molecules, and antibody ameliorates lupus glomerulonephritis in
extracellular matrix, and they may accelerate cell NZB/W F1 mice. The promotive role of IL-6 for
injury/death. Among the ILs, IL-1, IL-2, IL-4, mesangial proliferation or glomerulonephritis
IL-6, IL-8, IL-10, IL-11, IL-12, and IL-13 are may largely be mediated by activation of mono-
known to be involved in many aspects in glomer- cyte/macrophages.
ular injury. IL-8 is synthesized by macrophages, lympho-
IL-1, IL-6, IL-8, IL-12, and IL-18 primarily act cytes, epithelial cells, and endothelial cells. IL-8
to promote glomerular injury, whereas IL-4, stimulates the activation and chemotaxis of
IL-10, IL-11, and IL-13 have anti-inflammatory granulocytes via CXCR1/IL8RA. In addition,
actions. These molecules act on glomerulus by activated mesangial cells in glomerular injury
910 M. Nagata

synthesize IL-8, and neutralization of IL-8 by a glomerulonephritis with reduction of ED-1-posi-


specific antibody attenuates immune complex- tive macrophage influx [68]. The aggravation of
type glomerulonephritis, concomitant with the glomerulonephritis in IL-4-deficient mice is atten-
suppression of leukocytes. In lupus nephritis uated by treatment with recombinant IL-4. These
patients, urinary biomarkers such as TNF-like findings indicate a renoprotective role for IL-4 in
weak inducer of apoptosis (TWEAK), glomerulonephritis.
osteopontin, and MCP1 were positively correlated IL-10, a pluripotent cytokine, is produced by
with renal involvement but not that of IL-8 various activated immune cells, including
[63]. In poststreptococcal acute glomerulonephri- T-helper cells, B cells, monocytes/macrophages,
tis in children, increased levels of IL-8 were and keratinocytes. IL-10 downregulates the
detected in the urine and plasma particularly in costimulatory molecules that participate in T-cell
the acute phase, and upregulation of glomerular activation. IL-10 suppresses mesangial cell prolif-
IL-8 is associated with endocapillary proliferative eration in anti-Thy1 nephritis and also attenuates
lesions [64]. IL-8 may relate to the acute and glomerular injury in nephrotoxic serum nephritis.
active phases of glomerulonephritis. C-reactive protein suppresses nephrotoxic serum
IL-12, which is a heterodimeric molecule, com- nephritis in B6 mice, and the effect is abolished in
posed of the p35 and 40 subunits, plays a key role IL-10-deficient mice [69]. Renal dendritic cells
in uncommitted T cells of the Th1 phenotype. stimulate CD4+ T cells, inducing IL-10 production.
Antigen-presenting cells are the dominant source Nephrotoxic serum nephritis in mice depleted renal
of the IL-12, but intrinsic renal cells are also the dendritic cells by diphtheria toxin revealed
effector to produce IL-12. Anti-GBM antibody advanced tubulointerstitial and glomerular lesions,
nephritis induced in IL-12p40/ mice reveals without any effects on the macrophages. Produc-
attenuation of glomerulonephritis associated with tion of IL-10 by renal dendritic cells is mediated by
suppression of leukocyte infiltration [65]. promotion of the IL-10 inducer molecule, inducible
IL-18 is produced by macrophages and stimu- costimulatory molecule ligand (ICOSL). Long-
lates INF-γ synthesis by Th1 cells and NK cells, term expression of IL-10 by a recombinant
resulting in NK cell activation. IL-18 is adeno-associated virus modulated glomerulo-
overexpressed in patients with lupus nephritis sclerosis in a subtotal nephrectomy model accom-
associated with higher INF-γ and lower IL-4 pro- panied by suppressing the INF-α and IL-2 mRNA
duction [66]. Anti-GBM nephritis in IL18/ levels. This suggests a renoprotective role for IL-10
mice reveals reduction of local synthesis of TNF, in nonimmune-mediated chronic models of
IL1β, IFN-γ, and MIP1. IL-18 deposition in tis- glomerulosclerosis [70].
sues is observed in ANCA-related glomerulone- IL-11 is a bone marrow-derived,
phritis, and IL-18 is involved in the priming or multifunctional, anti-inflammatory cytokine that
recruiting of neutrophils in this disease [67]. IL- is involved in acute inflammatory responses medi-
18-primed neutrophils synthesize MPO and PR3 ated by acute-phase protein production. Rat
ANCA via p38 MAP kinase activation in vitro. mesangial cells and macrophages express the
IL-4 acts by promoting Th2 responses, and it IL-11 receptor α-chain. IL-11 suppresses the
inhibits the various proinflammatory effects of expression of the proinflammatory cytokines
macrophages. Macrophages primed with IL-4 TNF-α, IL-1β, and IL-12 in LPS-stimulated peri-
become unresponsive to proinflammatory cyto- toneal macrophages. Recombinant human IL-11
kines, including INF-γ and TNF-α. Glomerular- attenuates crescentic glomerulonephritis in WKY
resident cells, i.e., mesangial cells and podocytes, rats, showing reduced necrosis, macrophage infil-
synthesize IL-4. IL-4 ameliorated crescentic glo- tration, and apoptosis [71]. In human glomerulo-
merulonephritis in Wistar Kyoto rats, and this is nephritis, daily urinary IL-11 was correlated with
associated with macrophage suppression, and proteinuria and suggested IL-11 acts as a counter
transfection of IL-4-expressing macrophages in cytokine against Th1-mediated inflammatory glo-
rats with nephrotoxic serum nephritis prevented merular diseases [72].
29 Immune-Mediated Glomerular Injury in Children 911

Interferons local inflammatory injury and systemic immune-


regulatory functions in the kidney. Many experi-
Interferon (INF) stimulates the immune response, mental studies and clinical observations support
activating NK cells and macrophages and increas- roles for TNF-α in the pathogenesis of acute and
ing antigen presentation to lymphocytes, and is chronic renal disease. TNFR1 attenuates
now known to comprise three subtypes: α, β, and LPS-enhanced nephrotoxic nephritis with
γ. INF-γ is a key molecule for Th1 cells and is downregulation of IL-1β. Anti-GBM glomerulone-
synthesized by many cell types under various con- phritis is attenuated in TNF-α-deficient mice, and
ditions, although macrophages are the most impor- blocking TNF-α in nephrotoxic serum nephritis in
tant source in inflammation. INF-γ is important in Wistar Kyoto rats attenuated glomerulonephritis
the effector phases of nephritogenic immune accompanied by downregulation of urinary
responses, including IgG2a generation, T-cell and MCP-1 [74]. Wild-type bone marrow transplanta-
macrophage recruitment, and macrophage activa- tion to TNF-deficient recipients (intact bone mar-
tion. Evidence suggests that INF exerts different row but absent renal-derived TNF) resulted in
effects in various nephritogenic conditions. attenuation of anti-GBM nephritis, whereas the
In human lupus nephritis, laser microdissection effect was not seen in mice with TNF-deficient
of glomeruli shows upregulation of glomerular leukocytes but intact intrinsic renal cells. This sug-
INF-γ in association with IL-10, IL-12, and IL-18, gests intrinsic renal cells are the major source of
and that was correlated with glomerular leukocyte TNF and participate in TNF-mediated renal injury.
influx [73]. INF-γ is also upregulated in the mice Likewise TNF stimulates cytokine production in
model of lupus nephritis, and blocking INF-γ atten- mesangial cells in vitro. TNF-α probably activates
uated the kidney disease. INF-γ enhanced autoim- proinflammatory cytokines and participates in the
munity, generating damaging autoantibodies in the progression of glomerulonephritis.
lupus model. Lymphocyte depletion in the anti- Although TNF-α generally plays a stimulatory
Thy1 nephritis model revealed attenuation of glo- role in glomerulonephritis, it can also act as an
merular injury through the suppression of INF-γ. immune modulator. The fact that blockade of
Endogenous INF is pathogenic in anti-GBM anti- TNF-α in human rheumatoid arthritis or Crohn’s
body nephritis. In contrast, anti-GBM antibody disease leads to the development of autoanti-
nephritis in INF-γ receptor-deficient mice showed bodies, a lupus-like syndrome, and glomerulone-
aggravation of the nephritis and progressive cres- phritis in some patients suggests a B-cell
centic glomerulonephritis [74]. Both aggravating modulator of TNF-α [75].
and protective effects for INF-γ in glomerulone- Given these apparent dual functions of TNFs,
phritis are probably depending on the mechanistic balance between the proinflammatory and immu-
background of the diseases. INF-γ may be an effec- nosuppressive effects of TNF may participate in
tor for other cytokines or may modify the reactions immune-mediated glomerulonephritis.
of macrophages, rather than having direct actions
on glomerular cells.
Chemokines and Their Receptors

Tumor Necrosis Factor (TNF) Chemokines (chemotactic cytokines) are small


proteins (8–10 kDa) that are now known to con-
TNF is a monocyte-/macrophage-derived, stitute a superfamily of leukocyte subset-specific
proinflammatory cytokine and an important media- activating and/or chemoattractant cytokines. The
tor of inflammatory tissue damage. There are three function is to guide the migration of inflammatory
family members, TNF-α, TNF-β, and lymphotoxin- cells into the site of inflammation. The four clas-
β, and TNF-α is the best characterized TNF among ses of chemokines (CXC, CC, C, and CX3C) are
them. Two receptors for TNF (TNFR1 and TNFR2) classified based on similarities in their amino acid
have been identified and are involved in mediating sequences; specifically, they share patterns of
912 M. Nagata

Fig. 13 Chemokines and


their receptors. Four
subfamilies of chemokines
have been characterized,
including the CC subfamily,
the CXC subfamily, the XC
subfamily, and the CX
subfamily [76]

paired cysteine repeats and two internal disulfide are five receptors for CC chemokines. CCR1 is
bridges. Many chemokines bind multiple recep- expressed on monocytes, CCR3 and CCR5 are
tors, and most receptors bind multiple chemokines expressed on Th1 cells, and CCR3 and CCR4 are
(Fig. 13). In kidney diseases, chemokines bind expressed on Th2 cells. Th17 cells produce CCL20
variable receptors on the leukocyte surface and and induce monocyte and Th1 cells as well as
promote acute kidney injury and chronic kidney additional Th17 cells.
diseases (Fig. 14; [76]). MCP-1 accelerates glomerular injury. Expres-
The CC and CXC chemokines have been exten- sion of MCP-1 has been observed in proliferative
sively investigated in kidney diseases. In humans, glomerulonephritis and crescentic glomerulone-
urinary CC chemokines are detected in crescentic phritis. Urinary and serum MCP-1 levels are
glomerulonephritis and glomerulosclerosis. increased in poststreptococcal acute glomerulone-
Increased levels of urinary CXC chemokines are phritis (PSAGN) in children, and urinary but not
associated with glomerular deposition in cases of serum MCP-1 levels correlate with proteinuria in
active glomerulonephritis. The CC chemokines acute phase [64]. MCP-1 and MIP1-α have been
(β-chemokines), comprising to date a family of detected in human crescentic glomerulonephritis
27 members, are involved in the recruitment of and correlate with the degree of macrophage
monocytes/macrophages and T cells. Monocyte influx into the glomeruli [77]. MCP-1-deficient
chemoattractant protein (MCP-1/CCL2), macro- MRL-Fas/lpr mice showed decreased disease
phage inflammatory protein (MIP1/CCL3), and severity, with suppression of macrophage and
regulated upon activation normal T cells expressed T-cell recruitment, resulting in decreased local
and secreted (RANTES/CCL5) are the major CC levels of cytokines and chemokines. An MCP-1
chemokines involved in glomerulonephritis. There antagonist attenuated glomerulonephritis in
29 Immune-Mediated Glomerular Injury in Children 913

IL-1β, TNF-α, ROS, LPS,


IL-17, IL-23, IFN-γ, TGF-β...

CXCL1–3/5–8 CCL2–5/8/13–16 CXCL4/9–11/16 CCL17/19/21/22


CX3CL1 CCL1–5/7/8/11/12
CCL14–17/19/21–27

CCR1/2/5/8 CXCR3/6
CXCR1/2 CX3CR1 CCR2–10 CCR4/6–8

Neutrophils Monocytes T cells Dendritic cells

AKI CKD

Fig. 14 Interaction of chemokines and receptors on leu- expressed by the different subsets. Although infiltration
kocyte subsets during acute and chronic kidney injury. of neutrophils and monocytes mediates acute kidney
Kidney diseases are characterized by the accumulation of injury, activation of T cells, macrophages, and dendritic
various leukocyte subsets that are controlled by cells promotes progression of chronic kidney disease [76]
chemokines through their corresponding receptors

MRL-Fas/lpr mice, and a neutralizing antibody to on glomerular injury, suggesting the role of
MCP-1 attenuated the disease in a mouse model of CCR1 is different in the pathogenic background.
crescentic glomerulonephritis. A Spiegelmer CXC chemokines exert chemoattractant effects
(L-enantiomeric RNA oligonucleotide, mNOX- on PMNs by intracellular phosphatase activation,
E36) that binds to murine CCL inhibited its function via CXCR1 and CXCR2. IL-8 interacts with
and improved the survival rate of lupus mice by CXCR1 and CXCR 2, advancing PMN chemo-
modulating nephritis [78]. In addition, P38MAPK taxis. Immunohistochemistry revealed expression
ameliorates crescentic GN by suppressing MCP-1, of CXCR1 predominantly in MPGN and lupus
independently of TNF-α and IL-1β [79]. nephritis in humans and that CXCR-1-expressing
RANTES binds CCR1 and CCR5 in mono- cells are mainly PMNs [81]. The INF-γ-inducible
cytes. Inhibition of RANTES leads to protective protein of 10 kDa (IP-10) binds to CXCR3. Renal
effects in experimental glomerulonephritis, and microvascular endothelial injury model provided
CCR5 deficiency aggravates T-cell-dependent evidence that neutralizing antibody against IP-10
nephrotoxic serum nephritis via enhanced CCL3/ attenuated renal dysfunction through the reduced
CCL5-CCR1-driven renal T-cell recruitment [80]. number of infiltrating tubulointerstitial T cells
CCR1 antagonist prevents lupus nephritis by without affecting monocytes/macrophage
inhibiting interstitial inflammation but not affect migration [82].
914 M. Nagata

Cytokines stimulate glomerular-resident cells developed glomerular tufts, indicating a key role
to express chemokines, including IL-8, MCP-1, for PDGF in glomerular angiogenesis and cell
MIP-1α, and macrophage inflammatory protein migration. The intrarenal distribution of PDGF
(MIL)-1α, which can lead to local inflammation varies among species. PDGF-A is absent from
and glomerular injury. Chemokines are also the glomerulus in humans and rodents, whereas
involved in nonimmunologic renal injuries, PDGFR is expressed in mesangial cells and endo-
including ischemia-reperfusion models and inter- thelial cells. In experimental glomerular diseases,
stitial nephritis [83]. intense upregulation of PDGF-B and PDGFR-β is
observed. PDGFR-β is expressed in normal
human glomeruli, and this expression is
Growth Factors upregulated in all resident glomerular cells. Anti-
Thy1 nephritis shows expression of PDGF in
Growth factors are cytokines that function as cell- areas of mesangial proliferation, and blockade of
to-cell signaling molecules, regulating cellular PDGF by neutralizing antibodies or aptamers
processes, such as maturity, proliferation, differ- (PDGF receptor IgG) attenuates mesangial prolif-
entiation, and polarity. Growth factors bind to eration in the acute and chronic phase. PDGF
specific receptors on cell membranes. Several antagonists also prevent glomerular damage in
growth factors are involved in acute and chronic anti-Thy1 nephritis, nephrotoxic serum nephritis,
glomerular injuries and affect the survival, prolif- and lupus nephritis models. PDGF-C is expressed
eration, migration, and matrix production in both in podocytes and mesangial cells in areas of glo-
bone marrow-derived cells and intrinsic glomeru- merular injury. PDGF-D has a similar action as
lar cells. PDGF-B by β-receptor-mediated activation
[84]. Treatment with PDGF-D-neutralizing anti-
body in chronic progressive anti-Thy1 nephritis
Platelet-Derived Growth Factor (PDGF) resulted in attenuation of glomerulosclerosis
accompanied by suppression of monocytes/
PDGF acts primarily on mesenchymal cells, driv- macrophage influx and complement activation.
ing cell migration and mitogenesis, extracellular Stimulation of mesangial cells in vitro with differ-
matrix production, anti-inflammatory mediator ent PDGF ligands leads to cellular proliferation
production, tissue permeability, and hemody- and migration. PDGF induces extracellular matrix
namic regulation. Intrinsic glomerular cells that production in mesangial cells, parietal cells, and
are mesenchyme derived are plausible targets of tubular cells. PDGF-AB stimulates the production
PDGF. PDGF is released by activated platelets, of TGF-β, CCL2, CXC3CL1, plasminogen acti-
activated macrophages, endothelial cells, and vator inhibitor-1 (PAI-1), IL-6, endothelin, and
smooth muscle cells. PDGF exists in five isoforms iNOS in mesangial cells. The role of PDGF for
(A, B, C, D, AB), which bind two receptor chains kidney diseases is extensively reviewed [85].
(α, β) on the cell surface (receptor tyrosine
kinases). PDGF-B is a ligand for PDGF α-and
β -receptor, whereas PDGF-D binds β receptor. Transforming Growth Factor (TGF)
PDGF activates intranuclear proto-oncogenes
and immediate early gene expression, which are TGF is known to be a superfamily of cytokines,
effectors of the receptor tyrosine kinase and consisting of about 40 molecules, including the
responsible for particular downstream functions. TGF-β, activin, and BMP subfamilies. Several
PDGF-A and PDGF-B are secreted as homo- or family members involve many aspects in the kid-
heterodimers, whereas PDGF-C and PDGF-D are ney, i.e., morphogenesis, cell differentiation,
homodimers. The impressive glomerular struc- inflammation, and remodeling. TGF-β is pro-
tures seen in PDGF-B- and PDGFR-β-deficient duced by a variety of cell types in an inactive
mice reveal a lack of mesangium and aberrantly (latent) form, and proteolytic cleavage renders it
29 Immune-Mediated Glomerular Injury in Children 915

functional. TGF-β exerts its functions through its tissues. Among the 22 FGFs identified to date,
type I and type II receptors. An intracellular sig- FGF-2 (basic-FGF) is the major factor investi-
naling pathway, involving Smad proteins, trans- gated in glomerular injury.
duces the TGF-β signal to the target genes, As a potent angiogenic factor, FGF-2 mediates
including PAI-I and collagen type I (α2). TGF-β proliferation and matrix synthesis through binding
has pleiotropic functions, and the effects differ by with receptors FGF-R1 to FGF-R4. FGF-2 is
cell type and dosage. It acts as a growth inhibitor increased in mesangial proliferative glomerulone-
in epithelial cells, whereas it sometimes stimulates phritis. Recombinant FGF-2 injection in rats leads
cell proliferation and matrix synthesis in the to podocyte mitosis, without cytokinesis, resulting
mesenchyme. in severe nephrosis and focal segmental glomerulo-
TGF-mediated glomerular injury includes inhi- sclerosis. Parietal epithelial cells express FGF-R2,
bition of cell growth/proliferation, apoptosis, cell and FGF-2 stimulates matrix synthesis. FGF-1 and
differentiation, angiogenesis, and fibrosis. TGF-β its receptor are expressed in the glomeruli, but
stimulates proteoglycan, biglycan, and decorin pro- precise function is still unknown.
duction in mesangial cells in vitro. The role of
TGF-β in immune-mediated glomerular injury is
not well understood. Latent TGF-β transgenic Vascular Endothelial Growth Factor
mice have no renal disease, despite high levels of (VEGF)
circulating TGF-β. Overexpression of active TGF-β
results in severe renal damage. Neutralizing anti- VEGF is known to regulate vasculogenesis and
bodies to TGF-β attenuate anti-Thy1 nephritis. angiogenesis, through effects on endothelial
Latent TGF-β overexpression in anti-GBM anti- mitogenesis. It is involved in the chemotaxis of
body nephritis showed suppression of inflammatory macrophages and granulocytes. Vasomotor activity
cells and cytokines, by Smad7-mediated inhibition for VEGF has also been noted.
of NF-κB-dependent inflammation [86]. In contrast, The VEGF family consists of five members
blockade of TGF-β by adenovirus-mediated gene (A, B, C, D, PIGF), and VEGF-A is the most
transfer of the TGF-β IIR failed to attenuate anti- intensively investigated. VEGF-A has four
GBM antibody nephritis. Blockade of TGF-β sig- isoforms, VEGF121, 165, 189, and 206; the for-
naling in the T cells of Smad7 transgenic mice mer two are soluble and the latter two are cell
inhibited glomerulonephritis, only with a medium bound. In glomeruli, VEGF is constitutively and
dosage of anti-GBM antibodies. Smad7 gene ther- most abundantly expressed by podocytes and
apy ameliorates autoimmune crescentic glomerulo- occasionally by activated mesangial cells. The
nephritis [87]. The pleiotropic and complex VEGF receptors, flt and flk-1, are expressed on
functions of TGF-β are thought to be differentially endothelial cells [88]. Inhibition of VEGF
regulated in the phases and pathogenetic back- worsens anti-Thy1 nephritis, and treatment with
ground of the renal diseases. VEGF attenuates thrombotic microangiopathy.
Systemic administration of VEGF 165 in anti-
GBM antibody nephritis reduced inflammation
Fibroblast Growth Factor (FGF) and advanced glomerular repair [89]. Administra-
tion of sFlt-1 affected neither the infiltration of
The FGFs are a family of growth factors involved macrophages nor the crescents.
in angiogenesis, wound healing, and embryonic
development. The FGFs are heparin-binding pro-
teins that interact with cell-surface-associated Connective Tissue Growth Factor
heparan sulfate proteoglycans, which are a pre- (CTGF)
requisite for FGF signal transduction. FGFs are
key players in the processes of proliferation and CTGF (CCN2) is a secreted, cysteine-rich,
differentiation in a wide variety of cells and matrix-associated, heparin-binding protein of
916 M. Nagata

38 kDa. The primary function of CTGF is to that coagulation and the fibrinolysis system are
modulate and coordinate signaling responses involved in immune-mediated glomerulonephri-
involving cell-surface proteoglycans. CTGF tis. Local activation of the coagulation cascade of
operates the downstream of TGF-β and directly the extrinsic pathway eventually produces fibrin
enhances the TGF-β/Smad signaling pathway or or thrombi (Fig. 15). Glomerular thrombi are
independently acts to promote fibrosis and wound occasionally observed in the active phase of glo-
healing [90]. CTGF is expressed in the intrinsic merulonephritis, reflecting severe endothelial
glomerular cells and tubular cells and potentiates damage. Fibrin deposition is more frequently
to promote renal scar formation. TNF-α is an noted in glomerular injury, with tuft necrosis and
upregulator of CTGF, and CTGF rapidly activates thrombotic microangiopathy. The influences of
intracellular signaling and transcription of TGF-β these products in glomerular injury are to acceler-
inducible early gene through neurotrophin recep- ate inflammation, rather than ischemia due to glo-
tor TrkA in human mesangial cells. CTGF is merular capillary obstruction. In human
expressed in podocytes and PECs in normal glo- glomerulonephritis, localization of coagulation
meruli and binds perlican, a known component of system-related molecules has been described,
the GBM. Increased CTGF in PECs is associated and their functions have been tested in experimen-
with matrix production. Upregulation of CTGF tal models, using neutralizing antibodies or
mRNA is found during crescent formation in knockout mice.
humans and rats, and the expression is diminished
when the crescents become scars [91].
Platelets

Epidermal Growth Factor (EGF) Platelets are involved in glomerular injury,


through leukocyte recruitment, increased vascular
EGF is a 53-kDa aminopeptide that was initially permeability, and the vasoactive effects of their
identified in mice submandibular glands and intrinsic chemical substances. Platelets contain
human urine. The renal production sites of EGF two types of cytoplasmic granules, α and σ. The
are Henle’s loop and distal convoluted tubule. α granules express P-selectin on their membranes
EGF is known to be involved in cell survival and and contain fibrinogen, fibronectin, factors V and
the differentiation of tubular cells. Although the VIII, platelet-activating factor (PAF), platelet fac-
urinary levels of EGF are a marker of renal injury tor 4, PDGF, and TGF-αl. The σ granules contain
and urinary MCP-1/EGF seems to be a prognostic adenosine diphosphate (ADP), calcium ions, his-
factor in IgA nephropathy, the function(s) of EGF tamine, and serotonin. In local injury in glomeru-
in the glomeruli and its role in glomerular injury lar capillaries, platelets adhere to the extracellular
are shortly reviewed [92]. EGF induces mesangial matrices at sites of endothelial injury and become
cell contraction in vitro. Heparin-binding EGF is a activated. Activated platelets secrete granular
strong mitogen that stimulates mesangial prolifer- products and synthesize TXA2, PDGF, and PAF,
ation in vitro and involved in the crescentic which together stimulate clot formation, cell pro-
glomerulonephritis. liferation, and matrix production. PAF is bioactive
and increases leukocyte chemotaxis, vascular per-
meability, and vascular spasms. Platelets include
Platelets and the Coagulation Cascade phospholipid complexes that are important in the
intrinsic coagulation pathway. Injured or activated
As for vascular endothelial cells in general, glo- endothelial cells trigger aggregation of platelets,
merular endothelial cells normally possess forming a clot, and exposing tissue factor, which
antiplatelet, anticoagulant, and fibrinolytic prop- provokes capillary necrosis and fibrin exudates,
erties, to maintain the microcirculation in the glo- leading to GBM breaks and extraglomerular
merulus, and this is a very consistent background lesions and cellular crescent formation.
29 Immune-Mediated Glomerular Injury in Children 917

Fig. 15 Coagulation is the Tissue injury


cascade promoting the Resident cells
transformation of soluble macrophages
fibrinogen into insoluble Inflammatory
fibrin monomers. The cells
cascade is initiated in the
injured tissue by exposure
of tissue factor (TF) at the Tissue factor
surface of the injured
endothelium. TF synthesis TFPI
is stimulated by VIIa, Ca2+
inflammatory cells, which
activated resident cells or Factor X Factor Xa
macrophages for
production. Tissue factor Thrombin
activated conversion of X to
Xa, which further converts Factor V Factor Va Ca2+
prothrombin to thrombin. Xlll
Thrombin participate Ca2+
conversion of fibrinogen to
fibrin, which finally forms Prothrombin Thrombin Xllla
cross-linked fibrin by XIIIa. (II) (lla)
TF pathway inhibitor Ca2+
(TFPI) is a potent inhibitor
Fibrin
of this cascade. Fibrinogen
(IIa)
(I)

Cross-linked fibrin

In addition, platelets activate the complement as co-localization of factor V with fibrin in the
well as the intrinsic coagulation pathway and mesangium indicates local activation of the coag-
release vasoactive amines which potentially mod- ulation cascade in glomerulonephritis.
ulate pathological response in glomerular-resident Fibrin deposition is mediated largely by the
cells and inflammatory cells. stimulation of macrophage procoagulant activity.
Most of the fibrin deposition associated with
immunologic glomerular injury is mediated by
Fibrin activation of the extrinsic coagulation cascade,
commencing with tissue factor procoagulants
Fibrin is an insoluble fibrous protein that is the [94]. In crescentic glomerulonephritis, fibrin
end product of a locally accelerated coagulation deposition, renal pathology, and dysfunction are
system. Fibrin stabilizes and anchors aggregated synchronous with glomerular macrophage and
platelets. In glomerular diseases, fibrin is occa- T-cell influx. In experimental models, glomerulo-
sionally deposited in the capillary or mesangium. nephritis is attenuated by the administration of
In IgA glomerulonephritis soluble fibrin deposi- anti-factor V antibody. Defibrinogenation sup-
tion is located in the mesangial proliferation of presses proteinuria in crescentic glomerulonephri-
active stage [93]. Fibrin is detected in the capillary tis, although it has no effect on the inflammatory
necrosis in cellular crescentic formation. Cross- influx, indicating that the coagulation system pro-
linked fibrin, immunoglobulin deposition, and C3 motes crescentic glomerulonephritis indepen-
are occasionally seen in glomerulonephritis, and dently of the inflammatory reaction [95].
918 M. Nagata

Activation of the local coagulation system can be The Enzymatic Fibrinolysis System
initiated by immunologic effects, although sec-
ondary events, which include intrinsic glomerular In addition to the progression of coagulation pro-
cell proliferation, microcirculation defects, and cesses in tissues, the clotting cascade simulta-
cell necrosis, are promoted by bioactive mole- neously sets into motion a fibrinolytic cascade
cules of the coagulation system. In the activation that restricts final clot size, which may influence
of coagulation system, the fibrinolytic cascade tissue scarring. The fibrinolytic system consists of
also limits clot formation. Fibrinolytic agents an inactive proenzyme (plasminogen), which can
have shown relatively consistent efficacy in atten- be converted to an active enzyme (plasmin) that
uating immune-mediated glomerular injury in degrades fibrin into soluble fibrin products. This
several models. system is under the control of tissue-repair-related
molecules, including plasmin. Plasmin is derived
from the enzymatic breakdown of its inactive
Tissue Factor circulating precursor, plasminogen, either by a
factor XII-dependent pathway or by plasminogen
Tissue factor is a cellular lipoprotein that is present activators. Plasmin is involved in fibrinolysis.
at sites of tissue injury and activates extrinsic coag- Activation of plasmin significantly involves
ulation processes. In glomerular injury, inflamma- matrix degradation and fibrinolysis, which influ-
tory cell-derived cytokines induce tissue factor ences the fate of tissue injury. Activation of plas-
secretion by resident cells and macrophages. Endo- min inhibits fibrosis by enhancing matrix
thelial tissue factor release initiates glomerular metalloproteinase 2 activation, thereby promoting
fibrin deposition, through activation of the extrinsic ECM degradation. Plasminogen activator (PA) is
coagulation pathway, by binding circulating factor a serine protease that converts plasminogen to
VII and facilitating its allosteric conversion to fac- plasmin to regulate ECM tissue deposition. Two
tor VIIa. Tissue factor converts X to Xa, and factor plasminogen activators, tissue type (t-PA) and
V is a cofactor for the Xa, which is responsible for urokinase-type (u-PA), are known. t-PA is synthe-
prothrombin activation. Factor Va then converts sized primarily by endothelial cells and is active
prothrombin to thrombin, which further converts when attached to fibrin, which it dissolves. Plas-
fibrinogen to fibrin (Fig. 15). Tissue factor also has minogen binds to the plasminogen activator
fibrin-independent proinflammatory effects, medi- receptor (PAR), which belongs to the G protein-
ated by MHC class II protein expression and leu- coupled receptor family, and activation of PAR
kocyte accumulation. leads to cytoskeletal rearrangements, proliferation
Specific Xa inhibitor ameliorates anti-Thy of fibroblasts, and glomerular epithelial cell pro-
1 nephritis by inhibiting macrophage infiltration liferation. Thrombin also interacts with PAR.
and fibrin accumulation, suggesting that tissue PA and plasmin are suppressed in some glomer-
factor promotes mesangial proliferation by ulonephritis conditions and may be involved in
function of proinflammatory and procoagulant mesangial matrix accumulation via α1-antitrypsin,
mechanism of factor X [96]. Administration of α2-macroglobulin, or plasminogen activator inhib-
anti-factor V neutralizing antibody suppressed itor (PAI). PAI is synthesized by endothelial cells to
profibrotic cytokines in glomeruli. Inhibition of modulate the coagulation/anticoagulation balance
tissue factor activity by an antibody or tissue by blocking fibrinolysis. PAI-1 is a single glyco-
factor pathway inhibitor (TFPI) significantly protein of about 45 kDa consisting of 379 or
ameliorates the development of crescentic 381 amino acids. It is a member of the serpin family
glomerulonephritis, accompanied by decreased with reactive site peptide bound Arg345-Met346.
glomerular inflammatory cell influx. Infusion of Activation of PAI-1 requires complex processes.
recombinant TFPI ameliorates glomerular fibrin PAI-1 promotes fibrosis by inhibiting plasmin gen-
deposition and glomerular pathology in experi- eration, through the suppression of plasminogen
mental glomerulonephritis. activator. PAI-1 is also activated by interaction
29 Immune-Mediated Glomerular Injury in Children 919

with TGF-β, and it can modulate fibrosis and hemostasis. AA is a 20-carbon polyunsaturated
PAI-1, downregulating TGF-β activity in a nega- fatty acid that is present mainly in cell membrane
tive feedback loop. PAI-1 deficiency leads to path- phospholipids. Mechanical, chemical, and physi-
ogenic over-activation of TGF-β. ologic stimuli release AA from the cell membrane
PAI-1 is increased in glomerulonephritis, and into the cytoplasm, and specific enzymes then
plasmin-independent profibrotic action of PAI-1 synthesize the metabolites. The AA cascade
in glomerular injury involves the recruitment of branches into two major pathways, the cyclooxy-
fibroblasts and macrophages. Experimental genase and lipoxygenase pathways, both of which
cryoglobulin-associated mesangial proliferative are involved in glomerular injury (Fig. 16).
glomerulonephritis reveals upregulation of the The cyclooxygenase pathway products include
protease Nexin-1, t-PA, and PAI-1 [97]. Induction prostaglandins (PG) E2, PGD2, PDF2a, PGI2 (pros-
of crescentic glomerulonephritis in PAI-1-defi- tacyclin), and thromboxane (TX) A2. The
cient mice resulted in improvements in nephritis, lipoxygenase pathway begins with 5-lipoxygenase,
accompanied by depressed inflammatory infil- a well-characterized AA-metabolizing enzyme.
trates and collagen accumulation. Three major products, 5-hydroxyeicosatetraenoic
Protease-activated receptor 2 (PAR-2), receptor acid (HETE), the leukotriene (LT) family of com-
of factor Xa, is expressed in epithelial, mesangial, pounds, and the lipoxins, are known.
and endothelial cells and macrophages. PAR-2 is The restricted distribution of enzymes may
activated by serine proteases, such as trypsin, determine the cell-specific synthesis of certain
tryptase, and factors VIIa and Xa and increases substances and cell to cell transaction to produce
PAI-1 expression and macrophage-derived cytokine active end products. For example, the AA in neu-
induction. PAR-2 is involved in human mesangial trophils is converted into LTA4 followed by
cell proliferation. Experiments with PAR-2-defi- LTB4, which is the end product in neutrophils,
cient mice revealed that PAR-2 promoted glomeru- since neutrophils lack the further converting
lar fibrin deposition and the progression of enzyme of LTC4 synthase, and it is then secreted
crescentic glomerulonephritis, due to stimulation from the neutrophils. In contrast, platelets lack the
of renal PAI-1 expression and MMP-9 activation 5-lipoxygenase enzyme but have 12-lipoxygenase
[98] Thrombin-activatable fibrinolysis inhibitor and LTC4 synthase. Thus, in platelets, the LTB4
(TAFI) is a potent inhibitor of plasmin generation, derived from neutrophils is converted into lipoxin
and immune complex-mediated glomerulonephritis and LTC4, both of which are final platelet prod-
is ameliorated in TAFI-deficient mice. ucts. This transcellular interaction of AA metab-
Although the coagulation cascade/fibrinolysis olites promotes inflammation in situ, resulting in
system is substantially involved in the acute and biologic responses by the cells. Inflammatory
exudative phases of glomerular injury, by interacting mediators, such as complement components,
with inflammatory cells, its roles in chronic progres- also accelerate the release of these metabolites.
sive glomerulonephritis remain unknown. Further Common functions among the eicosanoids are
studies are needed to investigate the coagulation/ vasomotor effects; TXA2 and LTs cause vasocon-
fibrinolysis-independent actions of PAI-1 and tissue striction, whereas PGs, PGI2, and lipoxin partici-
factor, such as inflammatory responses, cell migra- pate in vasodilatation. LTB4 and lipoxins also
tion, cell proliferation, angiogenic actions, and promote chemotaxis and leukocyte adhesion.
matrix remodeling in glomerular injury. LTs affect vascular permeability. All these func-
tions of AA products are involved in the processes
of glomerular inflammation and renal function.
Eicosanoids Considering wide-ranged actions of AA
metabolites for inflammation and tissue damage/
Eicosanoids are short-acting, hormonelike sub- repair, their roles for glomerular injury are likely
stances. They are arachidonic acid (AA) metabo- important. AA involvement in glomerulonephritis
lites and have effects in inflammation and has been extensively investigated before 2000
920 M. Nagata

Fig. 16 The arachidonic acid cascade and metabolites. cyclooxygenase 1 and 2, HETE hydroxyeicosatetraenoic
Generation of arachidonic acid metabolites and their roles acid, HPETE hydroperoxyeicosatetraenoic acid The figure
in inflammation. Note enzymatic activities whose inhibi- was published in Kumar et al. Robbins Basic Pathology,
tion through pharmacologic intervention blocks major 8th edn. 2007, p. 48, Copyright Elsevier)
pathways (Denoted with COX-1 and COX-2

mainly using cell lines to demonstrate prolifera- Podocyte COX-2 expression is induced by glo-
tion and matrix production or administration of merular inflammation or various stresses and ren-
inhibitors in experimental models. ders podocytes susceptible to further injury [99].
There are two forms of cyclooxygenase, PGE2 is primarily produced by mesangial
COX-1 (constitutive) and COX-2 (inducible). cells. In acute glomerular inflammation, PGE2 is
COX-1 is constitutively expressed in the kidney, increased to maintain effective renal circulation
including in the mesangial cells, arterioles, PECs, by relaxing vascular smooth muscle cells.
and cortical and medullary collecting ducts. Another important function of PGE2 is its anti-
COX-2 is an inducible protein; its mRNA is inflammatory actions in suppressing inflamma-
detectable in the macula densa and the cortical tory cell recruitment, chemokine synthesis, and
thick ascending limb cells immediately adjacent NO production. PGI2 is a vasodilator and a potent
to the macula densa, whereas it is not detected in inhibitor of platelet aggregation. Glomerular dam-
the glomeruli. COX-2 expression in the glomeruli age with endothelial injury may result in
may appear in glomerulonephritis. In particular, decreased PGI2 in situ and may promote further
podocyte expression of COX-2 is prominent in glomerular injury. TXA2 has largely opposite
renal ablation models and anti-Thy1 nephritis. effects to PGI2 and is abundantly synthesized in
Furthermore, podocyte-specific COX-2 expres- nephritic glomeruli. TXA2 is produced by inflam-
sion, driven by the nephrin promoter, aggravates matory cells and intrinsic glomerular cells and
proteinuria and upregulates endogenous COX-2. accelerates glomerular injury, through platelet
29 Immune-Mediated Glomerular Injury in Children 921

aggregation and vasoconstriction. The LTs, a fam- activation of NADPH oxidase, which oxidizes
ily of compounds converted by 5-lipoxygenase, NADPH and reduces oxygen to the superoxide
include LTA4, LTB4, LTC4, LTD4, and LTE4. anion (O2). ROS are produced within the lyso-
LTB4 is a potent chemotactic compound that pro- some and may be released from leukocytes by
motes the recruitment of neutrophils and is ele- exposure to chemotactic agents, immune com-
vated in experimental models with glomerular plexes, complement, and other factors. In addi-
damage. LTC4, LTD4, and LTE4 participate in tion, ROS are occasionally released by intrinsic
vasoconstriction and increase vascular permeabil- glomerular cells. ROS are inherently unstable and
ity. Receptor antagonists of LTs attenuate glomer- generally decay spontaneously. The influence of
ular injury. oxygen-derived free radicals in inflammatory
Lipoxins are the products of a two-step reactions depends on the balance between the
lipoxygenation, i.e., 5-lipoxygenase followed by production and inactivation of the related
12-lipoxygenase. Lipoxins are produced locally at metabolites.
sites of inflammation by the transcellular interac- The biologic actions of ROS include the stim-
tion of neutrophils, platelets, and intrinsic cells. ulation of expression of chemokines, cytokines,
The platelets themselves cannot produce lipoxin, and endothelial leukocyte adhesion molecules.
so they take up neutrophil-derived LTA4 and con- In addition, ROS stimulate intracellular signaling
vert it into lipoxin. Lipoxins have both pro- and molecules that are involved in mitogenic path-
anti-inflammatory actions. Lipoxin A4 inhibits ways. The superoxide anion, hydrogen peroxide,
cytokine-induced mesangial cell proliferation and hydroxyl radical are major species of ROS.
in vitro and has been implicated as anti- The superoxide anion typically is the most abun-
inflammatory function in glomerulonephritis. dant, though its potential for tissue injury is lim-
Aspirin and nonsteroidal anti-inflammatory drugs ited. Hydroxyl radicals induced by the Haber
(NSAIDs) inhibit all the steps upstream of COX Weiss reaction promote considerable tissue dam-
activity. In acute poststreptococcal glomerulone- age. Activated leukocytes in the inflammatory
phritis in children, lipoxin A [5] and milieu synthesize ROS and stimulate endothelial
15-lipoxygenase (15-LO) were expressed in leuko- ROS production through xanthine oxidation,
cytes and glomeruli, and 15-LO products play anti- resulting in increased vascular permeability. Acti-
inflammatory roles in this disease [100]. Dietary vation of NADPH oxidase is involved in cell
manipulation, leading to changes in eicosanoid proliferation and extracellular matrix production,
metabolites, has been reported to affect glomerulo- mediated by intracellular signaling that includes
nephritis in humans. Dietary fish oil that contains the mitogen-activated protein kinase (MAPK) and
omega-3 fatty acids (eicosapentaenoic acid and activator protein 1 (AP-1) or TGF-β. In the acute
docosahexaenoic acid) modulates both COX and phase of anti-Thy1 nephritis, ROS- and NADPH-
lipoxygenase metabolism. In addition, dietary dependent oxidase activities are increased. Anti-
restriction of essential fatty acids impairs renal oxidants suppress proliferative anti-GBM glomer-
production of a specific lipid-derived macrophage ulonephritis [101]. Direct infusion of H2O2 in rat
chemoattractant and LTB4, resulting in decreased renal arteries causes proteinuria, and H2O2 deple-
production of cytokines, ROS, and NO in tion by catalase reduces neutrophil-dependent
macrophages. proteinuria, suggesting that ROS play a particu-
larly significant role in the promotion of
neutrophil-dependent glomerular injury. Like-
Reactive Oxygen Species (ROS) wise, ROS are involved in ANCA-associated
glomerulonephritis.
ROS are chemical species with a single unpaired Tissue fluids and the target cells of oxygen-
electron in an outer orbital. They are extremely derived radicals have antioxidant mechanisms to
unstable and readily react with various chemicals. protect against harmful species. Several enzy-
Generation of ROS can occur via the rapid matic and nonenzymatic antioxidant systems
922 M. Nagata

inactivate free radicals. Superoxide dismutase models of glomerular injury is not only a promo-
(SOD) significantly accelerates the spontaneous tive factor, but also has protective roles. In fact,
decay of ROS. Glutathione peroxidase also pro- iNOS inhibitor in Wistar Kyoto rats with nephro-
tects against ROS-mediated cell injury by catalyz- toxic serum nephritis showed reduced crescentic
ing free radical breakdown. Catalase, which is formation, and eNOS-deficient mice with anti-
present in cytoplasmic peroxisomes, directs the GBM nephritis reveal attenuation of nephritis indi-
degradation of hydrogen peroxide. Exogenous cating protective roles for NOS in glomerular
antioxidants (vitamins E, A, C, β-carotene) block injury. Anti-Thy1 nephritis shows an association
the formation of free radicals or scavenge such between NO synthesis and mesangiolysis, and sup-
products. The effects of these antioxidants on pression of NO resulted in reduced mesangiolysis.
glomerulonephritis have been reported, although Another experiment using anti-GBM antibody
presently there is insufficient evidence regarding nephritis in iNOS-deficient mice revealed no effect
their effects. on glomerular histology, macrophage-mediated
proteinuria, or glomerular tissue damage. In
humans, iNOS expression has been observed in
Nitric Oxide (NO) proliferative lupus nephritis and IgA glomerulone-
phritis, although iNOS expression is not associated
NO, which is a pleiotropic mediator of inflamma- with the presence of macrophages and its function
tion initially discovered as the endothelial cell- remains to be investigated more thoroughly. Endo-
derived factor relaxing vascular smooth muscle thelial NOS reduced glomerular injury through
cells, produces vasodilatation. NO is a soluble gas suppression of MCP-1 and ROS production in
that is produced by endothelial cells and macro- lupus glomerulonephritis model [102].
phages and is a typical cytotoxic substances of
macrophages. NO production is stimulated by Acknowledgment The author’s research and academic
acute inflammatory molecules, including TNF-α, efforts are supported by Grants-in-Aid for Scientific
Research (C) 19,590,932 and Scientific Research
ILs, and INFs. As the half-life of NO is extremely
(S) 16,109,005 from the Japan Society for the Promotion
short, NO action is limited to the immediate prox- of Science. Given a limited capacity for citation, the refer-
imity of the production site. NO acts on target cells ence includes recent review articles, despite numerous
through a paracrine mechanism and induces stable important original works published before 2005. The
author suggests the readers to reference Chap. 29 of fifth
products, such as nitrates, nitrites (NO2), and cyclic
and sixth edition of Pediatric Nephrology (Immune mech-
GMP-mediated intracellular alterations. anism of glomerular injury) which referenced earlier
Three NO synthase isoforms have been distin- published works.
guished in mammalian species: the constitutive
calcium-dependent neuronal NOS (nNOS) and
endothelial NOS (eNOS) and the inducible References
calcium-independent iNOS. In the glomerulus,
eNOS is constitutively expressed in endothelial 1. Couser WG. Basic and translational concepts of
immune-mediated glomerular diseases. J Am Soc
cells, and occasionally, iNOS transcripts are
Nephrol. 2012;23(3):381–99.
detected. 2. Borza DB, Hudson BG. Molecular characterization of
In acute-phase glomerulonephritis, macro- the target antigens of anti-glomerular basement mem-
phages are the major source of NO. iNOS is brane antibody disease. Springer Semin
Immunopathol. 2003;24:345–61.
induced in glomerular capillary inflammation, and
3. Ooi JD, Holdsworth SR, Kitching AR. Advances in
glomerular NO2 is increased in anti-GBM antibody the pathogenesis of Goodpasture’s disease: from epi-
nephritis models. NO2 expression is relatively topes to autoantibodies to effector T cells. J
acute and transient. Early studies showed that Autoimmun. 2008;22:1–6.
4. Debiec H, Guigonis V, Mougenot M, Decobert F,
decreasing NO production by L-arginine depletion
Haymann JP, Bensman A, et al. Antenatal membra-
in rat nephrotoxic serum nephritis exacerbated pro- nous glomerulonephritis due to anti-neutral endopep-
teinuria. Increased NO synthesis observed in acute tidase antibodies. N Engl J Med. 2002;346:2053–60.
29 Immune-Mediated Glomerular Injury in Children 923

5. Beck Jr LH, Bonegio RG, Lambeau G, Beck DM, expression of CD80 and CD86 is required for leuko-
Powell DW, Cummins TD, et al. M-type phospholi- cyte accumulation and injury in crescentic glomeru-
pase A2 receptor as target antigen in idiopathic lonephritis. J Am Soc Nephrol. 2005;16:2012–22.
membranous nephropathy. N Engl J Med. 20. Odobasic D, Kitching AR, Semple TJ, Holdsworth
2009;361(1):11–21. SR. Inducible co-stimulatory molecule ligand is pro-
6. Reiser J, von Gersdorff G, Loos M, Oh J, Asanuma K, tective during the induction and effector phases of
Giardino L, et al. Induction of B7-1 in podocytes is crescentic glomerulonephritis. J Am Soc Nephrol.
associated with nephrotic syndrome. J Clin Invest. 2006;17:1044–53.
2004;113:1390–7. 21. Reynolds J, Khan SB, Allen AR, Benjamin CD,
7. Shimizu M, Kondo S, Urushihara M, Takamatsu M, Pusey CD. Blockade of the CD154-CD40
Kanemoto K, Nagata M, et al. Role of integrin-linked costimulatory pathway prevents the development of
kinase in epithelial-mesenchymal transition in cres- experimental autoimmune glomerulonephritis. Kid-
cent formation of experimental glomerulonephritis. ney Int. 2004;66:1444–52.
Nephrol Dial Transplant. 2006;21:2380–90. 22. Summers SA, Steinmetz OM, Li M, Kausman JY,
8. Ueno T, Kobayashi N, Nakayama M, Takashima Y, Semple T, Edgtton KL, Borza DB, et al. Th1 and
Ohse T, Pastan I, et al. Aberrant Notch1-dependent Th17 cells induce proliferative glomerulonephritis. J
effects on glomerular parietal epithelial cells pro- Am Soc Nephrol. 2009;20(12):2518–24.
motes collapsing focal segmental glomerulosclerosis 23. Kitching AR, Holdsworth SR. The emergence of
with progressive podocyte loss. Kidney Int. TH17 cells as effectors of renal injury. J Am Soc
2013;83(6):1065–75. Nephrol. 2011;22(2):235–8.
9. Kitching RA, Holdsworth SR. The emergence of 24. Bagavant H, Tung KS. Failure of CD25 + T cells from
Th17 cells as effectors of renal injury. J Am Soc lupus-prone mice to suppress lupus glomerulonephri-
Nephrol. 2011;22:235–8. tis and sialoadenitis. J Immunol. 2005;175:944–50.
10. Couser WG, Steinmuller DR, Stilmant MM, Salant 25. Ostmann A, Paust HJ, Panzer U, Wegscheid C,
DJ, Lowenstein LM. Experimental glomerulonephri- Kapffer S, Huber S, et al. Regulatory T cell-derived
tis in the isolated perfused rat kidney. J Clin Invest. IL-10 ameliorates crescentic GN. J Am Soc Nephrol.
1978;62:1275–87. 2013;24(6):930–42.
11. Farquhar MG, Saito A, Kerjaschki D, Orlando 26. Paust HJ, Ostmann A, Erhardt A, Turner JE, Velden J,
RA. The Heymann nephritis antigenic complex: Mittr€ucker HW, et al. Regulatory T cells control the
megalin (gp330) and RAP. J Am Soc Nephrol. Th1 immune response in murine crescentic glomeru-
1995;6:35–47. lonephritis. Kidney Int. 2011;80(2):154–64.
12. Kitching AR, Holdsworth SR, Hickey MJ. Targeting 27. Holdsworth SR, Tipping PG. Leukocytes in glomer-
leukocytes in immune glomerular diseases. Med ular injury. Semin Immunopathol. 2007;29:355–74.
Chem. 2008;15:448–58. 28. Jennette JC, Xiao H, Falk RJ. Pathogenesis of vascu-
13. Wu J, Hicks J, Borillo J, Glass 2nd WF, Lou lar inflammation by anti-neutrophil cytoplasmic anti-
YH. CD4(+) T cells specific to a glomerular basement bodies. J Am Soc Nephrol. 2006;17:1235–42.
membrane antigen mediate glomerulonephritis. J Clin 29. Ruth AJ, Kitching AR, Kwan RY, Odobasic D, Ooi JD,
Invest. 2002;109:517–24. Timoshanko JR, et al. Anti-neutrophil cytoplasmic
14. Turner JE, Paust HJ, Steinmetz OM, Panzer U. The antibodies and effector CD4+ cells play nonredundant
Th17 immune response in renal inflammation. Kid- roles in anti-myeloperoxidase crescentic glomerulone-
ney Int. 2010;77(12):1070–5. phritis. J Am Soc Nephrol. 2006;17:1940–9.
15. Scholz J, Lukacs-Kornek V, Engel DR, Specht S, 30. Nakazawa D, Shida H, Tomaru U, Yoshida M,
Kiss E, Eitner F, et al. Renal dendritic cells stimulate Nishio S, Atsumi T, et al. Enhanced formation and
IL-10 production and attenuate nephrotoxic nephritis. disordered regulation of NETs in myeloperoxidase-
J Am Soc Nephrol. 2008;19:527–37. ANCA-associated microscopic polyangiitis. J Am
16. Tipping PG, Kitching AR. Glomerulonephritis, Soc Nephrol. 2014;25:990–7.
Th1 and Th2: what’s new? Clin Exp Immunol. 31. Xiao H, Heeringa P, Liu Z, Huugen D, Hu P,
2005;142:207–15. Maeda N, et al. The role of neutrophils in the induc-
17. Nitta K, Horita S, Ogawa S, Matsumoto M, Hara Y, tion of glomerulonephritis by anti-myeloperoxidase
Okano K, et al. Resistance of CD28-deficient mice to antibodies. Am J Pathol. 2005;167:39–45.
autologous phase of anti-glomerular basement mem- 32. Nolan SL, Kalia N, Nash GB, Kamel D, Heeringa P,
brane glomerulonephritis. Clin Exp Nephrol. Savage CO. Mechanisms of ANCA-mediated leuko-
2003;7:104–12. cyte-endothelial cell interactions in vivo. J Am Soc
18. Reynolds J, Tam FW, Chandraker A, Smith J, Karkar Nephrol. 2008;19:973–84.
AM, Cross J, et al. CD28-B7 blockade prevents the 33. Schreiber A, Xiao H, Falk RJ, Jennette JC. Bone
development of experimental autoimmune glomeru- marrow-derived cells are sufficient and necessary tar-
lonephritis. J Clin Invest. 2000;105:643–51. gets to mediate glomerulonephritis and vasculitis
19. Odobasic D, Kitching AR, Semple TJ, Timoshanko induced by anti-myeloperoxidase antibodies. J Am
JR, Tipping PG, Holdsworth SR. Glomerular Soc Nephrol. 2006;17:3355–64.
924 M. Nagata

34. Wang Y, Wang YP, Zheng G, Lee VW, Ouyang L, The role of complement in cryoglobulin-induced
Chang DH, et al. Ex vivo programmed macrophages immune complex glomerulonephritis. J Immunol.
ameliorate experimental chronic inflammatory renal 2005;175:6909–14.
disease. Kidney Int. 2007;72:290–9. 49. Schreiber A, Xiao H, Jennette JC, Schneider W, Luft
35. Isome M, Fujinaka H, Adhikary LP, Kovalenko P, FC, Kettritz R. C5a receptor mediates neutrophils
El-Shemi AG, Yoshida Y, et al. Important role of activation and ANCA-induced glomerulonephritis. J
macrophage in induction of crescentic anti-GBM glo- Am Soc Nephrol. 2008;58:289–98.
merulonephritis in WKY rats. Nephrol Dial Trans- 50. Mizuno M, Blanchin S, Gasque P, Nishikawa K,
plant. 2004;19:2997–3004. Matsuo S. High levels of complement C3a receptor
36. Ikezumi Y, Hurst LA, Masaki T, Atkins RC, Nikolic- in the glomeruli in lupus nephritis. Am J Kidney Dis.
Paterson DJ. Adoptive transfer studies demonstrate that 2007;49(5):598–606.
macrophages can induce proteinuria and mesangial cell 51. Pippin JW, Durvasula R, Petermann A, Hiromura K,
proliferation. Kidney Int. 2003;63:83–95. Couser WG, Shankland SJ. DNA damage is a
37. Gordon S, Taylor PR. Monocyte and macrophage novel response to sublytic complement C5b-9-
heterogeneity. Nat Rev Immunol. 2005;5:953–64. induced injury in podocytes. J Clin Invest.
38. Cao Q, Wang C, Zheng D, Wang Y, Lee VW, Wang 2003;111:877–85.
YM, et al. IL-25 induces M2 macrophages and 52. Miwa T, Zhou L, Tudoran R, Lambris JD, Madaio
reduces renal injury in proteinuric kidney disease. J MP, Nangaku M, et al. DAF/Crry double deficiency in
Am Soc Nephrol. 2011;22(7):1229–39. mice exacerbates nephrotoxic serum-induced protein-
39. Leng L, Chen L, Fan J, Greven D, Arjona A, Du X, uria despite markedly reduced systemic complement
et al. A small-molecule macrophage migration inhib- activity. Mol Immunol. 2007;4:139–46.
itory factor antagonist protects against glomerulone- 53. Appel GB, Cook T, Hageman G, Jennette JC,
phritis in lupus-prone NZB/NZW F1 and MRL/lpr Kashgarian M, Kirschfink M, et al. Membranoproli-
mice. J Immunol. 2011;186:527–38. ferative glomerulonephritis type II (dense deposit dis-
40. Hoi AY, Hickey MJ, Hall P, et al. Macrophage migra- ease): an update. J Am Soc Nephrol. 2005;16:
tion inhibitory factor deficiency attenuates macro- 1392–404.
phage recruitment, glomerulonephritis, and lethality 54. Licht C, Schlötzer-Schrehardt U, Kirschfink M,
in MRL/lpr mice. J Immunol. 2006;177:5687–96. Zipfel PF, Hoppe B. MPGN II-genetically determined
41. Kanamaru Y, Scandiuzzi L, Essig M, Yamana J, by defective complement regulation? Pediatr
O'Sullivan KM, Santos LL, et al. Mast cell-mediated Nephrol. 2007;22:2–9.
remodeling and fibrinolytic activity protect against 55. Pickering MC, D'Agati VD, Nester CM, Smith RJ,
fatal glomerulonephritis. J Immunol. 2006;176: Haas M, Appel GB, et al. C3 glomerulopathy: con-
5607–15. sensus report. Kidney Int. 2013;84(6):1079–89.
42. Pawar RD, Ramanjaneyulu A, Kulkarni OP, Lech M, 56. Rose KL, Paixao-Cavalcante D, Fish J, Manderson
Segerer S, Anders HJ. Inhibition of Toll-like recep- AP, Malik TH, Bygrave AE, et al. Factor I is required
tor-7 (TLR-7) or TLR-7 plus TLR-9 attenuates glo- for the development of membranoproliferative glo-
merulonephritis and lung injury in experimental merulonephritis in factor H-deficient mice. J Clin
lupus. J Am Soc Nephrol. 2007;18:1721–31. Invest. 2008;118:608–18.
43. Suzuki H, Suzuki Y, Narita I, Aizawa M, Kihara M, 57. Martínez-Barricarte R, Heurich M, Vazquez-Martul E,
Yamanaka T, et al. Toll-like receptor 9 affects severity Torreira E, Montes T, et al. Human C3 mutation
of IgA nephropathy. J Am Soc Nephrol. 2008;19: reveals a mechanism of dense deposit disease patho-
2348–95. genesis and provides insights into complement
44. Donadio ME, Loiacono E, Peruzzi L, Amore A, activation and regulation. J Clin Invest. 2010;120
Camilla R, Chiale F, et al. Toll-like receptors, (10):3702–12.
immunoproteasome and regulatory T cells in children 58. Pickering MC, Warren J, Rose KL, Carlucci F,
with Henoch-Schönlein purpura and primary IgA Wang Y, Walport MJ, et al. Prevention of C5 activa-
nephropathy. Pediatr Nephrol. 2014;29(9):1545–51. tion ameliorates spontaneous and experimental
45. Patole PS1, Pawar RD, Lichtnekert J, Lech M, glomerulonephritis in factor H-deficient mice. Proc
Kulkarni OP, Ramanjaneyulu A, et al. Coactivation Natl Acad Sci U S A. 2006;103:9649–54.
of Toll-like receptor-3 and -7 in immune complex 59. Dragon-Durey MA, Frémeaux-Bacchi V, Loirat C,
glomerulonephritis. J Autoimmun. 2007;29:52–9. Blouin J, Niaudet P, Deschenes G, et al. Homozygous
46. Berger SP, Daha MR. Complement in glomerular factor H deficiencies associated with hemolytic ure-
injury. Semin Immunopathol. 2007;29:375–84. mic syndrome or membranoproliferative glomerulo-
47. Hisano S, Matsushita M, Fujita T, Iwasaki H. Activa- nephritis: report and genetic analysis of 16 cases.
tion of lectin complement pathway in Henoch- J Am Soc Nephrol. 2004;15:787–95.
Schönlein purpura nephritis. Am J Kidney Dis. 60. Noris M, Brioschi S, Caprioli J, Todeschini M,
2005;45:295–302. Bresin E, Porrati F, et al. Familial haemolytic uraemic
48. Trendelenburg M, Fossati-Jimack L, Cortes- syndrome and an MCP mutation. Lancet.
Hernandez J, Turnberg D, Lewis M, Izui S, et al. 2003;362:1542–7.
29 Immune-Mediated Glomerular Injury in Children 925

61. Lapeyraque AL, Malina M, Fremeaux-Bacchi V, 73. Chan RW, Lai FM, Li EK, Tam LS, Chow KM, Lai
Boppel T, Kirschfink M, Oualha M, et al. Eculizumab KB, et al. Intrarenal cytokine gene expression in lupus
in severe Shiga-toxin-associated HUS. N Engl J Med. nephritis. Ann Rheum Dis. 2007;66:886–92.
2011;364(26):2561–3. 74. Khan SB, Cook HT, Bhangal G, Smith J, Tam FW,
62. Solà-Villà D, Camacho M, Solà R, Soler M, Diaz JM, Pusey CD. Antibody blockade of TNF-alpha reduces
Vila L. IL-1beta induces VEGF, independently of inflammation and scarring in experimental crescentic
PGE2 induction, mainly through the PI3-K/mTOR glomerulonephritis. Kidney Int. 2005;67:1812–20.
pathway in renal mesangial cells. Kidney Int. 75. Feldmann M, Pusey CD. Is there a role for TNF-alpha
2006;70:1935–41. in anti-neutrophil cytoplasmic antibody-associated
63. El-Shehaby A, Darweesh H, El-Khatib M, Momtaz M, vasculitis? Lessons from other chronic inflammatory
Marzouk S, El-Shaarawy N, et al. Correlations of diseases. J Am Soc Nephrol. 2006;17:1243–52.
urinary biomarkers, TNF-like weak inducer of 76. Chung AC, Lan HY. Chemokines in renal injury.
apoptosis (TWEAK), osteoprotegerin (OPG), J Am Soc Nephrol. 2011;22(5):802–9.
monocyte chemoattractant protein-1 (MCP-1), and 77. Liu ZH, Chen SF, Zhou H, Chen HP, Li
IL-8 with lupus nephritis. J Clin Immunol. 2011;31 LS. Glomerular expression of C-C chemokines in
(5):848–56. different types of human crescentic glomerulonephri-
64. Besbas N, Ozaltin F, Catal F, Ozen S, Topaloglu R, tis. Nephrol Dial Transplant. 2003;18:1526–34.
Bakkaloglu A. Monocyte chemoattractant protein-1 78. Kulkarni O, Pawar RD, Purschke W, Eulberg D,
and interleukin-8 levels in children with acute Selve N, Buchner K, et al. Spiegelmer inhibition of
poststreptococcal glomerulonephritis. Pediatr CCL2/MCP-1 ameliorates lupus nephritis in
Nephrol. 2004;19:864–8. MRL-(Fas)lpr mice. J Am Soc Nephrol. 2007;18:
65. Kitching AR, Turner AL, Wilson GR, Semple T, 2350–8.
Odobasic D, Timoshanko JR, et al. IL-12p40 and 79. Sheryanna A, Bhangal G, McDaid J, Smith J,
IL-18 in crescentic glomerulonephritis: IL-12p40 is Manning A, Foxwell BM, et al. Inhibition of p38
the key Th1-defining cytokine chain, whereas IL-18 mitogen-activated protein kinase is effective in the
promotes local inflammation and leukocyte recruit- treatment of experimental crescentic glomerulone-
ment. J Am Soc Nephrol. 2005;16:2023–33. phritis and suppresses monocyte chemoattractant
66. Calvani N, Tucci M, Richards HB, Tartaglia P, protein-1 but not IL-1beta or IL-6. J Am Soc Nephrol.
Silvestris F. Th1 cytokines in the pathogenesis of 2007;18:1167–79.
lupus nephritis: the role of IL-18. Autoimmun Rev. 80. Turner JE, Paust HJ, Steinmetz OM, Peters A, Meyer-
2005;4:542–8. Schwesinger C, Heymann F, et al. CCR5 deficiency
67. Hewins P, Morgan MD, Holden N, Neil D, Williams aggravates crescentic glomerulonephritis in mice. J
JM, Savage CO, et al. IL-18 is upregulated in the Immunol. 2008;181:6546–56.
kidney and primes neutrophil responsiveness in 81. Segerer S, Henger A, Schmid H, Kretzler M,
ANCA-associated vasculitis. Kidney Int. 2006;69: Draganovici D, Brandt U, et al. Expression of the
605–15. chemokine receptor CXCR1 in human glomerular
68. Kluth DC, Ainslie CV, Pearce WP, Finlay S, diseases. Kidney Int. 2006;69:1765–73.
Clarke D, Anegon I, et al. Macrophages transfected 82. Panzer U, Steinmetz OM, Reinking RR, Meyer TN,
with adenovirus to express IL-4 reduce inflammation Fehr S, Schneider A, et al. Compartment-specific
in experimental glomerulonephritis. J Immunol. expression and function of the chemokine IP-10/
2001;166:4728–36. CXCL10 in a model of renal endothelial microvascu-
69. Rodriguez W, Mold C, Kataranovski M, Hutt JA, lar injury. J Am Soc Nephrol. 2006;17:454–64.
Marnell LL, Verbeek JS, et al. C-reactive protein- 83. Wada T, Matsushima K, Kaneko S. The role of
mediated suppression of nephrotoxic nephritis: role chemokines in glomerulonephritis. Front Biosci.
of macrophages, complement, and Fcgamma recep- 2008;13:3966–74.
tors. J Immunol. 2007;178:530–8. 84. Ostendorf T, Rong S, Boor P, Wiedemann S,
70. Mu W, Ouyang X, Agarwal A, Zhang L, Long DA, Kunter U, Haubold U, et al. Antagonism of PDGF-
Cruz PE, et al. IL-10 suppresses chemokines, inflam- D by human antibody CR002 prevents renal scarring
mation, and fibrosis in a model of chronic renal dis- in experimental glomerulonephritis. J Am Soc
ease. J Am Soc Nephrol. 2005;16:3651–60. Nephrol. 2006;17:1054–62.
71. Lai PC, Smith J, Bhangal G, Chaudhry KA, Chaudhry 85. Floege J, Eitner F, Alpers CE. A new look at platelet-
AN, Keith Jr JC, et al. Interleukin-11 reduces renal derived growth factor in renal disease. J Am Soc
injury and glomerular NF-kappa B activity in murine Nephrol. 2008;19:12–23.
experimental glomerulonephritis. Nephron Exp 86. Huang XR, Chung AC, Zhou L, Wang XJ, Lan
Nephrol. 2005;101:e146–54. HY. Latent TGF-beta1 protects against crescentic
72. Chien JW, Chen WL, Tsui YG, Lee MC, Lin AY, Lin glomerulonephritis. J Am Soc Nephrol. 2008;19:
CY. Daily urinary interleukin-11 excretion correlated 233–42.
with proteinuria in IgA nephropathy and lupus 87. Ka SM, Huang XR, Lan HY, Tsai PY, Yang SM, Shui
nephritis. Pediatr Nephrol. 2006;21(4):490–6. HA, et al. Smad7 gene therapy ameliorates an
926 M. Nagata

autoimmune crescentic glomerulonephritis in mice. 96. Nomura K, Liu N, Nagai K, Hasegawa T,


J Am Soc Nephrol. 2007;18:1777–88. Kobayashi I, Nogaki F, et al. Roles of coagulation
88. Woolf AS. Angiopoietins: vascular growth factors pathway and factor Xa in rat mesangioproliferative
looking for roles in glomeruli. Curr Opin Nephrol glomerulonephritis. Lab Invest. 2007;87:150–60.
Hypertens. 2010;19(1):20–5. 97. Taneda S, Hudkins KL, M€ uhlfeld AS, Kowalewska J,
89. Shimizu A, Masuda Y, Mori T, Kitamura H, Pippin JW, Shankland SJ, et al. Protease nexin-1, tPA,
Ishizaki M, Sugisaki Y, et al. Vascular endothelial and PAI-1 are upregulated in cryoglobulinemic
growth factor165 resolves glomerular inflammation membranoproliferative glomerulonephritis. J Am
and accelerates glomerular capillary repair in rat anti- Soc Nephrol. 2008;19:243–51.
glomerular basement membrane glomerulonephritis. 98. Moussa L, Apostolopoulos J, Davenport P,
J Am Soc Nephrol. 2004;15:2655–65. Tchongue J, Tipping PG. Protease-activated recep-
90. Qi W, Chen X, Poronnik P, Pollock CA. Transforming tor-2 augments experimental crescentic glomerulone-
growth factor-beta/connective tissue growth factor phritis. Am J Pathol. 2007;171:800–8.
axis in the kidney. Int J Biochem Cell Biol. 99. Cheng H, Wang S, Jo Y, Hao CM, Zhang M, Fan X,
2008;40:9–13. et al. Overexpression of cyclooxygenase-2 predis-
91. Kanemoto K, Usui J, Tomari S, Yokoi H, poses to podocyte injury. J Am Soc Nephrol.
Mukoyama M, Aten J, et al. Connective tissue growth 2007;18:551–9.
factor participate in scar formation of crescentic glo- 100. Wu SH, Liao PY, Yin PL, Zhang YM, Dong L, et al.
merulonephritis. Lab Invest. 2003;83:1615–25. Elevated expressions of 15-lipoxygenase and lipoxin
92. Tang J, Liu N, Zhuang S. Role of epidermal growth A4 in children with acute poststreptococcal glomeru-
factor receptor in acute and chronic kidney injury. lonephritis. Am J Pathol. 2009;174(1):115–22.
Kidney Int. 2013;83(5):804–10. 101. Kondo S, Shimizu M, Urushihara M, Tsuchiya K,
93. Liu N, Mori N, Iehara N, Uemura K, Fukastu A, Yoshizumi M, Tamaki T, et al. Addition of the anti-
Kita T, et al. Soluble fibrin formation in the mesangial oxidant probucol to angiotensin II type I receptor
area of IgA nephropathy. Clin Exp Nephrol. antagonist arrests progressive mesangioproliferative
2007;11:71–6. glomerulonephritis in the rat. J Am Soc Nephrol.
94. Hertig A, Rondeau E. Role of the coagulation/fibri- 2006;17:783–94.
nolysis system in fibrin-associated glomerular injury. 102. Gilkeson GS, Mashmoushi AK, Ruiz P, Caza TN,
J Am Soc Nephrol. 2004;15(4):844–53. Perl A, Oates JC. Endothelial nitric oxide synthase
95. Cunningham MA, Kitching AR, Tipping PG, reduces crescentic and necrotic glomerular lesions,
Holdsworth SR. Fibrin independent proinflammatory reactive oxygen production, and MCP1 production
effects of tissue factor in experimental crescentic in murine lupus nephritis. PLoS One. 2013;8(5):
glomerulonephritis. Kidney Int. 2004;66:647–54. e64650.
Complement-Mediated Glomerular
Injury in Children 30
Zoltán Prohászka, Marina Vivarelli, and George S. Reusz

Contents Cell Death and Recovery in Response


to Complement Injury: The Lytic Terminal
Pathophysiology of the Complement Pathway Complement Complex . . . . . . . . . . . . . . . . . . . . . 944
System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 928
Outline of the Complement System . . . . . . . . . . . . . . . . . 928 Illustrative Examples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 945
Complement Activation Pathways . . . . . . . . . . . . . . . . . . 928 An Overview of the Mechanisms of
Control of Complement Activation . . . . . . . . . . . . . . . . . . 936 Complement-Mediated Glomerular Injury . . . . . . . . . . 945
Downstream Effects of Complement The Clinical Presentation of Glomerular
Activation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 938 Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 945
Complement Activation in Kidney Diseases . . . . . . . . 939 Antibody-Mediated Allograft Rejection
(AMR) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 952
Molecular Mechanisms of Complement- Tubulointerstitial Injury in Progressive
Mediated Renal Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 941 Kidney Disease: The Role of Complement . . . . . . . . . 953
Role of Structural Properties of
Kidney Components and of Local Production Therapeutic Inhibition of Complement
of Complement Factors and Regulators . . . . . . . . . . . . . 942 in Glomerular Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 953
Cellular Activation and Proliferation
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 954
in Response to Complement Activation:
The Non-lytic Terminal Pathway Complement
Complex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 942

Z. Prohászka (*)
3rd Department of Medicine, Faculty of Medicine,
Semmelweis University, Budapest, Hungary
e-mail: prohoz@kut.sote.hu
M. Vivarelli
Division of Nephrology and Dialysis, Children’s Hospital
Bambino Gesù-IRCCS, Rome, Italy
G.S. Reusz
1st Department of Pediatrics, Faculty of Medicine,
Semmelweis University, Budapest, Hungary

# Springer-Verlag Berlin Heidelberg 2016 927


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_26
928 Z. Prohászka et al.

Pathophysiology of the Complement role in platelet activation and cell contact sys-
System tem [7]. Moreover, complement components are
important regulators of cellular immunity mecha-
Outline of the Complement System nisms through their role in pathways involved
in pattern recognition (e.g., toll-like receptors)
The complement system comprises about 30–40 or can stimulate cell-surface receptors through
proteins, glycoproteins, and membrane receptors anaphylatoxins [8].
that are present in body fluids and on cell surfaces Overall, the complement system has general
(Tables 1, 2, and 3). Nearly all the soluble proteins sensing functions that trigger powerful biological
are produced by the liver, but most are also syn- actions under tight control from regulatory pro-
thesized in other tissues, including cells of the teins. It represents an essential triage mechanism
myeloid lineage and renal cells. Complement is that constantly protects the organism against
considered part of the immune system, as its main threats such as pathogenic microorganisms or
biological function is the lysis and removal of cell debris. When properly activated, complement
pathogens by opsonization. In the past decade, promotes cellular innate immunity and initiates
new studies have revealed a pivotal role of com- protective immunological processes. Improper or
plement in immune surveillance, the generation of insufficient activation can lead to loss of self-
inflammation, the initiation of antibody produc- tolerance, to the development of autoimmunity,
tion, and tissue repair [1–3]. or to tissue damage [3].
Complement recognition proteins can bind to a Genetic variants and differences in copy num-
variety of targets. They can trigger enzymatic ber of genes encoding for complement proteins
cascade reactions (complement activation path- and regulators together establish the complotype
ways), can opsonize and lyse particles or cells, of any given individual. The intensity of the
and can release several biologically active small complement response and of the downstream
proteins, termed anaphylatoxins. The activity of inflammatory reaction is largely dependent on
this system is tightly controlled by a number of this complex haplotype, which can predispose
soluble and surface-bound regulatory proteins. to a number of complement-related diseases,
The pathophysiological alterations of the comple- including autoimmune, inflammatory, auto-
ment system include overactivation and consump- inflammatory, degenerative, hematologic, ische-
tion, secondary to the sustained presence of mic, or renal disorders [9].
activators, deficiencies secondary to missing com- The aim of this chapter is to provide an over-
plement factors, or dysregulation, secondary to view of the complement system and of the molec-
altered regulatory proteins [3]. ular mechanisms of complement-mediated
All complement activation pathways result in glomerular damage, including diagnostic and
the formation of the C3 convertase complex, therapeutic tools.
which activates the central C3 component
(Fig. 1). This in turn triggers the formation of the
C5 convertase, resulting in C5 cleavage, which Complement Activation Pathways
marks the beginning of the terminal pathway.
Complement components also interact with The general mode of activation of the complement
other systems [4]. Activated proteases of the coag- system is similar to that of other plasma protease
ulation system, for example, can activate C3 cascades (e.g., to the coagulation system). Struc-
directly, bypassing traditional complement activa- tural changes in recognition complexes trigger the
tion pathways. Likewise, recognition molecules activation of proteases that in turn activate other
of the lectin pathway bind fibrinogen or fibrin zymogen proteases through limited proteolysis,
[5], whereas complement protease MASP-2 can leading to a cascade of reactions that result in the
activate prothrombin [6]. Other observations formation of heterocomplexes and in the release
also suggest that complement plays an important of anaphylatoxins (Fig. 1, Table 1). Complement
30 Complement-Mediated Glomerular Injury in Children 929

Table 1 Components of the complement pathways


Average
serum
Activator(s) of this This component concentration Molecular
Component Attributes component activates (mg/L) mass (kDa)
C3 The central C3 convertases of the C5 (as part of the C5 1,200–1,300 190
component of all classical/lectin convertase)
three (C4b2a) or alternative
complement (C3bBbP) pathways
pathways
Factor D Serine protease Activity of FD is FB 1 93
increased if bound to
C3bB
Factor B Serine protease, FD C3 200 25
binds to C3b
Properdin Recognition Binds to target surfaces and stabilizes the AP C3 25 55
component and convertase (monomeric
positive form)
regulator of the
AP. Does not
have enzymatic
activity
C1q 18 chain (6  IgG and IgM immune C1r 100–180 460
[ABC]) complexes, C-reactive
recognition protein, viral proteins,
component of cardiolipin,
the CP. Does not lipopolysaccharide,
have enzymatic polyanions, etc.
activity
C1r Serine protease Facilitation of C1s 50 85
autoactivation by the
conformational
change of C1q
C1s Serine protease C1r2 C4 50 85
C4 Common C1r2s2/MASP- Component of the C3 300–600 210
component of 12MASP-22 convertase
the classical and
lectin pathways
C2 Serine protease C1r2s2C4b or C3 20–25 110
MASP-12MASP-
22-C4b
MBL Carbohydrate Carbohydrates MASP-1 0.002–10 96 (trimer)
recognition (mannose > N-
macromolecule acetylglucosamine >
(2  (3  6) L-fucose > N-
chains) acetylmannosamine
> glucose)
Collectin Carbohydrate Carbohydrates MASP-1 0.28 200 (dimer
11 recognition (mannose > N- of trimeric
molecule acetylglucosamine > subunits)
L-fucose)
Ficolin-1 Recognition of Microbial surfaces, MASP-1 0.06 35
Ficolin-2 carbohydrate apoptotic bodies, 5 34
Ficolin-3 structures and altered self-structures 30 33
acetylated
molecules
(continued)
930 Z. Prohászka et al.

Table 1 (continued)
Average
serum
Activator(s) of this This component concentration Molecular
Component Attributes component activates (mg/L) mass (kDa)
MASP-1 Serine protease Facilitation of MASP-2 1.5–13 93
autoactivation by
MBL
MASP-2 Serine protease MASP-1 C4 0.5 76
C5 Binds C6 C5 is cleaved by the Components of the 70 190
C6 Binds C7 classical/lectin terminal pathway lack 60 110
C7 Binds C8 pathway (C4b2a3b) enzymatic activity, 55 10
or by the alternative conformational
C8 Binds C9 55 150
pathway (C3bBb3b) change of C5 upon
C9 Polymerization, C5 convertases cleavage results in 60 70
pore formation sequential binding of
C6–C9,
polymerization of C9
and pore formation

activation is heavily influenced by multiple fac- of the internal thioester bond, yielding C3(H2O).
tors, such as complement regulators, composition Once formed, C3b can bind covalently to cell-
of target surfaces, and blood flow. surface carbohydrates or to other macromole-
cules, via its reactive thioester group. C3b also
Complement Component 3 (C3): The has opsonizing and immunoregulatory functions.
Pivotal Molecule of Complement Surface-bound C3b is rapidly broken down by
Activation Pathways factor I into the inactivated form of C3b (iC3b,
Complement component C3 is a pivotal molecule see below) and into C3f (Fig. 2a). iC3b is further
that is common to the three main complement degraded by several proteases into C3dg and C3d,
pathways [10]. C3 is the most abundant comple- as well as into C3c (a large fragment cleaved off
ment protein in human plasma. It is produced from the surface). Complement regulators may
mainly by the liver but also by other cell types. facilitate the dissociation of the enzyme complex,
C3 plasma concentrations are approximately thereby accelerating its decay (Fig. 2b).
1.2–1.3 g/L. C3 is produced as a pre-pro single-
chain molecule including a 22-amino-acid leader The Alternative Pathway
peptide and a four-amino-acid cleavage site. The alternative pathway (AP) of complement is
Mature C3 has two chains, alpha and beta, that phylogenetically the oldest of the complement
are linked by a disulfide bond. Two major mech- pathways. Until recently, initiation of the AP
anisms for C3 activation have been recognized, was not attributed to the recognition of specific
namely spontaneous and proteolytic activation molecules [11]. The AP relies primarily on the
mechanisms, both resulting in conformational spontaneous, slow hydrolysis of C3 in the plasma
changes that are essential for the downstream to form C3(H2O), a mechanism also termed the
activation of the terminal pathway. The C3 tickover mechanism. Tickover of C3 results in
convertase activates C3 by cleaving the alpha conformational changes that increase its binding
chain, thereby releasing C3a anaphylatoxin and to factor B, allowing cleavage of factor B by factor
generating C3b. Active C3 convertases form as a D. Factor D is a self-inhibitory serine protease
result of the activation of all three complement that is produced by adipocytes [12]. Factor B is a
pathways. Alternatively, spontaneous activation serine protease that is produced in a zymogen
of C3 can also take place through the hydrolysis form. When bound to C3b, factor B becomes
30 Complement-Mediated Glomerular Injury in Children 931

Table 2 Soluble (fluid-phase) complement control proteins


Serum Molecular
concentration mass
Component Attributes Binds to Effect (mg/L) (kDa)
C1-inhibitor Serine protease C1r2s2 and Dissociation and 200–240 105
inhibitor (Serpin) MASPs covalent binding of
serine proteases of
recognition
complexes
C4b-binding protein CCP domain C4b, heparin Inactivation of CP C3 150–300 67 (alpha
containing convertase, cofactor chain)
molecule of factor I
(7 chains)
Factor H CCP domain Glycans with Inactivation of AP C3 200–600 150
containing terminal sialic convertase, cofactor
molecule acid, of factor I
(1 chains) C-reactive
protein, C3b,
heparin
Factor I Serine protease C3b and C4b Cofactor-dependent 30–50 88
cleavage of C4b and
C3b
Vitronectin Multidomain C5, C6, C7, Competes with MAC 300–500 85
regulator of C8, C9, and for binding to
coagulation, beta-1 membranes and
fibrinolysis, and integrins, inhibition of C9
complement heparin incorporation into
cascades MAC
Clusterin Highly conserved C7, C8, and Inhibition of MAC 50–100 80
heterodimer with C9 formation
five disulfide
bonds
Carboxypeptidase N Serum C3a, C4a, C5a Inactivation of 35 310
metalloprotease anaphylatoxins and
kinins by the
cleavage of terminal
basic amino acids

susceptible to cleavage by factor target surfaces. The composition of target cell


D. Conformational changes occur after formation surfaces, the presence of complement regulators,
of C3bB, resulting in an active enzyme form. This and the characteristics of blood flow over cell
results in the production of a large, proteolytically surfaces are all important determinants of magni-
active Bb molecule and of a smaller Ba tude of local alternative pathway activation.
fragment [1]. C3bBb is an unstable enzyme with a dissociation
The fluid-phase C3bBb protein complex forms half-life of Bb from C3b of approximately 2–3
the alternative pathway C3 convertase, which in min. Properdin binding to the C3bBb stabilizes
turn will cleave more C3 in the plasma. C3b the complex and prolongs its half-life to 20–30
molecules that are generated bind covalently to min. Properdin is a multimeric molecule (3 
cell-surface molecules. Factor B attaches to bound 7 monomers arranged in a triangular array) with
C3b to form C3bB. Factor D then produces a multiple binding sites that recognizes charge clus-
surface-bound alternative pathway convertase, ters and probably other motifs on zymosan, rabbit
which in turn can cleave more C3 and initiate erythrocytes, and late apoptotic or necrotic cells,
the alternative pathway amplification loop on which are all known activators of the AP. As first
932 Z. Prohászka et al.

Table 3 Cell-surface (membrane-bound) complement control proteins and receptors


Molecular
mass
Receptors Attributes Ligand(s) Function Expression (kDa)
CR1 (CD35) Single-chain C3b, C4b Cofactor for factor I Erythrocytes, cells of 190–260
transmembrane Decay-accelerating myeloid and lymphoid
receptor with activity lineages, dendritic
CCP domains cells, follicular
dendritic cells,
podocytes
Membrane Single-chain C3b, C4b Cofactor for factor I Nucleated cells 45
cofactor protein transmembrane
(MCP, CD46) receptor with
CCP domains
Thrombomodulin Single-chain Prothrombin, Increases CFH-dependent Endothelial cells 60
transmembrane protein C, and factor I-mediated
receptor with plasminogen degradation of C3b,
C-type lectin activator facilitates the degradation
domain inhibitor-1 of anaphylatoxins
Decay- GPI-anchored C3bBb and Facilitation of dissociation Nucleated cells 70
accelerating single-chain C4b2a of CP and AP C3 and C5
factor (DAF, molecule convertases
CD55)
CD59 (protectin) GPI-anchored C8, C9 Inhibits MAC formation, Nucleated cells 18
single-chain protection of “innocent
molecule bystander” cells from
action of MAC
CR2 (CD21) Single-chain C3d, C3dg Facilitation of antigen B lymphocytes, 120
transmembrane presentation and B-cell activated T
receptor with responses lymphocytes, follicular
CCP domains dendritic cells
CR3 (CD11b/ Heterodimer iC3b Facilitation of Monocytes, 165/95
CD18) phagocytosis macrophages,
neutrophil
granulocytes, NK cells,
and T cells
CR4 (CD11c/ Heterodimer iC3b Facilitation of Monocytes, 165/95
CD18) phagocytosis macrophages,
neutrophil
granulocytes, NK cells,
and T cells
C3aR 7TM GPCR C3a Cell activation (see the Mast cells, neutrophil, 53
text) eosinophil, basophil
granulocytes,
monocytes,
macrophages,
platelets, neurons
C5aR (CD88) 7TM GPCR C5a, Cell activation (see the Mast cells, basophil 40
C5a-desArg text) granulocytes,
monocytes,
macrophages, platelets
cC1qR CD91 and Collagen part Facilitation of Leukocytes 500 and 48
calreticulin of defense phagocytosis
lectins
GPI glycosylphosphatidylinositol, 7TM GPCR 7-transmembrane domain G-protein-coupled receptor
30 Complement-Mediated Glomerular Injury in Children 933

Activators of classical pathway: Activators of lectin pathway: Activators of alternative pathway:


Antibodies, acute phase proteins, Repetitive carbohydrate structures Surfaces allowing the binding of
Apoptotic and necrotic bodies Altered self structures properdin and C3b

C1q, C1r-C1s MBL, Ficolins, MASPs Properdin, Factor D


C1-INH

C4, C2 C3b, Factor B C3(H2O)

Factor I Factor Factor


C4BP
H I

MCP CR1

C3 convertases
Action of defence collagens, C3b, C3d, C3dg:
DAF Action of anaphylatoxins C3a, C5a:
Opsonisation, facilitation of phagocytosis
Proinflammatory action
Cell activation
Regulation of adaptive immunity
Regulation of adaptive immunity
C5 convertases CPN
Vitron-
ectin

CD59 Clusterin

Terminal pathway Action of membrane attack complex:


Induction of cell activation and proliferation
Membrane attack complex Lysis of target cells

Fig. 1 A simplified scheme of the complement system. level of C3 activation. The latter may give rise to the
The activators (boxes with arrows) of the complement formation of C5 convertase, the initiation of the terminal
recognition complexes of the classical and lectin pathways complement pathway, and the polymerization of C9, con-
induce conformational changes. These result in the auto- cluding by the formation of the membrane attack complex.
and hetero-activation of zymogen serine proteases (C1s, Multiple fluid-phase and plasma membrane-bound regula-
C1r, and MASPs) and in the formation of C3 convertase tors of complement activation (gray triangles) prevent the
enzyme complex of the CP and LP. The surfaces initiating uncontrolled activation of the cascade. The biological
alternative pathway activation are characterized by proper- actions of complement are mediated by surface-bound
din binding. The amplification of C3 activation may follow molecules (opsonins), small peptide mediators
– mediated by factor B and factor D (alternative pathway (anaphylatoxins), and the cellular effects of membrane
amplification loop), but C3 may undergo spontaneous con- attack complex (indicated in boxes). See the molecular
stant activation by the tickover mechanism as well. The details of complement components and activation in the
three complement activation pathways converge on the text and in Tables 1, 2, and 3

suggested by Pillemer at the end of the 1950s, changes in the hinge region and in the Fc domains
properdin acts as an initiator and amplificator of [10]. These changes enable the C1 complex (com-
the AP [13]. prising one recognition molecule C1q and two
If C3b molecules bind covalently to the C3 pairs of zymogen serine proteases C1s and C1r)
convertase, it generates the AP C5 convertase to bind to antibodies. Upon binding of the C1
(C3bBbC3b). complex to antibodies, C1q undergoes a confor-
mational change, which activates the C1r zymo-
The Classical Pathway gen. In turn, active C1r cleaves and activates C1s.
The classical pathway (CP) of complement is Activated C1 complex cleaves C4 and C2 and
initiated by the binding of immunoglobulins to generates the classical pathway C3 convertase
their target antigens, resulting in conformational (C4b2a). The complement component C4 is
934 Z. Prohászka et al.

Fig. 2 The principles of complement regulation. Panel Panel c: Control of active serine proteases by serpins
a: Control of C3 convertases by cofactor-mediated (serine protease inhibitors, example of C1 inhibitor).
cleavage. Factor I mediates the degradation of surface- Multiple serine protease inhibitors (including C1-inhibitor,
bound C3b by cofactor-mediated cleavage. Cofactors are antithrombin, alpha-2 macroglobulin) act on active serine
fluid-phase proteins (factor H or C4BP able to bind to neg- proteases of the complement system. The archetypal serpin
atively charged structures on cell surfaces) or transmembrane inhibitor of CP and LP is C1-INH acting in a canonical
complement regulators present on mammalian cells (CR1 (“suicidal”) manner on C1r, Cs, and MASPs. By forming
and MCP). The formation of a molecular complex with its covalent complexes with these active proteases, as well as by
cofactors enhances the protease activity of factor I in a facilitating the dissociation of recognition complexes,
substrate-induced (C3b or C4b) manner, and C3b (and C1-INH plays a pivotal role in the control of both the CP
C4b) degradation (release of C3f) occurs. Panel b: Control and the LP. Panel d: Control of terminal complement
of C3 convertases by decay-accelerating activity. The pathway (example of CD59). The mammalian cell-surface
molecules with decay-accelerating activity are factor H, regulator CD59 of the terminal pathway inhibits the incor-
DAF, and CR1. These mediate the destabilization of C3 poration and polymerization of C9 into the terminal comple-
convertases and facilitate the dissociation of Bb from C3b. ment complex, in order to protect host surfaces from attacks
30 Complement-Mediated Glomerular Injury in Children 935

structurally and functionally similar to C3 in that addition, MASPs may cleave proteins of the coag-
it has an internal thioester bond and it is capable of ulation pathway [6] and have a stimulatory activ-
covalent binding to surfaces. Activated C1 com- ity on endothelial cells [17].
plex splits C4 into a large C4b fragment that binds Activators of the lectin pathway interact with
to target surfaces and has an enhanced affinity to MBL in a calcium-dependent manner through the
C2 and a smaller C4a fragment that has weak carbohydrate recognition domain of MBLs.
anaphylatoxin activity. Complement component Repeat sugar motifs terminating with mannose,
C2 is a zymogen serine protease that, once glucosamine, or fucose can bind oligomeric
cleaved by activated C1 complex into C2a (large MBLs and induce conformational changes. Bac-
proteolytic fragment) and C2b, forms together terial and viral structures, for example, have been
with C4b the C3 convertase complex of the CP shown to activate MBLs. Surface-bound MBLs
(C4b2a). As for the AP, C3b can make covalent may also act as opsonins. The lectin pathway does
bonds with the C3 convertase to form the CP C5 not require antibodies; however, MBLs can bind
convertase complex (C4b2a3b). certain glycan structures of IgG and IgA [18].
In addition, C1q can also bind directly to repeat Similar to the collectins, ficolins are oligomeric
structures of non-immunoglobulin activators of recognition molecules of the lectin pathway. They
the CP, such as nucleic acids, chromatin, cyto- have collagen-like domains, form complexes with
plasm intermediate filaments, mitochondrial pro- zymogen MASPs, and, upon binding, facilitate
teins, cardiolipin, ligand-bound C-reactive their autoactivation. In contrast to collectins,
protein, protein aggregates (e.g., amyloids), bac- ficolins contain fibrinogen-like carbohydrate rec-
terial polysaccharides, and artificial surfaces (car- ognition domains and exhibit specific differences
bon nanotubes and polymers) [14]. These in target preference. Three ficolins have been
activators represent important potential mediators identified in humans until now, namely, ficolin-1,
of glomerular damage. ficolin-2, and ficolin-3 (formerly M-ficolin,
L-ficolin, and H-ficolin). Ficolin-3 is the most
The Lectin Pathway abundant, with a plasma concentration of approx-
Two collectins – collagen-containing C-type imately 30 mg/L. Few binding targets for ficolin-3
lectins (i.e., mannose-binding lectin, MBL) and have been shown, including N-acetylated struc-
collectin 11 – and three ficolins (fibrinogen colla- tures present on apoptotic cells [19] and other
gen lectins, i.e., ficolin-1, ficolin-2, and ficolin-3) bacterial targets. Ficolin-2 binds preferentially to
compose the recognition molecules of the lectin acetylated sugars, to some bacterial targets, and to
pathway [15]. These oligomeric molecules circu- C-reactive protein. Ficolin-1, the least abundant
late in the plasma as complexes formed with plasma ficolin, plays a major role in facilitating
zymogen serine proteases termed MBL-associ- phagocytosis [20].
ated serine proteases (MASPs). MASP-1 and Once recognition molecules of the lectin path-
MASP-3 are alternatively spliced products of the way have bound to their targets and conforma-
masp1/3 gene and have identical recognition tional changes have autoactivated MASPs, C4
domains but different serine protease domains, and C2 are cleaved. From there on, the lectin
whereas MASP-2 is encoded by the masp2 gene. pathway converges with the classical pathway to
Recognition molecules of the lectin pathway bind form the LP C3 convertase (C4b2a), subsequently
only one protease at a time, and in contrast to C1r activating the terminal pathway.
and C1s, activation of MASPs only involves
autoactivating mechanisms. The autoactivation The Terminal Pathway
of MASP-1 is much faster than that of MASP-2, The formation of the membrane attack complex
suggesting that MASP-1 is the primary player in (MAC, C5b-9) begins with the cleavage of C5 by
the activation of the lectin pathway [16]. Active C5 convertases. C3 convertases of all pathways
MASP-2 can cleave C4 and C2; the exact func- can cleave efficiently C3 but have weak activity
tions of MASP-1 and MASP-3 are unclear. In toward C5. However, after incorporating an
936 Z. Prohászka et al.

additional C3b molecule, C3 convertases form C5 include factor H, C4-binding protein, and surface-
convertases, and C5 is cleaved into C5a, the most bound regulators, such as the membrane cofactor
potent anaphylatoxin, and C5b. Since C5 lacks an protein (CD46 or MCP) and complement receptor
internal thioester bond, it does not bind covalently 1 (CD35 or CR1). A common structural property of
to the target surface. However, upon binding, these cofactors is the presence of multiple comple-
conformational change occurs, allowing subse- ment control protein (CCP) domains, each
quent aggregation of C5b with C6, C7, C8, and consisting of 60 amino acids. By way of
multiple copies of C9. Binding and fusion of their multiple binding domains, these cofactors
terminal pathway components in the lipid bilayers bring a regulatory protease, such as factor I, and
is reversible and involves ionic and hydrophobic C3b in close spatial vicinity and facilitate the
interactions. The pore assembly takes place proteolytic breakdown of C3b. Of notice, several
through well-defined heteropolymeric complexes, factor H-related proteins (CFHRs) belong to
collectively termed terminal complement com- factor H family [23]. These proteins lack strong
plexes (TCC). The final pore-forming complex complement inhibitory function. However, they
(C5b-9n), which can incorporate as many as may play an important role in fine-tuning
16 C9 molecules, is located deep in the lipid C3 activation.
bilayer and is a most effective inducer of cell A second mechanism for controlling C3 acti-
activation and death. vation relies on the decay-accelerating activity of
complement control proteins such as cofactors
(factor H, CR1) and the decay-accelerating factor
Control of Complement Activation (CD55 or DAF), which competitively dissociate
the C3 convertase enzyme complexes (Fig. 2b).
The complement system is strictly regulated to The control of the classical and of the lectin
prevent uncontrolled activation and tissue dam- pathway recognition molecules is mediated by the
age. Since the AP has the inherent ability of C3 serine protease C1 inhibitor (C1-INH), belonging
tickover-based amplification, tight control is of to the family of serpins. C1-INH can dissociate C1
particular importance. The major complement complexes (Fig. 2c) and can inactivate MASPs.
control proteins can be subdivided in fluid-phase Alpha-2-macroglobulin has very similar activities
and surface-bound molecules (Tables 2 and 3). on complement molecules. C4b, the common
Their activities include inhibition of active serine component of the classical and lectin pathways,
proteases, facilitation of C4 and C3 breakdown, is also regulated by factor I-mediated cleavage,
acceleration of C3 and C5 convertase decay, and similarly to C3. The C4-binding protein (C4BP)
prevention of MAC formation on target surfaces serves as cofactor. It circulates in complex with
(Fig. 2, panels a–d) [21]. The activity of comple- protein S, a vitamin K-dependent cofactor for the
ment control proteins is often mediated by com- breakdown of the coagulation factors Va and
plex molecular mechanisms that also require the VIIIa by protein C. Hence, C4BP represents a
presence of cofactors, of other proteases, and of link between regulation of the coagulation and of
surface receptors. the complement cascades.
Because of its central role, C3 is under partic- Finally, the control of the terminal pathway on
ularly strict control. First, C3b is rapidly cell surfaces is largely achieved by the abundant
inactivated by factor I, which after cleavage of expression of the membrane-bound glycosylpho-
three peptide bonds, generates iC3b from C3b, sphatidylinositol-linked protein CD59 (protectin)
which can no longer interact with factor B and and by two fluid-phase regulators, namely,
with C5 (Fig. 2a). Factor I is a serine protease that vitronectin and clusterin. These molecules control
requires cofactors to function [22]. The catalytic the formation of the membrane attack complex by
activity of factor I is regulated by an internal inhib- binding to various intermediate complexes
itory domain and by the temporary presence of its preventing the recruitment of C9 molecules
substrate (C3b and C4b). Cofactors of factor I (Fig. 2d). Furthermore, by inhibiting access to
30 Complement-Mediated Glomerular Injury in Children 937

a Endothelial injury mediated by immunoglobulin


induced complement activation

Swelling and
detachment of
endothelial cells

b Endothelial injury mediated by


dysregulation of complement activation on the
surface of endothelial cells

Enlarged area
Podocyte

Slit diaphragm Swelling and


detachment of
endothelial cells

Platelet
Pedicles of and fibrin
Endothelial podocytes rich thrombi
Glomerular glycocalyx
basement membrane c Deposition of complement fragments in the
Fenestrated endothelial cell GBM with or without immunoglobulins

Partial cross section


of a glomerular capillary

Thickening of GBM,
double contour
Heavy deposition of
complement fragments

d Local complement activation by deposited


autoantibodies and immune complexes

Penetration and
deposition of
antibodies to
variable depth
Damage of podocytes

Fig. 3 Schematic representation of complement- glomerular basement membrane, and the podocytes with their
mediated glomerular injury. A section of the three-layered interdigitating pedicles. The enlarged areas show the charac-
filtration barrier in the glomerulus is presented: exhibiting the teristic initial alterations of this filtration barrier in four distinct
fenestrated endothelium with the mesh-like glycocalyx, the types of complement-mediated glomerular injury. The yellow
938 Z. Prohászka et al.

binding sites on phospholipid bilayers, these reg- C3b-coated particles in the bloodstream bind to
ulators protect neighboring host cells from spread- CR1 on erythrocyte membranes. Since CR1 is a
ing of MACs from one cell to another. cofactor to factor I, C3b is gradually converted
into iC3b, which increases binding to complement
receptors 3 and 4 (CR3 and CR4) expressed by
Downstream Effects of Complement phagocytes. Uptake of iC3b-coated particles by
Activation phagocytic cells, mainly macrophages, occurs
predominantly in the spleen and in the liver.
Once particles or a cell surface are recognized by Impaired activation and degradation of C3 com-
complement recognition molecules, several mol- promises clearance of apoptotic bodies and
ecules of C3b attach. C3 activation results in immune complexes. Moreover, C3b-coated parti-
multiple biological effects aimed at defending cles can also be converted into C3d and C3dg,
the host, including opsonization, activation, and which are recognized by complement receptor 2
migration of inflammatory cells, release of inflam- (CR2) molecules present on the surfaces of B
matory molecules, and lysis of the target cells [3]. cells, activated T cells, and follicular dendritic
These aspects are briefly illustrated below. cells. Thus, opsonization of target particles also
C3 activation promotes clearance of apoptotic plays a pivotal role in antigen presentation, elicits
bodies and of immune complexes by opsonization. germinal center reactions, and contributes to the

Fig. 3 (continued) color marks the dominant site of depos- immunoglobulins are detected by immunofluorescence
ited complement fragments, as revealed by immunofluo- microscopy (immune-complex-mediated membranoproli-
rescence, and the green color marks immunoglobulins. Of ferative glomerulonephritis). On the other hand, fluid-
note, proliferative cells and mesangium are not represented phase complement activation (such as in the presence of
in the enlarged areas. Enlarged area A: Acute endothelial C3 nephritic factor) or the lack of an effective alternative
injury (endocapillary glomerulonephritis) is caused by pathway regulation (in various forms of complement-medi-
immunoglobulin-induced complement activation early in ated C3 glomerulopathies) may result in the presence of
the disease process, followed by IgG deposition. Cellular high amounts of activated C3 (C3b) in the plasma. This
infiltration is the consequence of the liberation of may accumulate (together with complement activation
complement-derived anaphylatoxins. Later, in the disease products of the terminal pathway, i.e., SC5b-9) within the
course, immunoglobulins and/or immune complexes may GBM. The result is similar thickening, deposition of
penetrate the basement membrane and form subepithelial electron-dense material, and the formation of the charac-
deposits as in postinfectious glomerulonephritis or in the teristic double contour of the GBM. Typically, the comple-
presence of donor-specific antibodies after transplanta- ment deposits (mostly activated and/or degraded forms of
tion. Enlarged area B: Failing complement regulation C3) without immunoglobulins can be detected by immuno-
due to loss of complement regulators in the endothelial fluorescence. Cell proliferation and mesangial cell interpo-
glycocalyx (such as in case of complement-mediated atyp- sition may accompany these processes. Enlarged area D:
ical HUS) with resulting complement activation may also Autoantibodies or immune complexes circulating in the
lead to capillary damage. Anaphylatoxins and the terminal blood penetrate through the endothelial fenestrae and the
complement complex both contribute to the activation and GBM. Subsequently, they can (1) get deposited on the GBM
damage of endothelial cells. This leads to changes in phe- in a linear pattern (e.g., in anti-GBM-mediated disease) or
notype, increased adhesion of leukocytes and platelets, loss (2) penetrate into and reside in the subepithelial space after
of cellular integrity, and detachment from the basal mem- an initial phase (such as in case of advanced lupus nephritis)
brane. Endothelial swelling, activated platelets, and fibrin- or (3) recognize their targets on the podocytes (such as in the
rich microvascular thrombi may occlude the lumen of case of anti-PLA2R autoantibodies in membranous
glomerular capillaries, reducing perfusion. As a rule, com- nephropathy). The deposited immunoglobulins, as well as
plement C3 deposits without immunoglobulins but with the components (C1q, C4, C3) and the activation products
fibrin. Enlarged area C: Immune complexes with specific (C4d, SC5b-9) of the classical and terminal pathways, can be
properties may bind to the basement membrane and cause detected in the subepithelial space by immunofluorescence.
complement activation, inflammation, mesangial cell Complement activation is largely separated from the capil-
interposition between the endothelium and the basement lary lumen; hence, recruitment of inflammatory cells is
membrane, and production of extracellular matrix leading limited but direct complement-mediated damage of the
to thickening of the basement membrane with the charac- podocytes occurs
teristic double contour appearance. Complement and
30 Complement-Mediated Glomerular Injury in Children 939

development of memory B cells. Additional It is increasingly clear that the complement


receptors for C1q and other recognition molecules system has multiple interactions with other sys-
of the lectin pathway promote attachment and/or tems, such as the immune and coagulation sys-
uptake by phagocytes and antigen-presenting tems. As new drugs that modulate the
cells. complement activity are being developed, their
Complement activation facilitates and effects on these pathways need to be carefully
enhances inflammatory reactions. The most scrutinized.
potent inflammatory mediators, anaphylatoxins
C5a and C3a, act on a wide variety of immune
and nonimmune cells. These small peptides with Complement Activation in Kidney
short half-lives exert their effects by binding to Diseases
7-transmembrane domain G-protein-coupled
receptors. Downstream effects on cells include Kidneys are particularly susceptible to
metabolic changes, degranulation, activation, complement-mediated attacks, due to their exten-
and enhanced cell migration. For the most part, sive endothelial surface, high blood flow, vascular
C3a and C5a have similar pro-inflammatory organization, and local production of complement
effects and can activate eosinophils, basophils, components. Broadly speaking, there are two
and mast cells. However, C3a has no effect on major groups of complement-mediated kidney
neutrophils, as opposed to C5a, which is a potent diseases. The first (1) is associated with increased
chemoattractant and activator of these cells. Acti- activation and consumption of complement and
vated mast cells release preformed TNF-alpha and involves the classical (and in certain cases the
increase histamine and eicosanoid production. lectin) pathway. The second (2) is characterized
This results in vasodilation, enhanced vascular by complement dysregulation and involves the
permeability, and recruitment of other immune alternative pathway. In the clinical practice, func-
cells at the site of complement activation. Further- tional, immunological, biochemical, and genetic
more, in conjunction with TNF-alpha, tests help distinguish these two disease mecha-
anaphylatoxins increase expression of cell adhe- nisms. This also affects protocols for treatment
sion molecules (e.g., E-selectin, ICAM-1, and monitoring. The term complement profile is
VCAM-1) on endothelial cells [24], facilitating often used to describe the multitude of comple-
adhesion and migration of leukocytes. The activ- ment parameters measured in a patient at a given
ities of anaphylatoxins are controlled by carboxy- time. This “snapshot” reflects the activity of com-
peptidase N, a serum metalloprotease that inhibits plement pathways (from the activation of a given
their biological effects by cleaving basic amino pathway to the polymerization of C9), along with
acids at the terminal ends. the levels of complement proteins and activation
Classically, cell lysis mediated by MAC is markers, and the presence of autoantibodies
considered the most important downstream effect against complement proteins (Table 4).
of complement activation. Deficiencies of termi- Mechanisms leading to complement activation
nal pathway components, however, do not support in kidney diseases are:
the view that MAC is a critical defense mecha-
nism in humans. Patients with C6, C7, C8, or C9 1. The sustained presence of antibodies, leading
deficiencies, for example, demonstrate increased to overactivation and consumption of the clas-
susceptibility only to a restricted number of bac- sical (and in certain cases the lectin) pathway.
teria, mainly some Neisseria species. Pathogens These antibodies may be generated in response
and tumor cells can in fact activate escape mech- to persistent stimulation by antigens (such as
anisms to prevent damage from complement in infections) and by malignant cells (mainly
attack. Sublytic amounts of terminal pathway in B-cell neoplasia) or may be secondary to
complexes, however, may play an important role autoimmune disease (immune complexes).
in nonlethal signaling events (see below). If present in excessive amount or proportion,
940 Z. Prohászka et al.

Table 4 Characteristic alterations and interpretation of complement parameters in pediatric kidney diseases
Interpretation of the
Package Tests Indication results Comments
Baseline C3, C4, and All pediatric Low levels of all three: Required for all patients
complement profile CH50 glomerulonephritides activation of CP
Decreased C3 with
normal C4: activation
of AP
Decreased C4 with
normal C3: suspected
cryoglobulins and/or
HCV infection
Functional Classical and Suspected May help to identify the Global functional
screening of alternative complement missing factor in the testing of complement
complement pathway deficiency case of complement
activity activity deficiency
Classical pathway “Baseline” Suspected (auto) Low levels of C1q, C3, May be particularly
and C1q, anti- antibody-mediated and C4 activity points helpful for the
C1q IgG, C2 glomerular to activation and differential diagnosis of
pathology consumption of CP antibody- versus
Low level of a single dysregulation-mediated
factor (C1q, C4 or C2) pathologies
and decreased CP
activity indicates factor
deficiency
Anti-C1q IgG positivity
causes acquired
deficiency of CP and is
indicative for lupus
nephritis or, in the
presence of urticaria, of
hypocomplementemic
urticarial vasculitis
syndrome
Alternative “Baseline” Thrombotic Low levels of C3, In the presence of AP
pathway and factors B, microangiopathies factor B, and AP dysregulation and TMA
I, H, anti- (TMA), C3 activity indicate or C3 glomerulopathy,
factor H IgG glomerulopathy dysregulation, genetic causes must be
activation, and investigated in
consumption of AP specialized laboratories
Anti-FH IgG positivity (see below)
causes acquired
dysregulation of AP
Proliferative C3 nephritic C3 glomerulopathy Low C3 and C3NeF These tests are available
glomerulonephritis factor, anti- positivity are frequently in specialized
autoantibodies factor B IgG, found in C3 laboratories only
anti-factor H glomerulopathy,
IgG particularly in DDD
Complement C3a, C3d, Differentiation Elevated levels indicate
activation products C5b-9 between complement ongoing complement
activation and activation
complement Low levels suggest
deficiency, therapy complement factor
monitoring deficiency
Low levels of C5b-9
imply therapeutic
efficacy of eculizumab
(continued)
30 Complement-Mediated Glomerular Injury in Children 941

Table 4 (continued)
Interpretation of the
Package Tests Indication results Comments
Genetic analysis of Bidirectional Thrombotic Indicated for all patients
complement sequencing of microangiopathies with atypical HUS
components and CFH, CFI, (TMA), C3 May be indicated for
AP regulators CD46, C3, glomerulopathy patients with C3
CFB, and glomerulopathy
THBD
Copy number
determination
of CFH-
related genes

such immunoglobulins may accumulate in circulating C3 levels are usually significantly


specific regions of the kidney. The local- and persistently reduced, as in C3
ization of these complement-activating sub- glomerulopathies (111). When the AP
stances (subendothelial vs. subepithelial) dysregulation involves membrane-bound com-
and of the resulting inflammatory reaction plement, circulating C3 may or may not be
(reflecting the extent of complement activation reduced, as happens in atypical hemolytic ure-
and cell proliferation) are decisive determi- mic syndrome (aHUS). Recent data show that
nants of the histological and clinical conse- in patients with aHUS, circulating C3 levels
quences. Overactivation of the CP in these are low in only 30–50 % of patients, excepting
forms of glomerulonephritis is accompanied those with C3 or CFB mutations who mostly
by hypocomplementemia. The characteristic (70–100 %) present low C3 [25].
alterations of the serum complement profile
include decreased C3, C4, CH50, and C1q Table 4 summarizes the complement tests cur-
levels. Typically, the complement regulators rently offered by specialized diagnostic laborato-
factor H and I are not affected (Table 4). ries, complete with comments for the
2. The impaired function of complement regula- interpretation of their findings.
tory proteins, leading to dysregulation of the The next two sections of this chapter outline
alternative pathway. As complement con- two aspects of complement-mediated glomerular
sumption affects just the one pathway, or a injury. The first section describes the molecular
few of its components controlled by the miss- mechanisms of damage to renal cells and function,
ing regulator, alterations of the serum comple- whereas the last one provides a brief clinical over-
ment profile are less marked and, if present, view of complement-mediated glomerular
indicate overactivation and consumption of the diseases.
alternative pathway – i.e., low C3 level. Defi-
ciencies of factor H or factor I are often accom-
panied by low factor B level (Table 4). It is
important to note that in this group of renal Molecular Mechanisms
diseases, circulating C3 levels may or may of Complement-Mediated Renal Injury
not be reduced, partly depending on whether
the complement alternative pathway The glomerular filtration barrier can be damaged
dysregulation is in the fluid phase or on the after local complement activation or by the depo-
cell membranes. When the alternative pathway sition of systemically activated complement
dysregulation involves the fluid phase, products.
942 Z. Prohászka et al.

Role of Structural Properties of Kidney proteins in podocytes and their positive regulation
Components and of Local Production by interleukin-1 and tumor-necrosis factor alpha
of Complement Factors and Regulators have been demonstrated [30, 31]. The crucial
importance of locally produced C3 in ischemia-
The glomerular filtration barrier is a three- reperfusion-induced kidney damage has been
layered structure separating the bloodstream and shown in a complement-deficient animal model
the urinary space (Fig. 3) within the glomerulus, a [32]. In addition to components of the comple-
tight aggregate of capillaries surrounded by the ment pathways, complement control proteins are
interdigitated extensions of podocytes. Two cel- also expressed and produced by the various cell
lular layers – one epithelial (belonging to the types present in the kidney. As reviewed in detail
podocyte) and one endothelial – are separated by elsewhere [33], the expression of MCP and CD59
the glomerular basement membrane (GBM), a is ubiquitous in glomerular and tubular cells,
specialized form of extracellular matrix. The whereas that of DAF appears to be lower. In
structure of these layers together with high glo- vitro, the glomerular endothelium has a lower
merular blood flow makes this filtration barrier baseline expression of CD59 and DAF when com-
susceptible to complement-mediated injury. pared to other endothelial cell types, and this
The glomerular capillaries are lined by special- partially explains renal susceptibility to
ized endothelial cells bearing large fenestrations complement-induced damage [34]. Of note, the
that allow the passage of fluid across the endothe- anticoagulant protein thrombomodulin – an acti-
lium. These fenestrations most probably allow the vator of the factor I-mediated degradation of C3b
direct flow of plasma (hence, also of complement – is another important constituent of the endothe-
components and activated fragments, if present) lial glycocalyx in the glomerulus. Hence, the com-
from the blood, across the proximal layer of the position of the endothelial glycocalyx with
filtration barrier, toward the GBM. Recent find- binding sites for soluble complement regulators
ings suggest a major role for the glomerular endo- and sufficient expression of membrane-bound
thelium and its glycocalyx in the maintenance of complement control proteins (including
an intact barrier, with the latter forming a mesh- thrombomodulin) may play an important role in
like, negatively charged, hydrated structure protecting the glomerulus against complement
[26]. This negatively charged glycocalyx is attacks. The composition of the glycocalyx is
important in recruiting fluid-phase complement subject to inflammatory changes [35]. Therefore,
regulators and in maintaining effective control of it can be supposed that the inflammatory process,
complement at the filtration barrier [27]. The complement activation, and related changes of the
GBM is unusually thick and formed by specific glycocalyx are among the most important mecha-
isoforms of proteins including laminin, type IV nisms driving complement-mediated kidney
collagen, nidogen, and heparan sulfate proteogly- disease.
cans. Importantly, the GBM does not contain
complement regulatory proteins [28], and hence,
its surface is susceptible to opsonization by com- Cellular Activation and Proliferation
plement. Besides, covalently fixed complement in Response to Complement Activation:
components may persist longer, since the cofac- The Non-lytic Terminal Pathway
tors necessary for C3b cleavage are missing. Complement Complex
Different cell types of the kidney appear to be a
rich local source of complement proteins, as Following complement activation, the terminal
shown by various in vitro gene or protein expres- complement complex is formed. The assembly
sion studies [21]. Genes of C1q and factor D are of the terminal pathway complement complex
strongly expressed in the glomeruli, whereas C2, (TCC or C5b-9) in the plasma membrane takes
C4, C3, and factor B are detected in the renal place in different ways. It is important to distin-
tubules [29]. The production of C2 and C3 guish lytic TCC (C5b-9n complexes with C9
30 Complement-Mediated Glomerular Injury in Children 943

Protective mechanisms Lytic terminal complement complex


Non-lytic
C5b-9n
against complement attack: terminal complement complex
Shedding or endocytosis of C5b-678
terminal complement complex
Mammalian cell surface
C9

Massive complement activation Moderate complement activation


High amount of lytic complex Low amount of lytic complex and non-lytic complex

++
Loss of ATP, ADP, AMP, increase of icCa Signal transduction
Loss of mitochondrial inner membrane potential Activation of Ras, PI3K and NFkB

Cell proliferation Increased expression


Cell death Transcription of cell cycle genes; of cell surface
Anti-apoptosis; Protein synthesis adhesion molecules

Facilitation of inflammation
Release of biologically active mediators,
growth factors and cytokines
Autocrine and paracrine effects

Fig. 4 Possible consequences of complement activation mediators, cytokines, and growth factors may significantly
mediated by the terminal complement complex. contribute to the consequences of complement attack.
Depending on the amount and composition of the terminal Nucleated cells possess effective defense mechanisms,
complement complex and on cell type, complement acti- including endocytosis and the shedding of membrane ves-
vation may result in cell death or proliferation. Moreover, icles, to protect themselves against complement attack
the autocrine and paracrine effects of biologically active

present in various concentrations), which forms activation in various types of nucleated cells and
pores within the cell membrane leading to cell in the proliferation of smooth muscle cells. More-
death, and non-lytic TCC (C5b-678 complexes over, C5b-9-induced growth factor release (such
without polymerization of C9), which triggers as the induction of platelet-derived growth factor
multiple signal transduction pathways that may and basic fibroblast growth factor in endothelial
induce cell activation, as well as upregulate cells) may also contribute to cell cycle activation
metabolism and proliferation [36, 37] (Fig. 4). [39]. Through its stimulatory action on nuclear
The non-lytic complex has been shown to activate factor kB, C5b-9 is also able to induce the secre-
the small G protein Ras, phosphatidylinositol tion of pro-inflammatory cytokines including IL-6
3-kinase, and also the ERK1, JNK1, p38, and in smooth muscle cells [40], as well as IL-8 and
MAPK pathways. MCP-1 in endothelial cells [41]. Further, it can
Depending on the composition of the TCC and promote surface adhesion molecule expression
on the cell type studied, these activities may con- such as neutrophil-endothelial adhesion via the
tribute to the cellular activation and to the initia- induction of osteoprotegerin [42] and intercellular
tion of cytoprotective mechanisms or lead to cell adhesion molecules (ICAMs), as shown in a por-
death. The non-lytic terminal pathway complex cine arterial endothelial cell model [43]. These
may, for example, induce tissue factor expression responses may be important for cell survival dur-
in macrophages after treatment with anti- ing inflammation accompanied by significant acti-
thymocyte globulin [38]. C5b-9 in sublytic con- vation of the terminal complement pathway, such
centration has been implicated in the cell cycle as in glomerular diseases with complement
944 Z. Prohászka et al.

activation. In addition to its endothelial and epi- number of C5b-9 complexes are efficiently elim-
thelial effects, the deposition of the C5b-9 com- inated from the plasma membrane through the
plex may play a dual role in mesangial and in formation of microvesicles. Therefore, a large
tubular cells. In particular, TCC may promote quantity of these complexes (i.e., intense or
tissue fibrosis via its stimulatory effect on uncontrolled complement activation) is
TGF-beta1 and collagen expression [44, 45] on required to inflict cell death via the terminal
one hand. On the other hand, it can also induce pathway, through a process best described as the
apoptosis of these cells [46, 47]. “multiple-hit” mechanism [52] (Fig. 4). This pro-
Parallel to the generation of C5b, equimolar cess is characterized by the loss of ATP,
quantities of the small anaphylatoxin C5a are ADP, and AMP and of the mitochondrial
also produced. Acting on its G-protein-coupled membrane potential before cell lysis by the
receptor, C5a elicits a multitude of biological C5b-9 complex occurs [53]. The rapid and
actions and changes that are essential in innate considerable intracellular calcium influx,
and adaptive immunity. These include an increase mediated by the C5b-9 channels, contributes to
of vascular permeability, the chemotaxis of the quick loss of inner mitochondrial
inflammatory cells (neutrophils), the induction of membrane potential, which is followed by acute
the respiratory burst, the release of cytokines and cell death. This process has several features
chemokines, and phagocytosis [48, 49]. C5a has resembling those of apoptosis, such as TUNEL
been shown to act on the expression of adhesion positivity and DNA fragmentation. Nevertheless,
molecules both in neutrophils and in endothelial the cell destruction initiated by the attack of the
cells, thereby enhancing their binding to each terminal complement pathway is primarily
other [50]. It is important to note that these necrotic in nature [54].
C5a-initiated responses are rather similar to In nucleated cells, several regulatory mecha-
those triggered by the terminal complement com- nisms exist to overcome the effects of a limited
plex and may significantly contribute to glomeru- complement attack. The two most important
lar damage through induction of cellular defense mechanisms are membrane shedding to
proliferation, fibrosis, and scarring in case of com- the extracellular space through the formation of
plement activation. Therefore, significant atten- microvesicles and endocytosis (Fig. 4). The time
tion has been paid to the pharmacological of removal of terminal complement complexes
targeting of the C5a receptor. In recent years, a shortens with the incorporation of C8 and multiple
variety of different compounds have been tested copies of C9 into the complex: C5b-8,9n has a
in clinical trials of patients with kidney half-life of 1–3 min only. By contrast, complexes
diseases [51]. without C8 and C9 persist in the membrane of
red blood cells for several hours [55]. The elimi-
nation rate of TC complexes also increases with
Cell Death and Recovery in Response the elevation of intracellular calcium level, lead-
to Complement Injury: The Lytic ing to activation of the PKC pathway which in
Terminal Pathway Complement turn induces membrane vesiculation and endocy-
Complex tosis. In case of a complement attack on
podocytes, the C5b-9 complex-containing mem-
A single terminal complement complex (with brane vesicles that are released into the extracel-
multiple copies of incorporated C9 proteins) lular space can be detected in the urine. Therefore,
forms the TCC lytic complex, which is able to measuring urinary C5b-9 levels may be useful for
lyse red blood cells in vitro; however, nucleated the assessment of complement activation in the
cells possess multiple protective mechanisms kidney, as has been shown recently in
against C5b-9n-mediated destruction. A limited preeclampsia [56].
30 Complement-Mediated Glomerular Injury in Children 945

Illustrative Examples complement-activating capacity. Its ability to


aggregate and the quantity of the deposits are
An Overview of the Mechanisms of additional influencing factors.
Complement-Mediated Glomerular As a variety of etiological factors may con-
Injury verge to similar final pathways capable of causing
glomerular damage, the clinical and histological
The complement system plays a key role in a appearance of the latter may be similar. The rec-
number of glomerular pathologies. The nature ognition of the underlying mechanisms may help
and the extent of complement-mediated glomeru- to understand the similarities and differences
lar damage will depend on the trigger and on the between the specific forms of nephropathy –
pathway of complement activation, as well as on such as the one occurring in systemic lupus
its localization to the subendothelial, mesangial, erythematosus – that may mimic other character-
or subepithelial space (Fig. 3). istic histological patterns, including membrano-
Soluble immune complexes formed in the cir- proliferative glomerulonephritis or membranous
culation may be trapped in subendothelial and nephropathy.
mesangial areas of the glomerulus. Glomerular Finally, complement activation may occur also
immune deposits may be formed by free circulat- in some nonimmune-complex-mediated renal dis-
ing antibodies binding to antigens trapped at eases that are not primary glomerular disorders.
subendothelial or mesangial sites or underneath Uncontrolled complement activation may either
the podocytes in the subepithelial space. Finally, lead to some forms of C3 glomerulopathies or to
autoantibodies may bind to normal glomerular renal diseases that are not primary glomerular
components like the Goodpasture’s glomerular diseases including among others hemolytic ure-
basement membrane antigen, as well as to mic syndrome, thrombotic thrombocytopenic pur-
podocyte antigens, such as the phospholipase A2 pura, and renal vasculitis.
receptor and neutral endopeptidase.
In immune-complex glomerulonephritis, the
structural properties of the immune complexes or The Clinical Presentation of Glomerular
of the trapped antigens will determine their final Injury
location within the glomerular structure and also
the biopsy findings and the clinical consequences. In the following section, we shall focus on the role
(a) Subendothelial distribution of the immune of the complement system in glomerular injury
complexes and activation of the complement cas- through some typical examples. For brevity, we
cade lead to massive endothelial damage and exu- have omitted ischemia-reperfusion injury, a model
dative infiltration by inflammatory cells. of complement-mediated injury that plays a central
(b) Immune-complex deposition in the role in acute kidney injury (reviewed in [59]) and in
mesangium induces mesangial cell proliferation renal graft rejection (reviewed in [60]).
and matrix expansion. (c) If the immune com-
plexes are deposited in the subepithelial area, the Poststreptococcal Glomerulonephritis
complement-generated chemotactic factors are Poststreptococcal glomerulonephritis (PSGN) is
isolated from the capillary space, and thus, the characterized by an acute glomerular lesion with
resulting lesion is rather a noninflammatory reac- proliferation of endothelial, mesangial, and epi-
tion with podocyte injury, effacement, and heavy thelial cells, as well as the presence of an inflam-
proteinuria [57, 58]. matory exudate. C3 deposition occurs early in the
The clinical consequences will be influenced disease process, followed by IgG deposition. This
also by the biologic properties of the antibody pattern of immune-complex deposition is seen
(or antigen), its Fc receptor affinity, and also in other forms of postinfectious GN [61, 62].
946 Z. Prohászka et al.

The discovery of streptococcal pyrogenic exo- from onset of urinary symptoms, especially if
toxin B (SpeB) may explain the possible mecha- there is no increasing trend, this could
nism of complement activation and binding in indicate that the “PSGN” was in fact the initial
PSGN. SpeB is a small, cationic cysteine protease event of a more chronic process such as C3
with complement-activating and plasmin-binding glomerulopathy (see below). Therefore, referral
properties [62, 63]. The level of the antibody to for diagnostic evaluation is justified in such
SpeB correlates with disease activity in PSGN, a case.
and the antibody co-localizes with IgG and C3 in
subepithelial humps, the histological hallmark of Immunoglobulin A Nephropathy (IgAN)
PSGN [64]. The peculiarity of SpeB is that it may Immunoglobulin A nephropathy (IgAN) is the
induce inflammation in the kidney, manifested by most common form of glomerulonephritis in
cytokine production, the expression of adhesion both children and adults. The clinical course is
molecules, and leukocyte proliferation followed variable, with episodes of recurrent gross hema-
by immune-complex deposition. SpeB exhibits turia concomitant with infectious (most frequently
plasmin-binding properties that might cause pro- upper respiratory tract) episodes. Following
teolysis in the glomerular basement membrane. macrohematuria, microscopic hematuria and
This may facilitate the transit of dissociated mild-to-severe proteinuria may persist. In some
subendothelial immune complexes, which can instances, IgA nephropathy may present
then form subepithelial humps. Some with heavy proteinuria and full-blown nephritis
subendothelial deposits may be present in PSGN [67, 68]. The histopathologic lesion is variable,
possibly because the antibody to SpeB also with focal or diffuse mesangial hypercellularity
exhibits molecular mimicry with endothelial and an increase in the mesangial matrix in most
cells [58, 62, 65]. cases. Endocapillary lesions are rare, just like
Other antigens such as nephritis-associated crescentic glomerulonephritis. In patients with
streptococcal plasmin have also been studied progressive disease, tubular injury often results
intensively as possible etiological factors of in fibrosis [69, 70]. Besides the IgA and C3, less
PSGN. Its nephritogenicity may be related to its intense IgG and/or IgM deposits are seen in up to
plasmin-binding activity, which induces inflam- 50 % of biopsies.
matory reaction and breakdown of the glomerular IgA nephropathy is the consequence of a sys-
basement membrane (GBM). It co-localizes in the temic abnormality in IgA synthesis, resulting in
glomeruli with plasmin, but, in contrast to SpeB, incompletely glycosylated IgA1 molecules that
not with IgG or C3 [66]. lack the galactose residues at the antibody’s
hinge region. Aberrant IgA1 has a strong tendency
Complement Profile and Diagnostics to spontaneous aggregation, which unmasks MBL
During the initial 2 weeks of the disease, the C3 binding sites. This leads to complement activation
level is significantly reduced in about 90 % of and binding to other molecules, such as fibronec-
patients. In contrast, C4 and C2 levels are tin, IgG, and collagen IV. Further, the aberrant
usually normal or only slightly decreased. C3 exposure of the misglycosylated IgA1 hinge
and CH50 return to normal within 4–8 weeks region may induce the production of antiglycan
after onset. It is important to stress the IgG autoantibodies, facilitated by concurrent
need for measuring C3 level early in the course infections by viruses (e.g., the Epstein-Barr
of PSGN. After the first 2 weeks of the onset of virus) or bacteria (e.g., streptococci) that express
urinary abnormalities, it may be impossible to GalNAc-containing moieties.
diagnose PSGN with certainty without a The circulating macromolecular form of
kidney biopsy, which may be indicated if a underglycosylated IgA evades removal from the
decrease in renal function, nephrotic-range circulation by asialoglycoprotein and CD89
proteinuria, or severe hypertension is present. If receptors, and this facilitates its mesangial
low circulating C3 persists more than 4–8 weeks deposition [71].
30 Complement-Mediated Glomerular Injury in Children 947

Complement Profile and Diagnostics Systemic Lupus Erythematosus (SLE)


Neither soluble monomeric nor dimeric human Kidney involvement in pediatric SLE is frequent
IgA can activate complement directly, through and may be more severe than in adult-onset dis-
the classical pathway. In IgA nephropathy, ease [78, 79]. A number of different mechanisms
C5b-9 is generated by complement activation leading to kidney damage are active in SLE. This
induced by the interaction of aberrantly explains some of the clinical and histological fea-
glycosylated IgA1 aggregates with MBL. Alter- tures shared with other glomerulopathies, such as
natively, in situ formation of immune membranous nephropathy or membranoproli-
complexes with IgG antiglycan antibodies may ferative glomerulonephritis.
occur [18, 72, 73]. Defective T-cell regulation, the persistence of
Thus, two distinct immunohistological pat- self-antigens in apoptotic bodies, and the loss of
terns may be observed in IgA nephropathy. In tolerance to such antigens trigger B-cell hyperac-
the majority of cases (75 %), biopsy specimens tivity and lead to an overproduction of autoanti-
show the deposition of IgA1, C3, and the terminal bodies. Typically, IgG anti-double-stranded DNA
complement complex, with the absence of other antibodies (anti-DNA) are found in the serum and
immunoglobulins or complement fragments, indi- in glomerular deposits of patients with SLE. Some
cating membrane-bound activation of the alterna- of the monoclonal anti-DNA antibodies also
tive pathway. Another pattern, showing the exhibit cross-reactivity with capillary wall anti-
presence of IgA1 and C3 in association with gens, especially laminin and alpha-actinin. When
MBL and mannose-associated serine protease internalized by cells, these may directly alter cell
1 (MASP-1), is seen in about 25 % of the cases. functions leading to apoptosis. Mesangial
The presence of MBL and MASP-1 correlates deposits have also been associated with antibodies
with disease severity, increased proteinuria, to mesangial cell constituents like annexin, which
decreased GFR, and poor histological prognostic co-localizes with IgG and C3 and correlates with
features [69, 74, 75]. disease activity [80]. Deposited anti-DNA may
C3 and mannose-binding lectin are also syn- also react with extracellular DNA present in the
thesized locally, by mesangial cells and form of nucleosomes. These consist of an anionic
podocytes. Therefore, the binding of polymeric segment of DNA associated with a highly cationic
IgA to mesangial cells may induce local comple- histone core, resulting in a net positive charge
ment activation independent of the systemic com- and a high affinity for glomerular anionic sites
plement activity [76]. Mesangial cells also [81, 82].
synthesize complement regulatory proteins, The complement system is involved in glomer-
which may explain why the generation of C5b-9 ular injury at different levels in SLE.
does not usually result in mesangiolysis in IgA First, deficiencies in the early components of
nephropathy. the classical pathway (C1q, C1r, C1s, C2, or C3)
As complement activation in IgAN is and low copy numbers of C4 predispose to the
primarily local and membrane bound, most development of SLE. Inherited or acquired C1q
frequently circulating levels of C3, C4, and deficiency is particularly important due to func-
CH50 are normal. However, there is some tional consequences including the defective clear-
evidence of systemic activation of ance of apoptotic bodies and reduced
complement in a subpopulation of patients with phagocytosis. The cellular debris may persist lon-
IgAN. Elevated levels of the split products ger and contribute to the loss of self-tolerance and
of activated C3 have been reported, suggesting to the development of autoimmunity. The pres-
activation of the alternative pathway in the fluid ence of C1q autoantibodies suggests lupus
phase [77]. The precise relationship between nephritis with a sensitivity and specificity of
systemic markers of complement activation and 90 % [83, 84].
disease activity is currently incompletely Once the complement system is activated in
understood. SLE, the activation products contribute directly
948 Z. Prohászka et al.

to glomerular injury (Fig. 3, enlarged area, D). partly due to hypocomplementemia-related


Proliferative lupus nephritis is associated with immunodeficiency, although the relative contribu-
IgG, IgM, and IgA (“full house”), and C3 deposi- tion of hypocomplementemia is unclear [87].
tion localized primarily in the mesangium The measurement of complement fragment
(mesangial lupus nephritis, classes I and II) in deposition (such as C3d and C4d) on peripheral
mild disease. The deposits are visible along the blood cells (i.e., red blood cells, platelets, and
subendothelial aspect of the capillary wall, lymphocytes) may serve as a biomarker of lupus
accompanied by increasing proliferation and disease activity [88, 89]. An acquired reduction in
inflammation (focal or diffuse proliferative lupus the numbers of complement receptor one (CR1,
nephritis class III and IV). In membranous lupus also known as CD35 and the C3b receptor) on the
nephritis (class V), by contrast, the deposits are erythrocytes commonly occurs in immune-
subepithelial. These could result from the dissoci- complex-mediated syndromes. This is a possible
ation of subendothelial immune complexes mov- marker of disease activity and particularly of renal
ing across the glomerular basement membrane flares [90, 91].
and reforming in the subepithelial space. Alterna- In pediatric patients with SLE, an in-depth
tively, lupus autoantibodies specific for podocyte analysis of the levels of complement factors and
antigens may occur just as in idiopathic membra- of complement pathway activity may identify
nous nephropathy [84–86]. some patients with inherited complement defi-
Subendothelial complement activation is a ciencies (Table 4).
major mediator of tissue injury through both the
intracapillary generation of chemotactic factors Atypical Hemolytic Uremic Syndrome
and the formation of C5b-9. The intense inflam- The hemolytic uremic syndrome (HUS) belongs
matory reaction with leukocyte infiltration and the to the group of thrombotic microangiopathies
destruction of glomerular structures are due to the (TMAs), defined by the clinical triad hemolytic
simplicity of the interaction between the anemia, thrombocytopenia, and renal failure. The
anaphylatoxins generated by the complement cas- typical, diarrhea-associated form of HUS is
cade and the circulating leukocytes. In membra- caused by the exotoxins of certain bacteria, most
nous lupus nephritis, the basement membrane frequently enterohemorrhagic Escherichia coli.
isolates the capillary space from the subepithelial By contrast, the atypical forms of HUS (aHUS)
site of complement activation and thus prevents include HUS caused by defects in the regulation
the recruitment of inflammatory cells [86]. of the alternative complement pathway on vascu-
lar endothelial cells [92]. Recently, mutations of
Complement Profile and Diagnostics diacylglycerol kinase epsilon have been described
In SLE, complement activation through the clas- in patients aged less than a year, who developed
sical pathway leads to low C3 and C4, whereas aHUS with persistent hypertension, hematuria,
factor B of the alternative pathway is usually and proteinuria [93]. Drug-mediated or disease-
normal. Low C3 values correlate closely with associated forms, collectively classified as sec-
disease severity, especially with that of ondary HUS, have also been reported.
renal involvement, and with the levels of anti- In patients with aHUS, the characteristic clin-
bodies to double-stranded DNA. The normaliza- ical and laboratory features of TMA result from
tion of low C4 and C3 levels during treatment is a the dysregulation of the complement alternative
good prognostic sign: therefore, C4 and C3 are pathway on cell surfaces (membrane bound). Sev-
valuable biomarkers of disease activity in eral studies have reported that about 50–60 % of
SLE [84]. patients with aHUS carry mutations in the genes
Moreover, patients undergoing immunosup- of complement regulators (CFH, CFI, MCP, and
pressive therapy for SLE may be susceptible to thrombomodulin) or of alternative pathway com-
infection by encapsulated bacteria. This may be ponents (CFB or C3). Functional characterization
30 Complement-Mediated Glomerular Injury in Children 949

of the mutations revealed the dysregulation of In addition, such complement activation frag-
complement activation (decreased expression of ments directly enhance the expression of surface
regulators, loss of surface binding of regulators, adhesion molecules as well as reduce the expres-
decreased cofactor-mediated degradation of C3b, sion of vascular endothelial growth factors and
low decay-accelerating activity) or increased their receptors on the endothelial cells.
activity of C3 convertase (resistance to degrada- The group of non-Shiga toxin-mediated HUS
tion, increased enzymatic activity). In addition to comprises diseases with different etiologies, and
mutations, anti-factor H autoantibodies in aHUS complement activation in the fluid phase does not
accelerate factor H clearance through immune- necessarily accompany the mutations of comple-
complex formation, the diminished binding of ment genes. Therefore, plasma and serum markers
factor H to cell surfaces, and the reduced factor of complement activation (i.e., low levels of cir-
H-mediated breakdown of C3b [25, 92]. However, culating C3) are seen in only about 30–50 % of
such genetic or acquired abnormalities should be patients with acute aHUS, excepting those with
regarded only as risk factors for the disease. Addi- C3 or CFB mutations who mostly (70–100 %)
tional disease triggers, which activate comple- present low C3 [25]. However, screening of the
ment, are required for aHUS to develop, complement profile with genetic analysis is indi-
including – among others – infections or preg- cated for all patients with acute aHUS. The first
nancy. If complement activation is not properly step in acute cases of thrombotic microangiopathy
controlled on the surface of endothelial cells, it is to investigate ADAMTS13 metalloproteinase
may promote endothelial damage and microvas- activity and EHEC microbiology [95]. For all
cular thrombosis characterized by the formation acute TMA patients with non-deficient
of fibrin- and platelet-rich thrombi (Fig. 3, ADAMTS13 activity, especially if age is below
enlarged area B). 2 and without acute gastroenteritis, investigation
The histological hallmark of acute aHUS is of the complete complement profile and molecu-
capillary thrombosis with wall thickening, endo- lar genetic analysis are indicated. The former
thelial swelling, and the deposition of a floccular should include appraisal of the activity of the
material between the glomerular basement mem- classical and alternative pathways by measuring
brane and the endothelial cell lining. circulating levels of C3and C4; factors H, I, and
Mesangiolysis commonly occurs. Platelet- and B; and autoantibodies to factor H. The genetic
fibrin-rich thrombi and fibrinoid necrosis of the analysis should comprise screening for mutations,
arterioles may occlude the lumen of capillaries the determination of the risk haplotypes of factors
and small arterioles. Fragmentation of red blood H and MCP (Table 4, [96]). Low C3 levels may be
cells may be observed within the vessel wall. present in patients with mutations in the CFH,
Immunofluorescence studies reveal immunoglob- CFI, MCP, C3, and CFB; however, a normal com-
ulin M and complement deposits primarily in the plement profile does not necessarily exclude
sclerotic areas [94]. complement-mediated aHUS. The presence of
anti-factor H autoantibody indicates the autoim-
Complement Profile and Diagnostics mune form of aHUS, and the level of the autoan-
In aHUS, complement activation may contribute tibody is a valuable laboratory marker of
to endothelial injury and thrombosis through mul- therapeutic efficacy. The complement activation
tiple mechanisms. Active fragments of C5b-9 and marker of the terminal pathway C5b-9 can
C5a (see above), generated during complement be elevated in acute complement-mediated
activation, may directly contribute to the activa- aHUS, whereas its decrease (together with the
tion, dysfunction, and destruction of the endothe- deficient activity of CP and AP) is a sensitive
lium. Moreover, the same factors may promote the marker of the therapeutic inhibition of the termi-
activation of platelets and neutrophils, further nal pathway by the anti-C5 monoclonal antibody
enhancing local thrombosis and inflammation. eculizumab [97].
950 Z. Prohászka et al.

Membranoproliferative autoimmune diseases with the prolonged, prefer-


Glomerulonephritis and C3 ential deposition of immune complexes within the
Glomerulopathy glomeruli. This leads to C1q deposition and acti-
The term “membranoproliferative glomerulone- vation of the classical pathway, resulting in the
phritis” (MPGN) denotes a spectrum of glomeru- chemoattraction of leukocytes and direct cell
lar pathologies. These are characterized by injury caused by terminal complement com-
mesangial and endocapillary hypercellularity, plexes. The cytokines and proteases produced by
accompanied by the thickening of the basement leukocytes stimulate mesangial cell proliferation
membrane and the interposition of cellular ele- and matrix expansion. These modify the integrity
ments between the glomerular basement mem- of the filtration barrier and cause nephritic syn-
brane and the endothelial cell layer and by the drome with occasional nephrotic features
formation of a new basement membrane [98]. [100–103].
MPGN is frequently idiopathic in children. In In general, there are no differences in the clin-
adults, however, it is commonly associated with ical presentation of the various forms of MPGN.
chronic viral, bacterial, or parasitic infections There is an exception, however, in that C3
(most commonly HBV, HCV, and HIV), malig- glomerulopathies may be associated with retinal
nant hematological diseases (multiple myeloma, drusen formation and with partial
B-cell chronic lymphoid leukemia), and lipodystrophy [104].
cryoglobulinemia [98]. Presumably, the uncontrolled activation of the
The original morphological classification alternative complement pathway in C3
(MPGN type I, subendothelial; type II, GBM; glomerulopathies is most often caused by differ-
and type III, subepithelial depositions) was ent mechanisms that stabilize the fluid-phase C3
based on histology and on typical electron micros- convertase. A specific autoantibody called the C3
copy features. However, recent advances in the nephritic factor (C3NeF) that binds C3 convertase
understanding of the pathogenesis of MPGN have is found in many patients, as well as acquired or
confirmed that different underlying causes may inherited deficiencies of the alternative
lead to similar EM findings, and a single pathoge- pathway components or of the regulatory proteins
netic pathway may result in different EM charac- [105, 106]. A C3 mutation that renders activated
teristics. Thus, the reclassification of the subtypes C3 resistant to factor H-mediated degradation has
of the conditions designated as MPGN is based on also been described [107]. Recently, mutations in
the underlying pathogenesis. Histologically, the the CFHR5 gene have been shown to cause an
pattern of immunofluorescence is most closely endemic form of MPGN in Cyprus (CFHR5
correlated with the underlying cause of disease. nephropathy) [108, 109].
Accordingly, MPGN may present with predomi- Chronic, dysregulated fluid-phase C3 activa-
nant immunoglobulin immunofluorescence tion generates a variety of complement activation
resulting from chronic immune-complex deposi- products of the alternative pathway. These accu-
tion with activation of the classical complement mulate along the inner aspect of glomerular
pathway (“immune-complex-mediated” form) or basement membrane and may form the classic
with predominant, albeit not exclusive, C3 immu- dense deposits seen by electron microscopy
nofluorescence resulting from uncontrolled acti- (Fig. 3, enlarged area C). The accumulated pro-
vation of the alternative pathway in the fluid phase teins damage the structure and the integrity of the
(“complement alternative pathway-mediated” filtration barrier. This may cause heavy protein-
form) [99]. These latter forms are defined C3 uria and lead to nephrotic syndrome. Dense
glomerulopathies (Fig. 5). deposit disease may be associated with other dis-
Immune-complex MPGN is the result of orders mediated by the alternative pathway, such
chronic inflammation caused by persistent viral as retinal drusen formation and partial
or bacterial infection, antigenemia, or lipodystrophy [106].
30 Complement-Mediated Glomerular Injury in Children 951

Clinical diagnosis of glomerulonephritis

Evaluation of baseline complement profile


Two times if C3 is low, 4-6 weeks apart

Temporary low C3 Prolonged low level of C3

Suspected C3-glomerulopathy
1. Full evaluation of complement with autoantibodies
2. Biopsy, immunofluorescence, electron microscopy
3. Molecular genetic investigations

C3, C4 and CH50 decreased


C3, AP activity and Factor B decreased
Complement and immunoglobulin
C3NeF, anti-FH or anti-FB can be positive
positive staining on immunofluorescence
Dominant complement staining on immunofluorescence
C3NeF or anti-FH negative
C4 not decreased
Factor B not decreased

Immune-complex-mediated
Poststreptococcal GN Dense deposit disease C3-glomerulonephritis
glomerulonephritis

C3-glomerulopathies
Antibody- and complement-mediated glomerular damage
Complement-mediated glomerular damage

Fig. 5 Role of laboratory evaluation of complement in may require full evaluation of complement system includ-
the classification of glomerulonephritis. Determination ing serum complement profile with C3 nephritic factor and
of baseline complement profile (circulating C3, C4) is anti-factor H autoantibody and genetic investigation of
indicated for all patients with glomerulonephritis (GN). complement regulators. Kidney biopsy, when performed,
Repeat measurements help differentiate between transient will show in immune-complex-mediated GN immuno-
and prolonged decrease of C3. Patients with evidence of globulin and complement positivity by immunofluores-
streptococcal disease (or other infections) and temporary cence with signs of classical pathway activation, whereas
C3 decrease most probably have poststreptococcal in C3 glomerulopathies (DDD or C3-GN), it will show
(or postinfectious) GN. Patients with prolonged decrease predominant C3 staining on immunofluorescence, even
of C3 and persistent glomerulonephritis despite therapy though immunoglobulin may be positive

Complement Profile and Diagnostics level does not exclude complement-mediated


Persisting hypocomplementemia (low C3 level) is MPGN, as normal C3 concentration is not excep-
common in all types of MPGN. tional in C3 glomerulopathies [110, 111]. Remark-
In immune-complex-mediated MPGN, com- ably, poststreptococcal glomerulonephritis is also
plement activation occurs via the classic pathway. characterized by this pattern, which can produce a
Typically, this is manifested by decreased (some- similar clinical syndrome. However, in C3
times mildly) serum C3, C1q, and C4 concentra- glomerulopathies, the decrease in C3 is usually
tion [108, 109]. persistent, whereas it is transient in poststrep-
In complement-mediated MPGN, i.e., C3 tococcal glomerulonephritis, resolving most fre-
glomerulopathy (including cases with C3NeF quently in about 8 weeks from initial presentation.
positivity), serum C3 levels are usually low
(often extremely low) due to dysregulation of the Membranous Nephropathy
alternative pathway, but C4 levels are normal. It Membranous nephropathy is characterized by
should be noted, however, that normal serum C3 heavy proteinuria, followed by the onset of
952 Z. Prohászka et al.

full-blown nephrotic syndrome. It is caused by the are usually present in the deposits. Another pos-
subepithelial accumulation of immune deposits, sibility could be that the IgG4 antibodies bound to
which triggers local complement activation. podocytes activate the lectin pathway and induce
Recently, phospholipase A2 receptor antibodies sublytic podocyte injury [117].
have been shown as the underlying cause in a
significant proportion of patients with idiopathic Complement Profile and Diagnostics
membranous nephropathy [112]. As heavy proteinuria in membranous nephropathy
In children, secondary membranous nephropa- may persist long after the immune response has
thy may occur more frequently than in adults. It is been inhibited and immune deposits do not form
caused by a variety of chronic infections, inflam- any longer, there is a need for validated bio-
mation, or antigenemia (HBV, HCV, syphilis, markers to monitor disease activity.
malaria, sickle-cell anemia, D-penicillamine treat- The glomerular deposition of C3 and C5b-9
ment, etc.) [113]. Lately, cationic bovine serum has been shown to correlate with the immune
albumin that may be absorbed in an undigested or deposit formation in progress. However, the direct
partially digested form from the gastrointestinal measurement of anti-PLA2R antibody, which
tract has been described as an environmental trig- reflects disease activity and the therapeutic
ger of early-childhood membranous nephropathy response, has now replaced their determination
[114, 115]. in clinical practice [118, 119].
As the basement membrane isolates the capil-
lary lumen from the subepithelial space, the acti-
vation products of the complement cascade Antibody-Mediated Allograft Rejection
cannot interact with circulating inflammatory (AMR)
cells. This in part explains the lack of any prolif-
erative reaction and the relatively bland urine Antibody-mediated rejection (AMR) of the renal
sediment seen in membranous nephropathy. graft is due largely to the presence of circulating
Podocyte injury occurring in membranous antibodies directed against the human leukocyte
nephropathy is entirely complement dependent antigens (HLA) of the donor kidney (donor-
[36]. In animal models, the inhibition of terminal specific antibodies, DSA).
complement complex formation prevents DSA-induced damage to the graft endothelium
podocyte injury and proteinuria but does not influ- and microvasculature depends on the magnitude
ence antibody deposition [112, 116]. of the injury and the repair capacity of the endo-
The terminal complement complex resulting thelial cells (EC). EC injury ranges from cell
from terminal pathway activation binds the necrosis to adaptive changes allowing the EC to
podocyte cell membrane. However, in contrast survive but with altered morphology and function
with erythrocytes and similar to endothelial cells, eventually leading to the occlusion of the
the podocyte has the capacity to shed the terminal microvasculature [74].
complement complex from its cell membrane and In early acute AMR, massive exposure of the
excrete it into the urine. Podocyte apoptosis may EC to DSA and the ensuing complement activa-
result from direct injury following insertion of the tion predominate with the formation of numerous
terminal complement complex. Alternatively, it terminal complement complexes (Fig. 3, enlarged
may be secondary to the toxic effects of locally area A). The EC can eliminate terminal comple-
produced cytokines and reactive oxygen radicals ment complexes. However, if the number of ter-
(Fig. 3, enlarged area D). minal complement complexes exceeds their
It is of interest that the majority of phospholi- eliminating capacity, the ECs may be destroyed.
pase A2 receptor antibodies belong to the IgG4 This leads to the exposure of the underlying
subclass with a poor complement fixing capacity. matrix and to the activation of the coagulation
A possible explanation for complement activation cascade and of the platelets, resulting subse-
could be that small quantities of IgG1 and IgG3 quently in mesangiolysis in the glomeruli and in
30 Complement-Mediated Glomerular Injury in Children 953

the formation of microthrombi throughout the intensity of C4d staining may vary significantly
microvasculature. The production of complement- if multiple biopsies are taken from the same
derived chemotactic factors induces the recruitment patient. This may reflect the concomitant activity
and adhesion of inflammatory cells, most com- of the processes of injury and repair [74, 124].
monly neutrophils, within the capillary lumina.
Histological features include swelling and necrosis
of the EC, denudation of the underlying matrix, Tubulointerstitial Injury in Progressive
platelet aggregation, thrombotic microangiopathy, Kidney Disease: The Role of
and neutrophil infiltration [120]. Complement
Sublytic injury, as detailed above, represents a
fragile equilibrium between damage and repair. As reviewed in Noris M et al. [21], complement
The cycles of cell injury and repair are character- components also appear to contribute to the
ized by EC swelling, loss of fenestrations, and the tubulointerstitial damage which is seen in all
expression of growth and mitogenic factors. All forms of progressive kidney disease, regardless
these result in proliferative changes manifested as of the initial pathogenetic mechanism. In
mesangial matrix expansion, duplication of the nonselective glomerular proteinuria, various pro-
glomerular basement membrane, and multilayering teins of the complement system appear in the
of the basal lumina of peritubular capillaries (trans- urine, leading to intratubular deposition of C3
plant glomerulopathy and capillaropathy) [121]. and of C5b-9. Their activation has been proposed
Throughout the course of AMR, lytic and sublytic to contribute to tubulointerstitial damage via
EC injury coexist and provide the basis for the cytotoxic, pro-inflammatory, and fibrogenic
morphologic and clinical heterogeneity of AMR. effects [125].

Complement Profile and Diagnostics


Activation of the classical pathway plays a pre- Therapeutic Inhibition of Complement
dominant role in AMR. The deposition of DSA in Glomerular Diseases
within the transplanted kidney leads to C1q bind-
ing. The presence and extent of peritubular C4d The improved understanding of the role of com-
staining is of diagnostic and prognostic impor- plement in a number of glomerular diseases stim-
tance. It should be noted, however, that ulated the development and clinical application of
C4d-negative AMR has also been reported various compounds targeting C3 or C5 or acting at
recently. This is more common in patients with different levels of complement activation
late-onset, chronic AMR and in AMR due to (Table 5) [128]. The availability of multiple drug
non-HLA DSA [122, 123]. Interestingly, the candidates ensures the potential for rapid progress

Table 5 Therapeutic targeting of complement, selected groups of drugs, and examples


Currently licensed
indications in pediatric
Target Name, type of the drug(s) kidney diseases References, comments
C5 (terminal Eculizumab (humanized anti-C5 Atypical hemolytic uremic [97, 126, 127]
complement monoclonal antibody) syndrome
pathway)
C3 Multiple compounds (recombinant None Preclinical and clinical
constructs, antibodies, minibodies, studies are in progress
etc.) [128]
C5a receptor Multiple compounds (peptidic, None Preclinical and clinical
(CD88) non-peptidic, small molecule receptor studies are in progress
inhibitors) [51]
954 Z. Prohászka et al.

in this field with clinical applicability in the next lectin-associated serine proteases leads to generation
few years. of a fibrin clot. Immunology. 2010;129(4):482–95.
7. Hamad OA, Back J, Nilsson PH, Nilsson B, Ekdahl
Eculizumab, a humanized monoclonal anti- KN. Platelets, complement, and contact activation:
body inhibiting the terminal complement pathway partners in inflammation and thrombosis. Adv Exp
at the level of C5, has revolutionized the treatment Med Biol. 2012;946:185–205.
of aHUS [97]. However, this agent has not proved 8. Zhang X, Kimura Y, Fang C, Zhou L, Sfyroera G,
Lambris JD, et al. Regulation of toll-like receptor-
similarly effective in all cases of C3 mediated inflammatory response by complement
glomerulopathy [129]. This heterogeneous group in vivo. Blood. 2007;110(1):228–36.
of complement-mediated glomerular diseases 9. Harris CL, Heurich M, Rodriguez de Cordoba S,
may benefit from complement inhibitors acting Morgan BP. The complotype: dictating risk for
inflammation and infection. Trends Immunol.
at the level of C3 [130]. The group of compounds 2012;33(10):513–21.
(peptidic, non-peptidic, and natural inhibitors) 10. Carroll MV, Sim RB. Complement in health and
that target C5a receptor are promising anti- disease. Adv Drug Deliv Rev. 2011;63(12):965–75.
inflammatory drugs that have the advantage of 11. Roumenina LT, Loirat C, Dragon-Durey MA,
Halbwachs-Mecarelli L, Sautes-Fridman C,
leaving the cascade intact for host defense Fremeaux-Bacchi V. Alternative complement path-
[51]. Improved understanding of the role of com- way assessment in patients with atypical HUS.
plement in the pathogenesis of glomerular dis- J Immunol Methods. 2011;365(1–2):8–26.
eases together with the improvement of 12. Forneris F, Burnley BT, Gros P. Ensemble refinement
shows conformational flexibility in crystal structures
complement diagnostics and therapy is of pivotal of human complement factor D. Acta Crystallogr D
importance in allowing better control of these Biol Crystallogr. 2014;70(Pt 3):733–43.
severe diseases in the near future. 13. Kemper C, Atkinson JP, Hourcade DE. Properdin:
emerging roles of a pattern-recognition molecule.
Acknowledgments The authors are grateful to Lilan Annu Rev Immunol. 2010;28:131–55.
Varga and Eniko Barnucz for their helpful suggestions. 14. Kang YH, Tan LA, Carroll MV, Gentle ME, Sim
Research in the author’s laboratories was supported by RB. Target pattern recognition by complement
the following grants: Hungarian Scientific Research Fund proteins of the classical and alternative pathways.
of Hungary OTKA 100909 to GSR, and OTKA 100687, Adv Exp Med Biol. 2009;653:117–28.
OTKA 110909 to ZP. 15. Kjaer TR, Thiel S, Andersen GR. Toward a structure-
based comprehension of the lectin pathway of com-
plement. Mol Immunol. 2013;56(4):413–22.
16. Megyeri M, Harmat V, Major B, Vegh A, Balczer J,
References Heja D, et al. Quantitative characterization of the
activation steps of mannan-binding lectin (MBL)-
1. Forneris F, Ricklin D, Wu J, Tzekou A, Wallace RS, associated serine proteases (MASPs) points to the
Lambris JD, et al. Structures of C3b in complex with central role of MASP-1 in the initiation of the com-
factors B and D give insight into complement plement lectin pathway. J Biol Chem. 2013;288
convertase formation. Science. 2010;330 (13):8922–34.
(6012):1816–20. 17. Megyeri M, Mako V, Beinrohr L, Doleschall Z,
2. Leslie M. Immunology. The new view of comple- Prohaszka Z, Cervenak L, et al. Complement protease
ment. Science. 2012;337(6098):1034–7. MASP-1 activates human endothelial cells: PAR4
3. Ricklin D, Lambris JD. Complement in immune and activation is a link between complement and endothe-
inflammatory disorders: pathophysiological mecha- lial function. J Immunol. 2009;183(5):3409–16.
nisms. J Immunol. 2013;190(8):3831–8. 18. Roos A, Bouwman LH, van Gijlswijk-Janssen DJ,
4. Oikonomopoulou K, Ricklin D, Ward PA, Lambris Faber-Krol MC, Stahl GL, Daha MR. Human IgA
JD. Interactions between coagulation and comple- activates the complement system via the mannan-
ment – their role in inflammation. Semin binding lectin pathway. J Immunol. 2001;167
Immunopathol. 2012;34(1):151–65. (5):2861–8.
5. Endo Y, Nakazawa N, Iwaki D, Takahashi M, 19. Kuraya M, Ming Z, Liu X, Matsushita M, Fujita
Matsushita M, Fujita T. Interactions of ficolin and T. Specific binding of L-ficolin and H-ficolin to apo-
mannose-binding lectin with fibrinogen/fibrin aug- ptotic cells leads to complement activation.
ment the lectin complement pathway. J Innate Immunobiology. 2005;209(9):689–97.
Immun. 2010;2(1):33–42. 20. Garlatti V, Martin L, Gout E, Reiser JB, Fujita T,
6. Gulla KC, Gupta K, Krarup A, Gal P, Schwaeble WJ, Arlaud GJ, et al. Structural basis for innate immune
Sim RB, et al. Activation of mannan-binding sensing by M-ficolin and its control by a
30 Complement-Mediated Glomerular Injury in Children 955

pH-dependent conformational switch. J Biol Chem. 37. Tegla CA, Cudrici C, Patel S, Trippe 3rd R, Rus V,
2007;282(49):35814–20. Niculescu F, et al. Membrane attack by complement:
21. Noris M, Remuzzi G. Overview of complement acti- the assembly and biology of terminal complement
vation and regulation. Semin Nephrol. 2013;33 complexes. Immunol Res. 2011;51(1):45–60.
(6):479–92. 38. Langer F, Spath B, Fischer C, Stolz M, Ayuk FA,
22. Nilsson SC, Sim RB, Lea SM, Fremeaux-Bacchi V, Kroger N, et al. Rapid activation of monocyte tissue
Blom AM. Complement factor I in health and disease. factor by antithymocyte globulin is dependent on
Mol Immunol. 2011;48(14):1611–20. complement and protein disulfide isomerase. Blood.
23. Jozsi M, Meri S. Factor H-related proteins. Methods 2013;121(12):2324–35.
Mol Biol. 2014;1100:225–36. 39. Benzaquen LR, Nicholson-Weller A, Halperin
24. Albrecht EA, Chinnaiyan AM, Varambally S, Kumar- JA. Terminal complement proteins C5b-9 release
Sinha C, Barrette TR, Sarma JV, et al. C5a-induced basic fibroblast growth factor and platelet-derived
gene expression in human umbilical vein endothelial growth factor from endothelial cells. J Exp Med.
cells. Am J Pathol. 2004;164(3):849–59. 1994;179(3):985–92.
25. Noris M, Caprioli J, Bresin E, Mossali C, Pianetti G, 40. Viedt C, Hansch GM, Brandes RP, Kubler W, Kreuzer
Gamba S, et al. Relative role of genetic complement J. The terminal complement complex C5b-9 stimu-
abnormalities in sporadic and familial aHUS and their lates interleukin-6 production in human smooth mus-
impact on clinical phenotype. Clin J Am Soc Nephrol. cle cells through activation of transcription factors
2010;5(10):1844–59. NF-kappa B and AP-1. FASEB J. 2000;14
26. Rostgaard J, Qvortrup K. Sieve plugs in fenestrae of (15):2370–2.
glomerular capillaries – site of the filtration barrier? 41. Kilgore KS, Schmid E, Shanley TP, Flory CM,
Cells Tissues Organs. 2002;170(2–3):132–8. Maheswari V, Tramontini NL, et al. Sublytic concen-
27. Morgan HP, Schmidt CQ, Guariento M, Blaum BS, trations of the membrane attack complex of comple-
Gillespie D, Herbert AP, et al. Structural basis for ment induce endothelial interleukin-8 and monocyte
engagement by complement factor H of C3b on a chemoattractant protein-1 through nuclear factor-
self surface. Nat Struct Mol Biol. 2011;18(4):463–70. kappa B activation. Am J Pathol. 1997;150
28. Pierchala BA, Munoz MR, Tsui CC. Proteomic anal- (6):2019–31.
ysis of the slit diaphragm complex: CLIC5 is a protein 42. Corallini F, Bossi F, Gonelli A, Tripodo C,
critical for podocyte morphology and function. Castellino G, Mollnes TE, et al. The soluble terminal
Kidney Int. 2010;78(9):868–82. complement complex (SC5b-9) up-regulates
29. Song D, Zhou W, Sheerin SH, Sacks osteoprotegerin expression and release by endothelial
SH. Compartmental localization of complement com- cells: implications in rheumatoid arthritis. Rheuma-
ponent transcripts in the normal human kidney. Neph- tology (Oxford). 2009;48(3):293–8.
ron. 1998;78(1):15–22. 43. Wood SC, Bushar G, Tesfamariam B. Inhibition of
30. Sheerin NS, Zhou W, Adler S, Sacks SH. TNF-alpha mammalian target of rapamycin modulates expres-
regulation of C3 gene expression and protein biosyn- sion of adhesion molecules in endothelial cells.
thesis in rat glomerular endothelial cells. Kidney Int. Toxicol Lett. 2006;165(3):242–9.
1997;51(3):703–10. 44. Gao L, Qiu W, Wang Y, Xu W, Xu J, Tong J. Sublytic
31. Falus A, Beuscher HU, Auerbach HS, Colten complement C5b-9 complexes induce
HR. Constitutive and IL 1-regulated murine comple- thrombospondin-1 production in rat glomerular
ment gene expression is strain and tissue specific. mesangial cells via PI3-k/Akt: association with acti-
J Immunol. 1987;138(3):856–60. vation of latent transforming growth factor-beta1.
32. Farrar CA, Zhou W, Lin T, Sacks SH. Local extravas- Clin Exp Immunol. 2006;144(2):326–34.
cular pool of C3 is a determinant of postischemic 45. Abe K, Li K, Sacks SH, Sheerin NS. The membrane
acute renal failure. FASEB J. 2006;20(2):217–26. attack complex, C5b-9, up regulates collagen gene
33. Nangaku M. Complement regulatory proteins in expression in renal tubular epithelial cells. Clin Exp
glomerular diseases. Kidney Int. 1998;54 Immunol. 2004;136(1):60–6.
(5):1419–28. 46. Chen Y, Zhang J, Guo G, Ruan Z, Jiang M, Wu S,
34. Richards A. Glomerular endothelial microvascular et al. Induced B7-H1 expression on human renal
heterogeneity and response to cytokines predispose tubular epithelial cells by the sublytic terminal com-
to the development of atypical HUS. Mol Immunol. plement complex C5b-9. Mol Immunol. 2009;46
2011;48(14):1732. (3):375–83.
35. Boels MG, Lee DH, van den Berg BM, Dane MJ, van 47. Nauta AJ, Daha MR, Tijsma O, van de Water B,
der Vlag J, Rabelink TJ. The endothelial glycocalyx Tedesco F, Roos A. The membrane attack complex
as a potential modifier of the hemolytic uremic syn- of complement induces caspase activation and apo-
drome. Eur J Intern Med. 2013;24(6):503–9. ptosis. Eur J Immunol. 2002;32(3):783–92.
36. Takano T, Elimam H, Cybulsky AV. Complement- 48. Manthey HD, Woodruff TM, Taylor SM, Monk
mediated cellular injury. Semin Nephrol. 2013;33 PN. Complement component 5a (C5a). Int J Biochem
(6):586–601. Cell Biol. 2009;41(11):2114–17.
956 Z. Prohászka et al.

49. Guo RF, Ward PA. Role of C5a in inflammatory 65. Ahn SY, Ingulli E. Acute poststreptococcal glomeru-
responses. Annu Rev Immunol. 2005;23:821–52. lonephritis: an update. Curr Opin Pediatr. 2008;20
50. Foreman KE, Glovsky MM, Warner RL, Horvath SJ, (2):157–62.
Ward PA. Comparative effect of C3a and C5a on 66. Batsford SR, Mezzano S, Mihatsch M, Schiltz E,
adhesion molecule expression on neutrophils and Rodriguez-Iturbe B. Is the nephritogenic antigen in
endothelial cells. Inflammation. 1996;20(1):1–9. post-streptococcal glomerulonephritis pyrogenic exo-
51. Woodruff TM, Nandakumar KS, Tedesco toxin B (SPE B) or GAPDH? Kidney Int. 2005;68
F. Inhibiting the C5-C5a receptor axis. Mol Immunol. (3):1120–9.
2011;48(14):1631–42. 67. Hogg RJ. Idiopathic immunoglobulin A nephropathy
52. Koski CL, Ramm LE, Hammer CH, Mayer MM, Shin in children and adolescents. Pediatr Nephrol. 2010;25
ML. Cytolysis of nucleated cells by complement: cell (5):823–9.
death displays multi-hit characteristics. Proc Natl 68. Haas M, Rahman MH, Cohn RA, Fathallah-Shaykh S,
Acad Sci U S A. 1983;80(12):3816–20. Ansari A, Bartosh SM. IgA nephropathy in children
53. Papadimitriou JC, Ramm LE, Drachenberg CB, and adults: comparison of histologic features and
Trump BF, Shin ML. Quantitative analysis of clinical outcomes. Nephrol Dial Transplant. 2008;23
adenine nucleotides during the prelytic phase of cell (8):2537–45.
death mediated by C5b-9. J Immunol. 1991;147 69. Coppo R, Troyanov S, Bellur S, Cattran D, Cook HT,
(1):212–17. Feehally J, et al. Validation of the Oxford classifica-
54. Ziporen L, Donin N, Shmushkovich T, Gross A, tion of IgA nephropathy in cohorts with different
Fishelson Z. Programmed necrotic cell death induced presentations and treatments. Kidney Int. 2014;86
by complement involves a Bid-dependent pathway. (4):828–836.
J Immunol. 2009;182(1):515–21. 70. A multicenter study of IgA nephropathy in children.
55. Ramm LE, Whitlow MB, Koski CL, Shin ML, A report of the Southwest Pediatric Nephrology Study
Mayer MM. Elimination of complement channels Group. Kidney Int. 1982;22(6):643–52. http://www.
from the plasma membranes of U937, a nucleated ncbi.nlm.nih.gov/pubmed/?term=6761487
mammalian cell line: temperature dependence 71. Coppo R, Feehally J, Glassock RJ. IgA nephropathy
of the elimination rate. J Immunol. 1983;131 at two score and one. Kidney Int. 2010;77(3):181–6.
(3):1411–15. 72. Imai H, Chen A, Wyatt RJ, Rifai A. Lack of comple-
56. Burwick RM, Fichorova RN, Dawood HY, Yama- ment activation by human IgA immune complexes.
moto HS, Feinberg BB. Urinary excretion of C5b-9 Clin Exp Immunol. 1988;73(3):479–83.
in severe preeclampsia: tipping the balance of com- 73. Wyatt RJ, Kanayama Y, Julian BA, Negoro N,
plement activation in pregnancy. Hypertension. Sugimoto S, Hudson EC, et al. Complement activa-
2013;62(6):1040–5. tion in IgA nephropathy. Kidney Int. 1987;31
57. Hogan J, Mohan P, Appel GB. Diagnostic tests and (4):1019–23.
treatment options in glomerular disease: 2014 update. 74. McCaughan JA, O’Rourke DM, Courtney AE. The
Am J Kidney Dis. 2014;63(4):656–66. complement cascade in kidney disease: from sideline
58. Couser WG. Basic and translational concepts of to center stage. Am J Kidney Dis. 2013;62
immune-mediated glomerular diseases. J Am Soc (3):604–14.
Nephrol. 2012;23(3):381–99. 75. Roos A, Rastaldi MP, Calvaresi N, Oortwijn BD,
59. McCullough JW, Renner B, Thurman JM. The role of Schlagwein N, van Gijlswijk-Janssen DJ,
the complement system in acute kidney injury. Semin et al. Glomerular activation of the lectin pathway of
Nephrol. 2013;33(6):543–56. complement in IgA nephropathy is associated with
60. Cravedi P, Heeger PS. Complement as a multifaceted more severe renal disease. J Am Soc Nephrol.
modulator of kidney transplant injury. J Clin Invest. 2006;17(6):1724–34.
2014;124(6):2348–54. 76. Montinaro V, Gesualdo L, Ranieri E, Monno R,
61. Eison TM, Ault BH, Jones DP, Chesney RW, Wyatt Grandaliano G, Schena FP. Renal cortical comple-
RJ. Post-streptococcal acute glomerulonephritis in ment C3 gene expression in IgA nephropathy. J Am
children: clinical features and pathogenesis. Pediatr Soc Nephrol. 1997;8(3):415–25.
Nephrol. 2011;26(2):165–80. 77. Seelen MA, Roos A, Daha MR. Role of complement
62. Rodriguez-Iturbe B, Musser JM. The current state of in innate and autoimmunity. J Nephrol. 2005;18
poststreptococcal glomerulonephritis. J Am Soc (6):642–53.
Nephrol. 2008;19(10):1855–64. 78. Ruggiero B, Vivarelli M, Gianviti A, Benetti E,
63. Rodriguez-Iturbe B, Batsford S. Pathogenesis of Peruzzi L, Barbano G, et al. Lupus nephritis in chil-
poststreptococcal glomerulonephritis a century after dren and adolescents: results of the Italian Collabora-
Clemens von Pirquet. Kidney Int. 2007;71 tive Study. Nephrol Dial Transplant. 2013;28
(11):1094–104. (6):1487–96.
64. Singh GR. Glomerulonephritis and managing the 79. Hiraki LT, Benseler SM, Tyrrell PN, Hebert D,
risks of chronic renal disease. Pediatr Clin North Harvey E, Silverman ED. Clinical and laboratory
Am. 2009;56(6):1363–82. characteristics and long-term outcome of pediatric
30 Complement-Mediated Glomerular Injury in Children 957

systemic lupus erythematosus: a longitudinal study. in DGKE cause atypical hemolytic-uremic syndrome.
J Pediatr. 2008;152(4):550–6. Nat Genet. 2013;45(5):531–6.
80. Ortega LM, Schultz DR, Lenz O, Pardo V, Contreras 94. Fogo AB, Bruijn JA, Cohen AH, Colvin RB, Jennette
GN. Review: lupus nephritis: pathologic features, JCE. Fundamentals of renal pathology. New York:
epidemiology and a guide to therapeutic decisions. Springer; 2006.
Lupus. 2010;19(5):557–74. 95. Cataland SR, Wu HM. How I treat: the clinical dif-
81. O’Flynn J, Flierman R, van der Pol P, Rops A, ferentiation and initial treatment of adult patients with
Satchell SC, Mathieson PW, et al. Nucleosomes and atypical hemolytic uremic syndrome. Blood.
C1q bound to glomerular endothelial cells serve as 2014;123(16):2478–84.
targets for autoantibodies and determine complement 96. Kavanagh D, Goodship TH, Richards A. Atypical
activation. Mol Immunol. 2011;49(1–2):75–83. hemolytic uremic syndrome. Semin Nephrol.
82. Ng KP, Manson JJ, Rahman A, Isenberg 2013;33(6):508–30.
DA. Association of antinucleosome antibodies with 97. Zuber J, Fakhouri F, Roumenina LT, Loirat C,
disease flare in serologically active clinically quies- Fremeaux-Bacchi V. Use of eculizumab for atypical
cent patients with systemic lupus erythematosus. haemolytic uraemic syndrome and C3
Arthritis Rheum. 2006;55(6):900–4. glomerulopathies. Nat Rev Nephrol. 2012;8
83. Crispin JC, Liossis SN, Kis-Toth K, Lieberman LA, (11):643–57.
Kyttaris VC, Juang YT, et al. Pathogenesis of human 98. Sethi S, Fervenza FC. Membranoproliferative glo-
systemic lupus erythematosus: recent advances. merulonephritis – a new look at an old entity. N
Trends Mol Med. 2010;16(2):47–57. Engl J Med. 2012;366(12):1119–31.
84. Chen M, Daha MR, Kallenberg CG. The complement 99. Sethi S, Nester CM, Smith
system in systemic autoimmune disease. J RJ. Membranoproliferative glomerulonephritis and
Autoimmun. 2010;34(3):J276–86. C3 glomerulopathy: resolving the confusion. Kidney
85. Markowitz GS, D’Agati VD. Classification of lupus Int. 2012;81(5):434–41.
nephritis. Curr Opin Nephrol Hypertens. 2009;18 100. Zand L, Fervenza FC, Nasr SH, Sethi
(3):220–5. S. Membranoproliferative glomerulonephritis associ-
86. Rahman A, Isenberg DA. Systemic lupus ated with autoimmune diseases. J Nephrol. 2014;27
erythematosus. N Engl J Med. 2008;358(9):929–39. (2):165–71.
87. Gilliland WR, Tsokos GC. Prophylactic use of anti- 101. Fakhouri F, Fremeaux-Bacchi V, Noel LH, Cook HT,
biotics and immunisations in patients with SLE. Ann Pickering MC. C3 glomerulopathy: a new classifica-
Rheum Dis. 2002;61(3):191–2. tion. Nat Rev Nephrol. 2010;6(8):494–9.
88. Kao AH, Navratil JS, Ruffing MJ, Liu CC, 102. Smith KD, Alpers CE. Pathogenic mechanisms in
Hawkins D, McKinnon KM, et al. Erythrocyte C3d membranoproliferative glomerulonephritis. Curr
and C4d for monitoring disease activity in systemic Opin Nephrol Hypertens. 2005;14(4):396–403.
lupus erythematosus. Arthritis Rheum. 2010;62 103. Sethi S, Fervenza FC. Membranoproliferative glo-
(3):837–44. merulonephritis: pathogenetic heterogeneity and pro-
89. Manzi S, Navratil JS, Ruffing MJ, Liu CC, posal for a new classification. Semin Nephrol.
Danchenko N, Nilson SE, et al. Measurement of 2011;31(4):341–8.
erythrocyte C4d and complement receptor 1 in sys- 104. D’Souza YB, Jones CJ, Short CD, Roberts IS,
temic lupus erythematosus. Arthritis Rheum. 2004;50 Bonshek RE. Oligosaccharide composition is similar
(11):3596–604. in drusen and dense deposits in membranoproli-
90. Birmingham DJ, Gavit KF, McCarty SM, Yu CY, ferative glomerulonephritis type II. Kidney Int.
Rovin BH, Nagaraja HN, et al. Consumption of eryth- 2009;75(8):824–7.
rocyte CR1 (CD35) is associated with protection 105. Barbour TD, Pickering MC, Terence Cook H. Dense
against systemic lupus erythematosus renal flare. deposit disease and C3 glomerulopathy. Semin
Clin Exp Immunol. 2006;143(2):274–80. Nephrol. 2013;33(6):493–507.
91. Cohen JH, Lutz HU, Pennaforte JL, Bouchard A, 106. Nasr SH, Valeri AM, Appel GB, Sherwinter J,
Kazatchkine MD. Peripheral catabolism of CR1 (the Stokes MB, Said SM, et al. Dense deposit
C3b receptor, CD35) on erythrocytes from healthy disease: clinicopathologic study of 32 pediatric and
individuals and patients with systemic lupus adult patients. Clin J Am Soc Nephrol. 2009;4
erythematosus (SLE). Clin Exp Immunol. 1992;87 (1):22–32.
(3):422–8. 107. Martinez-Barricarte R, Heurich M, Valdes-Canedo F,
92. Campistol JM, Arias M, Ariceta G, Blasco M, Vazquez-Martul E, Torreira E, Montes T, et al. Human
Espinosa M, Grinyo JM, et al. An update for atypical C3 mutation reveals a mechanism of dense deposit
haemolytic uraemic syndrome: diagnosis and treat- disease pathogenesis and provides insights into com-
ment. A consensus document. Nefrologia. 2013;33 plement activation and regulation. J Clin Invest.
(1):27–45. 2010;120(10):3702–12.
93. Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi M, 108. Alchi B, Jayne D. Membranoproliferative glomerulo-
Tang WH, Le Quintrec M, et al. Recessive mutations nephritis. Pediatr Nephrol. 2010;25(8):1409–18.
958 Z. Prohászka et al.

109. Nakopoulou L. Membranoproliferative glomerulone- 120. Racusen LC, Haas M. Antibody-mediated rejection in
phritis. Nephrol Dial Transplant. 2001;16 Suppl renal allografts: lessons from pathology. Clin J Am
6:71–3. Soc Nephrol. 2006;1(3):415–20.
110. Servais A, Noel LH, Roumenina LT, Le Quintrec M, 121. Drachenberg CB, Papadimitriou JC. Endothelial
Ngo S, Dragon-Durey MA, et al. Acquired and injury in renal antibody-mediated allograft rejection:
genetic complement abnormalities play a critical role a schematic view based on pathogenesis. Transplan-
in dense deposit disease and other C3 tation. 2013;95(9):1073–83.
glomerulopathies. Kidney Int. 2012;82(4):454–64. 122. Haas M. Pathologic features of antibody-mediated
111. Sethi S, Fervenza FC, Zhang Y, Nasr SH, Leung N, rejection in renal allografts: an expanding spectrum.
Vrana J, et al. Proliferative glomerulonephritis sec- Curr Opin Nephrol Hypertens. 2012;21(3):264–71.
ondary to dysfunction of the alternative pathway of 123. Reinsmoen NL, Lai CH, Heidecke H, Haas M, Cao K,
complement. Clin J Am Soc Nephrol. 2011; Ong G, et al. Anti-angiotensin type 1 receptor anti-
6(5):1009–17. bodies associated with antibody mediated rejection in
112. Ronco P, Debiec H. Membranous glomerulopathy: donor HLA antibody negative patients. Transplanta-
the evolving story. Curr Opin Nephrol Hypertens. tion. 2010;90(12):1473–7.
2010;19(3):254–9. 124. Regele H, Bohmig GA, Habicht A, Gollowitzer D,
113. Menon S, Valentini RP. Membranous nephropathy in Schillinger M, Rockenschaub S, et al. Capillary depo-
children: clinical presentation and therapeutic sition of complement split product C4d in renal allo-
approach. Pediatr Nephrol. 2010;25(8):1419–28. grafts is associated with basement membrane injury in
114. Liao MT, Chang MH, Lin FG, Tsai IJ, Chang YW, peritubular and glomerular capillaries: a contribution
Tsau YK. Universal hepatitis B vaccination reduces of humoral immunity to chronic allograft rejection.
childhood hepatitis B virus-associated membranous J Am Soc Nephrol. 2002;13(9):2371–80.
nephropathy. Pediatrics. 2011;128(3):e600–4. 125. Abbate M, Zoja C, Corna D, Rottoli D, Zanchi C,
115. Debiec H, Lefeu F, Kemper MJ, Niaudet P, Azzollini N, et al. Complement-mediated dysfunction
Deschenes G, Remuzzi G, et al. Early-childhood of glomerular filtration barrier accelerates progressive
membranous nephropathy due to cationic bovine renal injury. J Am Soc Nephrol. 2008;19(6):1158–67.
serum albumin. N Engl J Med. 2011;364 126. Legendre CM, Licht C, Muus P, Greenbaum LA,
(22):2101–10. Babu S, Bedrosian C, et al. Terminal complement
116. Ronco P, Debiec H. New insights into the pathogen- inhibitor eculizumab in atypical hemolytic-uremic
esis of membranous glomerulonephritis. Curr Opin syndrome. N Engl J Med. 2013;368(23):2169–81.
Nephrol Hypertens. 2006;15(3):258–63. 127. Zuber J, Le Quintrec M, Krid S, Bertoye C,
117. Glassock RJ. The pathogenesis of membranous Gueutin V, Lahoche A, et al. Eculizumab for atypical
nephropathy: evolution and revolution. Curr Opin hemolytic uremic syndrome recurrence in renal trans-
Nephrol Hypertens. 2012;21(3):235–42. plantation. Am J Transplant. 2012;12(12):3337–54.
118. Schlumberger W, Hornig N, Lange S, Probst C, 128. Ricklin D, Lambris JD. Complement in immune and
Komorowski L, Fechner K, et al. Differential diagno- inflammatory disorders: therapeutic interventions. J
sis of membranous nephropathy with autoantibodies Immunol. 2013;190(8):3839–47.
to phospholipase A2 receptor 1. Autoimmun Rev. 129. Vivarelli M, Emma F. Treatment of C3
2014;13(2):108–13. glomerulopathy with complement blockers. Semin
119. Hofstra JM, Wetzels JF. Anti-PLA(2)R antibodies in Thromb Hemost. 2014;40(4):472–7.
membranous nephropathy: ready for routine clinical 130. Bomback AS. Anti-complement therapy for glomerular
practice? Neth J Med. 2012;70(3):109–13. diseases. Adv Chronic Kidney Dis. 2014;21(2):152–8.
Acute Postinfectious
Glomerulonephritis in Children 31
Bernardo Rodríguez-Iturbe, Behzad Najafian,
Alfonso Silva, and Charles E. Alpers

Contents Shunt Nephritis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 974


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 960 Syphilis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 975
Acute Poststreptococcal Glomerulonephritis . . . . . 960 Other Infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 975
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 961
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 975
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 961
Clinical Characteristics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 963
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 967
Prognosis of APSGN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 969
IgA-Dominant Glomerulonephritis
Associated with Staphylococcal Infections . . . . . . . 969
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 969
Clinical Characteristics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 970
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 970
Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 970
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 970
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
Renal Disease Associated with Infective
Endocarditis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 971
Etiology and Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . 972
Clinical Characteristics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 972
Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 973
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 974

B. Rodríguez-Iturbe (*) • A. Silva


Nephrology Service, Hospital Universitario, Maracaibo,
Estado Zulia, Venezuela
e-mail: brodrigueziturbe@gmail.com; afsilvaes@yahoo.es
B. Najafian • C.E. Alpers
Department of Pathology, University of Washington,
Seattle, WA, USA
e-mail: najafian@uw.edu; calp@uw.edu

# Springer-Verlag Berlin Heidelberg 2016 959


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_27
960 B. Rodríguez-Iturbe et al.

Introduction capacity to remove the deposited complexes. In


situ formation of immune complexes results from
Postinfectious glomerulonephritis (PIGN) com- the deposition of the inciting antigen in the GBM
prises a large group of glomerulonephridities that and its penetration to the epithelial side of the
are caused by infectious agents. Acute glomerulo- GBM where it is met by the corresponding anti-
nephritis is a term that defines a pathological lesion body to form immune complexes that present
that may be asymptomatic or it may be manifested characteristic subepithelial electron-dense
clinically with the acute nephritic syndrome, or the deposits. In situ formation of immune complexes
nephrotic syndrome, or with a rapidly progressive occurs in conditions of antigen excess and is typ-
renal failure. In this chapter we will deal with the ical of cationic antigens that have electrostatic
most common glomerulonephridities that result charge-facilitated penetration of the polyanionic
from bacterial infections: poststreptococcal glomer- GBM [4]. In specific types of glomerulonephritis,
ulonephritis, IgA-dominant glomerulonephritis the pathogenesis involves the participation of bac-
associated with staphylococcal infections, and glo- terial wall superantigens causing intense T cell
merulonephritis associated with bacterial endocardi- activation that recruit cytokine-mediated poly-
tis, infected atrial ventricular shunts, syphilis, clonal B cell responses; this is the case in glomer-
and deep-seated infections. Since poststreptococcal ulonephritis associated with staphylococcal
glomerulonephritis is the best known of the acute infection and predominant IgA deposition.
postinfectious glomerulonephritis and its most Immune complexes may be predominantly
frequent clinical picture is the acute nephritic syn- located in the mesangium or in the glomerular
drome, the terms poststreptococcal glomerulone- basement membrane (GBM) or in both, and the
phritis, acute glomerulonephritis, and acute immunoglobulin and complement deposition is of
nephritic syndrome are often, and incorrectly, used granular (not linear) appearance as revealed by
interchangeably. The incidence of PIGN is declining immunofluorescence microscopy.
in affluent societies where most cases now occur in Some histological characteristics are often asso-
elderly individuals, many with comorbid debilitat- ciated with some clinical features: mesangial pro-
ing conditions such as alcoholism, HIV infection, liferative glomerulonephritis usually has a sell-
drug abuse, and diabetes. Nevertheless, PIGN, and limited course characterized by microscopic or
specifically poststreptococcal glomerulonephritis, is macroscopic hematuria and non-nephrotic protein-
still frequent in children in poor communities with uria. Heavy Ig deposition in the GBM and abun-
limited access to medical care. dance of electron-dense subepithelial deposits are
Postinfectious glomerulonephritis results from frequently associated with massive proteinuria.
antigen–antibody reactivity. Nephritogenic
immune complexes are formed in the circulation
or, in situ, in the glomerular basement membrane Acute Poststreptococcal
(GBM) and, in either case, cause local activation Glomerulonephritis
of the complement system and the coagulation
cascade. Classic experimental studies in the Dark urine and suppression of urine output was a
acute (one-shot) and chronic serum sickness feared complication of the convalescent period of
models [1–3] have demonstrated that glomerular scarlet fever as recorded in the medical literature
deposition of circulating immune complexes more than six centuries ago [5]. It was then a
depend on the size of the complexes (300–500 common disease, and the death of Mozart in
kDa) and on antigen load and the intensity of the 1791 was probably due to acute postscarlatinal
antibody response (antigen/antibody ratio). The glomerulonephritis [6]. The findings that acute
development of glomerular inflammation and glomerulonephritis followed streptococcal upper
severity, as well as the progression to chronic respiratory and skin infections [7, 8] permitted the
renal lesions, depends, on a large measure, on definition of the disease as acute poststreptococcal
the duration of antigen exposure and the host’s glomerulonephritis (PSGN), and the seminal
31 Acute Postinfectious Glomerulonephritis in Children 961

theoretical contribution of Clemens von Pirquet APSGN may occur in sporadic cases or in
[9], attributing acute PSGN (APSGN) to altered epidemic outbreaks. Earlier epidemics have been
reactivity (allergy) caused by “harmful” found to appear periodically in closed populations
antigen–antibody complexes of nephritogenic as the Red Lake Indian Reservation in Minnesota
potential (as opposed to protective antibodies and in specific areas in less developed countries as
induced by vaccination), defined the pathogenesis Port of Spain in Trinidad and in Maracaibo, Ven-
of this condition and opened the era of immune ezuela. The risk of nephritis in epidemics ranges
complex-mediated diseases [10]. from 5 % in throat infections to as high as 25 % in
pyoderma caused by M-type 49 streptococci
[21–23]. The most recent epidemic has been
Epidemiology reported from Nova Serrana, Brazil, and was
caused by Streptococcus zooepidemicus and asso-
In the last decades the epidemiology of APSGN ciated with the consumption of unpasteurized
has changed along with a decline in its incidence milk from cows with mastitis [24]. Similar etiol-
worldwide and particularly in industrialized coun- ogy has been found in clusters of cases reported in
tries where the disease is now rare and associated poor communities in industrialized countries [25].
with debilitating conditions such as alcoholism or
diabetes [11]. The reduction in the incidence of
APSGN is attributed to a variety of factors, Pathogenesis
including easier and earlier access to competent
medical treatment of streptococcal infections and The recognition that acute rheumatic fever and
the widespread use of fluorination of water since glomerulonephritis are noninfectious complica-
virulence factors in Streptococcus pyogenes are tions of streptococcal infection with different epi-
reduced with fluoride exposure [12]. Not only the demiological and biological characteristics led to
incidence but the severity of APSGN in children the concept that there existed rheumatogenic and
appears to be diminishing in developed countries nephritogenic streptococcal strains [26, 27].
[13], while in high-risk aboriginal populations, Among these differences, the rarity of recurrence
such as the French Polynesia aboriginal commu- of APSGN (in contrast to the frequency of recur-
nities, massive proteinuria, congestive heart fail- rence in rheumatic fever) suggested that
ure, and renal failure are frequent features of nephritogenic antigen(s) induced a long-lasting
APSGN [14]. protective immunity and raised the possibility of
The incidence of PSGN is also decreasing in a potential vaccination.
developing countries but is not uncommon. Streptococcal Antigens. APSGN has an
Endocapillary glomerulonephritis, assumed to be immune complex pathogenesis. The identity of
of poststreptococcal etiology, is the most common nephritogenic antigens has been the subject of
glomerulonephritis in children in underprivileged considerable investigation and controversy
societies [15] and particularly in aboriginal com- (reviewed in Rodriguez-Iturbe and Batsford
munities [16, 17]. Two studies [18, 19] have cal- [10]). Group A streptococci were traditionally
culated the global burden of APSGN and came to considered the culprit of APSGN. M types
similar conclusions, with lower estimates of 47, 49, 55, and 55 corresponded to streptococci
9.3–9.8 cases per 100,000 population per year in causing nephritis associated with pyodermitis,
developing countries and higher estimates that and M types 1, 2, 4, and 12 corresponded to
could be three times these values. The importance streptococci causing epidemics associated with
of educational programs has been evidenced by upper respiratory infection. However, more
the reduction in the incidence of APSGN recently, it has been found that glomerulonephritis
observed after the implementation of programs may result from group C streptococci (Streptococ-
designed to correct the high incidence of rheu- cus zooepidemicus) both in epidemic [24] and
matic fever in the Caribbean islands [20]. sporadic cases [25, 28] making evident that
962 B. Rodríguez-Iturbe et al.

antigenic fractions capable of causing nephritis and induce the production of pro-inflammatory
are not exclusively derived from group A cytokines [36]. SPEB/zymogen induces leukocyte
streptococci. infiltration in the glomerulus, possibly as a result of
Recent research has focused in two antigenic the induction of chemotactic and migration inhibi-
fractions: the so-called nephritis-associated plas- tion factor activities [37], and increases angiotensin
min receptor (NAPlr), identified as glyceraldehyde II production in mesangial cells [38]. The possibil-
3-phosphate dehydrogenase (GAPDH) [29, 30], ity of inducing a protective immunity to poststrep-
and the streptococcal pyrogenic exotoxin tococcal sequelae has been recently raised in
(erythrotoxin) B (SPEB) and its zymogen precur- studies that determined that the Fc portion of anti-
sor (zSPEB) [31, 32]. NAPlr and SPEB have been bodies directed to SPEB bind to the C-terminal
identified in early biopsies and elicit antibody domain (rSPEB 345–398) and immunization with
responses that may be detected in the vast majority this domain prevents group A streptococcal infec-
of convalescent sera. The frequency of the anti- tion in mice [39].
body response to SPEB/zSPEB has now been Finally, the possibility that more than one
shown to be the best antistreptococcal antibody streptococcal antigen is nephritogenic was made
response for the purposes of identifying a poten- evident by the demonstration that the gene
tially nephritogenic infection [33]. Simultaneous encoding for SPEB was absent in the
testing of both these fractions in renal biopsies of S. zooepidemicus causing the recent APSGN epi-
APSGN patients and convalescent sera indicated demic in Brazil which would indicate that this
that in the population of patients tested, antibody antigen was not involved in this epidemic [40].
response to and glomerular deposits of SPEB were Immune Reactivity. Immune complexes
frequent while those to NAPlr were unusual formed against nephritogenic streptococcal anti-
[34]. The discordant results may be due to the gen(s) may be formed in the circulation and
role of host-related factors in the development of deposited in the glomeruli or formed in situ
APSGN since NAPlr studies were done in a rela- against antigenic fractions planted in the glomer-
tively homogenous Japanese population while the uli. Both mechanisms are likely present in
SPEB studies were done in populations of patients APSGN. The relevance of the circulating immune
in Latin America and Europe. In fact, studies in complex mechanism derives from the classic
biopsies of Japanese patients did demonstrate the experiments of the glomerulonephritis that
deposits of both putative nephritogens [29]. develops in experimental models of acute serum
GAPDH is localized in the areas of the glomer- sickness [1, 2]. The participation of an in situ
uli with plasmin-like activity. Increased plasmin formation of immune complex is likely because
binding may promote the local inflammatory reac- the subepithelial electron-dense deposits (humps)
tivity and facilitate the penetration of nephritogenic that are a typical lesion of acute PSGN are difficult
antigen–antibody complexes. The finding of to produce experimentally by the injection of
increased urinary plasmin activity in APSGN is preformed immune complexes while they are
suggestive of increased urinary GAPDH common when cationic antigens are injected [4].
[35]. SPEB is co-localized in the glomeruli with Consequences of the deposition of the
complement and Ig deposits and is the only strep- antigen–antibody complexes are the local activa-
tococcal antigen that has been localized within the tion of the complement and coagulation cascades.
electron-dense subepithelial deposits (humps) [34] Complement Activation in PSGN. Reduction
that are characteristic of APSGN; the cationic of circulating complement levels is a constant fea-
(pk > 8.0) nature of this component favors the ture in APSGN. The classical pathway of comple-
penetration of the anionic barrier in the GBM. ment activation is partially blocked by Ig-binding
Histones are other cationic elements that may also proteins in streptococcal surface [41, 42] and
be relevant in the pathogenesis of APSGN since is the alternate pathway, the one that is usually
after streptococcal lysis they enter in circulation, activated, perhaps by a transient expression of
trigger an in situ formation of immune complexes, C3Nef autoantibody [43]. Yet, 15–30 % of patients
31 Acute Postinfectious Glomerulonephritis in Children 963

with APSGN have reduced levels of C1 and C4 renal inflammation [53]. Streptococcal neuramin-
components. Relevant to these findings are the idase was considered responsible for the simulta-
demonstration that Protein H, a surface protein of neous presentation of acute PSGN and thrombotic
Streptococcus pyogenes in combination with IgG, microangiopathy [54]. Another potential cause of
can activate the classic complement pathway [44]. anti-Ig reactivity is the binding of IgG to type II
The lectin pathway of complement activation may receptors in the streptococcal wall. This binding
also be activated in PSGN [45], but individuals produces intense anti-IgG reactivity that may play
genetically unable to activate this pathway may a role in PSGN [55].
still develop PSGN [46]. Additional autoimmune phenomena have been
Cellular Immunity and Cytokines. Cell- found in APSGN patients. Anti-DNA antibodies,
mediated mechanisms are also involved in the anti-C1q antibodies, and antineutrophil cytoplas-
development of APSGN. Glomerular infiltration mic antibodies (ANCA) are present in some
of lymphocytes and macrophages has long been patients. Interestingly, the latter has been found
recognized as a feature of the disease in two-thirds of the patients with azotemia and
[47]. Intercellular leukocyte adhesion molecules, 70 % of the patients with APSGN that develop
such as ICAM-1, are overexpressed in glomeruli crescentic glomerulonephritis [56].
and tubulointerstitial regions and correlated with Genetic Factors. The familial incidence of
the intensity of the inflammatory infiltration acute PSGN is indicated by the presentation of
[48]. Cytokine production plays an important the disease (clinical and subclinical) in 38 % of the
role in the renal injury in PSGN. SPEB, one of siblings of index cases [57]. This incidence is
the putative nephritogenic antigens, induces higher than the attack rate in the general popula-
mesangial proliferation, and increased IL-6 pro- tion in epidemics that range between 5 % and
duction [49] of glomerular IL-8 expression corre- 28 %. APSGN has been associated with
lates with neutrophil infiltration and transforming HLA-DR4 and DR-1 [23] but a definite genetic
growth factor –β with mesangial expansion in link has not been established. Genetic mutations
acute PSGN [50]. in the complement factor H (CFH) gene have been
Autoimmune Reactivity. A number of auto- suggested as a cause for inducing dense deposit
immune findings have been reported in APSGN; disease whose manifestations may be triggered by
among them, anti-IgG reactivity is the best stud- an episode of PSGN [58] with a clinical course
ied. High titers of rheumatoid factor are present in atypical for APSGN characterized by glomerulo-
about two-thirds of the patients in the first week of nephritis and chronic kidney disease persisting
the disease; furthermore, anti-IgG glomerular after streptococcal infection [59].
deposits are frequently found in biopsies (see
later), and anti-IgG reactivity has been
documented in the IgG eluted from the kidney in Clinical Characteristics
a fatal case of the disease [10, 23]. The anti-IgG
reactivity may be the result of autoantigenic mod- Children from 4 to 14 years are more frequently
ification of Ig that occurs with its desialization affected by acute PSGN. It is rare below the age of
caused by streptococcal neuraminidase. This pos- 2 and above the age of 20 and is twice more
sibility was raised by experimental studies show- frequent in males than in females. The usual
ing that injection of neuraminidase-treated sites of antecedent infection are the skin and the
autologous IgG causes the generation of anti- throat, although any location of streptococcal
IgG reactivity and received support from the dem- infection is possible. The latent period between
onstration of plasma neuraminidase activity and infection and nephritis is longer after skin infec-
increased free sialic acid levels in patients with tions (3–5 weeks) than after upper respiratory
APSGN [51, 52]. Since desialized leukocytes infections (7–15 days). During the latent period,
infiltrate more easily the glomeruli, neuramini- asymptomatic children may have microscopic
dase may have the additional effect of facilitating hematuria.
964 B. Rodríguez-Iturbe et al.

APSGN may have a subclinical course or may progression to chronic renal disease. Azotemia
present with the acute nephritic syndrome and occurs in 25–30 % of the children, but the need
more rarely with nephrotic syndrome or, excep- of dialysis is infrequent. A rapidly progressive
tionally, with a rapidly progressive (crescentic) azotemia may occur exceptionally and when pre-
glomerulonephritis [23]. Subclinical disease is sent is usually to the development of crescentic
characterized by a reduction of serum comple- glomerulonephritis.
ment, microscopic hematuria, and normal or Pathophysiology of the Acute Nephritic
increased blood pressure in asymptomatic Syndrome. The acute nephritic syndrome results
patients. In epidemic conditions this presentation from the reduction of the glomerular filtration rate
occurs 1.5 times more frequently than clinically caused by the inflammatory reaction in the glo-
apparent disease [21]. Prospective studies in meruli. The renal blood flow is usually normal,
household members of index cases have shown and therefore the filtration fraction is depressed,
that in non-epidemic situations the patients with- frequently below 1 %. The reabsorption is appro-
out symptoms are four to five times more frequent priately reduced in the proximal tubule and
than symptomatic patients [57, 60]. The most assumed to be maintained at the distal areas of
typical clinical picture in APSGN is the acute the nephron inaccessible to micropuncture. While
nephritic syndrome (hematuria, edema, hyperten- the reduction in glomerular filtration has long
sion, and moderate proteinuria). Glomerular been assigned a central role in the fluid retention
hematuria is an almost universal finding, and in acute glomerulonephritis, the participation of
gross hematuria is present in one-third of the additional undefined distal factor(s) is required. It
patients. The absence of red cells in the urine is is obvious that the reduction of glomerular filtra-
usually due to delays in the examination of the tion is a feature of renal conditions that are not
urinary sediment since red cells are rapidly necessarily associated with fluid retention and,
destroyed, especially in alkaline urine. As a rule, conversely, severe fluid retention may occur with-
red cell casts are present in association with dys- out reduction in glomerular filtration rate. Poten-
morphic of red cells, frequently presenting dough- tial influences that may modulate sodium and fluid
nut shape with one or more blebs. Macroscopic retention at more distal levels are endothelial or
hematuria usually disappears after a few days, but mesangial factors released by glomerular injury
microscopic hematuria may persist for a year and [61], overexpression of the epithelial sodium
occasionally exacerbates during febrile episodes channel [62], and interstitial inflammatory cells
and more rarely after strenuous exercise. Edema is capable of maintaining an increased intrarenal
the chief complaint more frequently in children angiotensin activity [63]. At any rate, the net result
(90 %) than in adult patients (75 %). Younger of the hemodynamic changes in glomeruli and
children tend to have anasarca more often than tubules is a tendency to sodium and water retention
adolescents or adult patients, but ascites is uncom- that results in expansion of the extracellular vol-
mon, except in the cases with nephrotic syndrome. ume, edema, and hypertension. The potential addi-
Hypertension is present in 60–80 % of the chil- tional effects of hypoalbuminemia are not present
dren and is severe enough to require specific anti- in the acute nephritic syndrome and, accordingly,
hypertensive treatment in about half of the cases. the plasma levels of hormones that regulate the
Edema and hypertension typically disappear in extracellular volume (renin–angiotensin, aldoste-
5–10 days. Oliguria is identified at initial presen- rone, and atrial natriuretic peptide) show an
tation by the patient or their family in less than appropriate response for an expanded extracel-
half of the patients. Other nonspecific symptoms lular volume. The suppression of plasma renin
include general malaise, weakness, headache, dull activity and aldosterone and the stimulation of
lumbar pain, and nausea. Massive proteinuria atrial natriuretic peptide are correlated with
with or without other features of the nephrotic the severity of edema. Renal prostaglandin
syndrome are found in about 2 % of the cases in and kallikrein activities are suppressed [23].
children, and its persistence is a risk factor for In concordance with this pathophysiology, the
31 Acute Postinfectious Glomerulonephritis in Children 965

improvement of volume retention with the demonstration of immune complexes in the glo-
development of spontaneous or induced diuresis meruli by immunofluorescence or electron
results in a prompt reduction of the edema and microscopy, preferably by both, and is key to
correction of the hypertension. establishing the diagnosis in a renal biopsy. By
Serological Findings. The most constant sero- light microscopy, all glomeruli are affected, and
logical finding is the reduction in serum comple- generally all to a similar extent. The glomerular
ment levels that occurs in more than 90 % of the capillaries are dilated and hypercellular. In many
cases. The activation of the complement system is cases there is an increase of endothelial and/or
usually via the alternative complement pathway mesangial cells, and the endothelial cells in par-
and reduced C3 with normal C1 and C4, but, as ticular appear swollen (a constellation of findings
mentioned earlier, the classic and the lectin path- sometimes referred to as “endocapillary prolifer-
ways of complement activation may also be ation” or “endocapillary hypercellularity”). Glo-
engaged in some patients. There are no clinical merular capillaries typically contain a prominent
characteristics associated to a specific comple- influx of inflammatory cells, especially neutro-
ment activation modality, and complement levels phils and monocytes (Fig. 1a). Because of the
return to normal usually within 1 month of the large numbers of neutrophils, the descriptive
development of acute glomerulonephritis. IgG term “exudative glomerulonephritis” has been
and IgM serum levels are elevated in 80–90 % applied to these lesions. Eosinophils and lympho-
of the patients with APSGN. Two-thirds of the cytes may be present, but not in large numbers.
patients with APSGN in the first week of the Overt necrosis is rarely identified, despite the
disease present cryoglobulins and elevated severe inflammation within the glomeruli. How-
IgG–anti-IgG rheumatoid factor titers [23]. ever, extracapillary proliferation with formation
Serum antistreptococcal antibody titers are of crescents and/or adhesions (synechiae) is
discussed later (see “Diagnosis”). often present in severe cases and presumably is a
Pathology. At the present time, renal biopsy is consequence of glomerular basement membrane
seldom done in patients with APSGN with a well- rupture due to the severe inflammatory injury.
defined clinical picture. Biopsy is indicated when Erythrocytes and sometimes neutrophils may be
specific features raise the possibility of a different present in Bowman’s space, often concurrently
diagnosis. For instance, if the serum complement with crescents. In renal biopsies obtained a few
is normal, a biopsy may define if glomerulone- weeks after the appearance of clinical symptoms,
phritis has a different etiology, such as IgA the picture may be less inflammatory, and the
nephropathy, and if the serum complement number of neutrophils will have decreased, the
remains low after 1 month, the biopsy may help swelling of endothelial cells will have subsided,
to rule out hypocomplementemic mesangio- and the number of humps seen by electron micros-
capillary glomerulonephritis or lupus nephritis. copy decreased. Nevertheless, at later stages in the
In patients with proteinuria in the nephrotic evolution of disease, diffuse mesangial hypercel-
range, a biopsy may serve to confirm the diagnosis lularity may still be seen, and this sometimes lasts
of APSGN and to raise awareness of a worse long- for several months. The combination of accentu-
term prognosis. Finally, if the patient has a rising ation of the normal glomerular lobular architec-
serum creatinine, the diagnosis of crescentic glo- ture as a result of glomerular cell proliferation, the
merulonephritis needs biopsy confirmation. overall hypercellularity of the glomeruli, and the
The classic pathologic manifestations of endocapillary changes involving the glomerular
postinfectious glomerulonephritis (PIGN) are suf- capillaries can lead to situations where PIGN is
ficiently distinctive at both the histological and difficult to distinguish from membranoproli-
ultrastructural studies that these disorders can ferative glomerulonephritis by light microscopy.
often be reliably diagnosed in a renal biopsy. Immunofluorescence and electron microscopy
APSGN is the prototype of the PIGN resulting usually allow the distinction between the two
from immune complex deposition and the diseases to be made. Other important
966 B. Rodríguez-Iturbe et al.

Fig. 1 Poststreptococcal glomerulonephritis. (a) Light mesangial regions of a glomerulus, showing in some
microscopy: global endocapillary hypercellularity/prolif- areas a “starry sky” pattern. (c) Electron microscopy:
eration with prominent influx of inflammatory cells, Characteristic subepithelial hump-like deposits (arrows)
including many neutrophils in capillary lumina and swol- over the subepithelial aspect of the glomerular basement
len endothelial cells (Jones methenamine silver stain, 40 membranes (asterisk). Abbreviations: PMN polymorpho-
objective). (b) Immunofluorescence: Coarse granular pat- nuclear, PC podocyte, E endothelial cell (11,000)
tern of C3 deposition in peripheral capillary walls and

considerations in the differential diagnosis associated with heavy proteinuria [67, 68]. The
include lupus nephritis, which usually can be dis- other types of immune deposits are not clearly
tinguished on clinical grounds and by the constel- related to clinical behavior or prognosis. The pat-
lation of associated immunofluorescence and terns of immune deposits likely represent the
electron microscopic findings. Some cases of changing distribution of immune complexes as
acute glomerulonephritis morphologically indis- the disease evolves over time.
tinguishable from postinfectious glomerulone- The ultrastructural changes in glomerular cells
phritis but with an atypical, non-resolving observed by electron microscopy closely corre-
clinical course have proven to be cases of the spond to the changes seen by light microscopy.
recently recognized entity C3 glomerulopathy Swelling of glomerular endothelial cells and
[59, 64]. increased mesangial cellularity are common. The
Immunofluorescence studies in biopsies taken glomerular basement membranes are usually of
during the first 2–3 weeks of the diseases most normal contour and thickness, although locally
often show diffuse, irregular, coarse granular some thickening may occur. The most consistent
deposits of immunoglobulin G (IgG), IgM, and and classic change is the presence of glomerular
C3 along the glomerular capillary walls (Fig. 1b). subepithelial dome-shaped electron-dense
The C3 deposits resolve later than immunoglobu- deposits, widely referred to as “humps” (Fig. 1c).
lin, and biopsies obtained late in the disease These are especially numerous during the first
course may show predominantly or only C3 by weeks of acute glomerulonephritis, and their num-
immunofluorescence. Antihuman IgG deposits ber decreases thereafter. Podocyte foot processes
can be demonstrated in 20–30 % of the cases overlying the humps are often effaced, with con-
and less extensive deposits of C1q may occasion- densation of cytoplasmic microfilaments.
ally be present [15, 65, 66]. Based on the distri- Early in the evolution of disease, the immune
bution and appearance of the immune deposits, deposits may form in the subendothelial aspect of
Sorger et al. [67] recognized by immunofluores- glomerular capillary walls and after undergoing
cence heavy “garland-type” immune deposits in some process of dissociation as the disease
the capillaries and immune deposits presenting a evolves reform as humps at the subepithelial sur-
“starry sky” and “mesangial” staining patterns. face of the glomerular capillary walls. This migra-
The garland type of immune deposits has been tion process is characteristic of the in situ immune
31 Acute Postinfectious Glomerulonephritis in Children 967

complex formation shown experimentally to demonstration of a rising titer of serum antistrep-


result from cationic antigens [69]. The early tococcal antibodies. Anti-NAPlr and antiSPEB-
subendothelial deposits are uncommonly visual- zSPEB titers have a greater specificity for
ized in electron micrographs of renal biopsies, but nephritogenic infections [33] but are not generally
in a minority of cases, these may be prominent and available. Antistreptolysin O titers and anti-
impart a predominantly membranoproliferative DNAse B titers are the most frequently elevated
pattern of injury. Such cases require careful clin- in upper respiratory infections and pyodermitis,
icopathological correlation to distinguish them respectively. Streptozyme test which includes
from other diagnostic entities, principally four antigens (DNAse B, streptolysin O, hyal-
membranoproliferative glomerulonephritis of uronidase, and streptokinase) is reported to be
noninfectious origin, lupus nephritis, and rarely positive in more than 80 % of the cases [23, 33].
IgA nephropathy. These cases also overlap with Complement levels are a first-line diagnostic
systemic or deep infection-related glomerulone- test that permits consideration of acute glomer-
phritis, such as nephritis due to infected ventricu- ulonephritides that usually present with low
loatrial shunts, deep tissue abscesses, and serum complement (APSGN, lupus nephritis,
endocarditis. In addition, some small irregular shunt nephritis, endocarditis, cryoglobulinemia,
electron-dense deposits may be seen in the lamina hypocomplementemic membranoproliferative
densa of glomerular basement membranes and the glomerulonephritis) and those that usually present
mesangium. with normal complement levels (IgA nephropa-
After resolution of the acute disease, residual thy, mesangioproliferative GN, hemolytic uremic
subepithelial electron-dense deposits may persist syndrome, Henoch–Schonlein purpura, vasculitis,
for extended periods of time. These are typically anti-GBM disease). The finding of profoundly
located at so-called mesangial “waists,” where the depressed C4 levels in association with normal
glomerular capillary walls overlay the mesangium, or mildly reduced C3 suggests the diagnosis of
and may result in an immunofluorescence pattern cryoglobulinemia type II and the reduction of C1,
of predominately mesangial staining of immune C4, and C3 levels, indicating activation of the
reactants. A few of these deposits may persist for classic pathway of complement, is characteristic
very long periods of time, and these are typically of lupus erythematosus. Serum complement
discovered as incidental findings in renal biopsies levels return to normal in less than 1 month, and
performed for other clinical indications unrelated to persistence of low complement levels should raise
PSGN [65, 70]. suspicion of a diagnosis different from APSGN.
Diagnosis. The typical clinical presentation is Physical examination findings suggestive of
that of a child of 4–15 years who suddenly nephrotic, rather than nephritic syndrome, are a
develops dark and scanty urine, swelling of the soft, paperlike consistency of the ear cartilage and
face and legs, and high blood pressure. The initial the existence of ascites, since both these signs are
diagnostic approach should attempt to define if the associated with long-standing hypoalbuminemia.
acute nephritic syndrome is associated with a sys- Inasmuch as the nephrotic syndrome is rare in
temic condition or if it is resulting from a primary APSGN, the demonstration of a massive protein-
renal disease. It is then necessary to establish an uria favors the diagnosis of lupus nephritis,
etiologic diagnosis of glomerulonephritis. A his- nephritis associated with visceral abscesses, and
tory of a precedent upper respiratory infection membranoproliferative glomerulonephritis.
and/or skin infection suggests a poststreptococcal
etiology that may be confirmed by a positive
culture or rising antistreptococcal antibody titers. Treatment
Positive streptococcal cultures are frequent in epi-
demic conditions but only in less than 25 % of the Antibiotic Treatment. The first question to be
sporadic cases; consequently, the evidence of pre- considered is when to give antibiotic treatment to
vious streptococcal infection usually resides in the a suspected nephritogenic streptococcal infection.
968 B. Rodríguez-Iturbe et al.

From a clinical viewpoint, the diagnosis of active 1.2 million units of benzathine penicillin in adults
skin infection is usually straightforward. However, and adolescents and half this dose in small chil-
clinical judgment may miss half of the streptococ- dren, or alternatively, oral phenoxymethyl or
cal pharyngitis and may incorrectly diagnose a sore phenoxymethyl penicillin G 125 mg, every 6 h
throat as due to streptococcal infection in 20–40 % for 7–10 days, are adequate treatment. Erythromy-
of the cases [71]. Several clinical scores have been cin (250 mg every 6 h and 40 mg per kg in children,
proposed to increase the accuracy of this diagnosis, for 7–10 days) is the treatment of choice in patients
and among the most popular of them is the one allergic to penicillin. It should be noted that the
proposed by McIsaac et al. [72]. This score has a existence of erythromycin-resistant strains may be
range from 0 to 4 and incorporates age as one of the increasing in developing countries. The incidence
criteria. The score gives a +1 to each one of the of resistance to erythromycin in isolates of Strep-
following: temperature >38 C, cervical tococcus pyogenes is 9.7 % in Japan [77], 21.7 % in
adenopathy, no cough, tonsillar exudate, and age Spain [78], 6.8 % in the United States [79], and
between 3 and 14 years. Age >44 years is assigned 11.6 % in bacterial isolates from several European
1 point. Sensitivity and specificity of the score is countries [80].
85 % and 95 %, respectively. Antibiotic treatment Preventive antibiotic treatment is indicated in
is recommended (without culture confirmation) epidemic situations and to household members of
when the score is 4, and antibiotic treatment is not index cases in non-epidemic conditions since
indicated (and cultures unnecessary) when scores most of them present evidence of recent infection
are 0–1. and about one-third of them develop nephritis
Rapid, high-sensitivity streptococcal tests are [57]. In high-risk communities the strategy of
good guides to treat if they are positive, but a treating household contacts has resulted in a
negative test requires confirmation [73]. Neverthe- decrease in the number of cases of PSGN [81].
less, a recent report indicates that a decision to Treatment of the Acute Nephritic Syn-
treat or not to treat based on the results of these drome. Patients with subclinical disease may be
tests is not associated with a higher incidence of followed as outpatients but patients with the acute
poststreptococcal complications [74]. More nephritic syndrome require hospitalization. Bed
recently, evaluation of a home score for the diag- rest is difficult to enforce and is of unproven
nosis of streptococcal pharyngitis in patients value, yet most children keep it on their own
15 years and older, based on patient-reported clin- while they are in the acute phase. Restrictions of
ical variables, has been calculated that avoided fluid and sodium intake are the cornerstones of the
230,000 doctor visits each year in the United treatment of patients with the acute nephritic syn-
States [75]. drome. Cases that present significant edema,
The diagnosis of PSGN carries with it the indi- hypertension, and circulatory congestion benefit
cation of treatment with penicillin or, in allergic from the administration of loop diuretics (40 mg
individuals, erythromycin. If infection is present at IV or orally every 12 h). This therapy facilitates
the time of diagnosis, it requires treatment. Early the resolution of edema and ameliorates the hyper-
administration of penicillin is reported to prevent or tension that is driven by extracellular volume
ameliorate the severity of acute glomerulonephri- expansion [82]. Diuretic therapy seldom if ever
tis, and at least one report suggests that APSGN is required for longer than 48 h. Other diuretics are
patients that receive antibiotic treatment have a without effect (thiazide diuretics) or dangerous
milder clinical course [76]. If infection is not appar- because of the possibility of hyperkalemia (aldo-
ent at the time of diagnosis, antibiotic treatment sterone antagonists).
should be given anyway because positive cultures Patients who present with severe hypertension
are sometimes obtained in apparently healthy may require antihypertensive treatment, and nifed-
patients and cross infection of household members ipine (5 mg in children, every 4–6 h) is usually
and siblings of index cases is very high [57]. effective. Parenteral hydralazine may be required
31 Acute Postinfectious Glomerulonephritis in Children 969

but the possibility of tachycardia requires close the various studies since the long-term prognosis
observation. Angiotensin-converting enzyme of APSGN is substantially worse in adults, partic-
inhibitors and type 1 receptor blockers carry the ularly those with persistent proteinuria in the
risk of hyperkalemia. Exceptionally, nitroprusside nephrotic range. An unfavorable course was also
is required to control hypertensive encephalopathy. observed in the patients in the Nova Serrana epi-
Another potential complication, frequently demic, most of whom were adults. Chronic renal
associated with hypertension, is the posterior failure developed in 8 % of these patients after
reversible leukoencephalopathy that has recently 5 years [86]. In contrast, children have in most
been reported in acute PSGN [83]. This compli- studies a much more favorable long-term progno-
cation is manifested clinically with mental distur- sis. A combined analysis of reported data indi-
bances, visual hallucinations, headache, and cates that 20 % of the children followed for 10–20
convulsions and may be confused with hyperten- years after acute PSGN have an abnormal urine
sive encephalopathy. The diagnosis requires the analysis but azotemia was found in less than 1 %
use of nuclear magnetic resonance image studies. of the patients, and in our own follow-up studies
Pulmonary edema is rare and should be treated of 110 children followed for 15–18 years after the
with oxygen therapy, rotating tourniquets and acute attack, non-nephrotic proteinuria was found
loop diuretics. Digitalis is not indicated because in 7.2 %, microhematuria in 5.4 %, hypertension
it is ineffective and may result in intoxication. in 3.0 %, and azotemia in 0.9 % [19]. Neverthe-
Rarely overt heart failure and pulmonary edema less, the subsequent contribution of additional risk
may complicate the clinical course. Hemodialysis factors, such as diabetes and obesity, worsens the
or peritoneal dialysis may be required occasion- late prognosis of these patients. Studies in
ally in children for the treatment of hyperkalemia, Australian aboriginal communities where these
uremia, or severe circulatory congestion. risk factors are prevalent indicate that patients
A rapidly progressive azotemia is usually asso- who had APSGN have an increased risk for albu-
ciated with crescentic APSGN. While of minuria (adjusted odds ratio 6.1 %, 95 % CI
unproven efficacy, there are anecdotal reports of 2.2–16.9) and hematuria (OR 3.7, CI 1.8–8.0) in
beneficial effects of pulse methylprednisolone relation to controls that did not have APSGN
therapy. [87]. The history of PSGN was significantly asso-
ciated (odds ratio 3–4) with increased incidence of
reduced (<60 ml/min) glomerular filtration
Prognosis of APSGN rate [88].

The short-term prognosis of APSGN is excellent


in children. Fatalities may occur as a result of IgA-Dominant Glomerulonephritis
hyperkalemia or pulmonary edema, but they are Associated with Staphylococcal
exceedingly rare. The long-term prognosis of Infections
APSGN has been the subject of many reports
since the initial studies in the first half of the Epidemiology
twentieth century reported essentially a complete
recovery, but the follow-up periods were short and In 1995 Koyama et al. [89] described a severe
the patients were only children in the majority of form of glomerulonephritis associated with meth-
the studies. Subsequent observations gave widely icillin-resistant Staphylococcus aureus (MRSA)
different results, and abnormal urinary findings in infections that was characterized by dominant or
patients followed for extended periods of time codominant IgA deposits and was caused by
ranged from 3.5 % [84] to 60 % [85]. The discrep- staphylococcal superantigens similar to those
ancies may be due in part to the lack of discrim- causing the toxic shock syndrome. The incidence
ination between adult and children populations in of IgA-dominant glomerulonephritis has been
970 B. Rodríguez-Iturbe et al.

estimated in 1.6 % of the biopsies in adult patients antigen-presenting cells and induce a massive
[90], but in recent reports from Japan, the fre- proliferation of T cells bearing specific V beta
quency appears to be decreasing, in association determinants. In some cases there may be a selec-
with the observed reduction in the number of tive IgA response or, in some cases, the intense
MRSA infections [91]. IgA-dominant glomerulo- T cell activation of B cells results in polyclonal
nephritis is usually observed in hospital-acquired IgA and IgG production. Koyama et al. [97]
staphylococcal infections in diabetic patients, but have identified an antigen in S. aureus that is
it may occur in community-acquired staphylococ- co-localized with glomerular IgA deposits and
cal infections [92] and is not exclusive of staphy- developed an experimental model of the disease
lococcal infections or diabetic patients [93]. immunizing Balb/c mice with this antigen [98].

Clinical Characteristics Pathology

The average age of the patients is 60 years; how- IgA-dominant postinfectious glomerulonephritis
ever, it has been reported in children [94]. The is defined by deposition of IgA in both glomerular
disease is three to four times more frequent in capillary walls and mesangial regions as the sole
males, and the usual clinical presentation is acute or predominant immune reactant (Fig. 2b), in
renal insufficiency. Massive proteinuria is present contrast to the typical pattern of IgG and/or C3
in 51 % of the patients and gross hematuria in deposition that is characteristic of other PIGN
20 % [66, 95]. Approximately half of the cases [89, 90, 95, 99]. By light microscopy (Fig. 2a)
occur in diabetic patients, and the existence of the histological characteristics may be quite
diabetes is a risk factor for the development of variable. They may be similar to those of PSGN
chronic renal disease: two-thirds of the patients or resemble mesangial proliferative GN or
that developed end-stage renal disease in this con- membranoproliferative GN with or without cres-
dition are diabetic. The most common site of cents. By electron microscopy, the characteristics
infection is the skin, followed by surgical wounds and distribution of electron-dense deposits in IgA-
and intravenous lines, but in some instances the dominant GN resemble those in PSGN ([95];
site of the original infection is not apparent. Fig. 2c). Since IgA-dominant PIGN may be
Serum complement levels are frequently low, superimposed on other chronic nephropathies,
but they may be normal and IgA serum levels are such as diabetic nephropathy, these cases may
frequently increased [93]. The number of circulat- present a hybrid morphologic appearance.
ing Vβ2(+) T cells expressing CD45RO, a marker The differential diagnosis between IgA-
of activation, is increased, and high levels of sev- dominant PIGN and IgA nephropathy may be
eral cytokines, including tumor necrosis factor- quite challenging, and one helpful feature is
alpha, interleukin-1 beta, IL-2, IL-6, IL-8, and the finding of hump-like subepithelial electron-
IL-10, may be found [94, 96]. dense deposits by electron microscopy in
IgA-dominant PIGN.

Pathogenesis
Diagnosis
Glomerulonephritis associated with staphylococ-
cal infections can result from immune complex- IgA-dominant glomerulonephritis caused by
mediated renal disease, but the pathogenesis MRSA infection may present similarities with
of the IgA-dominant glomerulonephritis is the conditions in which there are dominant or codom-
result of superantigen-induced immune reactivity. inant IgA deposits in the kidney such as
Staphylococcal superantigens are exoproteins Henoch–Schonlein purpura and IgA nephropathy.
that bind directly to MHC class II molecules in Staphylococcal infection frequently precedes
31 Acute Postinfectious Glomerulonephritis in Children 971

Fig. 2 IgA-dominant postinfectious glomerulonephri- Immunofluorescence – IgA: strong granular and predom-
tis in a patient with diabetic nephropathy. (a) Light inantly mesangial staining. (c) Immunofluorescence –
microscopy: Mesangial regions are expanded by increased C3: concomitant granular mesangial and peripheral capil-
matrix (as a feature of diabetic nephropathy) and cellular- lary wall staining. (d) Electron microscopy: subepithelial
ity. There is also an influx of inflammatory cells into hump-like deposits (red arrows) over the thickened glo-
glomerular capillaries and endothelial swelling, creating merular basement membrane (white asterisk). Abbrevia-
segmental endocapillary proliferation (arrowhead) (Jones tions: PMN polymorphonuclear, PC podocyte (11,000)
methenamine silver stain, 40 objective). (b)

Henoch–Schonlein nephropathy, but the finding


of a purpuric rash in the legs and pains in the Renal Disease Associated
abdomen and joints are absent in most patients with Infective Endocarditis
with IgA-dominant PIGN. On the other hand,
Henoch–Schonlein purpura seldom presents with Infective endocarditis is the underlying infectious
massive proteinuria or glomerular crescent process in 10 % of the cases of PIGN [65], and
formation [100]. renal involvement is present in 25–50 % of the
Patients with severe IgA nephropathy may be patients with infective endocarditis but is fre-
at times confused with IgA-dominant glomerulo- quently asymptomatic [102, 103]. The association
nephritis. Characteristics that may be helpful in of focal, embolic, and nonsuppurative glomerulo-
the differential diagnosis are the lack of associa- nephritis with bacterial endocarditis was first
tion with staphylococcal infection, the normal described by Max Lohlein in 1910 [104]. Subse-
serum complement levels, and the absence of quent descriptions two decades ago emphasized
massive proteinuria and of glomerular crescent the immune complex pathogenesis causing dif-
formation in IgA nephropathy. fuse glomerulonephritis [102, 105], and more
recent autopsy studies have disclosed that local-
ized infarcts, glomerulonephritis (without demon-
Treatment strable immune complex deposits), and interstitial
nephritis, mostly attributed to antibiotic therapy,
Antibiotic treatment of the staphylococcal infec- are all frequent renal lesions in endocarditis-
tion is the cornerstone of treatment in associated renal disease [106].
IgA-dominant PIGN resulting from staphylococ-
cal infection. Corticosteroid treatment is
contraindicated while there is active infection; Epidemiology
therefore, it is important to make the differential
diagnosis with IgA nephropathy in which treat- Approximately 15,000 new cases of infective
ment with corticosteroids may be indicated in endocarditis (hospital and community acquired)
severe cases. In selected cases with persisting or occur in the United States each year [107], and the
progressive deterioration of renal function, steroid incidence appears to be steadily increasing [108].
treatment and immunosuppression has been used However the incidence of community-acquired
when infection is no longer present [101]. native-valve endocarditis is essentially unchanged
972 B. Rodríguez-Iturbe et al.

at 1.7–6.2 cases per 100,000 person years in the lesions most frequently associated with
United States and Europe [109] but with impor- endocarditis [119].
tant epidemiologic changes. The proportion of When an immune complex pathogenesis is
adolescents and young adults that have endocarditis involved, bacterial antigens may be demonstrated
associated with rheumatic heart disease has in the glomeruli [120] and the nephritogenic mech-
decreased and, in contrast, the proportion of older anisms are similar to those operating in APSGN.
patients with endocarditis associated with prosthetic Polyclonal (type III) cryoglobulinemia may be
heart valves, implantable devices, and drug abuse found in roughly half of the patients. In addition,
has increased [110]. While the incidence of MRSA superantigen-driven T cell activation with poly-
infections has increased in community-acquired clonal gammopathy may occur in bacterial endocar-
cases [111], the incidence of methicillin-susceptible ditis caused by methicillin-resistant Staphylococcus
cases may be increasing in healthcare-associated aureus. In experimental studies, superantigen-
cases [112]. One of the most important epidemio- associated endocarditis is regularly associated with
logical changes in recent years has been the acute kidney injury [121]. Microembolization is
increment in cases associated with healthcare inter- responsible for the localized infarcts, and tubuloin-
ventions and, particularly, the cases associated with terstitial nephritis is usually secondary to antibiotic
hemodialysis and caused by staphylococcus. treatment. In the patients in hemodialysis programs,
Bacterial endocarditis is 20–60 times more com- the infection is usually located in the vascular access
mon in hemodialysis patients than in the general and when present synthetic arteriovenous grafts or
population and carries a near 50 % mortality risk venous dialysis catheters have been usually in place
[113, 114]. In the pediatric patient, congenital heart for more than 1 year [122, 123]. In these patients
disease and cardiac surgery represent the most com- calcifications of heart valves are risk factors and
mon risk factors, and in reports from a single insti- treatments directed to reduce the risk of ectopic
tution, the incidence of pediatric infective calcification are important preventive measures of
endocarditis has not changed over time but the infectious endocarditis [124].
mortality has decreased from 38 % in historical
cohorts to 4 % in modern age [115].
Clinical Characteristics

Etiology and Pathogenesis The classic clinical picture of subacute bacterial


endocarditis includes Osler’s nodules, Janeway
Infecting bacteria are Staphylococcus aureus, and lesions, and splinter hemorrhages; however,
Staphylococcus epidermidis, Streptococcus these findings are rare at the present time. Fever
viridans and pyogenes, Enterococcus faecalis, may be the only manifestation, accompanied or
and less commonly E. coli, Proteus, and microor- not with arthralgias, leukocytosis, increased sedi-
ganisms of the Bartonella and Candida species. In mentation rate, and purpura. The lack of classic
pediatric populations Streptococcus viridians and symptoms is likely the reason why 38 % of the
Staphylococcus aureus are the most common patients with endocarditis found at autopsy were
infecting agents and together represent more not diagnosed clinically [125].
than 50 % of the cases [115]. Peaks of bacteremia The cardiac lesion is best demonstrated by
as well as a cumulative exposure to low-level transesophageal echocardiography. Factors usu-
bacteremia are genuine risks for endocarditis ally associated with higher mortality are a vegeta-
[116]. In hospital-acquired endocarditis the most tion size 20 mm, age less than 1 year, the
common bacteria are Staphylococcus aureus and existence of heart failure, and Staphylococcus
Staphylococcus epidermidis, while in aureus as a causative organism [126]. Emboliza-
community-acquired endocarditis, streptococcal tion to the central nervous system with stroke may
infections are the most frequent [117, 118]. Mitral occur in 6 % of the children [127]. Systemic
valve disease and mitral prolapse are the heart embolism maybe microscopic or large; the latter
31 Acute Postinfectious Glomerulonephritis in Children 973

Fig. 3 Glomerulonephritis associated with endocardi- (c) Electron microscopy: Abundant subendothelial
tis. (a) Light microscopy: Widespread glomerular deposits (red arrows), associated with duplication of glo-
basement membrane duplication creates a membranopro- merular basement membrane (the original basement mem-
liferative pattern of injury with influx of inflammatory cells brane is marked by asterisk and the thin new membrane is
(less prominent than APSGN) and swollen endothelial shown by black arrows). There are also a few mesangial
cells (Jones methenamine silver stain, 40 objective). deposits (white arrow). Abbreviations: E endothelial cell,
(b) Immunofluorescence: Granular mesangial and M mesangial cell (11,000)
peripheral capillary wall staining for C3 and IgG.

usually corresponds to endocarditis caused by disease, immune complex glomerulonephritis,


fungus or Haemophilus. and drug-induced interstitial nephritis. Early
Microscopic hematuria and proteinuria are investigations come from autopsy studies that
indicative of a renal lesion. Urinary findings usu- described embolic complications [104, 134], and
ally persist for weeks or months. Azotemia may subsequent studies reported glomerulonephritis in
develop in one-thirds of the patients [128] but is nearly 80 % of the cases [102]. More recent stud-
usually mild except in cases that develop crescen- ies showed, in cases studied at autopsy, a higher
tic glomerulonephritis; nevertheless even mild incidence of localized embolic infarct than glo-
renal dysfunction is associated with a poor prog- merulonephritis, while in cases studied by renal
nosis [129]. The existence of eosinophilia and biopsy, acute glomerulonephritis was the most
eosinophiluria should raise the suspicion of frequent lesion and renal infarcts were absent
antibiotic-induced interstitial nephritis. Nephrotic [106, 125]. Of interest, 25 % of the renal biopsies
syndrome is unusual. in cases of infective endocarditis reported from
Serological findings include reduced C3 and the United Kingdom demonstrated acute intersti-
C4 (except in superantigen-mediated nephritis), tial nephritis [106].
high titers of rheumatoid factor, cryoglobulinemia Infective endocarditis-related PIGN differs
[130], and, on occasion, positive anti-PR3 ANCA from APSGN in that involvement of glomeruli
antibodies in the serum [131, 132]. When ANCA may not be uniform (i.e., the distribution of abnor-
are present in association with lung and kidney mal glomeruli may be focal rather than diffuse).
involvement, the clinical course resembles The histological alterations can be varied and
granulomatosis with polyangiitis (formerly range from a predominately mesangial prolifera-
known as Wegener’s granulomatosis) [133]. The tive glomerulonephritis to a membranoproli-
complement levels return to normal after the ferative pattern of injury (Fig. 3a). When
infection has been eradicated, and this finding is present, such a membranoproliferative pattern of
associated with good prognosis of the renal lesion. injury may be histologically indistinguishable
from membranoproliferative glomerulonephritis
type I due to other causes such as hepatitis C
Pathology virus infection and systemic autoimmune diseases
such as lupus. The glomeruli may have a mixed
Three different types of renal disease have been and predominately mononuclear leukocyte influx;
identified in infective endocarditis: embolic renal neutrophils may be present but their presence is
974 B. Rodríguez-Iturbe et al.

not typically florid as it is in APSGN. Features Shunt Nephritis


that overlap with those of APSGN include glo-
merular hypercellularity and endocapillary prolif- Congenital or acquired hydrocephalus may
eration with endothelial swelling. Foci of require the placement of a shunt between cerebral
segmental glomerular necrosis are not uncommon ventricles and the atrium (ventriculoatrial, VA) or
and are similar to those found in ANCA-positive the peritoneum (ventriculoperitoneal, VP) to ame-
vasculitis, but only a fraction of these patients liorate increased cerebrospinal fluid pressure.
have positive serum ANCA [106]. Crescents, Infection is a recognized complication of these
when present, usually involve a minority of glo- shunts that was initially reported to occur in
meruli and have been found in 10–22 % of the one-third of these patients [140, 141]. More recent
patients in various series [65, 102, 106, 135, 136]. studies have shown that the incidence of infection
Immunofluorescence studies typically show a in these shunts has decreased to 6.1 % [142]. The
granular pattern of deposition of immune com- development of nephrotic syndrome in a patient
plexes in peripheral glomerular capillary walls, with infected VA shunt was reported by Black and
with variable but usually less pronounced coworkers two decades ago [160], and glomeru-
involvement of mesangial regions (Fig. 3b). The lonephritis is now a recognized complication that
immune deposits typically contain some combi- develops a few months to many years after inser-
nation of IgG, IgM, and C3, but deposits of IgA tion in 0.7–2 % of the infected AV shunts. In
have also been described. contrast with VA shunts, VP shunts are rarely
Corresponding electron-dense deposits are complicated with glomerulonephritis.
found by electron microscopy, usually in The infecting microorganisms are usually
subendothelial locations in the capillary walls Staphylococcus epidermidis and, less frequently,
(Fig. 3c), particularly in cases where a membrano- Staphylococcus aureus. Propionibacterium acne,
proliferative glomerulonephritis pattern is pre- diphtheroids, and Pseudomonas and Serratia spe-
sent. However, subepithelial deposits, including cies are infecting organisms in rare occasions. The
occasional humps similar to those of PSGN, may etiology of the renal disease has been demonstrated
be found. All of the histological and immunoflu- by the finding of bacterial antigens in the glomeruli
orescence and electron microscopy changes are of patients with shunt nephritis [143, 144].
potentially reversible and may resolve with suc- The clinical manifestations are recurrent epi-
cessful treatment of the underlying infection. sodes of fever, anemia, hepatosplenomegaly, skin
rash, and cerebral symptoms related to increased
intracranial pressure. Renal involvement is indi-
Treatment cated by proteinuria that in most cases is in the
nephrotic range hematuria and hypertension. The
The high incidence of endocarditis in hemodialy- combination of skin and renal manifestation may
sis patients has caused a renewed emphasis on the lead to incorrect diagnosis, and the possible exis-
need for strict infection control policies and tence of urinary tract infection frequently accom-
prompt conversion to arteriovenous access from panying a neurogenic bladder that may afflict
catheters and appropriate antibiotic prescriptions these children may make the interpretation of the
[137]. Antibiotic treatment needs to be given for urinary findings difficult. Serological findings
4–6 weeks and the cessation of fever is not an include reduced C3 and C4 levels, indicating acti-
indication that antibiotics may be stopped before vation of the classic complement pathway,
this time [138]. cryoglobulinemia, anemia, elevated sedimenta-
In patients who present with crescentic glomer- tion rate, and high rheumatoid factor titers.
ulonephritis and a rapidly progressive course, pulse Serum anti-P3 ANCA titers are sometimes present
steroid therapy and plasmapheresis have been used [140, 145].
with success [139], but their overall value needs to The renal lesion demonstrated at biopsy is
be confirmed in more definite studies. indistinguishable from the range of lesions
31 Acute Postinfectious Glomerulonephritis in Children 975

encountered in settings of endocarditis, as Other Infections


described earlier.
Treatment of shunt nephritis requires intrave- Deep-seated infections such as osteomyelitis and
nous antibiotic therapy and removal of the intra-abdominal abscesses may be associated with
infected shunt and, if possible, substituting it for glomerulonephritis manifested clinically by the
a VP shunt. Rarely, if ever, are antibiotics alone nephrotic syndrome and progressive azotemia or
capable of eliminating the infection. The long- present only by mild urinary abnormalities [152].
term prognosis of the renal function is good if In children, pneumonia caused by Mycoplasma
the shunt is removed within a few weeks of pneumoniae infections [153, 154] and Streptococ-
detecting the infection [140]. If the removal of cus pneumoniae [155, 156] has been found to cause
the shunt is delayed, the renal damage may pro- immune complex-induced glomerulonephritis.
gress to irreversible end-stage renal failure. Pneumococcal antigen type 14 has been demon-
Hemodialysis is the preferred modality in strated in the glomeruli. Hematuria and
patients with end-stage renal disease because peri- non-nephrotic proteinuria are the usual clinical man-
tonitis associated with peritoneal dialysis carries ifestations in cases of glomerulonephritis associated
the risk of meningitis if a VP shunt is in place. with pneumonia. In infections with neuraminidase-
Given the high rate of complications in the VA producing pneumococcus, the patient may develop
shunts, the VP shunt is the preferred initial proce- hemolytic uremic syndrome due to exposure of the
dure, but VA shunts may be the selected method in Thomsen–Friedenreich antigen [157].
children with chronic renal failure in whom renal The most common histological pattern in these
transplantation is considered. glomerulonephritis is that of diffuse proliferative
glomerulonephritis, often with crescent formation
[152, 158]. Membranoproliferative glomerulone-
Syphilis phritis has also been found in some cases. Immune
deposits of C3 with little or no immunoglobulin
Congenital and acquired syphilis may cause glo- deposits are common findings [152, 159].
merulonephritis. Congenital syphilis is usually Glomerulonephritis improves with the treat-
manifested by rash, rhinitis, and osteochondritis, ment of infection if the therapy is started early
but 8–10 % of the patients may develop a full- and eradicates the infection completely [152].
blown nephrotic syndrome that presents as anasarca
4–12 weeks after birth [146, 147]. Later in life,
syphilis is a reversible cause of nephrotic syndrome
[148] that presents in 1–2 % of the patients with the
References
disease [149] and may masquerade as membranous 1. Germuth FG. Comparative histologic and immuno-
nephropathy. The most common histological logic study in rabbits of induced hypersensitivity of
appearance is that of membranous nephropathy, the serum sickness type. J Exp Med. 1953;97:257–82.
and electron microscopy shows electron-dense 2. Dixon FJ, Feldman JD, Vasquez JJ. Experimental
glomerulonephritis: the pathogenesis of a laboratory
deposits along the subepithelial aspect of the model resembling the spectrum of human glomerulo-
GBM [150]. Other types of glomerulonephritides, nephritis. J Exp Med. 1961;113:899–920.
including mesangioproliferative, membranoproli- 3. Germuth FG, Senterfit LB, Dressman GR. Immune
ferative, and diffuse proliferative glomerulonephri- complex disease. V. The nature of circulating com-
plexes associated with glomerular alterations in
tis, have been reported, especially in adult patients. chronic BSA-rabbit system. Johns Hopkins Med
Treponema antigens have been demonstrated in the J. 1972;130:344–57.
glomerular immune deposits [151]. Serological 4. Vogt A, Schmiedeke T, Stockl FW, Sugisaki Y,
tests for syphilis are positive and complement levels Mertz A, Batsford S. The role of cationic proteins in
the pathogenesis of immune complex glomerulone-
are normal. Treatment of the syphilis is associated phritis. Nephrol Dial Transplant. 1990;5 Suppl 1:6–9.
with improvement of the renal disease, but recovery 5. Becker CG, Murphy GE. The experimental induction
usually takes several months. of glomerulonephritis like that in man by infection
976 B. Rodríguez-Iturbe et al.

with Group A streptococci. J Exp Med. rheumatic fever in two French Caribbean Islands.
1968;127:1–23. Lancet. 1966;347:644–8.
6. Zegers RH, Weigl A, Steptoe A. The death of 21. Anthony BF, Kaplan EL, Wannamaker LW, Briese
Wolfgang Amadeus Mozart: an epidemiologic per- FW, Chapman SS. Attack rates of acute nephritis
spective. Ann Intern Med. 2009;151:274–8. after type 49 streptococcal infections of the skin and
7. Dochez AR, Sherman L. The significance of of the respiratory tract. J Clin Invest. 1969;48:
Streptococcus hemolyticus in scarlet fever and the 1697–704.
preparation of a specific antiscarlatinal serum by 22. Poon-King T, Mohammed I, Cox R, Potter EV,
immunization of the horse to Streptococcus Simon NM, Siegel AC, Earle DP. Recurrent epidemic
hemolyticus. J Am Med Assoc. 1924;82:542–4. nephritis in South Trinidad. N Engl J Med.
8. Futcher PH. Glomerular nephritis following skin 1967;277:728–33.
infections. Arch Intern Med. 1940;65:1192–210. 23. Rodriguez-Iturbe B. Epidemic poststreptococcal glo-
9. von Ergebn PC. Inn Med Kinderheilk. 1910;5: merulonephritis. Kidney Int. 1984;25:129–36.
459–539 translated into English in: von Pirquet 24. Balter S, Benin A, Pinto SW, Teixeira LM, Alvim
C. Allergy. Arch Int Med. 1911;7:259–88, 382–436. GG, Luna E, Jackson D, LaClaire L, Elliott J,
10. Rodriguez-Iturbe B, Batsford S. Pathogenesis of Facklam R, Schuchat A. Epidemic nephritis in Nova
poststreptococcal glomerulonephritis a century after Serrana, Brazil. Lancet. 2000;355:1776–80.
Clemens von Pirquet. Kidney Int. 2007;71:1094–104. 25. Nicholson ML, Ferdinand L, Sampson JS, Benin A,
11. Montseny JJ, Meyrier A, Kleinknecht D, Callard Balter S, Pinto SW, Dowell SF, Facklam RR, Carlone
P. The current spectrum of infectious glomerulone- GM, Beall B. Analysis of immunoreactivity to a
phritis. Experience with 76 patients and review of the Streptococcus equi subsp. zooepidemicus M-like pro-
literature. Medicine (Baltimore). 1995;74:63–73. tein to confirm an outbreak of poststreptococcal glo-
12. Thongboonkerd V, Luengpailin J, Cao J, Pierce WM, merulonephritis, and sequences of M-liked proteins
Cai J, Klein JB, Doyle RJ. Fluoride exposure attenu- from isolates obtained from different host species.
ates expression of Streptococcus pyogenes virulence J Clin Microbiol. 2000;38:4126–30.
factors. J Biol Chem. 2002;277:16599–605. 26. Rammelkamp CH, Weaver RS, Dingle JH. Signifi-
13. Ilyas M, Tolaymat A. Changing epidemiology of cance of the epidemiologic differences between acute
acute post-streptococcal glomerulonephritis in North- nephritis and acute rheumatic fever. Trans Assoc Am
east Florida: a comparative study. Pediatr Nephrol. Phys. 1952;65:168–75.
2008;23:1101–16. 27. Segal D, Earle DP. A consideration of certain
14. Becquet O, Pasche J, Gatti H, Chenel C, Abély M, biological differences between glomerulonephritis
Morville P, Pietrement C. Acute post-streptococcal and rheumatic fever. Am J Med Sci. 1941;201:
glomerulonephritis in children of French Polynesia: 528–39.
a 3-year retrospective study. Pediatr Nephrol. 28. Francis AJ, Nimmo GR, Efstratiou A, Galanis V,
2010;25:275–80. Nuttall N. Investigation of milk- borne Streptococcus
15. Rodriguez-Iturbe B, Mezzano S. Infections and kid- zooepidemicus infection associated with glomerulo-
ney diseases: a continuing global challenge. In: El nephritis in Australia. J Infect. 1993;27:317–23.
Nahas M, editor. Kidney diseases in the developing 29. Oda T, Yoshizawa N, Yamakami K, Tamura K,
world and ethnic minorities. London: M. Taylor & Kuroki A, Sugisaki T, Sawanobori E, Higashida K,
Francis; 2005. p. 59–82. Ohtomo Y, Hotta O, Kumagai H, Miura
16. Currie BJ, Brewster DR. Childhood infections in the S. Localization of nephritis-associated plasmin recep-
tropical north of Australia. J Paed Child Health. tor in acute poststreptococcal glomerulonephritis.
2001;37:326–30. Hum Pathol. 2010;41(9):1276–85.
17. Marshall CS, Cheng AC, Markey PG, Towers RJ, 30. Yoshizawa N, Yamakami K, Fujino M, Oda T,
Richardson LJ, Fagan PK, Scott L, Krause VL, Currie Tamura K, Matsumoto K, Sugisaki T, Boyle MD.
BJ. Acute post-streptococcal glomerulonephritis in Nephritis-associated plasmin receptor and acute
the Northern Territory of Australia: a review of poststreptococcal glomerulonephritis: characteriza-
16 years data and comparison with the literature. tion of the antigen and associated immune response.
Am J Trop Med Hyg. 2011;85(4):703–10. J Am Soc Nephrol. 2004;15:1785–93.
18. Carapetis JR, Steer AC, Mullholand EK, Steer AC, 31. Poon-King T, Bannan J, Viteri A, Cu G, Zabriskie
Mulholland EK, Weber M. The global burden of JB. Identification of an extracellular plasmin binding
group A streptococcal diseases. Lancet. 2005;5: protein from nephritogenic streptococci. J Exp Med.
685–94. 1993;178:759–63.
19. Rodriguez-Iturbe B, Musser JM. The current state of 32. Vogt A, Batsford S, Rodríguez-Iturbe B, García
poststreptococcal glomerulonephritis. J Am Soc R. Cationic antigens in poststreptococcal glomerulo-
Nephrol. 2008;19:1855–64. nephritis. Clin Nephrol. 1983;20:271–9.
20. Bach JF, Chalons S, Forier E, Elana G, Jouannelle J, 33. Parra G, Rodríguez-Iturbe B, Batsford S, Vogt A,
Kayemba S, Delbois D, Mosser A, Saint-Aime C, Mezzano S, Olavarría F, Exeni R, Laso M, Orta
Berchel C. 10-year educational programme aimed at N. Antibody to streptococcal zymogen in the serum
31 Acute Postinfectious Glomerulonephritis in Children 977

of patients with acute glomerulonephritis: a complement activation by IgG-coated targets. J Biol


multicentric study. Kidney Int. 1998;54:509–17. Chem. 1997;272:20774–81.
34. Batsford SR, Mezzano S, Mihatsch M, Schiltz E, 45. Oshawa I, Ohi H, Endo M, Fujita T, Matsushita M,
Rodríguez-Iturbe B. Is the nephritogenic antigen in Fujita T. Evidence of lectin complement pathway
post-streptococcal glomerulonephritis pyrogenic exo- activation in poststreptococcal glomerulonephritis.
toxin B (SPE B) or GAPDH? Kidney Int. Kidney Int. 1999;56:1158–9.
2005;68:1120–9. 46. Skattum L, Akesson P, Truedsson L, Sjöholm
35. Oda T, Tamura K, Yoshizawa N, Sugisaki T, AG. Antibodies against four proteins from a Strepto-
Matsumoto K, Hattori M, Sawai T, Namikoshi T, coccus pyogenes serotype M1 strain and levels of
Yamada M, Kikuchi Y, Suzuki S, Miura S. Elevated circulating mannan-binding lectin in acute poststrep-
urinary plasmin activity resistant to alpha2- tococcal glomerulonephritis. Int Arch Allergy
antiplasmin in acute poststreptococcal glomerulone- Immunol. 2006;140:9–19.
phritis. Nephrol Dial Transplant. 2008;23:2254–9. 47. Parra G, Platt JL, Falk RJ, Rodriguez-Iturbe B,
36. Zhang L, Ignatowski TA, Spengler RN, Noble B, Michael AF. Cell populations and membrane attack
Stinson MW. Streptococcal histone induces complex in glomeruli and patients with poststrep-
murine macrophages to produce interleukin-1 and tococcal glomerulonephritis: identification using
tumor necrosis factor alpha. Infect Immun. monoclonal antibodies by indirect immunofluores-
1999;67:6473–7. cence. Clin Exp Immunol Immunopathol. 1984;33:
37. Romero M, Mosquera J, Novo E, Fernandez L, Parra 324–32.
G. Erythrogenic toxin type B and its precursor iso- 48. Parra G, Romero M, Henríquez-LaRoche C,
lated from streptococci induce leukocyte infiltration Rodriguez-Iturbe B. Expression of adhesion mole-
in normal rat kidneys. Nephrol Dial Transplant. cules in poststreptococcal glomerulonephritis.
1999;14:1867–74. Nephrol Dial Transplant. 1994;9:1412–4.
38. Viera N, Pedreanez A, Rincon J, Mosquera J. Strep- 49. Pedreanez A, Viera N, Rincon J. Increased IL-6 in
tococcal zymogen type B induces angiotensin II in supernatant of rat mesangial cell cultures treated with
mesangial cells and leukocytes. Pediatr Nephrol. erythrogenic toxin type B and its precursor isolated
2009;24:1005–11. from nephritogenic streptococci. Am J Nephrol.
39. Tsao N, Cheng MH, Yang HC, Wang YC, Liu YL, 2006;26:75–81.
Kuo CF. Determining antibody- binding site of strep- 50. Mezzano S, Burgos ME, Olavarría F, Caorsi
tococcal pyrogenic exotoxin B to protect mice I. Immunohistochemical localization of IL-8 and
from group a streptococcus infection. PLoS One. TGF-beta in streptococcal glomerulonephritis. J Am
2013;8(1):e55028. Soc Nephrol. 1997;8:234–41.
40. Beres SB, Sesso R, Pinto SW, Hoe NP, Porcella SF, 51. Asami T, Tanaka A, Gunji T, Sakai K. Elevated serum
Deleo FR, Musser JM. Genome sequence of a and urine sialic acid levels in renal diseases of child-
Lancefield group C Streptococcus zooepidemicus hood. Clin Nephrol. 1985;23:112–9.
strain causing epidemic nephritis: new information 52. Rodriguez-Iturbe B, Katiyar VN, Coello
about an old disease. PLoS One. 2008;3:e3026. J. Neuraminidase activity and free sialic acid levels
doi:10.1371/. in the serum of patients with acute poststreptococcal
41. Perez-Caballero D, Garcia-Laorden I, Cortes G, glomerulonephritis. N Engl J Med. 1981;304:
Wessels MR, de Córdoba SR, Albertí S. Interaction 1506–10.
between complement regulators and Streptococcus 53. Marín C, Mosquera J, Rodríguez-Iturbe B. Neuramin-
pyogenes: binding of C4b-binding protein and factor idase promotes neutrophil, lymphocyte and macro-
H/factor H-like protein 1 to M18 strains involve two phage infiltration in the normal rat kidney. Kidney
different cell surface molecules. J Immunol. Int. 1995;47:88–95.
2004;173:6899–904. 54. Duvic C, Desramé J, Hérody M, Nédélec G. Acute
42. Them A, Stenberg L, Dahback B, Lindahl G. poststreptococcal glomerulonephritis associated with
Ig-binding surface proteins of Streptococcus thrombotic microangiopathy in an adult. Clin
pyogenes also bind human C4b-binding protein Nephrol. 2000;54:169–73.
(C4bP), a regulatory component of the complement 55. Burova LA, Nagornev VA, Pigarevsky PV,
system. J Immunol. 1995;154:375–86. Gladilina MM, Seliverstova VG, Schalen C,
43. Frémeaux-Bacchi V, Weiss L, Demouchy C, May A, Totolian AA. Triggering of renal tissue damage in
Palomera S, Kazatchkine MD. Hypocomple- the rabbit by IgG Fc receptor-positive group A strep-
mentaemia of poststreptococcal acute glomerulone- tococci. Acta Pathol Microb Immun Scand.
phritis is associated with a C3 nephritic factor 1998;106:277–87.
(C3NeF) IgG autoantibody activity. Nephrol Dial 56. Ardiles LG, Valderrama G, Moya P, Mezzano
Transplant. 1994;9:1747–50. SA. Incidence and studies on antigenic specificities
44. Berge A, Kihlberg BM, Sjöholm AG, Björck L. of antineutrophil-cytoplasmic autoantibodies
Streptococcal protein H forms soluble complement- (ANCA) in poststreptococcal glomerulonephritis.
activating complexes with IgG, but inhibits Clin Nephrol. 1997;47:1–5.
978 B. Rodríguez-Iturbe et al.

57. Rodriguez-Iturbe B, Rubio L, Garcia R. Attack rate complex glomerulonephritis in the rat induced by
of poststreptococcal glomerulonephritis in families. planted, cationized antigen. J Exp Med. 1982;55(2):
A prospective study. Lancet. 1981;1:401–3. 460–7.
58. Prasto J, Kaplan BS, Russo P, Chan E, Smith RJ, 70. Haas M. Incidental healed postinfectious glomerulo-
Meyers KE. Streptococcal infection as possible trig- nephritis: a study of 1012 renal biopsy specimens
ger for dense deposit disease (C3 glomerulopathy). examined by electron microscopy. Human Pathol.
Eur J Pediatr. 2014;173:767–72. 2003;34:3–10.
59. Vernon KA, Goicoechea de Jorge E, Hall AE, 71. Cebul DR, Poses RM. The comparative cost-
Fremeaux-Bacchi V, Aitman TJ, Cook HT, effectiveness of statistical decision rules and experi-
Hangartner R, Koziell A, Pickering MC. Acute pre- enced physicians in pharyngitis management. J Am
sentation and persistent glomerulonephritis following Med Assoc. 1986;256:3353–7.
streptococcal infection in a patient with heterozygous 72. McIsaac WJ, White D, Tannenbaum D. A clinical
complement factor H-related protein 5 deficiency. Am score to reduce unnecessary antibiotic use in patients
J Kidney Dis. 2012;60(1):121–5. with sore throat. Can Med Assoc J. 1998;158:75–83.
60. Dodge WF, Spargo BF, Travis LB. Occurrence 73. American Academy of Pediatrics, Committee on
of acute glomerulonephritis in sibling contacts of Infectious Diseases. Group A streptococcal infec-
children with sporadic acute glomerulonephritis. tions. In: The red book. Elk Grove Village: American
Pediatrics. 1967;40:1028–30. Academy of Pediatrics; 2000. p. 526–92.
61. Juncos LI. Intrarenal mechanisms of salt and water 74. Webb KH, Needham CA, Kurtz SR. Use of a high-
retention in the nephritic syndrome. Kidney Int. sensitivity rapid strep test without culture confirma-
2002;61:1182–95. tion of negative results: 2 years’ experience. J Fam
62. Kim SW, Frøkiaer J, Nielsen S. Pathogenesis of Pract. 2000;49:34–6.
oedema in nephrotic syndrome: role of epithelial 75. Fine AM, Nizet V, Mandl KD. Participatory medi-
sodium channel. Nephrology. 2007;12 Suppl 3: cine: a home score for streptococcal pharyngitis
S8–10. enabled real-time biosurveillance. Ann Intern Med.
63. Rodríguez-Iturbe B, Herrera-Acosta J, Johnson 2013;159:577–83.
RJ. Interstitial inflammation, sodium retention, and 76. Zoch-Zwierz W, Wasilewska A, Biernacka A,
the pathogenesis of nephrotic edema: a unifying Tomaszewska B, Winiecka W, Wierciński R, Porowski
hypothesis. Kidney Int. 2002;62:1379–84. T. The course of post-streptococcal glomerulonephritis
64. Pickering MC, D’Agati VD, Nester CM, Smith RJ, depending on methods of treatment of the preceding
Haas M, Appel GB, Alpers CE, Bajema IM, respiratory tract infection. Wiad Lek. 2001;54:56–63.
Bedrosian C, Braun M, Doyle M, Fakhouri F, 77. Billal DS, Hotomi M, Yamauchi K, Fujihara K,
Fervenza FC, Fogo AB, Frémeaux-Bacchi V, Gale Tamura S, Kuki K, Sugita R, Endou M,
DP, Goicoechea de Jorge E, Griffin G, Harris CL, Mukaigawa J, Yamanaka N. Macrolide-resistant
Holers VM, Johnson S, Lavin PJ, Medjeral-Thomas- genes of Streptococcus pyogenes isolated from the
N, Paul Morgan B, Nast CC, Noel LH, Peters DK, upper respiratory tract by polymerase chain reaction.
Rodríguez de Córdoba S, Servais A, Sethi S, Song J Infect Chem. 2004;10:115–20.
WC, Tamburini P, Thurman JM, Zavros M, Cook 78. Tamayo J, Pérez-Trallero E, Gómez-Garcés JL, Alós
HT. C3 glomerulopathy: consensus report. Kidney JI. Resistance to macrolides, clindamycin and
Int. 2013;84:1079–89. telithromycin in Streptococcus pyogenes isolated in
65. Nasr SH, Markowitz GS, Stokes MB, Said SM, Valeri Spain during 2004. J Antimicrob Chemother.
AM, D’Agati VD. Acute postinfectious glomerulone- 2005;56:780–2.
phritis in the modern era: experience with 86 adults 79. Richter SS, Heilmann KP, Beekmann SE, Miller NJ,
and review of the literature. Medicine (Baltimore). Miller AL, Rice CL, Doern CD, Reid SD, Doern
2008;87:21–32. GV. Macrolide-resistant Streptococcus pyogenes in
66. Nasr SH, D’Agati VD. IgA-dominant postinfectious the United States, 2002–2003. Clin Infect Dis.
glomerulonephritis: a new twist on an old disease. 2005;41:599–608.
Nephron Clin Pract. 2011;119:18–25. 80. Richter SS, Heilmann KP, Dohrn CL, Beekmann SE,
67. Sorger K, Gessler M, Hubner FK, Kohler H, Riahi F, Garcia-de-Lomas J, Ferech M, Goossens H,
Olbing H, Schulz W, Thoenes GH, Thoenes W. Doern GV. Increasing telithromycin resistance among
Follow-up studies of three subtypes of acute Streptococcus pyogenes in Europe. J Antimicrob
postinfectious glomerulonephritis ascertained by Chemother. 2008;61:603–11.
renal biopsy. Clin Nephrol. 1987;27:111–24. 81. Johnston F, Carapetis J, Patel MS, Wallace T, Spillane
68. Edelstein CL, Bates WD. Subtypes of acute P. Evaluating the use of penicillin to control outbreaks
postinfectious glomerulonephritis: a clinico- of acute poststreptococcal glomerulonephritis. Pediatr
pathological correlation. Clin Nephrol. 1992;38: Infect Dis J. 1999;18:327–32.
311–7. 82. Powell HR, McCredie D, Rotenberg F. Response to
69. Oite T, Batsford SR, Mihatsch MJ, Takamiya H, frusemide in acute renal failure. Dissociation of renin
Vogt A. Quantitative studies of in situ immune and diuretic responses. Clin Nephrol. 1980;14:55–9.
31 Acute Postinfectious Glomerulonephritis in Children 979

83. Ahn SY, Ingulli E. Acute poststreptococcal glomeru- 96. Yoh K, Kobayashi M, Yamaguchi N, Hirayama K,
lonephritis: an update. Curr Opin Pediatr. Ishizu T, Kikuchi S, Iwabuchi S, Muro K, Nagase S,
2008;20:157–62. Aoyagi K, Kondoh M, Takemura K, Yamagata K,
84. Potter EV, Lipschultz SA, Abidh S, Poon-King T, Koyama A. Cytokines and T-cell responses in
Earle DP. Twelve to seventeen- year follow up of superantigen- related glomerulonephritis following
patients with acute poststreptococcal glomerulone- methicillin-resistant Staphylococcus aureus infection.
phritis in Trinidad. N Engl J Med. 1982;307:725–9. Nephrol Dial Transplant. 2000;8:1170–4.
85. Baldwin DS, Gluck MC, Schacht RG, Gallo G. The 97. Koyama A, Sharmin S, Sakurai H, Shimizu Y,
long-term course of poststreptococcal glomerulone- Hirayama K, Usui J, Nagata M, Yoh K,
phritis. Ann Intern Med. 1974;80:342–58. Yamagata K, Muro K, Kobayashi M, Ohtani K,
86. Sesso R, Pinto SWL. Five-year follow-up of patients Shimizu T, Shimizu T. Staphylococcus aureus cell
with epidemic glomerulonephritis due to Streptococ- envelope antigen is a new candidate for the induction
cus zooepidemicus. Nephrol Dial Transplant. of IgA nephropathy. Kidney Int. 2004;66:121–32.
2005;20:1808–13. 98. Sharmin S, Shimizu Y, Hagiwara M, Hirayama K,
87. Hoy WE, Mathews JD, McCredie DA, Pugsley DJ, Koyama A. Staphylococcus aureus antigens induce
Hayhurst BG, Rees M, Kile E, Walker KA, Wang IgA-type glomerulonephritis in Balb/c mice.
Z. The multidimensional nature of renal disease: J Nephrol. 2004;17:504–11.
rates and association of albuminuria in an Australian 99. Nasr SH, Markowitz GS, Whelan JD, Albanese JJ,
Aboriginal community. Kidney Int. 1998;54: Rosen RM, Fein DA, Kim SS, D’Agati
1296–304. VD. IgA-dominant acute poststaphylococcal glomer-
88. Hoy WE, White AV, Dowling A, Sharma SK, ulonephritis complicating diabetic nephropathy. Hum
Bloomfield H, Tipiloura BT, Swanson CE, Pathol. 2003;34:1235–41.
Mathews JD, McCredie DA. Post-streptococcal 100. Satoskar AA, Molenda M, Scipio P, Shim R,
glomerulonephritis is a strong risk factor for chronic Zirwas M, Variath RS, Brodsky SV, Nadasdy GM,
kidney disease in later life. Kidney Int. Hebert L, Rovin B, Nadasdy T. Henoch-Schönlein
2012;81:1026–32. purpura-like presentation in IgA-dominant Staphylo-
89. Koyama A, Kobayashi M, Yamaguchi N, coccus infection – associated glomerulonephritis – a
Yamagata K, Takano K, Nakajima M, Irie F, diagnostic pitfall. Clin Nephrol. 2013;7(9):302–12.
Goto M, Igarashi M, Iitsuka T. Glomerulonephritis 101. Okuyama S, Wakui H, Maki N, Kuroki J, Nishinari T,
associated with MRSA infection: a possible role of Asakura K, Komatsuda A, Sawada K. Successful
bacterial superantigen. Kidney Int. 1995;47:207–16. treatment of post-MRSA infection glomerulonephri-
90. Satoskar AA, Nadasdy G, Plaza JA, Sedmak D, tis with steroid therapy. Clin Nephrol. 2008;70:
Shidham G, Hebert L, Nadasdy T. Staphylococcus 344–7.
infection-associated glomerulonephritis mimicking 102. Neugarten J, Baldwin DS. Glomerulonephritis
IgA nephropathy. Clin J Am Soc Nephrol. in bacterial endocarditis. Am J Med. 1984;77:
2006;1:1179–86. 297–304.
91. Usui J, Kobayashi M, Ebihara I, Koyama A, Yama- 103. Khalighi MA, Nguyen S, Wiedeman JA, Palma-Diaz
gata K. Methicillin-resistant Staphylococcus aureus MF. Bartonella endocarditis- associated glomerulone-
associated glomerulonephritis on the decline: phritis: a case report and review of the literature. Am J
decreased incidence since the 1990s. Clin Exp Kidney Dis. 2014;63:1060–5.
Nephrol. 2011;15:184–6. 104. Löhlein M. Über hämorrhagische Nierenalffek bei
92. Worawichawong S, Girard L, Trpkov K, Gough JC, chronischer ulzeröser Endokarditis embolische
Gregson DB, Benediktsson H. Immunoglobulin nichteiterige Herdnephritis. Med Klin. 1910;6:375–9.
A-dominant postinfectious glomerulonephritis: fre- 105. Eknoyan G. Renal complications of bacterial endo-
quent occurrence in nondiabetic patients with Staph- carditis. Am J Nephrol. 1985;5:457–69.
ylococcus aureus infection. Hum Pathol. 2011;42: 106. Majumdar A, Chowdhary S, Ferreira MA, Hammond
279–84. LA, Howie AJ, Lipkin GW, Littler WA. Renal patho-
93. Wen YK, Chen ML. IgA-dominant postinfectious logical findings in infective endocarditis. Nephrol
glomerulonephritis: not peculiar to staphylococcal Dial Transplant. 2000;15:1782–7.
infection and diabetic patients. Ren Fail. 107. Paterick TE, Paterick TJ, Nishimura RA, Nishimura
2011;33:480–5. RA, Steckelberg JM. Complexity and subtlety of
94. Kimata T, Tsuji S, Yoshimura K, Tsukaguchi H, infective endocarditis. Mayo Clin Proc.
Kaneko K. Methicillin- resistant Staphylococcus 2007;82:615–21.
aureus elated glomerulonephritis in a child. Pediatr 108. Bor DH, Woolhandler S, Nardin R, Brusch J,
Nephrol. 2012;11:2149–52. Himmelstein DU. Infective endocarditis in the U.S.,
95. Haas M, Racusen LC, Bagnasco SM. IgA-dominant 1998–2009: a nationwide study. PLoS One. 2013;8:
postinfectious glomerulonephritis: a report of e60033.
13 cases with common ultrastructural features. 109. Tleyjeh IM, Abdel-Latif A, Rahbi H, Scott CG,
Human Pathol. 2008;39:1309–16. Bailey KR, Steckelberg JM, Wilson WR,
980 B. Rodríguez-Iturbe et al.

Baddour LM. A systematic review of population- in chronic haemodialysis patients with infective endo-
based studies of infective endocarditis. Chest. carditis. Nephrol Dial Transplant. 2006;21:2184–90.
2007;132:1025–35. 124. Doulton T, Sabharwal N, Cairns HS, Schelenz S,
110. Tlevjeh IM, Steckelberg JM, Murad HS, Anavekar Eykyn S, O’Donnell P, Chambers J, Austen C,
NS, Ghomrawi HM, Mirzoyev Z, Moustafa SE, Goldsmith DJ. Infective endocarditis in dialysis
Hoskin TL, Mandrekar JN, Wilson WR, Baddour patients: new challenges and old. Kidney Int.
LM. Temporal trends in infective endocarditis: a pop- 2003;64:720–7.
ulation- based study in Olmsted County, Minnesota. 125. Fernández-Guerrero ML, Álvarez B, Manzarbeitia F,
J Am Med Assoc. 2005;293:3022–8. Renedo G. Infective endocarditis at autopsy: a review
111. David MZ, Daum RS. Community-associated methi- of pathologic manifestations and clinical. Medicine
cillin-resistant Staphylococcus aureus: epidemiology (Baltimore). 2012;91:152–64.
and clinical consequences of an emerging epidemic. 126. Yoshinaga M, Niwa K, Niwa A, Ishiwada N,
Clin Microb Rev. 2010;23:616–87. Takahashi H, Echigo S, Nakazawa M. Risk factors
112. David MZ, Boyle-Vavra S, Zychowski DL, Daum for in-hospital mortality during infective endocarditis
RS. Methicillin-susceptible Staphylococcus aureus in patients with congenital heart disease. Am J
as a predominantly healthcare-associated pathogen: Cardiol. 2008;101:114–8.
a possible reversal of roles? PLoS One. 2011;6(4): 127. Venkatesan C, Wainwright MS. Pediatric endocarditis
e18217. and stroke: a single-center retrospective review of
113. Hoen B. Infective endocarditis: a frequent disease in seven cases. Pediatr Neurol. 2008;38:243–7.
dialysis patients. Nephrol Dial Transplant. 2004;19: 128. Conlon PJ, Jefferies F, Krigman HR, Corey GR, Sex-
1360–2. ton DJ, Abramson MA. Predictors of prognosis and
114. Rekik S, Trabelsi I, Hentati M, Hammami A, Jemaa risk of acute renal failure in bacterial endocarditis.
MB, Hachicha J, Kammoun S. Infective endocarditis Clin Nephrol. 1998;49:96–101.
in hemodialysis patients: clinical features, echocar- 129. Koeda C, Tashiro A, Itoh T, Okabayashi H, Nakamura
diographic data and outcome: a 10-year descriptive M. Mild renal dysfunction on admission is an impor-
analysis. Clin Exp Nephrol. 2009;13:350–4. tant prognostic predictor in patients with infective
115. Johnson JA, Boyce TG, Cetta F, Steckelberg JM, endocarditis: a retrospective single-center study.
Johnson JN. Infective endocarditis in the pediatric Intern Med. 2013;52:1013–8.
patient: a 60-year single-institution review. Mayo 130. Agarwal A, Clemens J, Sedmark DD, Imler D,
Clin Proc. 2012;87:629–35. Nahman Jr NS, Orsinelli DA, Hebert LA. Subacute
116. Veloso TR, Amiguet M, Rousson V, Giddey M, bacterial endocarditis masquerading as type III essen-
Vouillamoz J, Moreillon P, Entenza JM. Induction of tial mixed cryoglobulinemia. J Am Soc Nephrol.
experimental endocarditis by continuous low- grade 1997;8:1971–6.
bacteremia mimicking spontaneous bacteremia in 131. Fukuda M, Motokawa M, Usami T, Oikawa T,
humans. Infect Immun. 2011;79:2006–11. Morozumi K, Yoshida A, Kimura G. PR3-ANCA-
117. Furuno JP, Johnson JK, Schweizer ML, Uche A, Stine positive crescentic necrotizing glomerulonephritis
OC, Shurland SM, Forrest GN. Community-associated accompanied by isolated pulmonic valve infective
methicillin-resistant Staphylococcus aureus bacteremia endocarditis, with reference to previous reports of
and endocarditis among HIV patients: a cohort study. renal pathology. Clin Nephrol. 2006;66:202–9.
BMC Infect Dis. 2011;11:298. 132. Choi HK, Lamprecht P, Niles JL, Gross WL, Merkel
118. Que YA, Moreillon P. Infective endocarditis. Nature PA. Subacute bacterial endocarditis with positive
Rev Cardiol. 2011;8:322–36. cytoplasmic antineutrophil cytoplasmic antibodies
119. Mylonakis E, Calderwood SB. Infective endocarditis and anti-proteinase 3 antibodies. Arthritis Rheum.
in adults. New Eng J Med. 2001;345:1318–30. 2000;43:226–31.
120. Yum M, Wheat LJ, Maxwell D, Edwards 133. Peng H, Chen WF, Wu C, Chen YR, Peng B, Paudel
JL. Immunofluorescent localization of Staphylococ- SD, Lou TQ. Culture-negative subacute bacterial
cus aureus antigen in acute bacterial endocarditis endocarditis masquerades as granulomatosis with
nephritis. Am J Clin Pathol. 1978;70:832–5. polyangiitis (Wegener’s granulomatosis) involving
121. Salgado-Pabón W, Breshears L, Spaulding AR, both the kidney and lung. BMC Nephrol.
Merriman JA, Stach CS, Horswill AR, Peterson ML, 2012;13:174.
Schlievert PM. Superantigens are critical for Staphy- 134. Baehr G. Glomerular lesions of subacute bacterial
lococcus aureus Infective endocarditis, sepsis, and endocarditis. J Exp Med. 1912;15:330–47.
acute kidney injury. MBio. 2013;4(4):pii: e00494- 135. Boulton-Jones JM, Sissons JGP, Evans DJ, Peters
13. doi:10.1128. DK. Renal lesions of subacute infective endocarditis.
122. McCarthy JT, Steckelberg JM. Infective endocarditis British Med J. 1974;2:11–4.
in patients receiving long-term hemodialysis. Mayo 136. Morel-Maroger L, Sraer JD, Herreman G, Godeau P.
Clin Proc. 2000;75:1008–14. Kidney in subacute endocarditis. Pathological
123. Nori US, Manoharan A, Thornby JI, Yee J, and immunofluorescence findings. Arch Pathol.
Parasuraman R, Ramanathan V. Mortality risk factors 1972;94:205–13.
31 Acute Postinfectious Glomerulonephritis in Children 981

137. Fitzgibbons LN, Puls DL, Mackay K, Forrest syndrome. BMJ Case Rep. 2013;pii: bcr2012008279.
GN. Management of gram-positive coccal bacteremia doi: 10.1136/bcr-2012-008279.
and hemodialysis. Am J Kidney Dis. 2011;57: 149. Hunte W, Ghraoui F, Cohen RJ. Secondary syphilis
624–40. and nephrotic syndrome. J Am Soc Nephrol.
138. Badour LM, Wilson WR, Bayer AS, Fowler Jr VG, 1993;3:1351–5.
Bolger AF, Levison ME, Ferrieri P, Gerber MA, 150. Soehardy Z, Hayati SN, Rozita M, et al. Subclinical
Tani LY, Gewitz MH, Tong DC, Steckelberg JM, acquired syphilis masquerading as membranous glo-
Baltimore RS, Shulman ST, Burns JC, Falace DA, merulonephritis. Med J Malaysia. 2006;61:484–6.
Newburger JW, Pallasch TJ, Takahashi M, Taubert 151. O’Regan S, Fong JS, de Chadarévian JP, Rishikof JR,
KA. Infective endocarditis. Diagnosis, antimicrobial Drummond KN. Treponemal antigens in congenital
therapy and management of complications. Circula- and acquired syphilitic nephritis: demonstration by
tion. 2005;111:3167–84. immunofluorescence studies. Ann Intern Med.
139. Konstantinov KN, Harris AA, Hartshorne MF, 1976;85:325–7.
Tzamaloukas AH. Symptomatic anti-neutrophil 152. Beaufils M, Morel-Maroger L, Sraer JD, Kanfer A,
cytoplasmic antibody-positive disease complicating Kourilsky O, Richet G. Acute renal failure of glomer-
subacute bacterial endocarditis: to treat or not to ular origin during visceral abscesses. New Engl J
treat? Case Rep Nephrol Urol. 2012;2:25–32. Med. 1976;295:185–9.
140. Haffner D, Schinderas F, Aschoff A. The clinical 153. Adra AL, Vigue MG, Dalla Vale F, Ichay L,
spectrum of shunt nephritis. Nephrol Dial Transplant. Raynaud P, Mariani A, Morin D. Favorable outcome
1997;12:1143–8. in a case of Mycoplasma pneumoniae-associated
141. Kubota M, Sakata Y, Saeki N, Yamaura A, Ogawa M. crescentic glomerulonephritis. Pediatr Nephrol.
A case of shunt nephritis diagnosed 17 years after 2010;25:1765–9.
ventriculoatrial shunt implantation. Clin Neurol 154. Siomou E, Kollios KD, Papadimitriou P, Kostoula A,
Neurosurg. 2001;103:245–6. Papadopoulou ZL. Acute nephritis and respiratory
142. Korinek AM, Fulla-Oller L, Boch AL, Golmard JL, tract infection caused by Mycoplasma pneumoniae:
Hadiji B, Puybasset L. Morbidity of ventricular cere- case report and review of the literature. Pediatr Infect
brospinal fluid shunt surgery in adults an 8- year Dis J. 2003;22:1103–6.
study. J Neurosurg Sci. 2011;55:161–3. 155. Carceller Lechón F, de la Torre Espí M, Porto Abal R,
143. Dobrin RS, Day NK, Quie PG, Moore HL, Vernier Écija Peiró JL. Acute glomerulonephritis associated
HL, Michael AF, Fish AJ. The role of complement, with pneumonia: a review of three cases. Pediatr
immunoglobulin and bacterial antigens in coagulase- Nephrol. 2010;25:161–4.
negative Staphylococcus shunt nephritis. Am J Med. 156. Phillips J, Palmer A, Baliga R. Glomerulonephritis
1975;59:660–73. associated with acute pneumococcal pneumonia: a
144. Groeneveld AB, Nommensen FE, Mullink H, Ooms case report. Pediatr Nephrol. 2005;20:1494–5.
EC, Bode WA. Shunt nephritis associated with 157. Krysan DJ, Flynn JT. Renal transplantation after
Propionibacterium acnes with demonstration of the Streptococcus pneumoniae- associated hemolytic ure-
antigen in the glomeruli. Nephron. 1982;32:365–9. mic syndrome. Am J Kidney Dis. 2001;37:E15.
145. Nagashima T, Hirata D, Yamamoto H, Okazaki H, 158. Whitworth JA, Morel-Maroger L, Mignon F,
Minota S. Antineutrophil cytoplasmic autoantibody Richet G. The significance of extracapillary prolifer-
specific for proteinase 3 in a patient with shunt nephri- ation. Clinicopathological review of 60 patients.
tis induced by Gemella morbillorum. Am J Kidney Nephron. 1976;16:1–19.
Dis. 2001;37(5):E38. 159. D’Agati VD, Jennette JC, Silva FG. Non-neoplastic
146. Basker M, Agarwal I, Bendon KS. Congenital kidney diseases. Atlas of nontumor pathology, First
nephrotic syndrome-a treatable cause. Ann Trop series; Fascicle 4. Washington, DC: American Regis-
Paediatr. 2007;27:87–90. try of Pathology; 2005. p. 269–96.
147. Tudorache E, Hogan J, Dourthe ME, Quinet B, 160. Black JA, Chaacombe DN, Ockenden BG. Nephrotic
Grimprel E, Sellier-Leclerc AL, Ulinski T. Congenital syndrome associated with bacteraemia after shunt
nephrotic syndrome with acute renal failure: ques- operations for hydrocephalus. Lancet. 1965;2:921–4.
tions. Pediatr Nephrol. 2012;47:49–50. 161. Ho CI, Wen YK, Chen M. Glomerulonephritis with
148. Handoko ML, Duijvestein M, Scheepstra CG, de acute renal failure related to osteomyelitis. J Chin
Fijter CW. Syphilis: a reversible cause of nephrotic Med Assoc. 2008;71:315–7.
Immunoglobulin A Nephropathies in
Children (Includes HSP) 32
Koichi Nakanishi and Norishige Yoshikawa

Contents Laboratory Investigation . . . . . . . . . . . . . . . . . . . . . . . . . 1003


Biomarkers in IgAN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1004
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 984
Differential Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1005
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 984 Relationship Between IgAN and HSP . . . . . . . . . . . . . 1005
Etiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 985 Chronic Liver Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1005
Predisposing Genetic Factors . . . . . . . . . . . . . . . . . . . . . . . 986 Idiopathic Nephrotic Syndrome . . . . . . . . . . . . . . . . . . . . 1006

Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 987 Natural History and Prognosis . . . . . . . . . . . . . . . . . . . 1006


Nature of Mesangial Immunoglobulin A Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1008
Deposits . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 988 Fish Oil/Omega 3 Fatty Acids . . . . . . . . . . . . . . . . . . . . . . 1009
Immunoglobulin A Glycosylation . . . . . . . . . . . . . . . . . 990 Coagulation Modifying Agents . . . . . . . . . . . . . . . . . . . . 1009
Immunoglobulin A Immune System . . . . . . . . . . . . . . . 991 Angiotensin-Converting Enzyme Inhibitors
Multi-hit Mechanism for Pathogenesis of IgAN . . . 993 and Angiotensin II Receptor Blockers . . . . . . . . . . . . . 1009
MicroRNAs (miRNAs) and Pathogenesis Corticosteroids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1009
of IgAN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 993 Immunosuppressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1010
Progression Mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . 993 Clinical Trials for Combination Therapy
Initiation and Progression Mechanism of Including Corticosteroids and
Glomerular Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 993 Immunosuppressant in Japan . . . . . . . . . . . . . . . . . . . . . . . 1010
Progression Mechanisms from Glomerular Injury Chinese Herbal Medicine (Sairei-to) . . . . . . . . . . . . . . . 1011
to Tubulointerstitial Injury . . . . . . . . . . . . . . . . . . . . . . . . . . 995 Tonsillectomy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011
Renal Fibrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 996 Japanese Guidelines for the Treatment
of Childhood IgAN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011
Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 996
Immunohistologic Findings . . . . . . . . . . . . . . . . . . . . . . . . 996 Clinical Manifestations and Laboratory
Light Microscopic Findings . . . . . . . . . . . . . . . . . . . . . . . . 997 Features in HSP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011
Electron Microscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 998 Extrarenal Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . 1011
Repeat Renal Biopsy Findings . . . . . . . . . . . . . . . . . . . . . 999 Skin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1011
Differences Between Childhood and Adult Joints . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012
Patients with IgAN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1000 Gastrointestinal Tract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012
The Oxford Classification of IgAN . . . . . . . . . . . . . . . . 1000 Renal Manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1012
Laboratory Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1013
Pathology in HSN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1001 Clinicopathologic Correlations . . . . . . . . . . . . . . . . . . . . . 1013
Clinical Features in IGAN . . . . . . . . . . . . . . . . . . . . . . . . 1003 Differential Diagnosis of HSP . . . . . . . . . . . . . . . . . . . . 1014
Treatment of HSP . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1014
K. Nakanishi (*) • N. Yoshikawa Prognosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1015
Department of Pediatrics, Wakayama Medical University, Recurrence in Renal Allograft . . . . . . . . . . . . . . . . . . . . . . 1017
Wakayama City, Japan
e-mail: knakanis@wakayama-med.ac.jp; References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1017
nori@wakayama-med.ac.jp

# Springer-Verlag Berlin Heidelberg 2016 983


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_28
984 K. Nakanishi and N. Yoshikawa

Introduction Table 1 Immunopathologic similarities between IgA


nephropathy and Henoch-Schönlein nephritis
Immunoglobulin A (IgA) nephropathy (IgAN) IgA Henoch-Schönlein
was first described in 1968 by Berger and Hinglais nephropathy nephritis
[1] and is now recognized as a distinct clinico- Tissue deposition
Glomeruli IgA1, J chain, IgA1, J chain
pathologic entity with a higher frequency world-
anionic
wide than any other primary glomerulopathy [2].
Skin IgA IgA (includes
It was initially considered a benign condition, but uninvolved skin)
extended follow-up of patients indicated that Serum IgA
20–50 % of adults would ultimately progress to Total " (50–70 % of " 50 % (early in
end-stage renal failure [2, 3]. Likewise, the favor- cases) disease)
able prognosis initially attributed to children Polymer " "
with IgAN must be questioned in the light of IgA1 " "
studies [4–8]. Circulating IgA
Henoch-Schönlein purpura (HSP) is a clinical Immune " "
complexes
syndrome and multisystem disorder that affects
IgA Present Present
predominantly the skin, joints, gastrointestinal rheumatoid
tract, and kidneys, although other organs can be factors
involved [9–12]. The triad is not necessarily pre- IgA1 O- Abnormal Abnormal
sent in every case. glycosylation
In 1801, Heberden reported a 5-year-old boy Polymeric IgA1 production
with abdominal pain, vomiting, melena, joint Duodenal # ?
mucosa
pain, petechial hemorrhages on his legs, and
Marrow " ?
urine “tinged with blood.” In 1837, Schönlein Tonsil " ?
first described the condition that he called IgA clearance Impaired ?
“peliosis rheumatica,” purpura rheumatica, in IgA response to immunization
which arthralgia was associated with purpura. In Systemic Exaggerated ?
1868, Henoch described the gastrointestinal man- Mucosal Reduced ?
ifestations of purpura rheumatica, and 30 years Transplant 50 % Common
later, he referred to nephritis as a complication. recurrence
Osler attributed the symptoms to anaphylaxis
[13], and the term anaphylactoid purpura, intro-
duced by Frank in 1915, remains popular in the and/or proteinuria) [17]. Recently, the second
United States [14], whereas most European and International Chapel Hill Consensus Conference
Japanese writers favor the eponymous term. Other (CHCC2012) was convened to improve the
terms used include purpura rhumatoide [15] and CHCC1994 nomenclature [18, 19]. HSP was
Schönlein-Henoch syndrome [16], the latter replaced with IgA vasculitis in CHCC2012.
acknowledging historical precedence. However, Although IgA vasculitis is a more descriptive
HSP remains popular as well as being traditional term, it does not still come into wide use.
and is used in this chapter. There are many similarities between IgAN and
The European League against Rheumatism/ Henoch-Schönlein nephritis (HSN) (Table 1).
Pediatric Rheumatology European Society
(EULAR/PReS) proposed as classification criteria
for HSP the presence of palpable purpura (man- Epidemiology
datory criterion) and at least one of the following
features: diffuse abdominal pain, any biopsy IgAN has been diagnosed all over the world, but
showing predominant IgA deposits, arthritis or its prevalence varies widely from one country to
arthralgia, and renal involvement (any hematuria another. In the Pacific Rim (e.g., Japan,
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 985

Singapore, Australia, and New Zealand), IgAN during winter [15, 32, 23]. Although HSP has a
accounts for as many as one half of cases of wide geographic distribution, there is appreciable
primary glomerulonephritis. In Europe, it variation. Clinical reports based on records of
accounts for between 20 % and 30 % of all pri- large numbers of patients have originated mostly
mary glomerulonephritis, whereas in North from Europe, United Kingdom [26, 33–35],
America, it is responsible for only 2–10 %. The France [15], and Finland [36], and Asia, Japan
explanation for this apparent variability in inci- [32, 27] and Singapore [24]. The condition
dence is uncertain, but it may be due to a racial appears to be less common in North America
difference or to differences in biopsy selection and Africa. These differences may be both envi-
practices [20]. ronmental and racial, because HSP is rare among
Genetic factors and environmental influences blacks in the southern United States [37, 38] and
could contribute to geographic differences in among persons from the Indian subcontinent
prevalence. A lower prevalence among blacks [39]. A familial instance of HSP is rare [26, 40],
than whites has been reported in the United States. suggesting that genetic factors are not of etiologic
However, in American children, similar significance. There appears to be no association
incidences of IgAN in Caucasian and African- between HLA antigens [41] or immunoglobulin
American children from Shelby County, Tennes- heavy-chain switch-region genes [42] and HSP.
see, have been reported [21]. In Australia [22], The proportion of patients with renal involve-
where the population is heterogeneous and ment has been reported to be 20–100 % [26, 32,
includes many immigrants from Third World 23, 43–47]. These variations may depend on the
countries, all racial groups seem to be equally method of detection of nephritis [48]. In many of
affected. the early studies, serial routine urinalysis was not
The high incidence of IgAN in certain coun- used, and transient microscopic hematuria proba-
tries may reflect the practice of routine urinalysis. bly was missed. Two studies in which routine
In Japan, all children between the ages of 6 and urinalysis was performed for all patients admitted
18 years are screened annually, and those found to to the hospital for HSP showed renal involvement
have urinary abnormalities are referred for further in 41 % [45] and 61 % [32] of cases.
investigation. Thus, IgAN is the most common
primary glomerulonephritis in children seen in
Kobe University and Wakayama Medical Univer- Etiology
sity Hospitals, detected in approximately 30 % of
biopsy specimens. The pathogenesis of IgAN is not fully uncovered.
HSP occurs mainly among children, but adults IgAN is generally considered to be an immune
can be affected [23–25]. It is rare among children complex-mediated or aggregated (polymerized)
younger than 2 years and has a peak incidence IgA-mediated glomerulonephritis. Because IgA
around 4–5 years of age, followed by a gradual is the main immunoglobulin directed against anti-
decline toward adolescence. The larger childhood gens (viral and bacterial) in the exocrine system,
series indicate a slight male preponderance and because of the frequent association between
[26, 15, 27, 28]. The annual incidences estimated upper respiratory tract or gastrointestinal infection
by Nielsen [29] in Denmark and by Stewart and the onset of macroscopic hematuria, it has
et al. [30] in Ireland were 1.4 and 13.5 cases per been suggested that certain viral or bacterial infec-
100,000 children, respectively. Forty years ago, tions may lead to IgAN. Considerable effort has
HSP was less than half as common as poststrep- been directed toward the search for antigens and
tococcal nephritis in the United Kingdom [31], for the antibody specificity of the mesangial IgA,
but whereas the latter is now uncommon in devel- but with limited success. Many antigens, includ-
oped countries, the incidence of HSP seems to ing herpes simplex virus, cytomegalovirus,
have remained stable. There is significant sea- Epstein-Barr virus nuclear antigen, adenovirus,
sonal variation in incidence, the peak occurring and milk antigen, have been identified. Although
986 K. Nakanishi and N. Yoshikawa

much of this work has been performed in adults, streptococcal infection has not been substantiated
there is no evidence to suggest that the findings [26, 15, 56, 32]. Our findings suggest that
cannot be extrapolated to children. Haemophilus parainfluenzae may be involved
Evidence that IgAN is not a disease restricted for a subset of patients [52]. Other infections
to the kidney but is a systemic disorder is provided implicated for a few children include varicella
by the findings of IgA deposits in unaffected skin [26], measles [16], rubella [26], adenovirus infec-
[49], recurrent mesangial IgA deposits in 50 % of tion [57], hepatitis A [58] and B [59], Yersinia
transplant recipients whose primary disease has infection [60], Shigella infection [61], Myco-
been IgAN [50], and the disappearance of IgA plasma pneumoniae infection [62], and staphylo-
from cadaver kidneys found to have unexpected coccal septicemia [63], but without strong
IgA deposits when these are transplanted into evidence of a causal relation. HSP has been
recipients with other primary renal disease [49]. reported to follow smallpox and influenza vacci-
The observation of numerous antigenic sub- nation [64, 65], insect bites [66], exposure to cold
stances in the glomeruli indicates that the anti- [67], and trauma [68]. Among adult patients, HSP
genic materials in IgAN may be heterogeneous. has been reported in association with human
The presence of Haemophilus parainfluenzae immunodeficiency virus infection [69], Behcet
antigens in a diffuse and global distribution in disease [70], uveitis [71], squamous cell carci-
the glomerular mesangium and the presence noma [72], and rheumatoid arthritis [73].
of IgA antibody against Haemophilus Interestingly, it has been reported 10 of 33 chil-
parainfluenzae in sera of Japanese patients with dren (30 %) with HSN showed segmental or
IgAN have been demonstrated [51, 52]. It has global mesangial staining with nephritis-
been reported that Staphylococcus aureus cell associated plasmin receptor (NAPlr, a group A
envelope antigen was localized in the glomeruli Streptococcal [GAS] antigen) antibody, whereas
in 68.1 % (79/116) of renal biopsy specimens only 2 of 48 children (4 %) with IgAN were
from patients with IgAN and the antigen was positive [74]. These findings suggest that the
colocalized with IgA antibody in the glomeruli. deposition of NAPlr in the mesangium, induced
Staphylococcus aureus cell envelope antigen may by Group A Streptococcal infection, may have a
be a new candidate for the induction of IgAN role in the pathogenesis of HSN. Schmitt
[53]. These findings remain unconfirmed in chil- et al. [75] have reported that deposits of the
dren with IgAN yet. Schmitt et al. have reported IgA-binding region of three different streptococ-
that antibody levels to the IgA-binding regions cal M proteins were detected in 7/13 HSP
were significantly higher in IgAN patients than kidneys as well as 10/16 IgAN kidneys. The
controls, particularly in patients with recent strep- IgA-binding region would access the circulation,
tococcal infection, suggesting that some children encounter circulatory IgA, and form a complex
with IgAN had a previous infection with a strep- with IgA-Fc that could deposit in tissues and
tococcal strain expressing an IgA-binding M contribute to the pathogenesis of HSP as well as
protein [54]. IgAN [75].
In order to present an antigen to T cells, the
antigen must first be degraded by proteasomes.
Coppo et al. have reported that a significant switch Predisposing Genetic Factors
in the expression of trypsin- and chymotrypsin-
like proteasome subunits to corresponding Predisposing genetic factors have been suggested
immuno-proteasome subunits was found in as important in the development of IgAN [76].
patients with IgAN as compared to healthy Moreover, it has been suggested that
controls [55]. genetic factors may not only determine suscepti-
The cause of HSP also remains unknown. The bility to glomerulonephritis but also influence the
illness is preceded by upper respiratory infection pathological severity and natural course of IgAN
in 30–50 % of cases [26, 15], but evidence of [77, 78].
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 987

Evidence for genetic factors in IgAN is pro- acetylhydrolase gene [97], constant region of the
vided by family studies [77–81]. Rambausek alpha chain for the T-cell receptor [98], miR-146a
et al. reported that 9.6 % of patients with [99], core 1 beta1,3-galactosyltransferase
mesangial IgAN in Germany had one or more (C1GalT1) [100, 101], and so on [102] have
siblings with glomerulonephritis [77]. Julian been reported. Associations among IgAN and
et al. described kindred from eastern Kentucky genetic polymorphisms of other molecules, in
in which six patients with IgAN descended from which relation to disease susceptibility and dis-
one ancestor and eight other patients belonged to ease progression are suggested, have been
potentially related pedigrees [79]. Moreover, they reported in sequence [103]. Although some asso-
indicated that at least 48 (60 %) of 80 IgAN ciations have emerged, they have been inconsis-
patients who were born in same region were tent [104]. Some of the discrepancies may be
related to at least one other patient [80]. Scolarim caused by different sample sizes and different
et al. reported that 26 (14 %) of 185 patients with geographical regions of the patients included in
IgAN investigated in Italy were related to at least the studies [105].
one other patient with the disease [81]. These In Japan, single-nucleotide polymorphisms
studies suggest that familial predisposition is a have been screened on a genome-wide scale to
very common finding and genetic factors are clarify various complex diseases, including IgAN.
involved in the pathogenesis of IgAN. Gharavi As a part of the benefit of this project, case-control
et al. [82] demonstrated linkage of IgAN to association studies were designed, using single-
6q22-23 under a dominant model of transmission nucleotide polymorphisms found in the selectin
with incomplete penetrance. Further linkage- gene [106], the class II region of MHC [107], and
based studies have identified significant or sug- the polymeric immunoglobulin receptor gene
gestive loci on chromosomes 4q26-31, 17q12-22 [108], and haplotypes of these genes that might
[83] and 2q36 [84], but no causal gene has yet serve to identify regions containing loci responsi-
been identified. Genome-wide association studies ble for IgAN phenotypes were estimated.
have been performed in IgAN [85–87]. Five dis- Several publications have focused on genetic
tinct susceptibility loci including three indepen- factors predisposing to HSP, many of which are
dent loci in the major histocompatibility complex polymorphisms in genes related to cytokines and
(MHC) as well as a common deletion of the com- cell adhesion molecules that are involved in the
plement factor H gene cluster (CFHR1 and pathways for inflammatory responses and endo-
CFHR3) at chromosome 1q32 and a locus at chro- thelial cell activation [109]. The associations of
mosome 22q12 were identified [86]. Another HLA-DRB1 01, 07, and 11 with HSP susceptibil-
study identified associations at 17p13 and 8p23 ity are reported [110]. Genetic regulation of endo-
that implicated the genes encoding tumor necrosis thelial function, such as polymorphisms in genes
factor (TNFSF13) and α-defensin (DEFA) as sus- coding RAS components [111], endothelial nitric
ceptibility genes. In addition, the study has found oxide synthases [112], intercellular adhesion mol-
multiple associations in MHC region and con- ecule 1 (ICAM1) [113], and vascular endothelial
firmed a previously reported association at growth factor (VEGF) [114], could also confer
22q12 [87]. effect on HSP. In addition, MEFV, whose muta-
Schena et al. [88] reported that familial IgAN tions cause familial Mediterranean fever, could be
had a poorer prognosis than sporadic IgAN, a candidate gene for HSP [115–118].
suggesting that genetic factors are implicated in
both disease susceptibility and disease progres-
sion [76]. Polymorphisms of Ig heavy-chain Pathogenesis
switch-region gene [89], Iα1 germ-line transcript
regulatory region gene [90], genes of the renin- Although the pathogenesis of IgAN remains
angiotensin system (RAS) [91–95], transforming uncertain, there is substantial evidence that it is
growth factor-beta1 [96], platelet activating factor an immune complex disease [2, 119]. Granular
988 K. Nakanishi and N. Yoshikawa

electron-dense deposits are observed in the glo- Nature of Mesangial Immunoglobulin A


merular mesangial areas by electron microscopy Deposits
and contain IgA and C3 by immunofluorescence
microscopy. Circulating IgA immune complexes IgA is the second most common immunoglobulin
have been detected by several different assays and contributes to immunity at the level of the
often associated with IgG immune complex. external secretory system. IgA exists in mono-
Many immunological abnormalities that may meric and polymeric forms. Monomeric IgA rep-
lead to the formation of IgA immune complex resents approximately 90 % of the serum IgA and
have been reported in patients with IgAN. is produced mainly by the circulating lympho-
Recurrence of IgAN frequently occurs in allo- cytes and plasma cells in the spleen and bone
grafts [120], and a rapid disappearance of glomer- marrow. Polymeric IgA is produced mostly by
ular IgA deposits is observed when kidneys with lymphocytes and plasma cells in the gastrointes-
mesangial IgA deposits are transplanted to tinal and respiratory tracts, where it is synthesized
patients without IgAN. These clinical observa- as monomers and then secreted as dimers linked
tions have provided strong support for the notion by the J-chain, which is also produced within the
that IgAN is a systemic disease. Although much plasma cells. During the passage of dimeric IgA
of this work was performed in adults, there is no molecules through the mucosal epithelium toward
evidence to suggest that the findings cannot be the external lumen, the secretory component is
extrapolated to children. Moreover, glomerular attached through specific noncovalent interac-
IgA deposits associated with histologic lesions tions; this component appears to protect the
similar to those of human IgAN can be induced dimeric IgA from the proteolytic enzymes present
in laboratory animals by passive administration of in the external secretions. IgA has two subclasses,
preformed IgA immune complex or by active IgA1 and IgA2. Approximately 90 % of serum
immunization [121–125]. IgA is composed of IgA1 mostly produced in the
Although the pathogenesis of HSP also bone marrow, whereas IgA2 is mostly derived
remains uncertain, there is substantial evidence from the local mucosa of the gastrointestinal and
that it is also an immune complex-mediated dis- respiratory tracts. Both IgA1 and IgA2 are pro-
ease. The mechanisms of formation of glomerular duced in the mucosae.
immune deposits likely are similar to those in The most prominent finding in the glomeruli of
IgAN, generally regarded as a kidney-limited var- renal biopsy specimens from patients with IgAN
iant of HSP. Granular electron-dense deposits in and HSN is mesangial IgA deposition. The major-
the glomerular mesangial areas are observed with ity of investigators have indicated that IgA1 is the
an electron microscope and contain IgA and C3 predominant subclass present in the glomeruli
by immunofluorescence microscopy. Circulating [133]. The J-chain has also been identified in the
IgA immune complexes have been detected, often mesangium of patients with IgAN [134]. The
associated with IgG immune complexes secretory component is not present in the
[126–128, 49]. Many immunologic abnormalities mesangial deposits, but it binds to the mesangial
that lead to formation of IgA immune complexes areas in vitro [135, 136]. These observations sug-
have been found in patients with HSP. Mesangial gest that the mesangial IgA deposits are poly-
IgA deposits recur frequently in allografted kid- meric, a hypothesis further supported by the
neys of patients with HSP [129–131]. This evi- immunochemical characterization of IgA eluted
dence indicates that the mesangial IgA must be of from renal biopsy sections [137].
host origin. Moreover, glomerular IgA deposits Most investigators have found that IgA1 is the
associated with histologic lesions similar to predominant subclass in the glomeruli of patients
those of human HSN can be induced among lab- with HSN [133, 138–140]. J-chain, independent
oratory animals by means of passive administra- of IgM, has been found in the mesangium of
tion of preformed polymeric IgA-concanavalin A patients with HSN [133, 141]. Secretory compo-
complexes [132]. nent is not present in the mesangial deposits
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 989

[139]. Although the capability of the mesangial mediated by IgA deposits in the glomeruli, and
deposits to bind the secretory component has not the mediator and the pathophysiologic signifi-
been examined in HSN, the mesangial IgA cance of this complement activation remain to be
deposits in HSN are also thought to be polymeric. determined.
The onset of IgAN may be associated with With regard to antibody specificity, IgA eluted
infections in the upper respiratory tract. It has from cryostat sections of IgAN biopsies has been
therefore been proposed that IgAN results from reported to react with mesangial areas of its own
hyperactivity of the mucosal immune system. and other IgAN patients’ biopsies, but not with
However, assessment of polymeric IgA1 produc- normal kidney [154]. Such eluates have also been
tion by in situ hybridization for J-chain messenger shown to contain antibodies that react with tonsil-
RNA in IgA plasma cells shows downregulation lar cells and cultured fibroblasts obtained from
in the mucosa [142] and upregulation in the bone patients with IgAN [155].
marrow [143]. Impaired mucosal IgA responses Because IgA is the main immunoglobulin
allowing enhanced antigen challenge to the mar- directed against viral and bacterial antigens in
row shown by de Fijter et al. [144] could be the the exocrine system, and because of the frequent
primary abnormality in IgAN, although this association between upper respiratory tract infec-
remains unproven [10]. tion and the development of HSP, it has been
A number of studies have suggested that the suggested that certain viral or bacterial infections
alternative complement pathway has a pathoge- may cause HSP and that IgA may be the antibody
netic role in IgAN and HSN. This hypothesis is to viral or bacterial antigens. Little effort has been
consistent with the typical immunohistologic directed at the search for antigens and for the
demonstration of C3 and properdin in a pattern antibody specificity of the mesangial IgA in
and distribution similar to that of IgA in the glo- HSP. Diffuse global deposition of
meruli and the absence of C1q and C4. The detec- H. parainfluenzae was found in one third of
tion of the membrane attack complex of patients with IgAN and HSN [52]. Patients with
complement further supports the role of comple- IgAN and HSN had significantly higher levels of
ment activation in this disease [145, 15, 24, 146]. plasma IgA1 antibody against H. parainfluenzae
Certain types of IgA aggregates or IgA from than did patients with other renal diseases
patients with myeloma have been shown to acti- [52]. Emancipator [156] described the develop-
vate complement in vitro [147]. IgA has been ment of intestinal lesions, purpura, glomerular
reported to activate the complement system via lesions, and hematuria in a bacterial polysaccha-
the mannan-binding lectin pathway [148]. How- ride model induced in mice and suggested that
ever, there is no direct evidence that IgA deposits HSP may be an expression of an antigen-
in the glomeruli mediate complement activation. dependent process. With regard to antibody spec-
Activation of C3 is observed in the majority of ificity, IgA eluted from cryostat sections of HSN
adult and pediatric patients with IgAN, but the obtained at biopsy has been reported to react with
mediator as well as the pathophysiologic signifi- mesangial areas of these biopsy specimens and
cance of this complement activation remains to be with biopsy specimens from other patients with
determined. C3 is deposited in the kidney but is HSN [157, 158].
also produced by mesangial cells in IgAN [149]. Despite intensive investigation, the mechanism
Detection of the membrane attack complex of underlying glomerular IgA deposition in IgAN
complement further supports the role of comple- has not been elucidated. Proposed possible mech-
ment activation in HSN [150]. The alternative anisms of glomerular IgA deposition are as fol-
complement pathway is activated in patients lows. Decreased IgA response to mucosal
with HSN [151, 152], and increased serum termi- antigens may promote increased production of
nal complement complex has been found during polymeric IgA1 by the bone marrow, leading to
the active phase of disease [153]. However, there increased serum levels of IgA1. Defective
is no direct evidence that the activation is galactosylation of IgA1 (later described in detail)
990 K. Nakanishi and N. Yoshikawa

may decrease hepatic clearance of IgA1 and pro- there is no proof that this abnormality directly
mote binding of IgA1 complexes to glomerular promotes mesangial IgA1 deposition. IgA1 is
mesangial cells. Aberrant IgA1 deposits in the unique among all Igs in its possession of a hinge
kidney trigger the production of a variety of cyto- region rich in proline, serine, and threonine and
kines and growth factors by renal cells and by characterized by O-glycosylation sites (Fig. 1).
circulating inflammatory cells, leading to the char- These O-glycosylation sites consist of N-acetylga-
acteristic histopathological features of mesangial lactosamine O-linked to the serine or threonine
cell proliferation and extracellular matrix deposi- residues of hinge region. In patients with IgAN,
tion [159]. Recently it has been proposed a model mesangial IgA1 and a small fraction of circulating
whereby two types of IgA receptors participate in IgA1 contain incompletely galactosylated
sequential steps to promote the development of O-linked glycans in the hinge region
IgAN, with soluble IgA-Fc receptor I (FcαRI/ [169–173]. This glycosylation abnormality is
CD89) being initially involved in the formation absent in other glycoproteins with O-linked gly-
and/or amplification of the size of circulating IgA cans, such as complement C1 inhibitor and IgD,
immune complexes and, subsequently, mesangial indicating a defect specific for IgA1 [171,
transferrin receptor (CD71), in mediating 174]. Abnormal galactosylated IgA1 increases
mesangial deposition of nephritogenic IgA affinity for glomerular fibronectin, laminin, and
immune complexes [160–164]. collagen IV [175] and may lead to accumulation
An animal study has demonstrated that of IgA in the mesangium [172]. Preliminary data
tetraspanin (CD37) controls the formation of indicate that deficient galactosylation of hinge
IgA-containing immune complexes and glomeru- region glycans may be detected even in family
lar IgA deposition, which induces influx of members of patients with IgAN [169]. Altered
inflammatory myeloid cells. Therefore, CD37 amino acid sequence of the IgA1 hinge region is
may protect against the development of a possible mechanism to consider for abnormal
IgAN [165]. galactosylation of IgA1. However, the hinge
In summary, the immunochemical nature of the region is a highly conserved region of IgA1 mol-
mesangial deposits in IgAN and HSN is consistent ecule. There is no evidence for any nucleotide
with antigen-polymeric IgA complexes predomi- sequence alteration or transcriptional abnormality
nantly of IgA1 subclass, and perhaps, of the hinge region in IgAN [176]. It has also been
multispecific for ubiquitous mucosally derived postulated that altered galactosylated IgA1 in
antigens. IgAN may be due to a deficiency of structural
modification of β 1,3-galactosyltransferase, the
enzyme responsible for the terminal
Immunoglobulin A Glycosylation galactosylation of GalNAc on O-linked glycans
[177]. This structural or functional deficiency may
IgA glycosylation has received recent attention as be genetically determined. The recent sequence of
a putative nonimmune feature of IgA, which may β1,3-galactosyltransferase may help us to under-
explain its abnormal behavior and glomerular stand the genetic basis of these abnormalities
deposition in IgAN [166] and HSN [167, 168]. [178, 179]. Circulating IgA1 has reduced terminal
IgA is a heavily glycosylated protein. IgA1 carries galactose on O-linked hinge region sugars in
sugars at the hinge region, which is most unusual IgAN [171], apparently because of a B-cell defect
in a circulating protein. IgA2 has no hinge region. in β1,3-galactosyltransferase, the enzyme respon-
Changes in these hinge region sugars may have sible for placing terminal galactose on O-linked
functional effects on the interaction of IgA1 with sugars [177]. The IgA1 O-glycan chains are
specific antigen and with effector mechanisms truncated in IgAN [180]. Circulating immune
such as complement and Fc receptors. Circulating complexes in IgAN consist of IgA1 with
IgA1s have a decreased amount of terminal galac- galactose-deficient hinge region, and the defi-
tose in the hinge region in IgAN and HSN. As yet ciency of galactose may result in the generation
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 991

CH1 β1,3 - Gal - α2,3 - Sialic acid


I
Pro
I
Ser←O-glycan → Ser/Thr -O- GalNAc
I
Thr ←O-glycan
I
Pro α2,6 - Sialic acid
I

Light Heavy Pro Normal


I
chain chain Thr ←O-glycan
I
Pro
I
CH1 Ser←O-glycan
I Ser/Thr-O-GalNAc
Hinge Pro
I
region Ser←O-glycan
I α2,6 -Sialic acid
Thr ←O-glycan
CH2 I
Gal-deficient O-linked glycan
Pro
I
Pro
I
CH3 Thr ←O-glycan
I
Pro
I
Ser←O-glycan
I
Pro
I
Ser←O-glycan
I
CH2

Fig. 1 IgA1 molecules with hinge region O-glycosylation O-linked to Ser or Thr residues of hinge region. The largest
sites. IgA1 molecules have two heavy chains with three O-linked saccharide of IgA1 is a tetrasaccharide with
constant region domains CH1 to CH3 and a hinge region GalNAc, galactose (Gal), and two sialic acid residues.
between CH1 and CH2. Each serine (Ser) and threonine Aberrant IgA1 molecules with Gal-deficient O-linked gly-
(Thr) residue is a potential site for an O-glycan side chain. cans can contain terminal GalNAc or sialylated GalNAc at
Although there are nine potential glycosylation sites, IgA1 one or more sites. Pro proline
contains up to 6 O-glycans per hinge region. O-glycosyl-
ation sites consist of N-acetylgalactosamine (GalNAc)

of antigenic determinants that are recognized by glycosylation clusters in most but not all families
naturally occurring IgG and IgA1 antibodies with IgAN suggest the possibility of IgAN
[170]. Sano et al. demonstrated that enzymatically patients with different pathogenic mechanisms of
deglycosylated human IgA1 molecules accumu- disease [184].
late and induced inflammatory cell reaction [181];
Amore et al. showed glycosylation of circulating
IgA-modulated mesangial proliferation in Immunoglobulin A Immune System
IgAN [182].
It has been showed that EBV-immortalized Serum IgA levels are increased in 50–70% of
IgA1-producing cells from peripheral blood cells patients with IgAN, with elevations in both mono-
in IgAN patients secreted mostly polymeric IgA1 meric and polymeric IgA. There is an increase in
with galactose-deficient O-linked glycans [183]. polymeric IgA1-producing plasma cells in the
Although it had not been previously known bone marrow [143] and in the tonsils [185] of
whether the aberrant glycosylation is the result patients with IgAN. The proportion of IgA-λ in
of an acquired or inherited defect or whether the serum IgA is also increased. Serum IgA is more
presence of aberrant IgA1 glycoforms alone can anionic, owing to the increased anionicity of
produce IgAN, to date studies have demonstrated λ-light chain compared with κ-light chain [185].
that abnormal IgA1 glycosylation is an inherited The binding of IgA to mesangial cells is charge
rather than acquired trait and that abnormal IgA1 dependent, and anionic charge may play an
992 K. Nakanishi and N. Yoshikawa

important role for in IgA1 deposition in the [205]. The presence of IgA rheumatoid factors has
mesangium [186]. In addition to the increased been related to disease activity [28]. IgA immune
levels of serum IgA, various types of autoanti- complexes are also frequently detected [126–128,
bodies of the IgA class have been recognized. 49]. Coppo et al. [206] found a high prevalence of
These IgA autoantibodies include rheumatoid fac- immune complexes during acute disease.
tor [187], antinuclear antibodies [188], and Levinsky and Barratt [126] found that immune
anticollagen antibody [189]. However the IgA complexes also contained IgG, and only the
may be polyspecific, indicating a polyclonal IgG-containing immune complexes were
increase rather than true antigen-specific autoan- nephritogenic in HSP. Codeposition of IgG in
tibodies [190]. IgA immune complexes are also the mesangium and the presence of circulating
frequently detected [191]. Cultured peripheral IgG-containing immune complexes suggested
blood lymphocytes from patients with the disease that IgG immune complexes may have an impor-
produce more IgA than those of normal individ- tant role in renal damage [207]. Not all patients
uals, either spontaneously or after polyclonal with HSP have circulating immune complexes.
stimulation in vitro. We also demonstrated an There are at least two explanations for the absence
increased spontaneous and pokeweed mitogen- of circulating immune complexes. These patients
stimulated IgA production by peripheral blood may have immune complexes only intermittently,
lymphocytes of children with IgAN [192]. This or formation of mesangial IgA deposits in patients
increased IgA production remained stable during without circulating immune complexes may be
the follow-up period in patients with persistent caused by in situ formation of the immune com-
urinary abnormalities, but decreases toward nor- plex directly in the mesangium. O’Donoghue
mal in patients with clinical remission. et al. [208] found IgG autoantibody binding
IgA production is T cell dependent, and the specificality to glomerular antigens present on
increased production in IgAN may indicate the mesangial cell only in IgAN and HSP. Circu-
altered T-cell function. An increased circulating lating IgA-fibronectin aggregates, macromole-
OKT4 to OKT8 cell ratio, due to increased OKT4 cules aggregated on a nonimmune basis, have
helper T lymphocytes and decreased OKT8 sup- been found in patients with HSP [209, 210]. The
pressor T lymphocytes, has been reported in presence of fibronectin in the circulating aggre-
patients [193]. Increased IgA-specific helper gates may have an important role in preferential
T-cell activity and decreased IgA-specific sup- deposition of nephritogenic IgA-containing
pressor T-cell activity have also been reported immune complexes in the mesangium [209].
[194, 195]. A large number of circulating IgA-producing
Defective clearance of immune complexes lymphocytes have been found during the acute
from the circulation may also be important phase of HSP [211–213], but not when the disease
[196], but this seems more likely to be a conse- is clinically inactive [213, 214]. Production of IgA
quence rather than the cause of the increased is highly T cell dependent, and the increased pro-
immune complex load. duction in HSP may indicate altered T-cell func-
Serum IgA levels are increased in one half tion. However, Casanueva et al. [213] found the
of patients with HSP during acute illness existence of persistent abnormalities in the cellu-
[15, 197, 198]. Serum IgA levels return to normal lar immune mechanisms that regulate immuno-
with recovery. In addition, various types of auto- globulin synthesis (impaired T-cell suppressor
antibodies of the IgA class have been found in activity) in IgAN, but not in HSP. Defective clear-
HSN. These autoantibodies include IgA rheuma- ance of immune complexes from the circulation
toid factor [199–201], IgA antineutrophil cyto- may be important, but this seems more likely to be
plasmic antibody [202, 203], IgA anti-IgG a consequence rather than the cause of the
antibody [204], and IgA anticardiolipin antibody increased immune complex load.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 993

Multi-hit Mechanism for Pathogenesis IgAN [220]. MiRNAs are important mediators of
of IgAN tissue fibrosis under various pathological condi-
tions and are of potential therapeutic relevance
A multi-hit mechanism for the pathogenesis of [221]. Although to date, limited data are available
IgAN has been proposed [215]. A high circulating on the role of miRNAs in the pathogenesis of
load of galactose-deficient IgA1 (Hit 1) alone does IgAN, miRNAs may have important roles in the
not induce the renal injury. Rather, several sequen- pathogenesis and progression of IgAN [220,
tial processes or hits are necessary for the clinical 222–227, 221, 228–230]. Some alterations seem
expression of IgAN. Synthesis and binding of anti- to be disease specific, whereas others are appar-
bodies directed against galactose-deficient IgA1 ently damage related [220]. As miRNAs in uri-
are required for formation of immune complexes nary sediment are relatively stable and easily
that accumulate in the glomerular mesangium (hits quantified, they have the potential to be used as
2 and 3). The detail is as follows [215]. Hit 1: biomarkers for the diagnosis and monitoring of
Production of galactose-deficient IgA1 by a sub- disease [220].
population of IgA1-secreting cells. IgA1 produc- A number of miRNAs are involved in the
tion may be affected by the IgAN-associated locus development of fibrosis and progression of
on chromosome 22q12.2 [86]. Hit 2: Formation of chronic kidney disease (CKD) in general [220].
anti-glycan antibodies with specific characteristics For example, miR21 [231], miR192 [232, 233],
of the variable region of the heavy chain that rec- miR29a [234, 235], miR377 [236], and miR377
ognize galactose-deficient IgA1. Hit 3: Formation [237] were reported. Although these miRNAs
of immune complexes from autoantigen may have a role in IgAN, none of them,
(galactose-deficient IgA1) and O-glycan-specific however, has been specifically studied in the
antibodies. Hits 2 and 3 may be regulated by the disease [220].
three MHC loci on chromosome 6p21 associated
with risk of IgAN [86]. Hit 4: Deposition of path-
ogenic immune complexes in the mesangium, acti- Progression Mechanism
vation of mesangial cells, and induction of
glomerular injury. Hits 3 and 4 may be affected Renal fibrosis is the final common manifestation
by genotype at the complement factor H locus on of CKDs [238]. There is little to suggest that the
chromosome 1q32 that regulates the alternative mechanisms of mesangial proliferative glomeru-
complement cascade [86]. The first pathway lonephritis, progression, and scarring are distinct
assumes formation of immune complexes in the in IgAN compared with other types of chronic
circulation and their subsequent mesangial deposi- glomerulonephritis. A simplified scheme of pro-
tion [216, 170, 217, 218]. An alternative theory gression mechanism in IgAN is shown in Fig. 2.
proposes that some of the aberrantly glycosylated
IgA1 molecules are in the mesangium as lanthanic
deposits and are later bound by newly generated Initiation and Progression Mechanism
anti-glycan antibodies to form immune complexes of Glomerular Injury
in situ that activate mesangial cells [219].
As described above, deposition of polymeric IgA,
but not of monomeric IgA, can induce the produc-
MicroRNAs (miRNAs) and Pathogenesis tion and local release of a variety of cytokines,
of IgAN growth factors, complements, and angiotensin II
by renal resident cells and by circulating inflam-
Accumulating evidence indicates that miRNAs matory cells leading to inflammatory injury, char-
have a role in many human diseases, including acteristic histopathological features of mesangial
994 K. Nakanishi and N. Yoshikawa

Glomerular injury
Events TGF-b /Smad
PDGF
Glomerular IgA1 deposition ↑Cytokines/growth factors Angiotensin II
↑Complements Interleukins
Reactive oxygen species
Mesangial activation Inflammatory cell infiltration MicroRNAs

Proteinuria
Tubulo-interstitial injury
Mesangial cell proliferation Glomerulotubular cross-talk
Fibroblast activation
Podocyte depletion Epithelial to mesenchymal transition

Imbalance of apoptosis

Mesangial-matrix expansion Imbalance of AT1R and AT2R

Extracellular-matrix deposition

Glomerulosclerosis Renal fibrosis Tubulo-interstitial fibrosis

End-stage renal disease

Fig. 2 A simplified scheme of progression mechanism in IgAN

cell proliferation, and extracellular matrix deposi- Studies in children with IgAN suggest that
tion [239–242]. IgA alone also appears to be mesangial proliferation may in part be the result
sufficient to provoke injury in susceptible individ- of local production of cytokines, interleukin-1,
uals [243]. Studies in vitro and in animal models interleukin-6, tumor necrosis factor (TNF),
of mesangial proliferative glomerulonephritis PDGF, TGF-β, and vascular permeability
have shown the key role of cytokines and growth factor/endothelial growth factor (VPF/VEGF)
factors, particularly platelet-derived growth factor [249–251]. C3 deposits in mesangium and activa-
(PDGF) and TGF-β, in the induction and progres- tion of complement proteins in mesangium [252]
sion of mesangial injury, and there is evidence that and excessive oxidant stress [253] may mediate
these are also involved in IgAN [244–246]. It has glomerular injury in IgAN.
been reported that the MAPK/ERK signaling Glomerular injury and proteinuria in IgAN are
pathway was activated in the mesangium of related to the degree of podocyte depletion in
patients presenting with heavy proteinuria [247]. humans [254]. Recent study has supported for
IgA1-dependent ERK activation required RAS, the concept that podocyte depletion could be a
and ERK activation alters mesangial cell major mechanism driving glomerulosclerosis in
podocyte cross talk, leading to renal dysfunction human glomerular diseases [255]. Dysregulation
in IgAN [247]. Although the role of infiltrating of apoptosis may have the important role in
activated monocyte/macrophage into glomeruli is podocyte depletion. It has been shown that
also thought to be important in glomerular downregulation of Bcl-2 by podocytes is associ-
injury [248], it has been shown that resident glo- ated with progressive glomerular injury and clin-
merular cells (mesangial and endothelial cells) are ical indices of poor renal prognosis in human
predominantly the major source of upregulated IgAN [256]. Recently, it has been reported that
growth factor production in IgAN [244]. IgA1 from IgAN patients may induce apoptosis of
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 995

podocytes through direct and indirect pathways activation, normal T cell expressed and secreted),
and that IgA1 may accelerate progression of IgAN an immunoregulatory cytokine with chemotactic
by inducing apoptosis of podocytes [257]. properties for monocytes and memory T cells in
It has been showed that enhanced gene expres- culture proximal tubular cells [263]. Albumin is
sion for RAS is detected in glomerular mesangial also a strong stimulus for tubular interleukin-
cells in IgAN [258]. Recent studies have demon- 8 expression, which occurs with nuclear factor-
strated an altered angiotensin II subtype 1 receptor kappaB-dependent pathways [262]. Consistent
expression in human mesangial cells in response with these in vitro studies, vasoactive and
to raised intrarenal angiotensin II in IgAN. In vitro proinflammatory molecules were enhanced in
studies have also support that an imbalance of renal tissue from rat models with proteinuric
angiotensin II subtypes 1 and 2 receptor activity renal disease, particularly at proximal tubular
in human mesangial cells following exposure to level [264, 265].
polymeric IgA plays a significant pathogenetic Activation of complement proteins in the prox-
role in the inflammatory injury in IgAN [259]. imal tubule has major proinflammatory potential
in interstitial damage with proteinuric conditions.
Intracellular C3 staining was detected in proximal
Progression Mechanisms from tubules of proteinuric rats with remnant kidneys
Glomerular Injury to Tubulointerstitial early after 5/6 nephrectomy and preceded the
Injury appearance of inflammation. C3 colocalized with
IgG to the same tubular cells [266]. Induction of
It remains in part unclear how mesangial IgA tubular C3 during protein overload has also been
deposition leads to tubulointerstitial injury in reported [267].
IgAN. Several mechanisms of tubulointerstitial Excessive protein reabsorption by proximal
injury may operate independently or synergisti- tubular cells promotes fibrogenesis by release
cally [242]. Recently, a glomerulotubular cross- of chemoattractants, which leads to local recruit-
talk has been proposed in addition to monocytic/ ment of mononuclear cells. Interstitial accumula-
macrophage infiltration, proteinuria, complement tion of inflammatory cells by release of TGF-β,
activation, and direct inflammatory effect of IgA PDGF, and other cytokines leads to interstitial
[260]. The importance of infiltrating inflamma- cell transformation into myofibroblasts. In addi-
tory cells in the tubulointerstitium in mediating tion, proximal tubular epithelial cells interact
tubular injury and renal fibrosis in IgAN has been with surrounding interstitial fibroblasts to pro-
demonstrated [261, 242]. Especially, recent atten- mote fibrogenesis by paracrine release of
tion has been focused on the role of proximal profibrogenic molecules such as TGF-β, PDGF,
tubular epithelial cells in organization inflamma- and endothelin-1 [268].
tory cell infiltration and renal fibrosis via produc- The other possible contributory factor is the
tion of inflammatory mediators upon direct toxic effect following tubular binding of
activation [242]. IgA. IgAN patients have increased urinary IgA
Proteinuria is the major stimulus of proximal concentration that correlates with serum creati-
tubular epithelial cell activation and subsequent nine concentration, as well as the urinary protein
chemotaxis of infiltrating immunocompetent cells excretion [269].
in most glomerular diseases [262]. Endothelin-1 A glomerulotubular cross-talk, a mechanism in
synthesis was enhanced in culture proximal tubu- which mesangial IgA deposition may lead to
lar cells exposed to high concentrations of albu- tubulointerstitial injury in IgAN, has been pro-
min, IgG, or transferrin. Similarly, monocyte posed. It has been documented that inflammatory
chemoattractant protein (MCP-1) gene was cytokines, including angiotensin II, are released
upregulated by albumin and transferrin. Very sim- from mesangial cells following binding to IgA
ilar findings were reported for albumin-induced from patients with IgAN. These mediators may
upregulation of RANTES (regulated upon alter the glomerular barrier pore size that allows
996 K. Nakanishi and N. Yoshikawa

the passage of these inflammatory mediators to Pathology


the tubular lumen. These mediators then activate
proximal tubular epithelial cells, which may Immunohistologic Findings
amplify the inflammatory cascade by local pro-
duction of chemotactic mediators, which attract The diagnostic immunopathological pattern of
more inflammatory cells. This glomerulotubular IgAN is the presence of IgA in the glomerular
cross-talk will generate a positive feedback loop mesangium as the sole or predominant Ig. IgA
of activation in the renal tubules that leads to deposits often extend just beyond the mesangio-
the overproduction of extracellular matrix capillary junctions into the adjacent capillary
components, resulting in fibrosis. This hypothesis walls (Fig. 3). There are also deposits of IgG
was tested by conducting an experiment in and/or IgM with the same staining pattern as IgA
which proximal tubular epithelial cells were cul- but with lesser intensity and frequency. In our
tured with medium prepared from mesangial cells series, mesangial IgA deposits were associated
incubated with IgA from IgAN patients [260]. with IgG in 32 % of patients, IgM in 8 %, and
Contrary to the absent stimulatory effect on both IgG and IgM in 11 % [275]. C3 deposits were
proximal tubular epithelial cells upon direct incu- observed in a similar distribution pattern in 64 %
bation with IgA, increased proliferation and of cases. The early components of the classical
enhanced expression of inflammatory mediators complement pathway, C4 or C1q, are absent.
(including interleukin-6, TNF-α, soluble Fibrin- or fibrinogen-related antigens are found
ICAM-1, and angiotensin II) in proximal tubular in a diffuse mesangial distribution in 25–70 % of
epithelial cells cultured with medium prepared patients and are believed to be one of the injurious
from mesangial cells incubated with IgA from agents in the glomeruli [276]. Although, in most
IgAN patients were observed. Tubular and patients, IgA is present only in the mesangial
interstitial ICAM-1-positive cells may regions, in approximately 10 % of patients it is
participate in adhesive interactions with also observed in the peripheral capillary walls.
interstitial leukocytes [270, 271]. Upregulation Such peripheral capillary wall deposits, whether
of renal interleukin-6 expression correlates well documented by immunofluorescence or electron
to the degree of tubulointerstitial damage in microscopy, have been associated with more
IgAN [272].

Renal Fibrosis

Glomerulosclerosis, tubulointerstitial fibrosis,


inflammatory infiltration, and loss of renal paren-
chyma characterized by tubular atrophy, capillary
loss, and podocyte depletion are constituent path-
ologic findings of renal fibrosis [238]. The cellular
events leading to these histologic findings include
mesangial and fibroblast activation, tubular EMT,
monocyte/macrophage/T-cell infiltration, and
apoptosis [238]. At present, renal fibrogenesis
process is thought to be similar to wound healing
response to injury [273, 274, 238]. The reason
why the difference between a healthy wound
Fig. 3 Immunofluorescence micrograph showing
healing and fibrotic response occurs remains mesangial IgA deposits in a patients with IgAN. Original
unknown. magnification,  400
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 997

Fig. 4 Light micrograph showing mesangial proliferation (c) The increase in matrix is more prominent than the
in patients with IgAN. Four types of mesangial change are mesangial cellularity. (d) Progression of IgAN is associ-
identified: (a) Mesangial hypercellularity is more promi- ated with the development of global glomerulosclerosis
nent than the increase in matrix. (b) The degrees of accompanied by reactive cell invasion. Original magnifi-
mesangial hypercellularity and matrix increase are similar. cation,  400

severe clinical manifestations and a poor renal 1. Minimal glomerular lesions. The majority of
outcome [277–280]. glomeruli appear optically normal, although a
few may show a slight increase of mesangial
matrix, with or without accompanying
Light Microscopic Findings hypercellularity. The number of mesangial
cells per peripheral mesangial area does not
Various glomerular changes are observed. The exceed three. There are also small foci of tubu-
most characteristic abnormality is mesangial lar atrophy and interstitial lymphocyte infiltra-
enlargement, caused by various combinations of tion in some patients.
hypercellularity and increase in matrix (Fig. 4). 2. Focal mesangial proliferation. Up to 80 % of
Occasionally, small eosinophilic and glomeruli show moderate or severe mesangial
PAS-positive fibrinoid mesangial deposits are cell proliferation (i.e., more than three cells per
also seen. Biopsies can be graded according to peripheral mesangial area). The degree of
the amount of mesangial cell proliferation on mesangial cell proliferation varies consider-
the basis of the World Health Organization ably among glomeruli as well as segmentally
criteria [281]: within individual glomeruli. The proliferation
998 K. Nakanishi and N. Yoshikawa

is usually associated with increased matrix. hypercellularity is characteristic of the early lesion
Small cellular or fibrocellular crescents are fre- of childhood IgAN and may disappear within a
quently found but rarely affect more than 20 % matter of months. An increase in mesangial cells,
of the glomeruli. Capsular adhesions are fre- although sometimes present, is seldom striking in
quently seen overlying lobules showing adult patients [282]. In contrast, biopsies with a
mesangial proliferation. Segmental capillary predominant matrix increase show a long interval
collapse is often observed in association with between onset of disease and biopsy and a high
crescents. A small number of glomeruli show- percentage of glomerular sclerosis. Serial patho-
ing global sclerosis are often present. Tubular logic observations reveal that this type of change
atrophy, interstitial fibrosis, and interstitial is usually seen in follow-up biopsies. An increase
lymphocyte infiltration are frequently present in the amount of mesangial matrix with duration
but are not extensive. of the disease has also been noted in adult patients
3. Diffuse mesangial proliferation. More than [283]. These findings suggest that progression of
80 % of glomeruli show moderate or severe IgAN leads to gradual resolution of mesangial
mesangial cell proliferation, which varies in hypercellularity and an increase of matrix associ-
intensity in different regions of the mesangium ated with the development of sclerosis [284].
in a given glomerulus as well as from one The severity of tubulointerstitial changes usu-
glomerulus to another. Mesangial cell prolifer- ally reflects the severity of glomerular damage.
ation is always accompanied by increased Vascular lesions, such as arterial or arteriolar scle-
mesangial matrix. Cellular and fibrocellular rosis, are reported to be common in adults [285]
crescents are often found, usually affecting but are very unusual in children with IgAN
less than 50 % of the glomeruli, although in [279]. This difference may be related to the age
approximately 10 % of patients, more than at biopsy and the duration of disease before
50 % are involved. Capsular adhesions are biopsy.
frequently seen in the absence of crescents. Endocapillary hypercellularity is defined as an
A small number of globally sclerosed glomer- increased number of cells within glomerular cap-
uli are often present. Tubular atrophy, intersti- illaries, causing narrowing of the lumina. It may
tial fibrosis, and interstitial lymphocyte be present, in either focal segmental or diffuse
infiltration are frequently present and are distribution, and is typically associated with
extensive in 10 % of patients. extension of deposits to peripheral loops. The
hypercellularity might reflect proliferation,
Four types of mesangial change are identified inflammatory cell infiltration, or endothelial cell
in children with IgAN [5] (Fig. 4): (a) mesangial swelling. Endocapillary lesions are more common
hypercellularity is more prominent than the in children and associated with active disease and
increase in matrix, (b) the degrees of mesangial high levels of proteinuria [286]. In IgAN, as in
hypercellularity and matrix increase are similar, other proliferative glomerulonephritides, many of
(c) the increase in matrix is more prominent than the glomerular cells are leukocytes [287]. The
the mesangial cellularity, and (d) progression of presence of glomerular macrophages correlates
IgAN is associated with the development of with endocapillary hypercellularity and
global glomerulosclerosis accompanied by reac- sclerosis [288].
tive cell invasion.
The first type of lesion is seen in biopsies in
which the interval between onset of disease and Electron Microscopy
biopsy is short. Serial pathologic observations
reveal that prominent mesangial hypercellularity Electron microscopic abnormalities are mainly
is almost exclusively seen in initial biopsies and observed in the mesangium, which is variably
disappears in follow-up biopsies. These observa- enlarged by a combination of increased cytoplasm
tions suggest that predominant mesangial and matrix. Electron-dense deposits in the
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 999

Fig. 5 Electron
micrograph showing
numerous electron-dense
deposits in the mesangium
in a patient with IgAN.
Original magnification,
 5,000

mesangium are the most constant and prominent Repeat Renal Biopsy Findings
feature and are seen in almost all patients (Fig. 5).
They are granular masses situated immediately There have been only a few reports on the results
beneath the lamina densa in the perimesangial of repeat renal biopsies [290, 291]. We previously
region and expanded mesangium. The size and reported our results in children with IgAN [6]. At
extent of mesangial deposits vary from patient to the time of the second biopsy, 23 patients had
patient; in some patients, they are large and pro- showed clinical remission, defined as complete
duce localized protrusions. Peripheral glomerular disappearance of proteinuria and hematuria with
capillary wall deposits are also found in the normal renal function, whereas 38 had persistent
subendothelial and subepithelial regions. urinary abnormalities with normal renal function.
Subendothelial deposits occur most frequently in There were no differences between the two groups
the capillary wall adjacent to the mesangium, with regard to the initial clinical findings and the
although they are also observed in the peripheral pathologic findings in the initial biopsy. The sec-
part of the loop. Subepithelial deposits are ond biopsy of patients who were in clinical remis-
reported to be unusual in adult patients but are sion showed improvement of the glomerular
frequently found in children with IgAN. They are changes on light microscopy, a disappearance of
generally small and flat and localized to a few or a decrease in mesangial IgA deposits, and a
capillary loops; the humps typical of acute decreased amount of electron-dense deposits.
poststreptococcal glomerulonephritis are never Conversely, light microscopy showed a progres-
observed. Lysis of the glomerular basement sion of histologic lesions and the persistence of
membrane is also seen quite frequently in both mesangial IgA deposits and electron-dense
children [289]. In affected areas of the glomerular deposits in patients with persistent urinary abnor-
capillary walls, the lamina densa is thin and irreg- malities. Clinical remission and histologic regres-
ular, and the epithelial aspect of the glomerular sion have been reported in adults with
basement membrane shows irregular segments of IgAN [292].
low electron density with an expanded, washed- Shima et al. [293] have retrospectively
out appearance. The epithelial foot processes are screened and analyzed 124 consecutive children
generally well preserved, but diffuse foot process (age 18 years at first biopsy) with newly diag-
effacement may be seen in patients with the nosed severe IgAN showing diffuse mesangial
nephrotic syndrome. proliferation, who received combination therapy
1000 K. Nakanishi and N. Yoshikawa

(prednisolone + azathioprine or mizoribine + war- [296, 301]. The Oxford classification involves ana-
farin + dipyridamole, 90 patients) or prednisolone lyses of data from patients with a wide variety of
alone (34 patients) for 2 years and underwent age [296, 301, 302]. This classification has
repeat biopsies. After 2 years of treatment, 27 of obtained a considerable level of worldwide accep-
the patients (21.8 %) showed disappearance of tance. However, the value of this classification
glomerular IgA. system remains to be definitively established [303].
The Oxford classification identified four path-
ological features (mesangial hypercellularity, M;
Differences Between Childhood endocapillary hypercellularity, E; segmental
and Adult Patients with IgAN glomerulosclerosis, S; and tubular atrophy/inter-
stitial fibrosis, T, resulting in a MEST score) that
Significant differences in the early glomerular predicted renal outcome independently of clinical
lesions of IgAN between children and adults indicators at the time of renal biopsy and during
have been demonstrated [294, 295]. Glomerular follow-up [296, 301]: M0 or M1, indicating
hypercellularity in mesangial area is prominent in mesangial hypercellularity in 50 % versus
children and significantly greater than in adults. In >50 % of glomeruli; E0 or E1, indicating
contrast, glomerular matrix expansion, crescent endocapillary hypercellularity in 0 versus 1 glo-
formation, and interstitial damage are more severe meruli; S0 or S1, indicating segmental sclerosis
in adults compared to children. Glomerular in 0 versus 1 glomeruli; and T0, T1, or T2,
hypercellularity correlated with proteinuria in indicating tubular atrophy/interstitial fibrosis in
children but not in adults, whereas glomerular 25 %, 26–50 %, or >50 % of renal cortex,
matrix correlated with proteinuria and renal func- respectively. These findings were generally valid
tion in adults but not in children [294]. both in adults and children [302]. However, the
limited number of patients (265 cases) and their
heterogeneous origin (from 11 countries in four
The Oxford Classification of IgAN continents) indicated a need for validation in other
cohorts.
During past decades, there have been many histo- Validation studies have demonstrated their
logic classifications for IgAN, and their utility in clinical benefits and limitations and highlighted
predicting renal outcome has been tested in stud- questions and difficulties of interpretation of the
ies. These classifications are mostly presented biopsy sample [304–315, 303]. Recently, the
using semiquantitative or single-grade systems VALIGA study including 1,147 patients from
[296, 297]. The former is generally well correlated 13 European countries provides a validation of
with renal outcome, but it limits its clinical utility the Oxford classification in a large European
due to a time-consuming process [298]. The two cohort of IgAN patients across the whole spec-
single-grade scoring systems widely used in clin- trum of the disease. However, the independent
ical practice are the Lee classification [299] and predictive value of pathology MEST score is
the Haas classification [300]. However, both clas- reduced by glucocorticoid/immunosuppressive
sifications have been criticized because of the lack therapy [316].
of reproducibility [297]. Although pathologic To date, none of the validation studies of the
classifications provide valuable information for Oxford classification including children as well as
determining prognosis, there is much controversy adults completely confirmed the results from the
as to whether histopathological features are original Oxford cohort regarding the predictive
superior to clinical factors in risk prediction of value of M, S, and T scores [303], and only one
IgAN [298]. In 2009, the Oxford classification study confirmed the association between E score
of IgAN in expectation of a high reproducibility and response to immunosuppressive therapy
and high predictive power of renal outcome [304]. Differences between studies are likely to
was published for international consensus be accounted for, at least in part, by differences in
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1001

inclusion or exclusion of patients with very mild prediction rule using clinical measures and the
or very severe disease (both excluded from the Oxford classification for developing ESRD in
original Oxford cohort), proportions of patients patients with IgAN and verified its validity in an
treated with angiotensin-converting enzyme independent cohort. This prediction rule provides
inhibitors and/or angiotensin receptor blockers a useful guide to estimate the individual risk for
and duration of this therapy, use of immunosup- ESRD in patients with IgAN and may be effective
pressive therapy, length of follow-up, and other at identifying those at high risk for the future
factors. Some of these differences appear to be development of ESRD.
potentially instructive. Notably, crescents, not a
significant predictor of outcomes in the original
Oxford cohort that excluded patients with rapidly Pathology in HSN
progressive glomerulonephritis, may in fact have
predictive value in patients with severe disease The renal histopathology of HSN is indistinguish-
(eGFR <30 ml/min) and in children. This ques- able from that of primary IgAN. Although as
tion deserves further study. IgAN various glomerular changes are observed
Recently, Shima et al. [317] have reported that at light microscopic examination of renal biopsy
a shorter time from IgAN onset to renal biopsy specimens, there are two basic types of glomerular
was associated with higher glomerular ratios lesions, mesangial proliferation and epithelial
of M, E, and crescents and lower ratios of S and crescent formation. The two lesions typically
G in 250 children with proteinuria 0.5 g/day/ coexist (Fig. 6).
1.73 m2 at biopsy, matching the inclusion criteria Severe mesangial proliferation may also be
for IgAN of the original Oxford cohort. These associated with mesangial interposition in which
findings indicate that acute lesions, such as M cell and matrix migrate into the capillary walls
and E, reflect disease activity, with shorter times between the basement membrane and endothelial
from onset to biopsy generally indicating more cytoplasm, giving the capillary walls a double
severe disease activity. Crescents also seem to contour appearance on silver staining. Mesangial
reflect disease activity, because crescents were interposition is only rarely diffuse in HSN, in
acute lesions in their cohort. In contrast, chronic which it may superficially mimic the appearance
lesions such as S and G tend to be absent early in of mesangiocapillary (membranoproliferative)
the course of disease, even in patients with severe glomerulonephritis [15].
acute lesions. Taken together, these findings sug- Because the mesangial proliferation usually
gest that each variable of the Oxford classification resolves without causing permanent damage, the
of IgAN may be influenced by the time from morphologic severity of HSN is best assessed on
disease onset to renal biopsy and that differences the basis of the extent of glomerular involvement
in the significance of S and crescents between the with crescents and segmental lesions, including
original Oxford classification and their data may necrosis and sclerosis. The classification used
be due, at least in part, to differences in the timing most is that evolved by the pathologists of the
of renal biopsy [317]. International Study of Kidney Disease in Children
Finally, the most important clinical question is (Table 2) [27, 33, 34]. Each of the severity grades
how we can make the Oxford classification more is based on the percentage of glomeruli with cres-
clinically relevant. One of key issues should be cents and segmental lesions. The clinicopatho-
considered is that there is sufficient evidence to logic correlations of this classification have been
warrant combining M, E, S, and T (and possibly effectively tested in follow-up studies (Table 3)
other) scores into a grading system (I, II, III, and [26, 27, 33–35].
so on) designed to indicate increasing levels of The characteristic immunopathologic pattern
disease severity and perhaps simplify this classi- of HSN is the same as that of IgAN with the
fication system for clinicians [303]. Tanaka presence of IgA in the glomerular mesangium
et al. [318] have developed and validated a new (Fig. 3). Although in most patients IgA is present
1002 K. Nakanishi and N. Yoshikawa

Fig. 6 Glomeruli from a


patient with HSN.
Circumferential crescents
are evident (Periodic acid-
methenamine-silver stain,
original magnification,
 200)

Table 2 Morphologic classification of Henoch-Schönlein Table 3 Correlations between ISKDC grade and clinical
nephritis evolved by the International Study of Kidney presentation/outcome in Henoch-Schönlein nephritis
Disease in Children
ISKDC Approximate risk
Minimal glomerular grade Clinical presentation of renal failure (%)
I abnormalities I Hematuria only 0
II Pure mesangial (a) focal or II Hematuria and <5
proliferation (b) diffuse proteinuria
III Minor glomerular (a) focal or III Hematuria and <10
abnormalities or (b) diffuse mesangial proteinuria
mesangial proliferation, proliferation Acute nephritic
with crescents or syndrome
segmental lesions
Nephrotic syndrome
(sclerosis, adhesions,
thrombosis, necrosis) in IV Hematuria and 25
fewer than 50 % of proteinuria
glomeruli Acute nephritic
IV As III but with crescents (a) focal or syndrome
or segmental lesions in (b) diffuse mesangial Nephrotic syndrome
50–75 % of glomeruli proliferation Rapidly progressive
V As III but with crescents/ (a) focal or nephritic syndrome
segmental lesions in more (b) diffuse mesangial V Hematuria and >50
than 75 % of glomeruli proliferation proteinuria
VI Membranoproliferative- (a) focal or Acute nephritic
like lesion (b) diffuse mesangial syndrome
proliferation Nephrotic syndrome
Rapidly progressive
nephritic syndrome
VI

only in the mesangial regions, in approximately


10 % of patients, it is also present in the peripheral
capillary walls. Such peripheral capillary wall Electron microscopic abnormalities of HSN
deposits, whether documented with immunofluo- are also the same as that of IgAN (Fig. 5).
rescence or electron microscopic examination, Niaudet et al. [319] found good correlation
have been associated with more severe clinical between histologic changes with time and both
manifestations and a poor renal outcome [34]. clinical status and outcome.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1003

Clinical Features in IGAN infection. The number of recurrences and the


intervals between different episodes are variable.
IgAN occurs at all ages but is most common The incidence of macroscopic hematuria is lower
during the second and third decades of life; it in adults (Southwest Pediatric Nephrology Study
affects boys more often than girls, with the Group [323, 324]). Emancipator et al. [3] summa-
reported male to female ratio varying from less rized the previous reports of IgAN, in which most
than 2:1 to 6:1 [2]. In a study of Japanese children the patients were adults, and reported that 43 %
[275], the mean age at presentation was 9.3 years had macroscopic hematuria. However, in Japan,
in boys and l0.3 years in girls, and the male to only 18–32 % adult patients have been reported to
female ratio was 3:2. The clinical presentation of have macroscopic hematuria [324, 325]. The rea-
IgAN varies. Some patients have asymptomatic son for the age-related differences in the incidence
microscopic hematuria with or without protein- of macroscopic hematuria has yet to be elucidated.
uria. Other patients have recurrent episodes of In asymptomatic patients, microscopic hema-
macroscopic hematuria. Some patients present turia is almost always present and persistent. Pro-
with acute nephritic syndrome and, more rarely, teinuria is common. The blood pressure and renal
with acute renal failure. function at onset are normal.
Sixty-two percent of our 258 Japanese children Patients with a nephritic or nephrotic onset have
were found to have microscopic hematuria with or the most severe glomerular damage. Hypertension
without asymptomatic proteinuria [275]. Twenty- is infrequent and usually mild to moderate. Malig-
six percent presented with macroscopic hematuria nant hypertension is not a presenting feature in
and 12 % with an acute nephritic syndrome or childhood. Nephrotic edema is reported in approx-
nephrotic syndrome. Several studies from Europe imately 10 % of patients. Acute renal failure is
and the United States reported that more than occasionally associated with episodes of macro-
80 % of the patients have episodes of macroscopic scopic hematuria and is usually reversible. How-
hematuria, and recurrent macroscopic hematuria ever, a number of investigators have documented a
is traditionally regarded as the hallmark of child- subset of patients with IgAN that is characterized
hood IgAN [320–322] (Southwest Pediatric by extensive crescents and a rapidly progressive
Nephrology Study Group [323]). However, it course [325–327]. A review of published cases of
was the initial feature in only 26 % of our series, crescentic IgAN revealed that 41 % of patients with
presumably because of the school screening pro- this rapidly progressive form of disease were
gram that detected a high prevalence of asymp- 16 years of age or younger [327].
tomatic urinary abnormalities rather than regional
variation in the expression of IgAN. During the
observation period, 60 % of patients had one or Laboratory Investigation
more episodes of macroscopic hematuria,
whereas the other 40 % remained asymptomatic. Serum IgA levels are increased in 30–50 % of
Macroscopic hematuria often occurs in associ- adult patients but in only 8–16 % in children
ation with upper respiratory tract infections; less with IgAN [275]. For this reason, it is seldom of
frequently, it occurs in association with other diagnostic significance. Serum complement com-
infections involving the mucosal system (e.g., ponent concentrations are usually normal, but the
diarrhea and sinusitis). Episodes of macroscopic C3 level should be measured routinely if the
hematuria are sometimes associated with loin patient has been referred for investigation after
pain. The interval between the precipitating infec- the first attack of hematuria to eliminate a diagno-
tion and the appearance of hematuria ranges from sis of postinfectious glomerulonephritis or
1 to 2 days compared with 1 or 2 weeks in acute membranoproliferative glomerulonephritis. Like-
postinfectious glomerulonephritis. Many patients wise, the antistreptococcal antibody titers should
have recurrent episodes of macroscopic hematu- be determined after initial hematuria. These inves-
ria, each often associated with the same type of tigations are of little value when hematuria is
1004 K. Nakanishi and N. Yoshikawa

known to have been present for more than Assays for serum levels of complement C3
3 months. The serum creatinine should be mea- may be readily available in clinical practice, either
sured routinely to estimate renal function; if nec- alone or in tandem with total serum IgA or serum
essary, the glomerular filtration rate should be galactose-deficient IgA1 levels, as prognostic bio-
determined. If present, proteinuria should be markers for patients with IgAN [328]. Several
quantified, as proteinuria is associated with histo- researchers have reported that serum C3 levels in
logic lesions and a risk of progression. The plasma patients with IgAN are significantly lower
proteins should be measured routinely in the pres- than those in patients with non-IgAN
ence of heavy proteinuria. [339–341]. Serum IgA levels in patients with
IgAN are significantly higher than those in
patients with non-IgAN, and the serum IgA/C3
Biomarkers in IgAN ratio seems to be a more reliable marker than
serum IgA to distinguish IgAN from non-IgAN
IgAN has been the focus of several studies aimed [339–345]. Recent studies have shown that ele-
at identifying specific biomarkers. Serum vated serum IgA/C3 ratio is not only a robust
galactose-deficient IgA1 level and glycan-specific diagnostic marker for IgAN but also a predictor
autoantibody levels are prime candidates to of prognostic grading in patients with IgAN in
become diagnostic biomarkers for IgAN because Japanese patients [339–345]. Komatsu
of their central role in the earliest stages of disease et al. have reported that IgAN patients with high
pathogenesis [328]. However, although the serum serum IgA/C3 (>4.5) have a significantly poorer
level of galactose-deficient IgA1 is frequently ele- renal outcome in Japanese patients [341]. How-
vated in patients with IgAN [329], the sensitivity ever, to date, these findings have not been con-
and specificity of this laboratory finding are insuf- firmed in children.
ficient for the test to replace kidney biopsy as the Urinary cytokine excretion may reflect histo-
diagnostic standard [330]. The serum level of logic changes in IgAN, and their measurement
glycan-specific IgG antibodies is correlated with can give information about disease outcome
the level of urinary protein excretion [216] and the [346]. Increased urinary excretion of epidermal
risk of progression to end-stage renal disease or growth factor (EGF) [347], MCP-1 [346], IL-6
death [331]. This biomarker may prove useful for [346], podocytes [348], low-molecular-mass pro-
monitoring disease progression or the response to teins [349], and mannose-binding lectin [350]
therapy [330]. Yanagawa et al. have reported that were reported in IgAN. Conversely, Stangou
serum levels of galactose-deficient IgA1-specific et al. have reported that decreased urinary EGF
antibodies are elevated in most IgAN patients, and levels may represent histology in IgAN and that
their assessment, together with serum levels of EGF excretion can be a predictive marker [346].
galactose-deficient-IgA1, improves the specificity Urinary proteomic data are also candidates for
of the assays. These observations suggest that a diagnostic biomarkers because this approach can
panel of serum biomarkers may be helpful in successfully differentiate patients with IgAN from
differentiating IgAN from other glomerular healthy controls and from patients with other
diseases [332]. forms of renal disease [328, 330]. Analyses of
It has reported that increased plasma levels of urinary samples by capillary electrophoresis with
activated complement C3 [333], advanced oxida- mass spectrometry have differentiated patients
tive protein products [334], and fibroblast growth with IgAN from healthy controls and patients
factor 23 [335]; an increased serum level of uric with minimal change disease or IgA immune
acid [336, 337] and decreased serum levels of complex nephritis due to chronic hepatitis C
CD89-IgA complexes [338] are associated with infection, even in association with nonpathologic
severe histologic changes, severe proteinuria, or a proteinuria [351–353]. Furthermore, the urinary
poor clinical outcome. However, these findings proteomic profile predicts the response to treat-
may not be unique to IgAN [330]. ment with an angiotensin-converting enzyme
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1005

inhibitor [354]. However, additional studies are Relationship Between IgAN and HSP
needed to determine the potential and cost-
effectiveness of urinary proteomic analysis in The relation between IgAN and HSP is complex.
establishing the diagnosis of IgAN and making There seems to be a close relationship between
decisions about treatment [330]. IgAN and HSP [355]. The morphologic and
immunopathologic features are similar in the two
conditions [355, 48]. The two disorders have been
Differential Diagnosis reported to coexist in different members of the
same family, including a pair of monozygotic
The diagnosis of IgAN is based on the presence of twins who developed the disorders simulta-
IgA as the sole or predominant Ig in the glomer- neously after a well-documented adenovirus
ular mesangium. Because diffuse mesangial IgA infection [11, 38, 356, 57]. Moreover, the evolu-
deposits are observed in a variety of other disor- tion of IgAN into HSN in the same patient is
ders (Table 4), the diagnosis of IgAN can be made described in both adults and children
only by exclusion. The IgA deposits are often [357–359]. It has been suggested that the two
incidental findings, and the pathogenesis and clin- conditions are variants of the same process and
ical significance are unclear. that IgAN is HSN without the rash [48, 57, 359].
Although there are similarities in their pathologic
and immunologic features, the two conditions are
clinically different, and the pathogenesis is not
Table 4 Diseases associated with diffuse mesangial IgA clear. Our study suggests that HSN is an acute
deposits disease, with glomerular lesions nonprogressive
Primary after the onset [27]. Therefore, in most patients,
IgA nephropathy the prognosis is associated with the severity of
Secondary glomerular change at the onset. In contrast,
Multisystem disease IgAN is a chronic, slowly progressive glomerular
Henoch-Schönlein purpura lesion, which may eventually lead to chronic renal
Systemic lupus erythematosus
failure, whatever the presentation. A few patients
Cystic fibrosis
with HSN have recurrent episodes of macroscopic
Celiac disease
hematuria and a progressive renal disease on
Crohn’s disease
repeat renal biopsies. Finally, HSN occurs mostly
Dermatitis herpetiformis
Ankylosing spondylitis
in young children and is rare in adulthood,
Neoplasms whereas IgAN affects mainly older children and
Carcinomas of the lung and colon young adults. It is therefore reasonable that IgAN
Monoclonal IgA gammopathy and HSN are treated as different clinicopathologic
Mycosis fungoides entities until pathogeneses of the two conditions
Non-Hodgkin’s lymphoma are better understood.
Infectious diseases
Mycoplasma infections
Leprosy Chronic Liver Disease
Toxoplasmosis
Others Glomerular IgA deposits may be observed in
Chronic liver disease patients with various types of chronic liver dis-
Thrombocytopenia eases [360]. Mesangial proliferation and IgA
Pulmonary hemosiderosis deposits are the most common findings. The
Mixed cryoglobulinemia
light, electron, and immunofluorescence micro-
Polycythemia
scopic features in patients with liver disease are
Scleritis
similar to those in patients with primary IgAN.
1006 K. Nakanishi and N. Yoshikawa

100%
9%
8%

21% Renal failure


Nephrotic syndrome
5%
Proteinura ≥ 1 g/day
50%
Proteinura < 1 g/day
Hematuria only
57%
Normal urine

0%
onset 5y 10y 15y
Follow-up period

Fig. 7 Long-term prognosis of the 169 Japanese children with IgAN followed more than 10 years

Most patients with chronic liver disease have clin- syndrome associated with mesangial IgA deposits
ically asymptomatic renal disease. Microscopic occurring in Asians and explains a favorable
hematuria and mild proteinuria may be observed, response to steroids, which is not the case in true
but macroscopic hematuria or a nephrotic syn- IgAN [363].
drome is rare. Renal functional impairment is
also rare. Glomerular lesions in children with
chronic liver disease are very unusual. The path- Natural History and Prognosis
ogenetic mechanisms that contribute to mesangial
IgA deposition in chronic liver diseases remain In adult series, the incidence of renal insufficiency
unknown. Significant elevations of the serum varies from less than 10 % to as high as 45 % in
monomeric and polymeric IgA levels have been patients followed up for more than 1 year. In long-
reported in patients with chronic liver disease term follow-up of adult patients, 30–35 % have
[361, 362]. Impaired hepatic clearance and been found to develop progressive renal insuffi-
increased synthesis of polymeric IgA, abnormali- ciency 20 years after the initial discovery of dis-
ties of IgA metabolism, and portosystemic ease [2, 364–366]. It can be estimated that 1–2 %
shunting of antigens and immune complexes of adult patients will enter end-stage renal failure
have been suggested as possible causes of each year from time of diagnosis [367]. The long-
mesangial IgA deposition in chronic liver term prognosis of the 169 Japanese children with
disease [362]. IgAN followed more than 10 years indicates that
9 % of the patients had developed chronic renal
failure by 15 years (Fig. 7).
Idiopathic Nephrotic Syndrome Because of the variable rate of progression to
chronic renal failure, there have been attempts to
A few patients with steroid-sensitive nephrotic identify features present at the time of diagnosis
syndrome show mesangial deposits of IgA on that would predict the outcome. The following
renal biopsy. They are classified by some as clinical findings are regarded as poor prognostic
IgAN, whereas others consider that mesangial indicators in adult patients [2]: persistent hyper-
IgA in patients with minimal changes (i.e., with- tension, persistent heavy proteinuria, and reduced
out cellular proliferation) is coincidental. This glomerular filtration rate at presentation
probably applies to idiopathic nephrotic [364–366, 368–373]. In children, several studies
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1007

have shown that the degree of proteinuria corre- in a study by Tsuboi et al., suggesting that glo-
lates with the severity of morphologic glomerular merular density may serve as an early histopatho-
lesion [322, 374, 279], and heavy proteinuria at logical marker of long-term renal prognosis in
the time of biopsy predicts a poor outcome [280, IgAN [376]. Activation of the lectin pathway of
375]. In contrast, slight proteinuria or its absence complement is associated with more severe renal
at the time of biopsy predicts a favorable outcome. disease. Glomerular deposition of C4d is a marker
Acute renal failure at onset is usually transient and of activation of the lectin pathway of complement.
associated with macroscopic hematuria and Espinosa et al. have reported that negative
reversible tubular lesions. Male gender has also mesangial C4d staining in glomeruli in patients
been considered an unfavorable prognostic fea- with IgAN helps to identify patients with a good
ture by some investigators [365], but we [280] long-term prognosis [377, 378].
and others [278] could not confirm it in large Recurrence of mesangial IgA deposits is often
cohort of adult and pediatric patients. Schena observed in transplant recipients whose original
et al. reported an increased risk of end-stage disease was IgAN [120, 50]. Recurrence of IgA
renal disease in familial IgAN [88]. deposits is most often clinically asymptomatic,
Several pathologic features are associated with and graft survival is considered good [120].
a poor outcome: diffuse mesangial proliferation; a At the beginning of the 1990s, the use of
high proportion of glomeruli showing sclerosis, angiotensin-converting enzyme inhibitors for
crescents, or capsular adhesions; the presence of focal mesangial proliferation and combined ther-
moderate or severe tubulointerstitial changes; the apies including corticosteroids for diffuse
presence of subepithelial electron-dense deposit; mesangial proliferation increased dramatically in
and lysis of the glomerular basement membrane Japan. Our recent retrospective cohort study of
by electron microscopy [280]. Patients with dif- 500 children with IgAN has clarified an improved
fuse mesangial proliferation have been reported to renal survival in Japanese children with IgAN
have a significantly worse prognosis than those [379]. Among all patients, the actuarial renal sur-
with focal proliferation or minimal lesions by light vival was 96.4 % at 10 years, 84.5 % at 15 years,
microscopy in adults [364, 369, 300]. In many and 73.9 % at 20 years. Diagnosed in 1976–1989,
adult studies [278, 364, 372, 285], glomerular the renal survival was 94.0 % at 10 years, 80.1 %
sclerosis and crescents have also been associated at 15 years, and 70.1 % at 20 years. Diagnosed in
with poor renal outcome. Levy and associates 1990–2004, the renal survival was 98.8 % at
found that mesangial proliferative glomerulone- 10 years and 98.8 % at 15 years ( p = 0.008).
phritis with crescents was associated with poor With diffuse mesangial proliferation, both the
prognosis in children [322]. As the severity of 10- and 13-year renal survivals were 97.8 % in
the tubulointerstitial changes is usually related to 1990–2004, compared with 78.5 % and 68.6 %,
the severity of the glomerular changes, tubuloin- respectively, in 1976–1989 ( p = 0.0003). In the
terstitial changes in IgAN are believed to be sec- same study, prognostic factors for end-stage renal
ondary to the glomerular injury. Vascular lesions, disease-free survival were analyzed [379].
such as arterial or arteriolar sclerosis, have been Mesangial proliferation degree and initial renal
reported to play an important role in the progres- biopsy year were significant in both the univariate
sion of IgAN in adults. However, vascular and the multivariate analysis. For children with
changes are very unusual in children [5] (South- IgAN, the most influential prognostic variable
west Pediatric Nephrology Study Group was mesangial proliferation degree. Proteinuria
[323, 326]). This difference may be related to the at diagnosis was significant in the univariate, but
age at biopsy and the duration of disease before not in the multivariate analysis. Multivariate anal-
biopsy. ysis showed that initial renal biopsy year was a
A strong predictive relationship of low glomer- significant factor for renal survival independently
ular density (nonsclerotic glomerular number per of mesangial proliferation degree, proteinuria at
renal cortical area) with progression was observed diagnosis, and estimate creatinine clearance at
1008 K. Nakanishi and N. Yoshikawa

diagnosis (hazard ratio = 0.08, 95 % CI is the selection of appropriate patients in whom


0.004–0.43). the treatment is to be evaluated. Patients with
Some patients with IgAN achieve spontaneous heavy proteinuria at biopsy and the most severe
remission even when not receiving medication. glomerular lesions on renal biopsy appear to be at
The retrospective study of 96 children with greatest risk of progressive renal deterioration
minor glomerular abnormalities or focal and, therefore, the most appropriate candidates
mesangial proliferation who did not receive med- for specific therapeutic interventions. Patients
ication from the 555 patients with childhood with long-standing disease and extensive, irre-
IgAN by Shima et al. [380] showed that versible glomerular damage are unsuitable for
57 (59.4 %) achieved spontaneous remission. such treatments.
The cumulative spontaneous remission rates In a randomized controlled trial, another
among these patients were 57.5 and 77.4 % at important issue is to select adequate endpoints.
5 and 10 years, respectively, from onset. The Although in a clinical trial of progressive IgAN
mean time from onset to remission was 5.9  the ultimate endpoint is development of chronic
0.4 years. Of the 57 patients with spontaneous renal insufficiency, most pediatric patients do not
remissions, ten (17.5 %) also developed a recur- develop it during the study period. Thus, studies
rence of urinary abnormalities. The cumulative of pediatric patients with IgAN may differ mark-
recurrence-free rates were 79.9 and 67.9 % at edly from studies of adults with regard to the
5 and 10 years, respectively, after remission. The apparent risk of progressive disease [381]. There-
spontaneous remission rate in childhood IgAN fore, due to the long time period from clinical
with minor glomerular abnormalities or focal onset of disease until progression to end-stage
mesangial proliferation was higher than expected renal disease, surrogate markers of outcome
[380]. These results suggest that physicians must be used to evaluate efficacy of therapy for
should consider the potential for spontaneous IgAN in clinical trials. It is a noteworthy fact that
remission and refrain from aggressive treatment validation of these surrogate markers may be
in IgAN patients with minor glomerular abnor- lacking, resulting in the potential for inappropriate
malities or focal mesangial proliferation. conclusion with regard to therapeutic efficacy.
Therefore, the careful investigation in detailed
long-term outcome of previous randomized con-
trolled trials is important.
Treatment With regard to treatment of IgAN, it is impor-
tant to consider the differences in the nature of
IgAN is a leading cause of chronic renal disease IgAN between children and adults. An “evidence-
and end-stage renal disease in adult patients, and based” therapy is important in both children and
recent long-term studies assessing the prognosis adults. However, available evidence for treatment
in children have challenged earlier views that the is partially, clearly, different between children
condition represents a benign disorder. Thus, and adults. Generally and roughly speaking, the
IgAN presents a therapeutic challenge in both evidence for treatments of IgAN in adults sup-
adults and children. The most appropriate treat- ports relatively passive treatments, whereas that
ment for patients with IgAN is still a matter of in children supports relatively active treatments.
controversy [381]. At present, there is no curative The reason of this difference is unknown.
therapy for IgAN [382]. Because of the variable Differences in the diagnosis timing and
rate of progression to renal failure and because of histological differences may be associated.
the probable multifactorial pathogenesis of the Although we should refrain from radical treat-
disease, the effectiveness of any treatment can ments that are not based on evidence, however,
only be properly evaluated by means of a random- appropriate active treatments that are based on
ized controlled trial [381]. When considering evidence are important in treatments for children
treatment protocols, an issue of great importance with IgAN.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1009

Fish Oil/Omega 3 Fatty Acids children and young people when it was assessed
as a composite end point of 30 % reduction in
Controlled double-blind trial in adult patients with GFR or an increase of proteinuria over the
IgAN [383–385] showed that treatment with fish nephrotic range. It has also showed the benefit of
oil for 2 years retarded the rate at which renal angiotensin-converting enzyme inhibitor on
function was lost, but a meta-analysis showed remission of proteinuria below what is generally
that there was only a 75 % probability that fish considered a harmful level [399]. In adult setting a
oil was beneficial [386]. A randomized, placebo- randomized controlled trial has showed that com-
controlled, double-blind trial by the Southwest bination therapy with angiotensin-converting
Pediatric Nephrology Study Group in the United enzyme inhibitor and angiotensin II receptor
States and Canada evaluated the role of omega blocker has an additive dose-dependent
3 fatty acids in children and young adults with antiproteinuric effect compared to monotherapies
IgAN, and the treatment group did not showed in patients with IgAN [400]. The COOPERATE
benefit over the placebo group with respect to time trial, a large randomized study of 336 patients
to failure, defined as estimated GFR <60 % of with nondiabetic renal disease, in which 50 % of
baseline [387]. Furthermore, from analysis of subjects had IgAN, has demonstrated that combi-
these clinical trials, it was reported that efficacy nation treatment safely retards disease progres-
of omega-3 fatty acids in children and adults with sion compared with monotherapy [401]. A
IgAN is dosage and size dependent [388]. randomized controlled trial in 109 adults with
IgAN showed that valsartan significantly slowed
renal deterioration compared with placebo
Coagulation Modifying Agents [402]. Although there is no randomized controlled
trial for angiotensin II receptor blockers in chil-
Warfarin, urokinase, and antiplatelet agents have dren with IgAN, some studies have demonstrated
all been assessed for the treatment of IgAN. their antiproteinuric effect in combination thera-
At present there is no sufficient evidence to sup- pies with angiotensin-converting enzyme inhibi-
port the use of coagulation modifying agents tor and angiotensin II receptor blocker [403, 404].
[389, 390]. However, coagulation modifying
agents may have a role in combination therapy.
Corticosteroids

Angiotensin-Converting Enzyme Corticosteroids have been widely used to treat


Inhibitors and Angiotensin II Receptor moderate to severe IgAN, particularly in pediatric
Blockers patients. To date, information concerning not only
the effectiveness but also safety of corticosteroid
As already demonstrated in nondiabetic chronic therapy over a long time course has been largely
nephropathies [391] (GISEN Group [392, 393]), defective. It has been difficult to assess the results
studies including randomized controlled trials of treatment trials with these agents in terms of
have indicated that angiotensin-converting preservation of renal function, due in part to wide
enzyme inhibitors reduce urinary protein excre- variations in the length of therapy and the dosing
tion [394, 395] and preserve renal function [396] regimens employed and also to the use of cortico-
in adult patients with IgAN. However, there is no steroids in combination with other drugs [381].
randomized controlled study only in children with At present, however, some evidence has been
IgAN demonstrating that angiotensin-converting obtained for the role of corticosteroids in the treat-
enzyme inhibitors preserve renal function [397, ment of IgAN [405–407]. In adults with IgAN, an
398]. A randomized controlled trial has proved a Italian prospective randomized controlled trial
significant benefit of angiotensin-converting demonstrated that a 6-month course of steroid
enzyme inhibitor on the progression of IgAN in treatment protected against deterioration of renal
1010 K. Nakanishi and N. Yoshikawa

function with no notable adverse effects during due in part to wide variations in the length of
follow-up [405]. Recently, the long-term follow- therapy and the dosing regimens employed and
up data of the trial showed that corticosteroids also to the use of corticosteroids in combination
significantly reduced proteinuria and protected with other drugs [381]. There is currently insuffi-
against renal function deterioration [406]. cient evidence to support the use of immunosup-
An English single-center randomized controlled pressants alone for treatment of children with
trial has demonstrated that the value of combined IgAN. Recently a meta-analysis of immunosup-
immunosuppressive treatment with prednisolone pressive treatments for IgAN suggested a benefit
and cytotoxic agents in reducing the occurrence of of corticosteroids and immunosuppressants [412].
renal failure in IgAN [407]. A homogeneous
cohort of 38 patients who had mean blood pres-
sure within 10 % of current targets and estimated Clinical Trials for Combination Therapy
GFR 50 % normal but losing 10 % estimated Including Corticosteroids and
GFR/year were randomly assigned; no patient Immunosuppressant in Japan
had crescentic disease. Renal survival improved
12-fold at 5 year, with remission of nephritis by Prospective trials by The Japanese Pediatric IgAN
urinalysis. Treatment Study Group have provided some use-
With regard to children, our previous studies ful data regarding the treatment of children with
[389, 408] are the only randomized controlled IgAN [389, 408]. In the trials, treatment was
trials so far to have demonstrated that treatment started early in the course of disease because the
including corticosteroid for 2 years early in the duration of the disease before treatment was short
course of disease reduces immunologic renal and the extent of glomerulosclerosis was low as a
injury and prevent any further increase of glomer- result of the Japanese school screening program.
ular sclerosis. Up to now, however, it has been In the trials, the majority of patients presented
unclear whether corticosteroid alone is sufficient with asymptomatic proteinuria and microscopic
for treatment of IgAN in children, and there has hematuria detected by this school screening
been no reliable evidence for its effectiveness in program.
this group of patients [381]. High-dose intrave- A randomized controlled trial by The Japanese
nous methylprednisolone were shown to delay Pediatric IgAN Treatment Study Group demon-
development of renal failure in a randomized con- strated that treatment of children with severe
trolled trial in adult patients [405]. However, no IgAN showing diffuse mesangial proliferation
convincing evidence has been published to date to with prednisolone, azathioprine, heparin-warfa-
support the use of high-dose intravenous methyl- rin, and dipyridamole for 2 years early in the
prednisolone for the treatment of children with course of disease prevents immunologic renal
IgAN. injury and progression of the disease [389]. The
follow-up study of this randomized controlled
trial revealed that 2-year combination therapy
Immunosuppressants not only ameliorated the activity of the acute
phase of nephritis but also improved the long-
The use of immunosuppressants other than corti- term outcome of severe childhood IgAN [413].
costeroids for treatment of IgAN has not been In a randomized controlled trial carried out
sufficiently evaluated, even in adult patients. sequentially by The Japanese Pediatric IgAN
Although the results of prospective trials of cyclo- Treatment Study Group, the effects of predniso-
phosphamide [407] and mycophenolate mofetil lone, azathioprine, warfarin, and dipyridamole
[409–411] for IgAN have been published, their (combination) with those of prednisolone alone
efficacy is controversial. It has been difficult to were compared in 80 children with newly diag-
assess the results of treatment trials with these nosed IgAN showing diffuse mesangial prolifera-
agents in terms of preservation of renal function, tion [408]. Patients were randomly assigned to
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1011

receive either the combination or prednisolone cyclophosphamide, antiplatelets, and warfarin.


alone for 2 years. It was concluded that the com- At present studies provide conflicting data. The
bination treatment may be better for severe IgAN results of a randomized controlled trial for adults
than prednisolone alone [408]. in Japan, which compares tonsillectomy com-
Based on the two previous randomized con- bined with steroid pulses versus steroid pulses
trolled trials and the follow-up study described monotherapy, indicated that tonsillectomy has no
above, the immunosuppressant is considered to beneficial effect over steroid pulses alone to atten-
be important for the treatment in children with uate hematuria and to increase the incidence of
IgAN. However, azathioprine regimen had often clinical remission. Although the antiproteinuric
to be stopped due to toxicity. Therefore, a differ- effect was significantly greater in combined ther-
ent but effective immunosuppressant may be apy, the difference was marginal, and its impact on
worth trying. Mizoribine, like azathioprine, is an the renal functional outcome remains to be clari-
antimetabolite that exerts its immunosuppressant fied [418]. Therefore, it cannot be recommended
effect by inhibiting lymphocyte proliferation. In a for widespread use for treatment of IgAN patients,
recent prospective pilot study by The Japanese especially for children with IgAN [419].
Pediatric IgAN Treatment Study Group,
mizoribine was administered instead of azathio-
prine as part of the combination therapy for treat- Japanese Guidelines for the Treatment
ment of 23 children with severe IgAN. The of Childhood IgAN
efficacy and safety of the mizoribine combination
seem to be acceptable for the treatment of children Recently, the Japanese Society for Pediatric
with severe IgAN [414]. Nephrology has developed “Guidelines for the
treatment of childhood IgAN.” The disease sever-
ity has been divided into two categories, i.e., mild
Chinese Herbal Medicine (Sairei-to) [397] and severe [389, 408, 414, 413] IgAN. A
summary of the guidelines is shown in Table 5.
To determine the effect of the Chinese herbal
medicine, Sairei-to (TJ-114), in children with
newly diagnosed IgAN showing focal/minimal Clinical Manifestations and Laboratory
mesangial proliferation, a randomized controlled Features in HSP
trial was undertaken by The Japanese Pediatric
IgAN Treatment Study Group [415]. One hundred Extrarenal Manifestations
and one patients were randomly assigned to
receive Sairei-to for 2 years or no drug for The extrarenal manifestations of HSP are charac-
2 years. The trial has demonstrated that 2-year teristic and can occur in any order and at any time
Sairei-to treatment early in the course of disease over several days or weeks.
is effective in children with IgAN showing focal/
minimal mesangial proliferation.
Skin

Tonsillectomy The skin lesion typically begins with erythema-


tous macules, some of which develop into slightly
Several retrospective studies have analyzed the raised (palpable), urticarial papules, which soon
role of tonsillectomy in the treatment of IgAN become purpuric (Fig. 8). The eruption is of sym-
mainly in adult [416, 417]. Treatment was not, metric distribution and predominantly affects the
however, homogeneous between the study buttocks and the extensor surfaces of the lower
groups; patients who underwent tonsillectomy legs and forearms but spares the trunk. Purpura
were also treated with steroid pulses, occasionally affects the earlobes, nose, and
1012 K. Nakanishi and N. Yoshikawa

Table 5 Summary of guidelines for the treatment of child- associated with the recurrence of abdominal and
hood IgA nephropathy (ver. 1.0, by the Japanese Society joint symptoms.
for Pediatric Nephrology)
Mild cases
Definition: patients who meet both clinical findings AND Joints
histological findings as follows
Clinical findings: slight proteinuria (early morning
urinary protein to creatinine ratio <1.0) Approximately 70 % of children [26, 15, 420]
Histological findings: <80 % of glomeruli showing have joint involvement, and joint symptoms may
moderate or severe mesangial cell proliferation, crescent be the initial manifestation. Joint involvement
formation, adhesion, or sclerosis and <30 % of glomeruli mainly affects the knees, ankles, elbows, and
showing crescent formation
wrists. This consists of arthralgia and periarticular
Treatments: Either lisinopril or Sairei-to should be given
for more than 2 years edema without overlying erythema, tenderness, or
Severe cases joint effusions. It is transient and leaves no resid-
Definition: patients who meet either clinical findings OR ual damage.
histological findings
Clinical manifestations: heavy proteinuria (early
morning urinary protein to creatinine ratio 1.0) Gastrointestinal Tract
Histological findings: 80 % of glomeruli showing
moderate or severe mesangial cell proliferation, crescent
formation, adhesion, or sclerosis or 30 % of glomeruli Gastrointestinal manifestations occur among
showing crescent formation 50–70 % of affected children [420], but the inci-
Treatments: the combination therapy of prednisolone, dence increases to more than 90 % among those
azathioprine or mizoribine, warfarin, and dipyridamole who have renal involvement [26, 15]. The most
should be given for 2 years common symptom is abdominal colic, which
often is severe and accompanied by vomiting,
although melena occurs in one half of cases
[420]. A mass occasionally is palpable in the
upper abdomen, suggesting intussusception.
When the abdominal symptoms precede the
other manifestations, laparotomy sometimes is
considered necessary [26]. Intussusception due
to severe purpura of the intestine is common
among older children [48]. Protein-losing enter-
opathy has been reported [421, 422] and may
account for the occasional observation of
hypoalbuminemic edema in the absence of
Fig. 8 Palpable purpura in a patient with HSP heavy proteinuria. Vasculitic lesions have been
found in many other organs and may explain
rare manifestations such as hemorrhage in the
external genitalia but in typical, mild cases is calf, the testis, the lung, acute hemorrhagic pan-
confined to the ankles. Among older children, creatitis or parotitis, neurologic manifestations,
purpura can be the sole cutaneous manifestation. and myocardial or muscle involvement [464].
Among preschool children, urticaria can occur
without purpura, although localized edema of the
dorsal surfaces of the hands and feet, face, and Renal Manifestations
scalp can occur [16, 420]. As the primary lesions
fade, new crops of purpura commonly recur as Renal manifestations occur at any time. The first
long as 3 months after onset and occasionally urinary abnormality usually is noticed after other
much longer [26, 15, 23] and sometimes are symptoms, but hematuria sometimes is the initial
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1013

feature. Among 80 % of children with a urinary and hypoalbuminemic edema [26]. Such clinical
abnormality, the first abnormality is detected presentation is most often associated with severe
within 4 weeks of onset of the illness. In most of crescentic glomerulonephritis.
the remainder, the urinary abnormality develops
within the next 8 weeks [15]. Recurrences of renal
manifestations are common and appear to be par- Laboratory Features
ticularly so among patients with severe renal dam-
age. The relapses can occur at the time of an upper There are no specific laboratory findings of HSP.
respiratory tract infection. There are no correla- Laboratory investigation needed is mainly
tions between the severity of extrarenal manifes- directed at assessment of the extent of renal
tations and the severity of renal symptoms. involvement. If purpura is absent but has been
Several studies have intended to identify pre- suggested by the patient’s recent history, the
dictive factors for developing of nephritis in child- feces are tested for occult blood, the finding of
hood HSP [423–427]. These studies revealed that which supports a diagnosis of HSP. If there is any
an older age at onset, persistent purpura, severe doubt about the cause of the purpura, a full blood
bowel angina, and relapse were identified as fac- cell count and coagulation screen is performed.
tors associated with the occurrence of nephritis. The level of clotting factor XIII, a fibrinstabilizing
A recent prospective study [428] clarified that the factor, is markedly low among children with HSP.
risk factors for developing nephritis were age over Although the serum level IgA is high in a large
8 years at onset, abdominal pain, and recurrence number of cases of HSP, measuring IgA for
of HSP disease. This study also showed that individual patients does not contribute to
patients with two or three risk factors developed management.
nephritis in 63 % and 87 % of cases, In every case of HSP, the urine is routinely
respectively [428]. tested for blood and protein at onset and at least
Minimal renal involvement may cause no more weekly until systemic signs have resolved. If uri-
than transient microscopic hematuria, detectable nary abnormalities are found, proteinuria is quan-
only with routine urinalysis during the acute ill- tified, and serum levels of total protein and
ness. The illness also presents with initial macro- albumin are measured. Serum creatinine level is
scopic hematuria that usually lasts a few days but measured to estimate renal function.
can last several weeks followed by microscopic
hematuria that can persist for months or years.
Macroscopic hematuria may be accompanied by Clinicopathologic Correlations
transient heavy proteinuria. Persistence of heavy
proteinuria of more than 1 g/day per square meter Renal biopsy rarely is needed for diagnosis, but it
of body surface area with microscopic hematuria is used to assess the severity of glomerulonephri-
usually is usually associated with significant his- tis. Biopsy is required only in the care of patients
tologic lesions. In these cases, blood pressure and with severe initial manifestations (nephritic,
renal function are normal. Impairment of renal nephrotic, or decreased renal function) or with
function is rare and occurs with acute nephritic heavy proteinuria >1 g/day/m2 that persists lon-
syndrome associated with combination ger than 1 month. The biopsy indication for with
hypervolemia, oliguria, and hypertension [26]. slight proteinuria <1 g/day/m2 that persists longer
Massive proteinuria can cause nephrotic syn- than 3 month may be considered. The more severe
drome (serum level of albumin less than 25 g/l) the clinical presentation, the larger is the percent-
with pitting edema of the face and ankles and age of glomeruli affected by crescents and seg-
sometimes ascites. The most severe clinical pre- mental lesions. The findings [26, 27, 33, 35],
sentation is mixed nephritic-nephrotic syndrome summarized in Table 6, show that (a) patients
in which hematuria, hypertension, and impair- with initial macroscopic or persistent microscopic
ment renal function are combined with proteinuria hematuria who do not have persistent heavy
1014 K. Nakanishi and N. Yoshikawa

Table 6 Clinicopathologic correlations in Henoch- Among older children and adults, the combi-
Schönlein nephritis nation of joint symptoms and purpura makes it
Approximate necessary to consider systemic lupus
Biopsy risk of renal erythematosus and microscopic polyangiitis as
Clinical presentation grade failure (%)
alternative diagnoses [23]. The light microscopic
Macroscopic or I–II, <5
microscopic hematuria, rarely findings of glomeruli in patients with systemic
proteinuria minimal or III lupus erythematosus and those with HSN are sim-
absent ilar and may be indistinguishable. In systemic
Hematuria and I–IV 15 lupus erythematosus, however, glomerular IgA
persistent heavy deposits, when present, are less prominent than
proteinuria
IgG deposits, and C1q deposits almost always are
Acute nephritic II–IV 15
syndrome present. Antinuclear antibodies usually are absent
Nephrotic syndrome II–IV, 40 from patients with HSN. The differential diagno-
rarely I sis between HSN and the microscopic polyangiitis
or V is difficult. Mesangial IgA deposits are absent in
Nephritic-nephrotic II–V, >50 polyarteritis nodosa. Therefore the diagnosis of
syndrome mostly
V the microscopic polyangiitis is considered when
From White and Yoshikawa [463]
any patient has clinical signs of HSP if mesangial
IgA deposits are not found [48].

proteinuria have glomerular lesions of grade III or


less; (b) those with nephritis or nephrosis at onset Treatment of HSP
of disease or persistent heavy proteinuria have a
10–20 % likelihood of having grade IV to V Although corticosteroids enhance the rate of res-
lesions; (c) a mixed nephritic-nephrotic presenta- olution of the arthritis and abdominal pain, their
tion carries a 60 % risk of development of grade effectiveness in the prevention of nephritis is
IV to V lesions; and (d) most grade I to III lesions debated. Saulsburry in an uncontrolled study of
resolve, whereas an increasing proportion of severe HSP cases concluded the corticosteroid
grade IV to V lesions do not. therapy does not prevent nephritis [429]. Con-
versely, Mollica et al. found that early prednisone
treatment reduced the incidence of microscopic
Differential Diagnosis of HSP hematuria, but this symptom is not associated
with a high risk of progression [430]. Ronkainen
The distribution of the rash and the accompanying et al. performed a randomized control study and
gastrointestinal and articular lesions usually is so concluded that prednisone does not prevent the
characteristic that the diagnosis is not in doubt. development of renal symptoms but is effective in
Difficulty arises in rare instances when the skin treating them [431]. Dudley et al. in a large ran-
eruption consists entirely of urticarial lesions domized, double-blind, placebo-controlled trial
without purpura. Although it is not feasible to found no evidence that early treatment with pred-
diagnose HSP in the absence of cutaneous mani- nisolone reduces the prevalence of proteinuria
festations, there is anecdotal evidence that it 12 months after disease onset [432]. Therefore,
occurs without skin lesions, albeit rarely [464]. the existing evidence does not support prednisone
Hypoalbuminemic edema sometimes is found in in the prevention of nephritis.
the absence of heavy proteinuria and can be The clinical spectrum of HSN ranges from the
caused by proteinlosing enteropathy [421]. Quan- relatively common transient isolated microscopic
titative assessment of proteinuria therefore is hematuria to nephrotic syndrome, rapidly pro-
essential. gressive glomerulonephritis, and renal failure.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1015

In mild cases, no treatment is needed if the serum albumin >2.5 g/dl) were treated with
urine is adequately evaluated, and ambulatory angiotensin-converting enzyme inhibitors and/or
care may suffice. The prolonged bed rest custom- angiotensin receptor blockers. Patients (n = 19)
ary in the past occasionally led to complications with HSN exceeding grade III or Alb 2.5 g/dl
such as femoral venous thrombosis and psycho- received combination therapy comprising pred-
social problems [26]. Although severe abdominal nisolone, immunosuppressants, warfarin, and
pain and vomiting are self-limiting, resolution can dipyridamole. All patients showed resolution of
be hastened by means of administration of gluco- proteinuria without renal dysfunction during the
corticoids [433]. Utani et al. [434] found that observation period (3.8  0.4 years). Their find-
factor XIII concentrate replacement resolved ings support those of some earlier reports that
severe abdominal symptoms in the care of adults treatment strategies for HSN should depend on
with HSP associated with a decreased level of the histologic and clinical severity. Furthermore,
factor XIII activity. aggressive therapies, particularly combination
There is no therapy of proven value for HSN, therapies, are unnecessary for moderate to
although some regimens have been proposed and severe HSN.
tested, with controversial results. These include A recent meta-analyses [449, 450] has evaluated
(a) glucocorticoids and immunosuppressive drugs the benefits and harms of different agents (used
and (b) plasma exchange to remove circulating singularly or in combination) compared with pla-
IgA immune complexes. Retrospective studies cebo or no treatment or another agent for the pre-
suggested that oral glucocorticoids were of no vention or treatment of kidney disease in patients
benefit [435, 26, 15, 33]. Similarly, oral glucocor- with HSP. It has revealed that data from randomized
ticoids with azathioprine, chlorambucil, or cyclo- controlled trials for any intervention used in improve
phosphamide do not seem to be beneficial [15, 26, kidney outcomes in children with HSP are very
435, 436]. A beneficial effect of methylpredniso- sparse except for short-term prednisone and that
lone pulse therapy followed by oral prednisone there was no evidence of benefit of prednisone in
was reported in a prospective case series of severe preventing serious long-term kidney disease in HSP.
HSN in comparison with historical controls at the The Kidney Disease Improving Global Out-
same institution [437]. The beneficial effect of come (KDIGO) initiative published guidelines for
methylprednisolone pulses in combination with the treatment of HSN [465]. Due to the lack of
immunosuppressive agents such as cyclophos- evidence-based data for HSN treatment and simi-
phamide or mizoribine has also been reported larities between HSN and primary IgAN, the
[438, 439]. Cyclosporine A therapy has given KDIGO guidelines proposed the same treatment
promising results in case series, although some for both diseases when the clinical conditions are
patients become cyclosporine dependent similar. Although the KDIGO guidelines have been
[440–443]. Plasma exchange combined with glu- introduced, clinicians following the KDIGO guide-
cocorticoids and immunosuppressive drugs may lines on the treatment of HSN may face the risk of
be of value in the care of patients with rapidly delaying the initiation of effective treatment and
progressive crescentic disease [444–447]. increasing the risk of CKD over the long term.
The use of angiotensin-converting enzyme Consequently, it should be expected that pediatric
inhibitor or angiotensin receptor blockers may be nephrologists would be reluctant to follow those
useful in patients with moderate HSN. Recently, guidelines for fear of undertreatment [451, 452].
Ninchoji et al. [448] reported the results of retro-
spective study for the efficacy of treatment for
HSN. Renal biopsy was performed in patients Prognosis
with nephrotic syndrome or persistent proteinuria
for more than 3 months, and patients were classi- The long-term morbidity and mortality of HSP are
fied by treatment. Patients (n = 31) with moder- almost exclusively attributable to renal disease.
ately severe HSN (histological grade I–III and Several centers have reported the correlation of
1016 K. Nakanishi and N. Yoshikawa

Table 7 Correlations between biopsy grade and outcome in Henoch-Schönlein nephritis


ISKDC biopsy Total number of Slight proteinuria, Heavy Renal
grade patients Normal urine hematuria, or both proteinuria failure
I 22 14 8 0 0
II 22 15 6 1 0
III 53 38 10 4 1
IV 12 5 3 1 3
V 12 0 2 1 9
Total 121 72 29 7 13
All values are numbers of patients

both clinical presentation and renal morphology between histologic grade and outcome among our
with outcome [15, 26, 32, 33, 35, 289, 121 children with HSN who underwent biopsy
453–457]. We found 14 (11 %) of 122 children and whose conditions were followed for at least
with renal involvement [27] to have chronic renal 2 years is summarized in Table 7. The table shows
failure or to have died as a result. A further seven a progressively worsening outcome with increas-
patients (6 %) had active disease. The overall ing histologic severity, as judged by the percent-
prognosis of HSP is better appreciated with results age of patients in each grade with either active
from two large series in which the conditions of disease or chronic renal failure: 0 % for grade I,
patients with or without nephritis were followed; 4 % for grade II, 9 % for grade III, 33 % for grade
the estimated incidences of end-stage renal failure IV, and 83 % for grade V. Of the 72 patients who
were 2 % [36] and 5 % [32]. completely recovered, 67 had fewer than 50 % of
The results of long-term follow-up studies their glomeruli affected by crescents or segmental
show that the clinical presentation has no prog- lesions. In contrast, among the 13 patients with
nostic value for individual patients. It is clear, chronic renal failure, 12 had more than 50 % of
however, that children who come to medical glomeruli affected by crescents or segmental
attention with slight proteinuria or hematuria lesions.
have an excellent prognosis and that patients Goldstein et al. [35] observed that of 78 chil-
with persistent heavy proteinuria, acute nephritic dren followed for 23 years, 17 (22 %) had deteri-
syndrome, or nephrotic syndrome are at risk of orated clinically. Included in this group were
chronic renal failure. Results of a 23-year follow- seven children who had apparently completely
up study by Goldstein et al. [35] indicated that recovered at 10 years. Furthermore, 36 % of
approximately 15 % of patients with nephritis at 44 pregnancies were complicated by hypertension
onset of HSP or persistent heavy proteinuria, 40 % and/or persistent proteinuria. The late develop-
of those with nephrosis, and fewer than 50 % of ment of hypertension and proteinuria after a
those with a mixed nephritic-nephrotic onset have period of normality and the proteinuria of preg-
ongoing urinary abnormalities and that many of nancy, together with the progressive decline in
these patients ultimately have renal failure renal function that can follow initial improve-
(Table 6). As noteworthy facts, it has been ment, are consistent with a sequence of glomeru-
reported that some children with mild renal symp- lar hyperfiltration followed by secondary
toms have poor long-term outcome [458–460]. glomerulosclerosis [461] in which glomerular
The proportion of glomeruli with crescents or destruction due to the original disease has been
segmental lesions (sclerosis, adhesions, thrombo- extensive. Because most patients found by Gold-
sis, or necrosis) seems to be the most important stein et al. [35] to have residual abnormalities had
prognostic indicator. The larger the number of no symptoms, it follows that children who have
glomeruli affected by crescents or segmental HSN with a severe clinical onset or biopsy grade
lesions, the poorer is the prognosis. The relation need almost indefinite follow-up care.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1017

Recurrence in Renal Allograft 10. White RHR, Yoshikawa N, Feehally J. IgA nephrop-
athy and Henoch-Schönlein nephritis. In: Barratt TM,
Avner ED, Harmon WE, editors. Pediatric nephrol-
In HSN, clinical recurrence in renal allografts is ogy. 4th ed. Baltimore: Lippincott Williams &
rare, but recurrence of the mesangial IgA deposits Wilkins; 1999. p. 691–706.
is frequent [129–131, 462]. Recurrent disease 11. Yoshikawa N, Nakanishi K, Iijima K. Henoch-
appears to be strongly associated with living- Schönlein purpura. In: Neilson EG, Couser WG, edi-
tors. Immunologic renal diseases. 2nd ed. Philadelphia:
related donor transplantation. The significance is Lippincott Williams & Wilkins; 2001. p. 1127–40.
unclear, but the association of disease recurrence 12. Rook A. William Heberden’s cases of anaphylactoid
with living-related donor transplantation may purpura. Arch Dis Child. 1958;33(169):271.
imply a genetic predisposition. In a report by 13. Osler W. The visceral lesions of purpura and allied
conditions. Br Med J. 1914;1(2775):517–25.
Hasegawa et al. [131], there were 9 histologic 14. Bunchman TE, Mauer SM, Sibley RK, Vernier RL.
recurrences among 12 living-related donor grafts, Anaphylactoid purpura: characteristics of 16 patients
5 of them associated with clinical manifestations. who progressed to renal failure. Pediatr Nephrol.
In contrast, clinical recurrence was not observed 1988;2(4):393–7.
15. Levy M, Broyer M, Arsan A, Levy-Bentolila D,
in 5 cadaveric grafts, and 4 of these had no IgA Habib R. Anaphylactoid purpura nephritis in child-
deposits at biopsy. Habib et al. [48] found asymp- hood: natural history and immunopathology. Adv
tomatic IgA deposits in 11 of the 13 cadaveric Nephrol Necker Hosp. 1976;6:183–228.
grafts, but only 1 of 13 patients had a clinical 16. Gairdner D. The Schonlein-Henoch syndrome (ana-
phylactoid purpura). Q J Med. 1948;17(66):95–122.
recurrence. 17. Ozen S, Ruperto N, Dillon MJ, Bagga A, Barron K,
Davin JC, et al. EULAR/PReS endorsed consensus
criteria for the classification of childhood vasculiti-
References des. Ann Rheum Dis. 2006;65(7):936–41.
18. Jennette JC, Falk RJ, Andrassy K, Bacon PA, Churg J,
Gross WL, et al. Nomenclature of systemic vasculit-
1. Berger J, Hinglais N. Les depots intercapillaire d’lgA- ides. Proposal of an international consensus confer-
IgG. J Urol Nephrol. 1968;74:694–5. ence. Arthritis Rheum. 1994;37(2):187–92.
2. D’Amico G. The commonest glomerulonephritis in 19. Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC,
the world: IgA nephropathy. Q J Med. 1987;64(245): Ferrario F, et al. 2012 revised international Chapel
709–27. Hill consensus conference nomenclature of vasculiti-
3. Emancipator SN, Gallo GR, Lamm ME. IgA des. Arthritis Rheum. 2013;65(1):1–11.
nephropathy: perspectives on pathogenesis and clas- 20. Rodicio JL. Idiopathic IgA nephropathy. Kidney Int.
sification. Clin Nephrol. 1985;24(4):161–79. 1984;25(4):717–29.
4. Yoshikawa N, Ito H, Yoshiara S, Nakahara C, 21. Sehic AM, Gaber LW, Roy 3rd S, Miller PM,
Yoshiya K, Hasegawa O, et al. Clinical course of Kritchevsky SB, Wyatt RJ. Increased recognition of
immunoglobulin A nephropathy in children. J Pediatr. IgA nephropathy in African-American children.
1987;110(4):555–60. Pediatr Nephrol. 1997;11(4):435–7.
5. Yoshikawa N, Iijima K, Maehara K, Yoshiara S, 22. Clarkson AR, Woodroffe AJ, Aarons I. IgA nephrop-
Yoshiya K, Matsuo T, et al. Mesangial changes in athy and Henoch-Schönlein purpura. In: Schrier R,
IgA nephropathy in children. Kidney Int. 1987; Gottschalk C, editors. Diseases of the kidney. Boston:
32(4):585–9. Little, Brown; 1996. p. 1645–70.
6. Yoshikawa N, Iijima K, Matsuyama S, Suzuki J, 23. Cream JJ, Gumpel JM, Peachey RD. Schonlein-
Kameda A, Okada S, et al. Repeat renal biopsy in Henoch purpura in the adult. A study of 77 adults
children with IgA nephropathy. Clin Nephrol. with anaphylactoid or Schonlein-Henoch purpura. Q J
1990;33(4):160–7. Med. 1970;39(156):461–84.
7. Wyatt RJ, Kritchevsky SB, Woodford SY, Miller PM, 24. Sinniah R, Feng PH, Chen BT. Henoch-Schoenlein
Roy 3rd S, Holland NH, et al. IgA nephropathy: long- syndrome: a clinical and morphological study of renal
term prognosis for pediatric patients. J Pediatr. biopsies. Clin Nephrol. 1978;9(6):219–28.
1995;127(6):913–9. 25. Fogazzi GB, Pasquali S, Moriggi M, Casanova S,
8. Hogg RJ, Silva FG, Wyatt RJ, Reisch JS, Argyle JC, Damilano I, Mihatsch MJ, et al. Long-term outcome
Savino DA. Prognostic indicators in children with of Schonlein-Henoch nephritis in the adult. Clin
IgA nephropathy – report of the Southwest Pediatric Nephrol. 1989;31(2):60–6.
Nephrology Study Group. Pediatr Nephrol. 1994; 26. Meadow SR, Glasgow EF, White RH, Moncrieff MW,
8(1):15–20. Cameron JS, Ogg CS. Schonlein-Henoch nephritis. Q J
9. White RH. Henoch-Schonlein nephritis. Br Med Med. 1972;41(163):241–58.
J. 1971;2(5762):653–4.
1018 K. Nakanishi and N. Yoshikawa

27. Yoshikawa N, Ito H, Yoshiya K, Nakahara C, 43. Philpott MG. The Schoenlein-Henoch syndrome
Yoshiara S, Hasegawa O, et al. Henoch-Schoenlein in childhood with particular reference to the occur-
nephritis and IgA nephropathy in children: a compar- rence of nephritis. Arch Dis Child. 1952;27(135):
ison of clinical course. Clin Nephrol. 1987;27(5): 480–1.
233–7. 44. Derham RJ, Rogerson MM. The Schonlein-Henoch
28. Knight JF. The rheumatic poison: a survey of some syndrome with particular reference to renal sequelae.
published investigations of the immunopathogenesis Arch Dis Child. 1956;31(159):364–8.
of Henoch-Schonlein purpura. Pediatr Nephrol. 45. Koskimies O, Rapola J, Savilahti E, Vilska J. Renal
1990;4(5):533–41. involvement in Schonlein-Henoch purpura. Acta
29. Nielsen HE. Epidemiology of Schonlein-Henoch pur- Paediatr Scand. 1974;63(3):357–63.
pura. Acta Paediatr Scand. 1988;77(1):125–31. 46. Ilan Y, Naparstek Y. Schonlein-Henoch syndrome in
30. Stewart M, Savage JM, Bell B, McCord B. Long term adults and children. Semin Arthritis Rheum. 1991;
renal prognosis of Henoch-Schonlein purpura in an 21(2):103–9.
unselected childhood population. Eur J Pediatr. 47. Zurowska AM, Wrzolkowa T, Uszycka-Karcz M.
1988;147(2):113–5. Henoch-Schonlein nephritis in children – a clinico-
31. Lewis IC. The Schonlein-Henoch syndrome (anaphy- pathological study. Int J Pediatr Nephrol. 1985;
lactoid purpura) compared with certain features of 6(3):183–8.
nephritis and rheumatism. Arch Dis Child. 1955; 48. Habib R, Niaudet P, Levy M. Schönlein-Henoch pur-
30(151):212–6. pura nephritis and IgA nephropathy. In: Tisher CBB,
32. Kobayashi O, Wada H, Okawa K, Takeyama editor. Renal pathology: with clinical and functional
I. Schonlein-Henoch’s syndrome in children. Contrib correlations. 2nd ed. Philadelphia: JB Lippincott;
Nephrol. 1975;4:48–71. 1994. p. 472–523.
33. Counahan R, Winterborn MH, White RH, Heaton JM, 49. Feehally J. Immune mechanisms in glomerular IgA
Meadow SR, Bluett NH, et al. Prognosis of Henoch- deposition. Nephrol Dial Transplant. 1988;
Schonlein nephritis in children. Br Med J. 1977; 3(4):361–78.
2(6078):11–4. 50. Odum J, Peh CA, Clarkson AR, Bannister KM, Sey-
34. Yoshikawa N, White RH, Cameron AH. Prognostic mour AE, Gillis D, et al. Recurrent mesangial IgA
significance of the glomerular changes in Henoch- nephritis following renal transplantation. Nephrol
Schoenlein nephritis. Clin Nephrol. 1981;16(5): Dial Transplant. 1994;9(3):309–12.
223–9. 51. Suzuki S, Nakatomi Y, Sato H, Tsukada H,
35. Goldstein AR, White RH, Akuse R, Chantler C. Arakawa M. Haemophilus parainfluenzae antigen
Long-term follow-up of childhood and antibody in renal biopsy samples and serum of
Henoch-Schonlein nephritis. Lancet. 1992;339 patients with IgA nephropathy. Lancet. 1994;
(8788):280–2. 343(8888):12–6.
36. Koskimies O, Mir S, Rapola J, Vilska J. 52. Ogura Y, Suzuki S, Shirakawa T, Masuda M,
Henoch-Schonlein nephritis: long-term prognosis of Nakamura H, Iijima K, et al. Haemophilus
unselected patients. Arch Dis Child. 1981;56(6): parainfluenzae antigen and antibody in children with
482–4. IgA nephropathy and Henoch-Schonlein nephritis.
37. Galla JH, Kohaut EC, Alexander R, Mestecky J. Am J Kidney Dis. 2000;36(1):47–52.
Racial difference in the prevalence of IgA-associated 53. Koyama A, Sharmin S, Sakurai H, Shimizu Y,
nephropathies. Lancet. 1984;2(8401):522. Hirayama K, Usui J, et al. Staphylococcus aureus
38. Levy M. Do genetic factors play a role in Berger’s cell envelope antigen is a new candidate for the induc-
disease? Pediatr Nephrol. 1987;1(3):447–54. tion of IgA nephropathy. Kidney Int. 2004;
39. Bhuyan UN, Tiwari SC, Malaviya AN, Srivastava 66(1):121–32.
RN, Dash SC, Malhotra KK. Immunopathology & 54. Schmitt R, Lindahl G, Karpman D. Antibody
prognosis in Henoch-Schonlein glomerulonephritis. response to IgA-binding streptococcal M proteins in
Indian J Med Res. 1986;83:33–40. children with IgA nephropathy. Nephrol Dial Trans-
40. Farley TA, Gillespie S, Rasoulpour M, Tolentino N, plant. 2010;25(10):3434–6.
Hadler JL, Hurwitz E. Epidemiology of a cluster of 55. Coppo R, Camilla R, Alfarano A, Balegno S,
Henoch-Schonlein purpura. Am J Dis Child. Mancuso D, Peruzzi L, et al. Upregulation of the
1989;143(7):798–803. immunoproteasome in peripheral blood mononuclear
41. Ostergaard JR, Storm K, Lamm LU. Lack of associ- cells of patients with IgA nephropathy. Kidney Int.
ation between HLA and Schoenlein-Henoch Purpura. 2009;75(5):536–41.
Tissue Antigens. 1990;35(5):234–5. 56. Ayoub EM, Hoyer J. Anaphylactoid purpura: strepto-
42. Jin DK, Kohsaka T, Kobayashi N. The polymorphism coccal antibody titers and beta1c-globulin levels.
of the immunoglobulin heavy chain switch region J Pediatr. 1969;75(2):193–201.
gene in IgA nephropathy, Henoch-Schonlein nephri- 57. Meadow SR, Scott DG. Berger disease: Henoch-
tis and idiopathic nephrotic syndrome. Pediatr Schonlein syndrome without the rash. J Pediatr.
Nephrol. 1993;7(4):449–51. 1985;106(1):27–32.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1019

58. Garty BZ, Danon YL, Nitzan M. Schoenlein-Henoch 75. Schmitt R, Carlsson F, Morgelin M, Tati R,
purpura associated with hepatitis A infection. Am J Lindahl G, Karpman D. Tissue deposits of
Dis Child. 1985;139(6):547. IgA-binding streptococcal M proteins in IgA
59. Maggiore G, Martini A, Grifeo S, De Giacomo C, nephropathy and Henoch-Schonlein purpura. Am J
Scotta MS. Hepatitis B virus infection and Schonlein- Pathol. 2010;176(2):608–18.
Henoch purpura. Am J Dis Child. 1984;138(7): 76. Hsu SI, Ramirez SB, Winn MP, Bonventre JV, Owen
681–2. WF. Evidence for genetic factors in the development
60. Rasmussen NH. Henoch-Schonlein purpura after and progression of IgA nephropathy. Kidney Int.
yersiniosis. Arch Dis Child. 1982;57(4):322–3. 2000;57(5):1818–35.
61. Roza M, Galbe M, Gonzalez Baschwitz C, 77. Rambausek MH, Waldherr R, Ritz E. Immunogenetic
Fernandez M, Miguell MA. Henoch-Schoenlein findings in glomerulonephritis. Kidney Int Suppl.
purpura after shigellosis. Clin Nephrol. 1983;20 1993;39:S3–8.
(5):269. 78. Schmidt S, Ritz E. Genetic factors in IgA nephropa-
62. Liew SW, Kessel I. Letter: mycoplasmal pneumonia thy. Ann Med Interne. 1999;150(2):86–90.
preceding Henoch-Schonlein purpura. Arch Dis 79. Julian BA, Quiggins PA, Thompson JS, Woodford
Child. 1974;49(11):912–3. SY, Gleason K, Wyatt RJ. Familial IgA nephropathy.
63. Alwar AJ. Schonlein-Henoch syndrome: a case report Evidence of an inherited mechanism of disease. N
and literature review. East Afr Med J. 1982;59(5): Engl J Med. 1985;312(4):202–8.
357–60. 80. Wyatt RJ, Rivas ML, Julian BA, Quiggins PA,
64. Jimenez EL, Dorrington HJ. Vaccination and Henoch- Woodford SY, McMorrow RG, et al. Regionalization
Schoenlein purpura. N Engl J Med. 1968;279 in hereditary IgA nephropathy. Am J Hum Genet.
(21):1171. 1987;41(1):36–50.
65. Patel U, Bradley JR, Hamilton DV. Henoch- 81. Scolari F, Amoroso A, Savoldi S, Mazzola G, Prati E,
Schonlein purpura after influenza vaccination. Br Valzorio B, et al. Familial clustering of IgA nephrop-
Med J (Clin Res Ed). 1988;296(6639):1800. athy: further evidence in an Italian population. Am J
66. Burke DM, Jellinek HL. Nearly fatal case of Kidney Dis. 1999;33(5):857–65.
Schoenlein-Henoch syndrome following insect bite. 82. Gharavi AG, Yan Y, Scolari F, Schena FP, Frasca GM,
AMA Am J Dis Child. 1954;88(6):772–4. Ghiggeri GM, et al. IgA nephropathy, the most com-
67. Rogers PW, Bunn Jr SM, Kurtzman NA, White mon cause of glomerulonephritis, is linked to 6q22-
MG. Schonlein-Henoch syndrome associated with 23. Nat Genet. 2000;26(3):354–7.
exposure to cold. Arch Intern Med. 1971;128(5): 83. Bisceglia L, Cerullo G, Forabosco P, Torres DD,
782–6. Scolari F, Di Perna M, et al. Genetic heterogeneity
68. Talbot D, Craig R, Falconer S, Tomson D, Milne in Italian families with IgA nephropathy: suggestive
DD. Henoch-Schonlein purpura secondary to trauma. linkage for two novel IgA nephropathy loci. Am J
Arch Dis Child. 1988;63(9):1114–5. Hum Genet. 2006;79(6):1130–4.
69. Kimmel PL, Phillips TM, Ferreira-Centeno A, 84. Paterson AD, Liu XQ, Wang K, Magistroni R,
Farkas-Szallasi T, Abraham AA, Garrett CT. Brief Song X, Kappel J, et al. Genome-wide linkage scan
report: idiotypic IgA nephropathy in patients with of a large family with IgA nephropathy localizes a
human immunodeficiency virus infection. N Engl J novel susceptibility locus to chromosome 2q36. J Am
Med. 1992;327(10):702–6. Soc Nephrol. 2007;18(8):2408–15.
70. Furukawa T, Hisao O, Furuta S, Shigematsu H. 85. Feehally J, Farrall M, Boland A, Gale DP, Gut I,
Henoch-Schonlein purpura with nephritis in a patient Heath S, et al. HLA has strongest association with
with Behcet’s disease. Am J Kidney Dis. IgA nephropathy in genome-wide analysis. J Am Soc
1989;13(6):497–500. Nephrol. 2010;21(10):1791–7.
71. Yamabe H, Ozawa K, Fukushi K, Kubota H, 86. Gharavi AG, Kiryluk K, Choi M, Li Y, Hou P, Xie J,
Ohsawa H, Akitsu H, et al. IgA nephropathy and et al. Genome-wide association study identifies sus-
Henoch-Schonlein purpura nephritis with anterior ceptibility loci for IgA nephropathy. Nat Genet.
uveitis. Nephron. 1988;50(4):368–70. 2011;43(4):321–7.
72. Cairns SA, Mallick NP, Lawler W, Williams G. Squa- 87. Yu XQ, Li M, Zhang H, Low HQ, Wei X, Wang JQ,
mous cell carcinoma of bronchus presenting with et al. A genome-wide association study in Han Chi-
Henoch-Schonlein purpura. Br Med J. 1978; nese identifies multiple susceptibility loci for IgA
2(6135):474–5. nephropathy. Nat Genet. 2012;44(2):178–82.
73. Mitsuhashi H, Mitsuhashi M, Tsukada Y, Yano S, 88. Schena FP, Cerullo G, Rossini M, Lanzilotta SG,
Naruse T. Henoch-Schonlein purpura in rheumatoid D’Altri C, Manno C. Increased risk of end-stage
arthritis. J Rheumatol. 1994;21(6):1138–40. renal disease in familial IgA nephropathy. J Am Soc
74. Masuda M, Nakanishi K, Yoshizawa N, Iijima K, Nephrol. 2002;13(2):453–60.
Yoshikawa N. Group A streptococcal antigen in the 89. Shimomura M, Yoshikawa N, Iijima K, Nakamura H,
glomeruli of children with Henoch-Schonlein nephri- Miyazaki M, Sakai H. Polymorphism of immuno-
tis. Am J Kidney Dis. 2003;41(2):366–70. globulin heavy chain switch region gene in children
1020 K. Nakanishi and N. Yoshikawa

with severe IgA nephropathy. Clin Nephrol. 1995;43 103. Beerman I, Novak J, Wyatt RJ, Julian BA, Gharavi
(4):211–5. AG. The genetics of IgA nephropathy. Nat Clin Pract
90. Yano N, Asakura K, Endoh M, Abe Y, Nomoto Y, Nephrol. 2007;3(6):325–38.
Sakai H, et al. Polymorphism in the Ialpha1 germ-line 104. Schena FP, D’Altri C, Cerullo G, Manno C, Gesualdo
transcript regulatory region and IgA productivity in L. ACE gene polymorphism and IgA nephropathy: an
patients with IgA nephropathy. J Immunol. 1998;160 ethnically homogeneous study and a meta-analysis.
(10):4936–42. Kidney Int. 2001;60(2):732–40.
91. Tanaka R, Iijima K, Murakami R, Koide M, 105. Xue C, Nie W, Xu J, Dai B, Mei CL. C509T and
Nakamura H, Yoshikawa N. ACE gene polymor- T869C polymorphisms of transforming growth factor
phism in childhood IgA nephropathy: association beta1 and the risk of IgA nephropathy: a meta-
with clinicopathologic findings. Am J Kidney Dis. analysis. Chin Med J (Engl). 2013;126(23):4562–7.
1998;31(5):774–9. 106. Takei T, Iida A, Nitta K, Tanaka T, Ohnishi Y,
92. Maruyama K, Yoshida M, Nishio H, Shirakawa T, Yamada R, et al. Association between single-
Kawamura T, Tanaka R, et al. Polymorphisms of nucleotide polymorphisms in selectin genes and
renin-angiotensin system genes in childhood IgA immunoglobulin A nephropathy. Am J Hum Genet.
nephropathy. Pediatr Nephrol. 2001;16(4):350–5. 2002;70(3):781–6.
93. Nakanishi K, Sako M, Yata N, Aoyagi N, Nozu K, 107. Akiyama F, Tanaka T, Yamada R, Ohnishi Y,
Tanaka R, et al. A-20C angiotensinogen gene poly- Tsunoda T, Maeda S, et al. Single-nucleotide poly-
morphism and proteinuria in childhood IgA nephrop- morphisms in the class II region of the major histo-
athy. Pediatr Nephrol. 2004;19(2):144–7. compatibility complex in Japanese patients with
94. Qin YH, Zhou TB, Su LN, Lei FY, Huang WF, Zhao immunoglobulin A nephropathy. J Hum Genet.
YJ. Association between ACE polymorphism and 2002;47(10):532–8.
risk of IgA nephropathy: a meta-analysis. J Renin 108. Obara W, Iida A, Suzuki Y, Tanaka T, Akiyama F,
Angiotensin Aldosterone Syst. 2011;12(3):215–23. Maeda S, et al. Association of single-nucleotide poly-
95. Zhou TB, Ou C, Qin YH, Luo W. A meta-analysis of morphisms in the polymeric immunoglobulin recep-
the association between angiotensin-converting tor gene with immunoglobulin A nephropathy (IgAN)
enzyme insertion/deletion gene polymorphism and in Japanese patients. J Hum Genet. 2003;48(6):
Henoch-Schonlein purpura nephritis risk in Asian 293–9.
children. Clin Exp Rheumatol. 2012;30(2):315–6. 109. Brogan PA. What’s new in the aetiopathogenesis of
96. Vuong MT, Lundberg S, Gunnarsson I, Wramner L, vasculitis? Pediatr Nephrol. 2007;22(8):1083–94.
Seddighzadeh M, Hahn-Zoric M, et al. Genetic vari- 110. He X, Yu C, Zhao P, Ding Y, Liang X, Zhao Y,
ation in the transforming growth factor-beta1 gene is et al. The genetics of Henoch-Schonlein purpura: a
associated with susceptibility to IgA nephropathy. systematic review and meta-analysis. Rheumatol Int.
Nephrol Dial Transplant. 2009;24(10):3061–7. 2013;33(6):1387–95.
97. Tanaka R, Iijima K, Xu H, Inoue Y, Murakami R, 111. Amoroso A, Danek G, Vatta S, Crovella S, Berrino M,
Shirakawa T, et al. Role of platelet-activating factor Guarrera S, et al. Polymorphisms in angiotensin-
acetylhydrolase gene mutation in Japanese childhood converting enzyme gene and severity of renal disease
IgA nephropathy. Am J Kidney Dis. 1999;34(2): in Henoch-Schoenlein patients. Italian Group of
289–95. Renal Immunopathology. Nephrol Dial Transplant.
98. Li R, Xue C, Li C, Lou T, Tao Y, Li Y, et al. TRAC 1998;13(12):3184–8.
variants associate with IgA nephropathy. J Am Soc 112. Di B, Li X, Song L, Wang Q, Liu S. Association study
Nephrol. 2009;20(6):1359–67. of ACE and eNOS single nucleotide polymorphisms
99. Lin J, Huang Y, Zhang X, Chen J, Sheng H. Associ- with Henoch-Schonlein purpura nephritis. Mol Med
ation of miR-146a rs2910164 with childhood IgA Rep. 2012;6(5):1171–7.
nephropathy. Pediatr Nephrol. 2014;29:1979–86. 113. Amoli MM, Mattey DL, Calvino MC, Garcia-Porrua-
100. Zhu L, Tang W, Li G, Lv J, Ding J, Yu L, C, Thomson W, Hajeer AH, et al. Polymorphism at
et al. Interaction between variants of two glycosyl- codon 469 of the intercellular adhesion molecule-1
transferase genes in IgA nephropathy. Kidney Int. locus is associated with protection against severe gas-
2009;76(2):190–8. trointestinal complications in Henoch-Schonlein pur-
101. Pirulli D, Crovella S, Ulivi S, Zadro C, Bertok S, pura. J Rheumatol. 2001;28(5):1014–8.
Rendine S, et al. Genetic variant of C1GalT1 contrib- 114. Rueda B, Perez-Armengol C, Lopez-Lopez S, Garcia-
utes to the susceptibility to IgA nephropathy. J Porrua C, Martin J, Gonzalez-Gay MA. Association
Nephrol. 2009;22(1):152–9. between functional haplotypes of vascular endothelial
102. Woo KT, Lau YK, Wong KS, Zhao Y, Chan growth factor and renal complications in Henoch-
CM. Parallel genotyping of 10,204 single nucleotide Schonlein purpura. J Rheumatol. 2006;33(1):69–73.
polymorphisms to screen for susceptible genes for 115. Gershoni-Baruch R, Broza Y, Brik R. Prevalence and
IgA nephropathy. Ann Acad Med Singapore. significance of mutations in the familial Mediterra-
2009;38(10):894–9. nean fever gene in Henoch-Schonlein purpura. J
Pediatr. 2003;143(5):658–61.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1021

116. Tunca M, Akar S, Onen F, Ozdogan H, 132. Davin JC, Dechenne C, Lombet J, Rentier B, Foidart
Kasapcopur O, Yalcinkaya F, et al. Familial Mediter- JB, Mahieu PR. Acute experimental glomerulone-
ranean fever (FMF) in Turkey: results of a nationwide phritis induced by the glomerular deposition of circu-
multicenter study. Medicine. 2005;84(1):1–11. lating polymeric IgA-concanavalin A complexes.
117. Ozcakar ZB, Yalcinkaya F, Cakar N, Acar B, Virchows Arch A Pathol Anat Histopathol.
Kasapcopur O, Uguten D, et al. MEFV mutations 1989;415(1):7–20.
modify the clinical presentation of Henoch-Schonlein 133. Conley ME, Cooper MD, Michael AF. Selective
purpura. J Rheumatol. 2008;35(12):2427–9. deposition of immunoglobulin A1 in immunoglobu-
118. He X, Lu H, Kang S, Luan J, Liu Z, Yin W, lin A nephropathy, anaphylactoid purpura nephritis,
et al. MEFV E148Q polymorphism is associated and systemic lupus erythematosus. J Clin Invest.
with Henoch-Schonlein purpura in Chinese children. 1980;66(6):1432–6.
Pediatr Nephrol. 2010;25(10):2077–82. 134. Komatsu N, Nagura H, Watanabe K, Nomoto Y,
119. Sakai H. Current views on pathogenesis and treat- Kobayashi K. Mesangial deposition of J chain-linked
ment. In: Andreucci VE, editor. International year polymeric IgA in IgA nephropathy. Nephron.
book of nephrology. Boston: Kluwer; 1989. p. 23–46. 1983;33(1):61–4.
120. Andresdottir MB, Hoitsma AJ, Assmann KJ, Wetzels 135. Bene MC, Faure G, Duheille J. IgA nephropathy:
JF. Favorable outcome of renal transplantation in characterization of the polymeric nature of mesangial
patients with IgA nephropathy. Clin Nephrol. deposits by in vitro binding of free secretory compo-
2001;56(4):279–88. nent. Clin Exp Immunol. 1982;47(3):527–34.
121. Yoshikawa N, Iijima K, Ito H. IgA nephropathy in 136. Valentijn RM, Radl J, Haayman JJ, Vermeer BJ,
children. Nephron. 1999;83(1):1–12. Weening JJ, Kauffmann RH, et al. Macromolecular
122. Yoshikawa N, Tanaka R, Iijima K. Pathophysiology IgA in the circulation and mesangial deposits in
and treatment of IgA nephropathy in children. Pediatr patients with primary IgA nephropathy. Contrib
Nephrol. 2001;16(5):446–57. Nephrol. 1984;40:87–92.
123. Rifai A, Small Jr P, Ayoub EM. Experimental IgA 137. Monteiro RC, Halbwachs-Mecarelli L, Berger J,
nephropathy. Factors governing the persistence of Lesavre P. Characteristics of eluted IgA in primary
IgA-antigen complexes in the circulation of mice. IgA nephropathy. Contrib Nephrol. 1984;40:107–11.
Contrib Nephrol. 1984;40:37–44. 138. Tomino Y, Endoh M, Suga T, Miura M, Kaneshige H,
124. Isaacs K, Miller F. Dextran-induced IgA nephropathy. Nomoto Y, et al. Prevalence of IgAl deposits in
Contrib Nephrol. 1984;40:45–50. Henoch-Schoenlein purpura (HSP) nephritis. Tokai J
125. Emancipator SN, Gallo GR, Lamm Exp Clin Med. 1982;7(4):527–32.
ME. Experimental IgA nephropathy induced by oral 139. Rajaraman S, Goldblum RM, Cavallo
immunization. J Exp Med. 1983;157(2):572–82. T. IgA-associated glomerulonephritides: a study
126. Levinsky RJ, Barratt TM. IgA immune complexes in with monoclonal antibodies. Clin Immunol
Henoch-Schonlein purpura. Lancet. 1979;2 Immunopathol. 1986;39(3):514–22.
(8152):1100–3. 140. Russell MW, Mestecky J, Julian BA, Galla
127. Kauffmann RH, Herrmann WA, Meyer CJ, Daha MR, JH. IgA-associated renal diseases: antibodies to envi-
Van Es LA. Circulating IgA-immune complexes in ronmental antigens in sera and deposition of immu-
Henoch-Schonlein purpura. A longitudinal study of noglobulins and antigens in glomeruli. J Clin
their relationship to disease activity and vascular Immunol. 1986;6(1):74–86.
deposition of IgA. Am J Med. 1980;69(6):859–66. 141. Egido J, Sancho J, Mampaso F, Lopez Trascasa M,
128. Coppo R, Basolo B, Martina G, Rollino C, De March- Sanchez Crespo M, Blasco R, et al. A possible com-
i M, Giacchino F, et al. Circulating immune com- mon pathogenesis of the mesangial IgA glomerulone-
plexes containing IgA. IgG and IgM in patients with phritis in patients with Berger’s disease and
primary IgA nephropathy and with Henoch- Schonlein-Henoch syndrome. Proc Eur Dial Trans-
Schoenlein nephritis. Correlation with clinical and plant Assoc. 1980;17:660–6.
histologic signs of activity. Clin Nephrol. 1982;18 142. Harper SJ, Pringle JH, Wicks AC, Hattersley J,
(5):230–9. Layward L, Allen A, et al. Expression of J chain
129. Baliah T, Kim KH, Anthone S, Anthone R, mRNA in duodenal IgA plasma cells in IgA nephrop-
Montes M, Andres GA. Recurrence of Henoch- athy. Kidney Int. 1994;45(3):836–44.
Schonlein purpura glomerulonephritis in transplanted 143. Harper SJ, Allen AC, Pringle JH, Feehally
kidneys. Transplantation. 1974;18(4):343–6. J. Increased dimeric IgA producing B cells in the
130. Nast CC, Ward HJ, Koyle MA, Cohen AH. Recurrent bone marrow in IgA nephropathy determined by in
Henoch-Schonlein purpura following renal transplan- situ hybridisation for J chain mRNA. J Clin Pathol.
tation. Am J Kidney Dis. 1987;9(1):39–43. 1996;49(1):38–42.
131. Hasegawa A, Kawamura T, Ito H, Hasegawa O, 144. de Fijter JW, Eijgenraam JW, Braam CA, Holmgren J,
Ogawa O, Honda M, et al. Fate of renal grafts with Daha MR, van Es LA, et al. Deficient IgA1 immune
recurrent Henoch-Schonlein purpura nephritis in chil- response to nasal cholera toxin subunit B in primary
dren. Transplant Proc. 1989;21(1 Pt 2):2130–3. IgA nephropathy. Kidney Int. 1996;50(3):952–61.
1022 K. Nakanishi and N. Yoshikawa

145. Rauterberg EW, Lieberknecht HM, Wingen AM, Ritz 159. Donadio JV, Grande JP. IgA nephropathy. N Engl J
E. Complement membrane attack (MAC) in idio- Med. 2002;347(10):738–48.
pathic IgA-glomerulonephritis. Kidney Int. 1987;31 160. Launay P, Grossetete B, Arcos-Fajardo M, Gaudin E,
(3):820–9. Torres SP, Beaudoin L, et al. Fcalpha receptor (CD89)
146. Evans DJ, Williams DG, Peters DK, Sissons JG, mediates the development of immunoglobulin A
Boulton-Jones JM, Ogg CS, et al. Glomerular depo- (IgA) nephropathy (Berger’s disease). Evidence for
sition of properdin in Henoch-Schonlein syndrome pathogenic soluble receptor-Iga complexes in patients
and idiopathic focal nephritis. Br Med J. 1973;3 and CD89 transgenic mice. J Exp Med. 2000;191
(5875):326–8. (11):1999–2009.
147. Julian BA, Wyatt RJ, McMorrow RG, Galla 161. Moura IC, Centelles MN, Arcos-Fajardo M,
JH. Serum complement proteins in IgA nephropathy. Malheiros DM, Collawn JF, Cooper MD,
Clin Nephrol. 1983;20(5):251–8. et al. Identification of the transferrin receptor as a
148. Roos A, Bouwman LH, van Gijlswijk-Janssen DJ, novel immunoglobulin (Ig)A1 receptor and its
Faber-Krol MC, Stahl GL, Daha MR. Human IgA enhanced expression on mesangial cells in IgA
activates the complement system via the mannan- nephropathy. J Exp Med. 2001;194(4):417–25.
binding lectin pathway. J Immunol. 2001;167 162. Haddad E, Moura IC, Arcos-Fajardo M, Macher MA,
(5):2861–8. Baudouin V, Alberti C, et al. Enhanced expression of
149. Abe K, Miyazaki M, Koji T, Furusu A, Ozono Y, the CD71 mesangial IgA1 receptor in Berger disease
Harada T, et al. Expression of decay accelerating and Henoch-Schonlein nephritis: association between
factor mRNA and complement C3 mRNA in human CD71 expression and IgA deposits. J Am Soc
diseased kidney. Kidney Int. 1998;54(1):120–30. Nephrol. 2003;14(2):327–37.
150. Kawana S, Shen GH, Kobayashi Y, Nishiyama 163. Moura IC, Arcos-Fajardo M, Sadaka C, Leroy V,
S. Membrane attack complex of complement in Benhamou M, Novak J, et al. Glycosylation and size
Henoch-Schonlein purpura skin and nephritis. Arch of IgA1 are essential for interaction with mesangial
Dermatol Res. 1990;282(3):183–7. transferrin receptor in IgA nephropathy. J Am Soc
151. Garcia-Fuentes M, Martin A, Chantler C, Williams Nephrol. 2004;15(3):622–34.
DG. Serum complement components in Henoch- 164. Moura IC, Arcos-Fajardo M, Gdoura A, Leroy V,
Schonlein purpura. Arch Dis Child. 1978;53 Sadaka C, Mahlaoui N, et al. Engagement of transfer-
(5):417–9. rin receptor by polymeric IgA1: evidence for a posi-
152. Petersen S, Taaning E, Soderstrom T, Pilgaard B, tive feedback loop involving increased receptor
Tranebjaerg L, Christensen T, et al. Immunoglobulin expression and mesangial cell proliferation in IgA
and complement studies in children with Schonlein- nephropathy. J Am Soc Nephrol. 2005;16
Henoch Syndrome and other vasculitic diseases. Acta (9):2667–76.
Paediatr Scand. 1991;80(11):1037–43. 165. Rops AL, Figdor CG, van der Schaaf A, Tamboer WP,
153. Kawana S, Nishiyama S. Serum SC5b-9 (terminal Bakker MA, Berden JH, et al. The tetraspanin CD37
complement complex) level, a sensitive indicator of protects against glomerular IgA deposition and renal
disease activity in patients with Henoch-Schonlein pathology. Am J Pathol. 2010;176(5):2188–97.
purpura. Dermatology. 1992;184(3):171–6. 166. Allen AC, Feehally J. IgA1 glycosylation and the
154. Tomino Y, Endoh M, Nomoto Y, Sakai H. Specificity pathogenesis of IgA nephropathy. Am J Kidney Dis.
of eluted antibody from renal tissues of patients with 2000;35(3):551–6.
IgA nephropathy. Am J Kidney Dis. 1982;1 167. Kiryluk K, Moldoveanu Z, Sanders JT, Eison TM,
(5):276–80. Suzuki H, Julian BA, et al. Aberrant glycosylation of
155. Tomino Y, Sakai H, Endoh M, Suga T, Miura M, IgA1 is inherited in both pediatric IgA nephropathy
Kaneshige H, et al. Cross-reactivity of IgA antibodies and Henoch-Schonlein purpura nephritis. Kidney Int.
between renal mesangial areas and nuclei of tonsillar 2011;80(1):79–87.
cells in patients with IgA nephropathy. Clin Exp 168. Sediva A, Stejskal J, Bartunkova J, Smetana Jr K,
Immunol. 1983;51(3):605–10. Gabius HJ. Detection of alpha(beta)-N-acetyl-D-
156. Emancipator SN. Immunoregulatory factors in the galactosamine-binding sites in kidney – relation to
pathogenesis of IgA nephropathy. Kidney Int. Henoch-Schonlein-associated IgA nephropathy.
1990;38(6):1216–29. Folia Biol. 1999;45(4):147–50.
157. Tomino Y, Sakai H, Endoh M, Miura M, Suga T, 169. Tomana M, Matousovic K, Julian BA, Radl J,
Kaneshige H, et al. Cross-reactivity of eluted anti- Konecny K, Mestecky J. Galactose-deficient IgA1 in
bodies from renal tissues of patients with henoch- sera of IgA nephropathy patients is present in com-
Schonlein purpura nephritis and IgA nephropathy. plexes with IgG. Kidney Int. 1997;52(2):509–16.
Am J Nephrol. 1983;3(6):315–8. 170. Tomana M, Novak J, Julian BA, Matousovic K,
158. Tomino Y, Endoh M, Miura M, Nomoto Y, Sakai Konecny K, Mestecky J. Circulating immune com-
H. Immunopathological similarities between IgA plexes in IgA nephropathy consist of IgA1 with
nephropathy and Henoch-Schoenlein purpura (HSP) galactose-deficient hinge region and antiglycan anti-
nephritis. Acta Pathol Jpn. 1983;33(1):113–22. bodies. J Clin Invest. 1999;104(1):73–81.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1023

171. Allen AC, Harper SJ, Feehally J. Galactosylation of 184. Gharavi AG, Moldoveanu Z, Wyatt RJ, Barker CV,
N- and O-linked carbohydrate moieties of IgA1 and Woodford SY, Lifton RP, et al. Aberrant IgA1 Glyco-
IgG in IgA nephropathy. Clin Exp Immunol. sylation Is Inherited in Familial and Sporadic IgA
1995;100(3):470–4. Nephropathy. J Am Soc Nephrol. 2008;19:1008–14.
172. Allen AC, Bailey EM, Brenchley PE, Buck KS, 185. Harper SJ, Allen AC, Bene MC, Pringle JH, Faure G,
Barratt J, Feehally J. Mesangial IgA1 in IgA nephrop- Lauder I, et al. Increased dimeric IgA-producing B
athy exhibits aberrant O-glycosylation: observations cells in tonsils in IgA nephropathy determined by in
in three patients. Kidney Int. 2001;60(3):969–73. situ hybridization for J chain mRNA. Clin Exp
173. Hiki Y, Odani H, Takahashi M, Yasuda Y, Immunol. 1995;101(3):442–8.
Nishimoto A, Iwase H, et al. Mass spectrometry 186. Leung JC, Tang SC, Lam MF, Chan TM, Lai
proves under-O-glycosylation of glomerular IgA1 in KN. Charge-dependent binding of polymeric IgA1
IgA nephropathy. Kidney Int. 2001;59(3):1077–85. to human mesangial cells in IgA nephropathy. Kidney
174. Smith AC, de Wolff JF, Molyneux K, Feehally J, Int. 2001;59(1):277–85.
Barratt J. O-glycosylation of serum IgD in IgA 187. Sinico RA, Fornasieri A, Oreni N, Benuzzi S,
nephropathy. J Am Soc Nephrol. 2006;17(4):1192–9. D’Amico G. Polymeric IgA rheumatoid factor in idi-
175. Kokubo T, Hiki Y, Iwase H, Tanaka A, Toma K, opathic IgA mesangial nephropathy (Berger’s dis-
Hotta K, et al. Protective role of IgA1 glycans against ease). J Immunol. 1986;137(2):536–41.
IgA1 self-aggregation and adhesion to extracellular 188. Nomoto Y, Suga T, Miura M, Nomoto H, Tomino Y,
matrix proteins. J Am Soc Nephrol. 1998;9 Sakai H. Characterization of an acidic nuclear protein
(11):2048–54. recognized by autoantibodies in sera from patients
176. Greer MR, Barratt J, Harper SJ, Allen AC, Feehally with IgA nephropathy. Clin Exp Immunol. 1986;65
J. The nucleotide sequence of the IgA1 hinge region (3):513–9.
in IgA nephropathy. Nephrol Dial Transplant. 189. Cederholm B, Wieslander J, Bygren P, Heinegard
1998;13(8):1980–3. D. Patients with IgA nephropathy have circulating
177. Allen AC, Topham PS, Harper SJ, Feehally anti-basement membrane antibodies reacting with
J. Leucocyte beta 1,3 galactosyltransferase activity structures common to collagen I, II, and IV. Proc
in IgA nephropathy. Nephrol Dial Transplant. Natl Acad Sci U S A. 1986;83(16):6151–5.
1997;12(4):701–6. 190. O’Donoghue DJ, Feehally J. Autoantibodies in IgA
178. Amado M, Almeida R, Carneiro F, Levery SB, nephropathy. Contrib Nephrol. 1995;111:93–103;
Holmes EH, Nomoto M, et al. A family of human discussion 4–6.
beta3-galactosyltransferases. Characterization of four 191. Hall RP, Stachura I, Cason J, Whiteside TL, Lawley
members of a UDP-galactose:beta-N-acetyl-glucos- TJ. IgA-containing circulating immune complexes in
amine/beta-nacetyl-galactosamine beta-1,3- patients with igA nephropathy. Am J Med. 1983;74
galactosyltransferase family. J Biol Chem. 1998;273 (1):56–63.
(21):12770–8. 192. Iijima K, Yoshikawa N, Shiozawa S, Matsuyama S,
179. Zhou D, Berger EG, Hennet T. Molecular cloning of a Kobayashi K, Yoshiya K, et al. Immune abnormalities
human UDP-galactose:GlcNAcbeta1,3GalNAc and clinical course in childhood IgA nephropathy.
beta1, 3 galactosyltransferase gene encoding an Nephron. 1990;56(3):255–60.
O-linked core3-elongation enzyme. Eur J Biochem. 193. Kameda A, Yoshikawa N, Shiozawa S, Doi K,
1999;263(2):571–6. Nakamura H. Lymphocyte subpopulations and func-
180. Allen AC, Bailey EM, Barratt J, Buck KS, Feehally tion in childhood IgA nephropathy. Nephron. 1991;59
J. Analysis of IgA1 O-glycans in IgA nephropathy by (4):546–51.
fluorophore-assisted carbohydrate electrophoresis. J 194. Sakai H, Nomoto Y, Arimori S. Decrease of
Am Soc Nephrol. 1999;10(8):1763–71. IgA-specific suppressor T cell activity in patients
181. Sano T, Hiki Y, Kokubo T, Iwase H, Shigematsu H, with IgA nephropathy. Clin Exp Immunol. 1979;38
Kobayashi Y. Enzymatically deglycosylated human (2):243–8.
IgA1 molecules accumulate and induce inflammatory 195. Sakai H, Endoh M, Tomino Y, Nomoto Y. Increase of
cell reaction in rat glomeruli. Nephrol Dial Trans- IgA specific helper T alpha cells in patients with
plant. 2002;17(1):50–6. IgA nephropathy. Clin Exp Immunol. 1982;50
182. Amore A, Cirina P, Conti G, Brusa P, Peruzzi L, (1):77–82.
Coppo R. Glycosylation of circulating IgA in patients 196. Roccatello D, Picciotto G, Torchio M, Ropolo R,
with IgA nephropathy modulates proliferation and Ferro M, Franceschini R, et al. Removal systems of
apoptosis of mesangial cells. J Am Soc Nephrol. immunoglobulin A and immunoglobulin A
2001;12(9):1862–71. containing complexes in IgA nephropathy and cirrho-
183. Suzuki H, Moldoveanu Z, Hall S, Brown R, Vu HL, sis patients. The role of asialoglycoprotein receptors.
Novak L, et al. IgA1-secreting cell lines from patients Lab Invest. 1993;69(6):714–23.
with IgA nephropathy produce aberrantly 197. Trygstad CW, Stiehm ER. Elevated serum IgA glob-
glycosylated IgA1. J Clin Invest. 2008;118 ulin in anaphylactoid purpura. Pediatrics. 1971;47
(2):629–39. (6):1023–8.
1024 K. Nakanishi and N. Yoshikawa

198. Simila S, Kouvalainen K, Lanning M. Serum immu- 212. Casanueva B, Rodriguez-Valverde V, Merino J,
noglobulin levels in the course of anaphylactoid pur- Arias M, Garcia-Fuentes M. Increased
pura in children. Acta Paediatr Scand. 1977;66 IgA-producing cells in the blood of patients with
(5):537–40. active Henoch-Schonlein purpura. Arthritis Rheum.
199. Saulsbury FT. The role of IgA1 rheumatoid factor in 1983;26(7):854–60.
the formation of IgA-containing immune complexes 213. Casanueva B, Rodriguez-Valverde V, Luceno
in Henoch-Schonlein purpura. J Clin Lab Immunol. A. Circulating IgA producing cells in the differential
1987;23(3):123–7. diagnosis of Henoch-Schonlein purpura. J
200. Saulsbury FT. IgA rheumatoid factor in Henoch- Rheumatol. 1988;15(8):1229–33.
Schonlein purpura. J Pediatr. 1986;108(1):71–6. 214. Casanueva B, Rodriguez-Valverde V, Farinas MC,
201. Saulsbury FT. Heavy and light chain composition of Vallo A, Rodriguez-Soriano J. Autologous mixed
serum IgA and IgA rheumatoid factor in Henoch- lymphocyte reaction and T-cell suppressor activity
Schonlein purpura. Arthritis Rheum. 1992;35 in patients with Henoch-Schonlein purpura and IgA
(11):1377–80. nephropathy. Nephron. 1990;54(3):224–8.
202. Saulsbury FT, Kirkpatrick PR, Bolton WK. IgA 215. Suzuki H, Kiryluk K, Novak J, Moldoveanu Z, Herr
antineutrophil cytoplasmic antibody in Henoch- AB, Renfrow MB, et al. The pathophysiology of IgA
Schonlein purpura. Am J Nephrol. 1991;11 nephropathy. J Am Soc Nephrol. 2011;22
(4):295–300. (10):1795–803.
203. Ronda N, Esnault VL, Layward L, Sepe V, Allen A, 216. Suzuki H, Fan R, Zhang Z, Brown R, Hall S, Julian
Feehally J, et al. Antineutrophil cytoplasm BA, et al. Aberrantly glycosylated IgA1 in IgA
antibodies (ANCA) of IgA isotype in adult Henoch- nephropathy patients is recognized by IgG antibodies
Schonlein purpura. Clin Exp Immunol. 1994;95 with restricted heterogeneity. J Clin Invest. 2009;119
(1):49–55. (6):1668–77.
204. Blanco Quiros A, Blanco C, Alvarez J, Solis P, 217. Julian BA, Novak J. IgA nephropathy: an
Conde F, Gomez S. Anti-immunoglobulin antibodies update. Curr Opin Nephrol Hypertens. 2004;13
in children with Schonlein-Henoch syndrome. (2):171–9.
Absence of serum anti-IgA antibodies. Eur J Pediatr. 218. Mestecky J, Tomana M, Moldoveanu Z, Julian BA,
1994;153(2):103–6. Suzuki H, Matousovic K, et al. Role of aberrant
205. Burden AD, Gibson IW, Rodger RS, Tillman glycosylation of IgA1 molecules in the pathogenesis
DM. IgA anticardiolipin antibodies associated with of IgA nephropathy. Kidney Blood Press Res.
Henoch-Schonlein purpura. J Am Acad Dermatol. 2008;31(1):29–37.
1994;31(5 Pt 2):857–60. 219. Glassock RJ. The pathogenesis of IgA nephropathy.
206. Coppo R, Basolo B, Piccoli G, Mazzucco G, Bulzomi Curr Opin Nephrol Hypertens. 2011;20(2):153–60.
MR, Roccatello D, et al. IgA1 and IgA2 immune 220. Szeto CC, Li PK. MicroRNAs in IgA nephropathy.
complexes in primary IgA nephropathy and Henoch- Nat Rev Nephrol. 2014;10(5):249–56.
Schonlein nephritis. Clin Exp Immunol. 1984;57 221. Lorenzen JM, Haller H, Thum T. MicroRNAs as
(3):583–90. mediators and therapeutic targets in chronic kidney
207. Waldo FB, Cochran AM. Mixed IgA-IgG aggregates disease. Nat Rev Nephrol. 2011;7(5):286–94.
as a model of immune complexes in IgA nephropathy. 222. Serino G, Sallustio F, Cox SN, Pesce F, Schena
J Immunol. 1989;142(11):3841–6. FP. Abnormal miR-148b expression promotes aber-
208. O’Donoghue DJ, Darvill A, Ballardie FW. Mesangial rant glycosylation of IgA1 in IgA nephropathy. J Am
cell autoantigens in immunoglobulin A nephropathy Soc Nephrol. 2012;23(5):814–24.
and Henoch-Schonlein purpura. J Clin Invest. 223. Fang Y, Yu X, Liu Y, Kriegel AJ, Heng Y, Xu X,
1991;88(5):1522–30. et al. miR-29c is downregulated in renal interstitial
209. Jennette JC, Wieslander J, Tuttle R, Falk RJ. Serum fibrosis in humans and rats and restored by HIF-alpha
IgA-fibronectin aggregates in patients with IgA activation. Am J Physiol Renal Physiol. 2013;304
nephropathy and Henoch-Schonlein purpura: diag- (10):F1274–82.
nostic value and pathogenic implications. The Glo- 224. Caubet C, Lacroix C, Decramer S, Drube J, Ehrich
merular Disease Collaborative Network. Am J JH, Mischak H, et al. Advances in urinary proteome
Kidney Dis. 1991;18(4):466–71. analysis and biomarker discovery in pediatric renal
210. Cederholm B, Linne T, Wieslander J, Bygren P, disease. Pediatr Nephrol. 2010;25(1):27–35.
Heinegard D. Fibronectin-immunoglobulin com- 225. Wang G, Kwan BC, Lai FM, Chow KM, Kam-Tao
plexes in the early course of IgA and Henoch- Li P, Szeto CC. Expression of microRNAs in the
Schonlein nephritis. Pediatr Nephrol. 1991;5 urinary sediment of patients with IgA nephropathy.
(2):200–4. Dis Markers. 2010;28(2):79–86.
211. Kuno-Sakai H, Sakai H, Nomoto Y, Takakura I, 226. Wang G, Kwan BC, Lai FM, Chow KM, Li PK, Szeto
Kimura M. Increase of IgA-bearing peripheral blood CC. Elevated levels of miR-146a and miR-155 in
lymphocytes in children with Henoch-Schoenlein kidney biopsy and urine from patients with IgA
purpura. Pediatrics. 1979;64(6):918–22. nephropathy. Dis Markers. 2011;30(4):171–9.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1025

227. Tan K, Chen J, Li W, Chen Y, Sui W, Zhang Y, IgA nephropathy upregulates transforming growth
et al. Genome-wide analysis of microRNAs expres- factor-beta synthesis and signal transduction in
sion profiling in patients with primary IgA nephropa- human mesangial cells via the renin-angiotensin sys-
thy. Genome. 2013;56(3):161–9. tem. J Am Soc Nephrol. 2003;14(12):3127–37.
228. Muratsu-Ikeda S, Nangaku M, Ikeda Y, Tanaka T, 242. Lai KN, Chan LY, Leung JC. Mechanisms of
Wada T, Inagi R. Downregulation of miR-205 modu- tubulointerstitial injury in IgA nephropathy. Kidney
lates cell susceptibility to oxidative and endoplasmic Int Suppl. 2005;94:S110–5.
reticulum stresses in renal tubular cells. PLoS One. 243. Allen AC, Barratt J, Feehally J. Immunoglobulin A
2012;7(7):e41462. nephropathy. In: Neilson EG, Couser WG, editors.
229. Puhr M, Hoefer J, Schafer G, Erb HH, Oh SJ, Immunologic renal diseases. Philadelphia: Lippincott
Klocker H, et al. Epithelial-to-mesenchymal transi- Williams & Wilkins; 2001. p. 931–47.
tion leads to docetaxel resistance in prostate cancer 244. Niemir ZI, Stein H, Noronha IL, Kruger C,
and is mediated by reduced expression of miR-200c Andrassy K, Ritz E, et al. PDGF and TGF-beta con-
and miR-205. Am J Pathol. 2012;181(6):2188–201. tribute to the natural course of human IgA glomeru-
230. Bao H, Chen H, Zhu X, Zhang M, Yao G, Yu Y, lonephritis. Kidney Int. 1995;48(5):1530–41.
et al. MiR-223 downregulation promotes glomerular 245. Stein-Oakley AN, Maguire JA, Dowling J, Perry G,
endothelial cell activation by upregulating importin Thomsom NM. Altered expression of fibrogenic
alpha4 and alpha5 in IgA nephropathy. Kidney Int. growth factors in IgA nephropathy and focal and
2014;85(3):624–35. segmental glomerulosclerosis. Kidney Int. 1997;51
231. Chau BN, Xin C, Hartner J, Ren S, Castano AP, (1):195–204.
Linn G, et al. MicroRNA-21 promotes fibrosis of the 246. Taniguchi Y, Yorioka N, Masaki T, Asakimori Y,
kidney by silencing metabolic pathways. Sci Transl Yamashita K, Yamakido M. Localization of
Med. 2012;4(121):121ra18. transforming growth factors beta1 and beta2 and epi-
232. Chung AC, Huang XR, Meng X, Lan HY. miR-192 dermal growth factor in IgA nephropathy. Scand J
mediates TGF-beta/Smad3-driven renal fibrosis. Urol Nephrol. 1999;33(4):243–7.
J Am Soc Nephrol. 2010;21(8):1317–25. 247. Tamouza H, Chemouny JM, Raskova Kafkova L,
233. Krupa A, Jenkins R, Luo DD, Lewis A, Phillips A, Berthelot L, Flamant M, Demion M, et al. The IgA1
Fraser D. Loss of MicroRNA-192 promotes immune complex-mediated activation of the MAPK/
fibrogenesis in diabetic nephropathy. J Am Soc ERK kinase pathway in mesangial cells is associated
Nephrol. 2010;21(3):438–47. with glomerular damage in IgA nephropathy. Kidney
234. Du B, Ma LM, Huang MB, Zhou H, Huang HL, Int. 2012;82(12):1284–96.
Shao P, et al. High glucose down-regulates miR-29a 248. Nagata M, Akioka Y, Tsunoda Y, Komatsu Y,
to increase collagen IV production in HK-2 cells. Kawaguchi H, Yamaguchi Y, et al. Macrophages in
FEBS Lett. 2010;584(4):811–6. childhood IgA nephropathy. Kidney Int. 1995;48
235. Liu Y, Taylor NE, Lu L, Usa K, Cowley Jr AW, Ferreri (2):527–35.
NR, et al. Renal medullary microRNAs in Dahl salt- 249. Yoshioka K, Takemura T, Murakami K, Okada M,
sensitive rats: miR-29b regulates several collagens Yagi K, Miyazato H, et al. In situ expression of
and related genes. Hypertension. 2010;55(4):974–82. cytokines in IgA nephritis. Kidney Int. 1993;44
236. Wang Q, Wang Y, Minto AW, Wang J, Shi Q, Li X, (4):825–33.
et al. MicroRNA-377 is up-regulated and can lead to 250. Iijima K, Yoshikawa N, Connolly DT, Nakamura
increased fibronectin production in diabetic nephrop- H. Human mesangial cells and peripheral blood
athy. FASEB J. 2008;22(12):4126–35. mononuclear cells produce vascular permeability fac-
237. Fleissner F, Jazbutyte V, Fiedler J, Gupta SK, Yin X, tor. Kidney Int. 1993;44(5):959–66.
Xu Q, et al. Short communication: asymmetric 251. Noguchi K, Yoshikawa N, Ito-Kariya S, Inoue Y,
dimethylarginine impairs angiogenic progenitor cell Hayashi Y, Ito H, et al. Activated mesangial cells
function in patients with coronary artery disease produce vascular permeability factor in early-stage
through a microRNA-21-dependent mechanism. mesangial proliferative glomerulonephritis. J Am
Circ Res. 2010;107(1):138–43. Soc Nephrol. 1998;9(10):1815–25.
238. Liu Y. Renal fibrosis: new insights into the pathogen- 252. Tomino Y, Endoh M, Nomoto Y, Sakai H. Activation
esis and therapeutics. Kidney Int. 2006;69(2):213–7. of complement by renal tissues from patients with IgA
239. Stad RK, Bruijn JA, van Gijlswijk-Janssen DJ, van Es nephropathy. J Clin Pathol. 1981;34(1):35–40.
LA, Daha MR. An acute model for IgA-mediated 253. Kashem A, Endoh M, Nomoto Y, Sakai H, Nakazawa
glomerular inflammation in rats induced by monoclo- H. Fc alpha R expression on polymorphonuclear leu-
nal polymeric rat IgA antibodies. Clin Exp Immunol. kocyte and superoxide generation in IgA nephropa-
1993;92(3):514–21. thy. Kidney Int. 1994;45(3):868–75.
240. Floege J, Feehally J. IgA nephropathy: recent devel- 254. Lemley KV, Lafayette RA, Safai M, Derby G,
opments. J Am Soc Nephrol. 2000;11(12):2395–403. Blouch K, Squarer A, et al. Podocytopenia and dis-
241. Lai KN, Tang SC, Guh JY, Chuang TD, Lam MF, ease severity in IgA nephropathy. Kidney Int.
Chan LY, et al. Polymeric IgA1 from patients with 2002;61(4):1475–85.
1026 K. Nakanishi and N. Yoshikawa

255. Wharram BL, Goyal M, Wiggins JE, Sanden SK, 267. Tang S, Leung JC, Tsang AW, Lan HY, Chan TM, Lai
Hussain S, Filipiak WE, et al. Podocyte depletion KN. Transferrin up-regulates chemokine synthesis by
causes glomerulosclerosis: diphtheria toxin-induced human proximal tubular epithelial cells: implication
podocyte depletion in rats expressing human diphthe- on mechanism of tubuloglomerular communication in
ria toxin receptor transgene. J Am Soc Nephrol. glomerulopathic proteinura. Kidney Int. 2002;61(5):
2005;16(10):2941–52. 1655–65.
256. Qiu LQ, Sinniah R, I-Hong Hsu S. Downregulation of 268. Zoja C, Benigni A, Remuzzi G. Cellular responses to
Bcl-2 by podocytes is associated with progressive protein overload: key event in renal disease progres-
glomerular injury and clinical indices of poor renal sion. Curr Opin Nephrol Hypertens. 2004;13(1):
prognosis in human IgA nephropathy. J Am Soc 31–7.
Nephrol. 2004;15(1):79–90. 269. Galla JH, Spotswood MF, Harrison LA, Mestecky J.
257. Wang C, Peng H, Tang H, Liu X, Chen Z, Lou T, Urinary IgA in IgA nephropathy and Henoch-
et al. Serum IgA1 from IgA nephropathy patients Schoenlein purpura. J Clin Immunol. 1985;5(5):
induces apoptosis in podocytes through direct and 298–306.
indirect pathways. Clin Invest Med. 2007;30(6): 270. Arrizabalaga P, Sole M, Abellana R, de las Cuevas X,
E240–9. Soler J, Pascual J, et al. Tubular and
258. Dillon JJ. Angiotensin-converting enzyme inhibitors interstitial expression of ICAM-1 as a marker of
and angiotensin receptor blockers for IgA nephropa- renal injury in IgA nephropathy. Am J Nephrol.
thy. Semin Nephrol. 2004;24(3):218–24. 2003;23(3):121–8.
259. Lai KN, Chan LY, Tang SC, Tsang AW, Li FF, 271. Arrizabalaga P, Sole M, Quinto IL, Ascaso C.
Lam MF, et al. Mesangial expression of Intercellular adhesion molecule-1 mediated
angiotensin II receptor in IgA nephropathy and its interactions and leucocyte infiltration in IgA
regulation by polymeric IgA1. Kidney Int. 2004; nephropathy. Nephrol Dial Transplant. 1997;12(11):
66(4):1403–16. 2258–62.
260. Chan LY, Leung JC, Tsang AW, Tang SC, Lai 272. Ranieri E, Gesualdo L, Petrarulo F, Schena
KN. Activation of tubular epithelial cells by FP. Urinary IL-6/EGF ratio: a useful prognostic
mesangial-derived TNF-alpha: glomerulotubular marker for the progression of renal damage in IgA
communication in IgA nephropathy. Kidney Int. nephropathy. Kidney Int. 1996;50(6):1990–2001.
2005;67(2):602–12. 273. Eddy AA. Molecular basis of renal fibrosis. Pediatr
261. Wu Q, Jinde K, Endoh M, Sakai H. Clinical signifi- Nephrol. 2000;15(3–4):290–301.
cance of costimulatory molecules CD80/CD86 274. Hirschberg R. Wound healing in the kidney: complex
expression in IgA nephropathy. Kidney Int. interactions in renal interstitial fibrogenesis. J Am Soc
2004;65(3):888–96. Nephrol. 2005;16(1):9–11.
262. Tang SC, Leung JC, Abe K, Chan KW, 275. Yoshikawa N, Ito H, Nakamura H. IgA nephropathy
Chan LY, Chan TM, et al. Albumin stimulates in children from Japan. Clinical and pathological fea-
interleukin-8 expression in proximal tubular epithelial tures. Child Nephrol Urol. 1988;9(4):191–9.
cells in vitro and in vivo. J Clin Invest. 2003;111(4): 276. Miura M, Tomino Y, Yagame M, Endoh M, Suga T,
515–27. Nomoto Y, et al. Immunofluorescent studies on alpha
263. Zoja C, Donadelli R, Colleoni S, Figliuzzi M, 2-plasmin inhibitor (alpha 2-PI) in glomeruli from
Bonazzola S, Morigi M, et al. Protein overload stim- patients with IgA nephropathy. Clin Exp Immunol.
ulates RANTES production by proximal tubular cells 1985;62(2):380–6.
depending on NF-kappa B activation. Kidney Int. 277. Yoshikawa N, Ito H, Nakahara C, Yoshiara S,
1998;53(6):1608–15. Yoshiya K, Matsuo T, et al. Glomerular electron-
264. Abbate M, Zoja C, Corna D, Capitanio M, Bertani T, dense deposits in childhood IgA nephropathy.
Remuzzi G. In progressive nephropathies, overload of Virchows Arch A Pathol Anat Histopathol.
tubular cells with filtered proteins translates glomer- 1985;406(1):33–43.
ular permeability dysfunction into cellular signals of 278. D’Amico G, Minetti L, Ponticelli C, Fellin G,
interstitial inflammation. J Am Soc Nephrol. Ferrario F, Barbiano di Belgioioso G, et al. Prognostic
1998;9(7):1213–24. indicators in idiopathic IgA mesangial nephropathy.
265. Wang Y, Chen J, Chen L, Tay YC, Rangan GK, Harris Q J Med. 1986;59(228):363–78.
DC. Induction of monocyte chemoattractant protein-1 279. Andreoli SP, Yum MN, Bergstein JM. IgA nephropa-
in proximal tubule cells by urinary protein. J Am Soc thy in children: significance of glomerular basement
Nephrol. 1997;8(10):1537–45. membrane deposition of IgA. Am J Nephrol.
266. Abbate M, Zoja C, Rottoli D, Corna D, Perico N, 1986;6(1):28–33.
Bertani T, et al. Antiproteinuric therapy while 280. Yoshikawa N, Ito H, Nakamura H. Prognostic indica-
preventing the abnormal protein traffic in proximal tors in childhood IgA nephropathy. Nephron.
tubule abrogates protein- and complement-dependent 1992;60(1):60–7.
interstitial inflammation in experimental renal dis- 281. Churg J, Bernstein J, Glassock RJ. Renal disease:
ease. J Am Soc Nephrol. 1999;10(4):804–13. classification and atlas of glomerular diseases.
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1027

Tokyo: Igaku-shoin Medical Publishers; 1995. clinical outcomes. Nephrol Dial Transplant.
p. 86–8. 2008;23:2537–45.
282. Zimmerman SW, Burkholder PM. Immunoglobulin A 296. Cattran DC, Coppo R, Cook HT, Feehally J, Roberts
nephropathy. Arch Intern Med. 1975;135(9): IS, Troyanov S, et al. The Oxford classification of IgA
1217–23. nephropathy: rationale, clinicopathological correla-
283. Hara M, Endo Y, Nihei H, Hara S, Fukushima O, tions, and classification. Kidney Int. 2009;76(5):
Mimura N. IgA nephropathy with subendothelial 534–45.
deposits. Virchows Arch A Pathol Anat Histol. 297. Park KS, Han SH, Kie JH, Nam KH, Lee MJ, Lim BJ,
1980;386(3):249–63. et al. Comparison of the Haas and the Oxford classi-
284. Suzuki J, Yoshikawa N, Nakamura H. A quantitative fications for prediction of renal outcome in patients
analysis of the mesangium in children with IgA with IgA nephropathy. Hum Pathol. 2014;45(2):
nephropathy: sequential study. J Pathol. 1990; 236–43.
161(1):57–64. 298. Haas M, Reich HN. Morphologic markers of progres-
285. Nicholls KM, Fairley KF, Dowling JP, Kincaid-Smith sive immunoglobulin A nephropathy. Adv Chronic
P. The clinical course of mesangial IgA associated Kidney Dis. 2012;19(2):107–13.
nephropathy in adults. Q J Med. 1984;53(210): 299. Lee SM, Rao VM, Franklin WA, Schiffer MS,
227–50. Aronson AJ, Spargo BH, et al. IgA nephropathy:
286. D’Amico G. Natural history of idiopathic IgA morphologic predictors of progressive renal disease.
nephropathy and factors predictive of disease out- Hum Pathol. 1982;13(4):314–22.
come. Semin Nephrol. 2004;24(3):179–96. 300. Haas M. Histologic subclassification of IgA nephrop-
287. Wu Q, Tanaka H, Hirukawa T, Endoh M, Fukagawa athy: a clinicopathologic study of 244 cases. Am J
M. Characterization and quantification of proliferat- Kidney Dis. 1997;29(6):829–42.
ing cell patterns in endocapillary proliferation. 301. Roberts IS, Cook HT, Troyanov S, Alpers CE,
Nephrol Dial Transplant. 2012;27(8):3234–41. Amore A, Barratt J, et al. The Oxford classification
288. Ikezumi Y, Suzuki T, Karasawa T, Hasegawa H, of IgA nephropathy: pathology definitions, correla-
Yamada T, Imai N, et al. Identification of alternatively tions, and reproducibility. Kidney Int. 2009;76(5):
activated macrophages in new-onset paediatric and 546–56.
adult immunoglobulin A nephropathy: potential role 302. Coppo R, Troyanov S, Camilla R, Hogg RJ, Cattran
in mesangial matrix expansion. Histopathology. DC, Cook HT, et al. The Oxford IgA nephropathy
2011;58(2):198–210. clinicopathological classification is valid for children
289. Yoshikawa N, Yoshiara S, Yoshiya K, Matsuo T, as well as adults. Kidney Int. 2010;77(10):921–7.
Okada S. Lysis of the glomerular basement 303. Haas M, Rastaldi MP, Fervenza FC. Histologic clas-
membrane in children with IgA nephropathy and sification of glomerular diseases: clinicopathologic
Henoch-Shonlein nephritis. J Pathol. 1986;150(2): correlations, limitations exposed by validation stud-
119–26. ies, and suggestions for modification. Kidney Int.
290. Droz D. Natural history of primary glomerulonephri- 2014;85(4):779–93.
tis with mesangial deposits of IgA. Contrib Nephrol. 304. Herzenberg AM, Fogo AB, Reich HN, Troyanov S,
1976;2:150–6. Bavbek N, Massat AE, et al. Validation of the Oxford
291. Ibels LS, Gyory AZ. IgA nephropathy: analysis of the classification of IgA nephropathy. Kidney Int.
natural history, important factors in the progression of 2011;80(3):310–7.
renal disease, and a review of the literature. Medicine. 305. Shi SF, Wang SX, Jiang L, Lv JC, Liu LJ, Chen YQ,
1994;73(2):79–102. et al. Pathologic predictors of renal outcome and
292. Hotta O, Furuta T, Chiba S, Tomioka S, Taguma therapeutic efficacy in IgA nephropathy: validation
Y. Regression of IgA nephropathy: a repeat biopsy of the oxford classification. Clin J Am Soc Nephrol.
study. Am J Kidney Dis. 2002;39(3):493–502. 2011;6(9):2175–84.
293. Shima Y, Nakanishi K, Kamei K, Togawa H, Nozu K, 306. Katafuchi R, Ninomiya T, Nagata M, Mitsuiki K,
Tanaka R, et al. Disappearance of glomerular IgA Hirakata H. Validation study of oxford classification
deposits in childhood IgA nephropathy showing dif- of IgA nephropathy: the significance of extracapillary
fuse mesangial proliferation after 2 years of combina- proliferation. Clin J Am Soc Nephrol. 2011;6(12):
tion/prednisolone therapy. Nephrol Dial Transplant. 2806–13.
2011;26(1):163–9. 307. Edstrom Halling S, Soderberg MP, Berg
294. Ikezumi Y, Suzuki T, Imai N, Ueno M, Narita I, UB. Predictors of outcome in paediatric IgA nephrop-
Kawachi H, et al. Histological differences in athy with regard to clinical and histopathological
new-onset IgA nephropathy between children and variables (Oxford classification). Nephrol Dial Trans-
adults. Nephrol Dial Transplant. 2006;21(12): plant. 2012;27(2):715–22.
3466–74. 308. Shima Y, Nakanishi K, Hama T, Mukaiyama H,
295. Haas M, Rahman MH, Cohn RA, Fathallah-Shaykh S, Togawa H, Hashimura Y, et al. Validity of the Oxford
Ansari A, Bartosh SM. IgA Nephropathy in children classification of IgA nephropathy in children. Pediatr
and adults: comparison of histologic features and Nephrol. 2012;27(5):783–92.
1028 K. Nakanishi and N. Yoshikawa

309. Zeng CH, Le W, Ni Z, Zhang M, Miao L, Luo P, 323. A multicenter study of IgA nephropathy in children.
et al. A multicenter application and evaluation of the A report of the Southwest Pediatric Nephrology Study
oxford classification of IgA nephropathy in adult Chi- Group. Kidney Int. 1982;22(6):643–52.
nese patients. Am J Kidney Dis. 2012;60(5):812–20. 324. Ueda Y, Sakai O, Yamagata M, Kitajima T,
310. El Karoui K, Hill GS, Karras A, Moulonguet L, Kawamura K. IgA glomerulonephritis in Japan.
Caudwell V, Loupy A, et al. Focal segmental Contrib Nephrol. 1975;4:37–47.
glomerulosclerosis plays a major role in the progres- 325. Nakamoto Y, Asano Y, Dohi K, Fujioka M, Iida H,
sion of IgA nephropathy. II. Light microscopic and Kida H, et al. Primary IgA glomerulonephritis and
clinical studies. Kidney Int. 2011;79(6):643–54. Schonlein-Henoch purpura nephritis: clinicopatho-
311. Alamartine E, Sauron C, Laurent B, Sury A, Seffert A, logical and immunohistological characteristics. Q J
Mariat C. The use of the Oxford classification of IgA Med. 1978;47(188):495–516.
nephropathy to predict renal survival. Clin J Am Soc 326. Silva FG, Hogg RJ. IgA nephropathy. In: Tisher CC,
Nephrol. 2011;6(10):2384–8. Brenner BM, editors. Renal pathology: with clinical
312. Lee H, Yi SH, Seo MS, Hyun JN, Jeon JS, Noh H, and functional correlations. Philadelphia: Lippincott;
et al. Validation of the Oxford classification of IgA 1989. p. 434–93.
nephropathy: a single-center study in Korean adults. 327. Abuelo JG, Esparza AR, Matarese RA, Endreny RG,
Korean J Intern Med. 2012;27(3):293–300. Carvalho JS, Allegra SR. Crescentic IgA nephropa-
313. Le W, Zeng CH, Liu Z, Liu D, Yang Q, Lin RX, thy. Medicine. 1984;63(6):396–406.
et al. Validation of the Oxford classification of IgA 328. Hastings MC, Moldoveanu Z, Suzuki H, Berthoux F,
nephropathy for pediatric patients from China. BMC Julian BA, Sanders JT, et al. Biomarkers in IgA
Nephrol. 2012;13:158. nephropathy: relationship to pathogenetic hits. Expert
314. Kang SH, Choi SR, Park HS, Lee JY, Sun IO, Hwang Opin Med Diagn. 2013;7(6):615–27.
HS, et al. The Oxford classification as a predictor of 329. Moldoveanu Z, Wyatt RJ, Lee JY, Tomana M,
prognosis in patients with IgA nephropathy. Nephrol Julian BA, Mestecky J, et al. Patients with IgA
Dial Transplant. 2012;27(1):252–8. nephropathy have increased serum galactose-deficient
315. Roberts IS. Oxford classification of immunoglobulin IgA1 levels. Kidney Int. 2007;71(11):1148–54.
A nephropathy: an update. Curr Opin Nephrol 330. Wyatt RJ, Julian BA. IgA nephropathy. N Engl J Med.
Hypertens. 2013;22(3):281–6. 2013;368(25):2402–14.
316. Coppo R, Troyanov S, Bellur S, Cattran D, Cook HT, 331. Berthoux F, Suzuki H, Thibaudin L, Yanagawa H,
Feehally J, et al. Validation of the Oxford classifica- Maillard N, Mariat C, et al. Autoantibodies targeting
tion of IgA nephropathy in cohorts with different galactose-deficient IgA1 associate with progression
presentations and treatments. Kidney Int. 2014;86: of IgA nephropathy. J Am Soc Nephrol.
828–36. 2012;23(9):1579–87.
317. Shima Y, Nakanishi K, Hama T, Sato M, 332. Yanagawa H, Suzuki H, Suzuki Y, Kiryluk K, Gharavi
Mukaiyama H, Togawa H, et al. Biopsy timing and AG, Matsuoka K, et al. A panel of serum biomarkers
Oxford classification variables in Childhood/Adoles- differentiates IgA nephropathy from other renal dis-
cent IgA nephropathy. Pediatr Nephrol. 2014. eases. PLoS One. 2014;9(5):e98081.
318. Tanaka S, Ninomiya T, Katafuchi R, Masutani K, 333. Zwirner J, Burg M, Schulze M, Brunkhorst R,
Tsuchimoto A, Noguchi H, et al. Development and Gotze O, Koch KM, et al. Activated complement
validation of a prediction rule using the Oxford clas- C3: a potentially novel predictor of progressive IgA
sification in IgA nephropathy. Clin J Am Soc nephropathy. Kidney Int. 1997;51(4):1257–64.
Nephrol. 2013;8(12):2082–90. 334. Camilla R, Suzuki H, Dapra V, Loiacono E, Peruzzi L,
319. Niaudet P, Levy M, Broyer M, Habib R. Clinicopath- Amore A, et al. Oxidative stress and galactose-
ologic correlations in severe forms of Henoch- deficient IgA1 as markers of progression in IgA
Schonlein purpura nephritis based on repeat biopsies. nephropathy. Clin J Am Soc Nephrol. 2011;6(8):
Contrib Nephrol. 1984;40:250–4. 1903–11.
320. Michalk D, Waldherr R, Seelig HP, Weber HP, 335. Lundberg S, Qureshi AR, Olivecrona S,
Scharer K. Idiopathic mesangial Gunnarsson I, Jacobson SH, Larsson TE. FGF23,
IgA-glomerulonephritis in childhood. Description of albuminuria, and disease progression in patients
19 pediatric cases and review of the literature. Eur J with chronic IgA nephropathy. Clin J Am Soc
Pediatr. 1980;134(1):13–22. Nephrol. 2012;7(5):727–34.
321. Linne T, Aperia A, Broberger O, Bergstrand A, 336. Shi Y, Chen W, Jalal D, Li Z, Chen W, Mao H,
Bohman SO, Rekola S. Course of renal function in et al. Clinical outcome of hyperuricemia in IgA
IgA glomerulonephritis in children and adolescents. nephropathy: a retrospective cohort study and ran-
Acta Paediatr Scand. 1982;71(5):735–43. domized controlled trial. Kidney Blood Press Res.
322. Levy M, Gonzalez-Burchard G, Broyer M, 2012;35(3):153–60.
Dommergues JP, Foulard M, Sorez JP, 337. Cheng GY, Liu DW, Zhang N, Tang L, Zhao ZZ, Liu
et al. Berger’s disease in children. Natural history ZS. Clinical and prognostic implications of serum uric
and outcome. Medicine. 1985;64(3):157–80. acid levels on IgA nephropathy: a cohort study of
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1029

348 cases with a mean 5-year follow-up. Clin prognostic markers in IgA nephropathy. Neth J Med.
Nephrol. 2013;80(1):40–6. 2009;67(2):54–61.
338. Vuong MT, Hahn-Zoric M, Lundberg S, 350. Liu LL, Jiang Y, Wang LN, Liu N. Urinary mannose-
Gunnarsson I, van Kooten C, Wramner L, et al. Asso- binding lectin is a biomarker for predicting the pro-
ciation of soluble CD89 levels with disease progres- gression of immunoglobulin (Ig)A nephropathy. Clin
sion but not susceptibility in IgA nephropathy. Exp Immunol. 2012;169(2):148–55.
Kidney Int. 2010;78(12):1281–7. 351. Haubitz M, Wittke S, Weissinger EM, Walden M,
339. Tomino Y, Suzuki S, Imai H, Saito T, Kawamura T, Rupprecht HD, Floege J, et al. Urine protein patterns
Yorioka N, et al. Measurement of serum IgA and C3 can serve as diagnostic tools in patients with IgA
may predict the diagnosis of patients with IgA nephropathy. Kidney Int. 2005;67(6):2313–20.
nephropathy prior to renal biopsy. J Clin Lab Anal. 352. Julian BA, Wittke S, Novak J, Good DM, Coon JJ,
2000;14(5):220–3. Kellmann M, et al. Electrophoretic methods for anal-
340. Nakayama K, Ohsawa I, Maeda-Ohtani A, ysis of urinary polypeptides in IgA-associated renal
Murakoshi M, Horikoshi S, Tomino Y. Prediction of diseases. Electrophoresis. 2007;28(23):4469–83.
diagnosis of immunoglobulin A nephropathy prior to 353. Julian BA, Wittke S, Haubitz M, Zurbig P, Schiffer E,
renal biopsy and correlation with urinary sediment McGuire BM, et al. Urinary biomarkers of IgA
findings and prognostic grading. J Clin Lab Anal. nephropathy and other IgA-associated renal diseases.
2008;22(2):114–8. World J Urol. 2007;25(5):467–76.
341. Komatsu H, Fujimoto S, Hara S, Sato Y, Yamada K, 354. Rocchetti MT, Centra M, Papale M, Bortone G,
Eto T. Relationship between serum IgA/C3 ratio and Palermo C, Centonze D, et al. Urine protein profile
progression of IgA nephropathy. Intern Med. of IgA nephropathy patients may predict the response
2004;43(11):1023–8. to ACE-inhibitor therapy. Proteomics. 2008;8(1):
342. Ishiguro C, Yaguchi Y, Funabiki K, Horikoshi S, 206–16.
Shirato I, Tomino Y. Serum IgA/C3 ratio may predict 355. Davin JC, Ten Berge IJ, Weening JJ. What is the
diagnosis and prognostic grading in patients with IgA difference between IgA nephropathy and Henoch-
nephropathy. Nephron. 2002;91(4):755–8. Schonlein purpura nephritis? Kidney Int. 2001;
343. Maeda A, Gohda T, Funabiki K, Horikoshi S, 59(3):823–34.
Shirato I, Tomino Y. Significance of serum IgA levels 356. Montoliu J, Lens XM, Torras A, Revert L. Henoch-
and serum IgA/C3 ratio in diagnostic analysis of Schonlein purpura and IgA nephropathy in father and
patients with IgA nephropathy. J Clin Lab Anal. son. Nephron. 1990;54(1):77–9.
2003;17(3):73–6. 357. Araque A, Sanchez R, Alamo C, Torres N, Praga
344. Shen P, He L, Jiang Y, Wang C, Chen M. Useful M. Evolution of immunoglobulin A nephropathy
indicators for performing renal biopsy in adult into Henoch-Schonlein purpura in an adult patient.
patients with isolated microscopic haematuria. Int J Am J Kidney Dis. 1995;25(2):340–2.
Clin Pract. 2007;61(5):789–94. 358. Silverstein DM, Greifer I, Folkert V, Bennett B, Corey
345. Zhang J, Wang C, Tang Y, Peng H, Ye ZC, Li CC, HE, Spitzer A. Sequential occurrence of IgA nephrop-
et al. Serum immunoglobulin A/C3 ratio predicts athy and Henoch-Schonlein purpura: support for
progression of immunoglobulin A nephropathy. common pathogenesis. Pediatr Nephrol. 1994;8(6):
Nephrology (Carlton). 2013;18(2):125–31. 752–3.
346. Stangou M, Alexopoulos E, Papagianni A, 359. Weiss JH, Bhathena DB, Curtis JJ, Lucas BA, Luke
Pantzaki A, Bantis C, Dovas S, et al. Urinary levels RG. A possible relationship between Henoch-
of epidermal growth factor, interleukin-6 and mono- Schonlein syndrome and IgA nephropathy (Berger’s
cyte chemoattractant protein-1 may act as predictor disease). An illustrative case. Nephron. 1978;22
markers of renal function outcome in immunoglobu- (4–6):582–91.
lin A nephropathy. Nephrology (Carlton). 2009; 360. Pouria S, Feehally J. Glomerular IgA deposition in
14(6):613–20. liver disease. Nephrol Dial Transplant. 1999;14(10):
347. Torres DD, Rossini M, Manno C, Mattace-Raso F, 2279–82.
D’Altri C, Ranieri E, et al. The ratio of epidermal 361. Kalsi J, Delacroix DL, Hodgson HJ. IgA in alcoholic
growth factor to monocyte chemotactic peptide-1 in cirrhosis. Clin Exp Immunol. 1983;52(3):499–504.
the urine predicts renal prognosis in IgA nephropathy. 362. Sancho J, Egido J, Sanchez-Crespo M, Blasco R.
Kidney Int. 2008;73(3):327–33. Detection of monomeric and polymeric IgA
348. Asao R, Asanuma K, Kodama F, Akiba-Takagi M, containing immune complexes in serum and kidney
Nagai-Hosoe Y, Seki T, et al. Relationships between from patients with alcoholic liver disease. Clin Exp
levels of urinary podocalyxin, number of urinary Immunol. 1982;47(2):327–35.
podocytes, and histologic injury in adult patients 363. Habib R, Girardin E, Gagnadoux MF, Hinglais N,
with IgA nephropathy. Clin J Am Soc Nephrol. Levy M, Broyer M. Immunopathological findings in
2012;7(9):1385–93. idiopathic nephrosis: clinical significance of glomer-
349. Peters HP, van den Brand JA, Wetzels JF. Urinary ular “immune deposits”. Pediatr Nephrol. 1988;2(4):
excretion of low-molecular-weight proteins as 402–8.
1030 K. Nakanishi and N. Yoshikawa

364. D’Amico G, Imbasciati E, Barbiano Di Belgioioso G, children with IgA nephropathy. Pediatr Nephrol.
Bertoli S, Fogazzi G, Ferrario F, et al. Idiopathic IgA 2013;28(1):71–6.
mesangial nephropathy. Clinical and histological 381. Wyatt RJ, Hogg RJ. Evidence-based assessment of
study of 374 patients. Medicine. 1985;64(1):49–60. treatment options for children with IgA nephropa-
365. Droz D, Kramar A, Nawar T, Noel LH. Primary IgA thies. Pediatr Nephrol. 2001;16(2):156–67.
nephropathy: prognostic factors. Contrib Nephrol. 382. Schlondorff D, Dendorfer U, Brumberger V,
1984;40:202–7. Keuneke C. Limitations of therapeutic approaches to
366. D’Amico G. Natural history of idiopathic IgA glomerular diseases. Kidney Int Suppl. 1995;50:
nephropathy: role of clinical and histological prog- S19–25.
nostic factors. Am J Kidney Dis. 2000;36(2):227–37. 383. Donadio Jr JV, Bergstralh EJ, Offord KP, Spencer DC,
367. Glassock RJ, Adler SG, Ward HJ, Cohen AH. Pri- Holley KE. A controlled trial of fish oil in IgA
mary glomerular diseases. In: Brenner BM, Rector nephropathy. Mayo Nephrology Collaborative
FCJ, editors. The kidney. 4th ed. Philadelphia: Group. N Engl J Med. 1994;331(18):1194–9.
Saunders; 1991. p. 1182–279. 384. Donadio Jr JV, Grande JP, Bergstralh EJ, Dart RA,
368. Hood SA, Velosa JA, Holley KE, Donadio Jr JV. Larson TS, Spencer DC. The long-term outcome of
IgA-IgG nephropathy: predictive indices of progres- patients with IgA nephropathy treated with fish oil in
sive disease. Clin Nephrol. 1981;16(2):55–62. a controlled trial. Mayo Nephrology Collaborative
369. Kobayashi Y, Tateno S, Hiki Y, Shigematsu H. IgA Group. J Am Soc Nephrol. 1999;10(8):1772–7.
nephropathy: prognostic significance of proteinuria 385. Donadio Jr JV, Larson TS, Bergstralh EJ, Grande JP.
and histological alterations. Nephron. 1983;34(3): A randomized trial of high-dose compared with
146–53. low-dose omega-3 fatty acids in severe IgA nephrop-
370. Katz A, Walker JF, Landy PJ. IgA nephritis with athy. J Am Soc Nephrol. 2001;12(4):791–9.
nephrotic range proteinuria. Clin Nephrol. 1983; 386. Dillon JJ. Fish oil therapy for IgA nephropathy: effi-
20(2):67–71. cacy and interstudy variability. J Am Soc Nephrol.
371. Chida Y, Tomura S, Takeuchi J. Renal survival rate of 1997;8(11):1739–44.
IgA nephropathy. Nephron. 1985;40(2):189–94. 387. Hogg RJ, Lee J, Nardelli N, Julian BA, Cattran D,
372. Neelakantappa K, Gallo GR, Baldwin DS. Proteinuria Waldo B, et al. Clinical trial to evaluate omega-3 fatty
in IgA nephropathy. Kidney Int. 1988;33(3):716–21. acids and alternate day prednisone in patients with
373. Bartosik LP, Lajoie G, Sugar L, Cattran DC. IgA nephropathy: report from the Southwest Pediatric
Predicting progression in IgA nephropathy. Am J Nephrology Study Group. Clin J Am Soc Nephrol.
Kidney Dis. 2001;38(4):728–35. 2006;1(3):467–74.
374. Hattori S, Karashima S, Furuse A, Terashima T, 388. Hogg RJ, Fitzgibbons L, Atkins C, Nardelli N, Bay RC.
Hiramatsu M, Murakami M, et al. Clinicopathological Efficacy of omega-3 fatty acids in children and adults
correlation of IgA nephropathy in children. Am J with IgA nephropathy is dosage- and size-dependent.
Nephrol. 1985;5(3):182–9. Clin J Am Soc Nephrol. 2006;1(6):1167–72.
375. Hogg RJ. Prognostic indicators and treatment of 389. Yoshikawa N, Ito H, Sakai T, Takekoshi Y, Honda M,
childhood IgA nephropathy. Contrib Nephrol. Awazu M, et al. A controlled trial of combined ther-
1995;111:194–9; discussion 9–200. apy for newly diagnosed severe childhood IgA
376. Tsuboi N, Kawamura T, Koike K, Okonogi H, nephropathy. The Japanese Pediatric IgA Nephropa-
Hirano K, Hamaguchi A, et al. Glomerular density thy Treatment Study Group. J Am Soc Nephrol.
in renal biopsy specimens predicts the long-term 1999;10(1):101–9.
prognosis of IgA nephropathy. Clin J Am Soc 390. Barratt J, Feehally J. Treatment of IgA nephropathy.
Nephrol. 2010;5(1):39–44. Kidney Int. 2006;69(11):1934–8.
377. Espinosa M, Ortega R, Gomez-Carrasco JM, Lopez- 391. Maschio G, Alberti D, Janin G, Locatelli F, Mann JF,
Rubio F, Lopez-Andreu M, Lopez-Oliva MO, et al. Motolese M, et al. Effect of the angiotensin-
Mesangial C4d deposition: a new prognostic factor in converting-enzyme inhibitor benazepril on the pro-
IgA nephropathy. Nephrol Dial Transplant. 2009; gression of chronic renal insufficiency. The
24(3):886–91. Angiotensin-Converting-Enzyme Inhibition in Pro-
378. Espinosa M, Ortega R, Sanchez M, Segarra A, gressive Renal Insufficiency Study Group. N Engl J
Salcedo MT, Gonzalez F, et al. Association of c4d Med. 1996;334(15):939–45.
deposition with clinical outcomes in IgA 392. Randomised placebo-controlled trial of effect of
nephropathy. Clin J Am Soc Nephrol. 2014;9(5): ramipril on decline in glomerular filtration rate and
897–904. risk of terminal renal failure in proteinuric,
379. Yata N, Nakanishi K, Shima Y, Togawa H, Obana M, non-diabetic nephropathy. The GISEN Group
Sako M, et al. Improved renal survival in Japanese (Gruppo Italiano di Studi Epidemiologici in
children with IgA nephropathy. Pediatr Nephrol. Nefrologia). Lancet. 1997;349(9069):1857–63.
2008;23:905–12. 393. Ruggenenti P, Perna A, Gherardi G, Garini G,
380. Shima Y, Nakanishi K, Hama T, Mukaiyama H, Zoccali C, Salvadori M, et al. Renoprotective proper-
Togawa H, Sako M, et al. Spontaneous remission in ties of ACE-inhibition in non-diabetic nephropathies
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1031

with non-nephrotic proteinuria. Lancet. 1999; 406. Pozzi C, Andrulli S, Del Vecchio L, Melis P,
354(9176):359–64. Fogazzi GB, Altieri P, et al. Corticosteroid effective-
394. Maschio G, Cagnoli L, Claroni F, Fusaroli M, ness in IgA nephropathy: long-term results of a ran-
Rugiu C, Sanna G, et al. ACE inhibition reduces domized, controlled trial. J Am Soc Nephrol.
proteinuria in normotensive patients with IgA 2004;15(1):157–63.
nephropathy: a multicentre, randomized, placebo- 407. Ballardie FW, Roberts IS. Controlled prospective
controlled study. Nephrol Dial Transplant. 1994; trial of prednisolone and cytotoxics in progressive
9(3):265–9. IgA nephropathy. J Am Soc Nephrol. 2002;13(1):
395. Perico N, Remuzzi A, Sangalli F, Azzollini N, 142–8.
Mister M, Ruggenenti P, et al. The antiproteinuric 408. Yoshikawa N, Honda M, Iijima K, Awazu M,
effect of angiotensin antagonism in human IgA Hattori S, Nakanishi K, et al. Steroid treatment for
nephropathy is potentiated by indomethacin. J Am severe childhood IgA nephropathy: a randomized,
Soc Nephrol. 1998;9(12):2308–17. controlled trial. Clin J Am Soc Nephrol. 2006;1(3):
396. Praga M, Gutierrez E, Gonzalez E, Morales E, 511–7.
Hernandez E. Treatment of IgA nephropathy with 409. Maes BD, Oyen R, Claes K, Evenepoel P, Kuypers D,
ACE inhibitors: a randomized and controlled trial. Vanwalleghem J, et al. Mycophenolate mofetil in IgA
J Am Soc Nephrol. 2003;14(6):1578–83. nephropathy: results of a 3-year prospective placebo-
397. Nakanishi K, Iijima K, Ishikura K, Hataya H, controlled randomized study. Kidney Int. 2004;65(5):
Awazu M, Sako M, et al. Efficacy and safety of 1842–9.
lisinopril for mild childhood IgA nephropathy: a 410. Frisch G, Lin J, Rosenstock J, Markowitz G,
pilot study. Pediatr Nephrol. 2009;24(4):845–9. D’Agati V, Radhakrishnan J, et al. Mycophenolate
398. Cheng J, Zhang W, Zhang XH, He Q, Tao XJ, Chen mofetil (MMF) vs placebo in patients with moder-
JH. ACEI/ARB therapy for IgA nephropathy: a meta ately advanced IgA nephropathy: a double-blind ran-
analysis of randomised controlled trials. Int J Clin domized controlled trial. Nephrol Dial Transplant.
Pract. 2009;63(6):880–8. 2005;20(10):2139–45.
399. Coppo R, Peruzzi L, Amore A, Piccoli A, Cochat P, 411. Tang S, Leung JC, Chan LY, Lui YH, Tang CS, Kan
Stone R, et al. IgACE: a placebo-controlled, random- CH, et al. Mycophenolate mofetil alleviates persistent
ized trial of angiotensin-converting enzyme inhibitors proteinuria in IgA nephropathy. Kidney Int.
in children and young people with IgA nephropathy 2005;68(2):802–12.
and moderate proteinuria. J Am Soc Nephrol. 412. Samuels JA, Strippoli GF, Craig JC, Schena FP,
2007;18(6):1880–8. Molony DA. Immunosuppressive treatments for
400. Russo D, Minutolo R, Pisani A, Esposito R, immunoglobulin A nephropathy: a meta-analysis of
Signoriello G, Andreucci M, et al. Coadministration randomized controlled trials. Nephrology (Carlton).
of losartan and enalapril exerts additive 2004;9(4):177–85.
antiproteinuric effect in IgA nephropathy. Am J Kid- 413. Kamei K, Nakanishi K, Ito S, Saito M, Sako M,
ney Dis. 2001;38(1):18–25. Ishikura K, et al. Long-term results of a randomized
401. Nakao N, Yoshimura A, Morita H, Takada M, controlled trial in childhood IgA nephropathy. Clin J
Kayano T, Ideura T. Combination treatment of Am Soc Nephrol. 2011;6(6):1301–7.
angiotensin-II receptor blocker and angiotensin- 414. Yoshikawa N, Nakanishi K, Ishikura K, Hataya H,
converting-enzyme inhibitor in non-diabetic renal Iijima K, Honda M. Combination therapy with
disease (COOPERATE): a randomised controlled mizoribine for severe childhood IgA nephropathy: a
trial. Lancet. 2003;361(9352):117–24. pilot study. Pediatr Nephrol. 2008;23:757–63.
402. Li PK, Leung CB, Chow KM, Cheng YL, Fung SK, 415. Yoshikawa N, Ito H, Sakai T, Takekoshi Y, Honda M,
Mak SK, et al. Hong Kong study using valsartan in Awazu M, et al. A prospective controlled study of
IgA nephropathy (HKVIN): a double-blind, random- sairei-to in childhood IgA nephropathy with focal/
ized, placebo-controlled study. Am J Kidney Dis. minimal mesangial proliferation. Japanese Pediatric
2006;47(5):751–60. IgA Nephropathy Treatment Study Group. Nippon
403. Bhattacharjee R, Filler G. Additive antiproteinuric Jinzo Gakkai Shi. 1997;39(5):503–6.
effect of ACE inhibitor and losartan in IgA nephrop- 416. Appel GB, Waldman M. The IgA nephropathy treat-
athy. Pediatr Nephrol. 2002;17(4):302–4. ment dilemma. Kidney Int. 2006;69(11):1939–44.
404. Yang Y, Ohta K, Shimizu M, Nakai A, Kasahara Y, 417. Locatelli F, Vecchio LD, Pozzi C. IgA glomerulone-
Yachie A, et al. Treatment with low-dose angiotensin- phritis: beyond angiotensin-converting enzyme inhib-
converting enzyme inhibitor (ACEI) plus angiotensin itors. Nat Clin Pract Nephrol. 2006;2(1):24–31.
II receptor blocker (ARB) in pediatric patients with 418. Kawamura T, Yoshimura M, Miyazaki Y,
IgA nephropathy. Clin Nephrol. 2005;64(1):35–40. Okamoto H, Kimura K, Hirano K, et al. A multicenter
405. Pozzi C, Bolasco PG, Fogazzi GB, Andrulli S, randomized controlled trial of tonsillectomy com-
Altieri P, Ponticelli C, et al. Corticosteroids in IgA bined with steroid pulse therapy in patients with
nephropathy: a randomised controlled trial. Lancet. immunoglobulin A nephropathy. Nephrol Dial Trans-
1999;353(9156):883–7. plant. 2014;29:1546–53.
1032 K. Nakanishi and N. Yoshikawa

419. Hogg RJ. IgA nephropathy: what’s new? Pediatr 434. Utani A, Ohta M, Shinya A, Ohno S, Takakuwa H,
Nephrol. 2007;22(11):1809–14. Yamamoto T, et al. Successful treatment of
420. Allen DM, Diamond LK, Howell DA. Anaphylactoid adult Henoch-Schonlein purpura with factor XIII
purpura in children (Schonlein-Henoch syndrome): concentrate. J Am Acad Dermatol. 1991;24(3):
review with a follow-up of the renal complications. 438–42.
AMA J Dis Child. 1960;99:833–54. 435. White RH, Cameron JS, Trounce JR. Immunosup-
421. Jones NF, Creamer B, Gimlette TM. pressive therapy in steroid-resistant proliferative
Hypoproteinaemia in anaphylactoid purpura. Br glomerulonephritis accompanied by the nephrotic
Med J. 1966;2(5523):1166–8. syndrome. Br Med J. 1966;2(5518):853–60.
422. Reif S, Jain A, Santiago J, Rossi T. Protein losing 436. Shin JI, Park JM, Shin YH, Kim JH, Lee JS, Kim PK,
enteropathy as a manifestation of Henoch-Schonlein et al. Can azathioprine and steroids alter the progres-
purpura. Acta Paediatr Scand. 1991;80(4):482–5. sion of severe Henoch-Schonlein nephritis in chil-
423. Kaku Y, Nohara K, Honda S. Renal involvement in dren? Pediatr Nephrol. 2005;20(8):1087–92.
Henoch-Schonlein purpura: a multivariate analysis of 437. Niaudet P, Habib R. Methylprednisolone pulse ther-
prognostic factors. Kidney Int. 1998;53(6):1755–9. apy in the treatment of severe forms of Schonlein-
424. Sano H, Izumida M, Shimizu H, Ogawa Y. Risk fac- Henoch purpura nephritis. Pediatr Nephrol.
tors of renal involvement and significant proteinuria 1998;12(3):238–43.
in Henoch-Schonlein purpura. Eur J Pediatr. 438. Kawasaki Y, Suyama K, Hashimoto K, Hosoya M.
2002;161(4):196–201. Methylprednisolone pulse plus mizoribine in children
425. Rigante D, Candelli M, Federico G, Bartolozzi F, with Henoch-Schoenlein purpura nephritis. Clin
Porri MG, Stabile A. Predictive factors of renal Rheumatol. 2011;30(4):529–35.
involvement or relapsing disease in children with 439. Kawasaki Y, Suzuki J, Suzuki H. Efficacy of methyl-
Henoch-Schonlein purpura. Rheumatol Int. 2005; prednisolone and urokinase pulse therapy combined
25(1):45–8. with or without cyclophosphamide in severe
426. Shin JI, Park JM, Shin YH, Hwang DH, Kim JH, Henoch-Schoenlein nephritis: a clinical and histo-
Lee JS. Predictive factors for nephritis, relapse, and pathological study. Nephrol Dial Transplant.
significant proteinuria in childhood Henoch- 2004;19(4):858–64.
Schonlein purpura. Scand J Rheumatol. 2006;35(1): 440. Ronkainen J, Autio-Harmainen H, Nuutinen M.
56–60. Cyclosporin A for the treatment of severe Henoch-
427. Bogdanovic R. Henoch-Schonlein purpura nephritis Schonlein glomerulonephritis. Pediatr Nephrol.
in children: risk factors, prevention and treatment. 2003;18(11):1138–42.
Acta Paediatr. 2009;98(12):1882–9. 441. Shin JI, Park JM, Shin YH, Kim JH, Kim PK, Lee JS,
428. Jauhola O, Ronkainen J, Koskimies O, et al. Cyclosporin A therapy for severe Henoch-
Ala-Houhala M, Arikoski P, Holtta T, et al. Renal Schonlein nephritis with nephrotic syndrome. Pediatr
manifestations of Henoch-Schonlein purpura in a Nephrol. 2005;20(8):1093–7.
6-month prospective study of 223 children. Arch Dis 442. Jauhola O, Ronkainen J, Autio-Harmainen H,
Child. 2010;95(11):877–82. Koskimies O, Ala-Houhala M, Arikoski P, et al.
429. Saulsbury FT. Corticosteroid therapy does not prevent Cyclosporine A vs. methylprednisolone for Henoch-
nephritis in Henoch-Schonlein purpura. Pediatr Schonlein nephritis: a randomized trial. Pediatr
Nephrol. 1993;7(1):69–71. Nephrol. 2011;26(12):2159–66.
430. Mollica F, Li Volti S, Garozzo R, Russo G. Effective- 443. Park JM, Won SC, Shin JI, Yim H, Pai KS. Cyclo-
ness of early prednisone treatment in preventing the sporin A therapy for Henoch-Schonlein nephritis with
development of nephropathy in anaphylactoid pur- nephrotic-range proteinuria. Pediatr Nephrol.
pura. Eur J Pediatr. 1992;151(2):140–4. 2011;26(3):411–7.
431. Ronkainen J, Koskimies O, Ala-Houhala M, 444. Kauffmann RH, Houwert DA. Plasmapheresis in rap-
Antikainen M, Merenmies J, Rajantie J, et al. Early idly progressive Henoch-Schoenlein glomerulone-
prednisone therapy in Henoch-Schonlein purpura: a phritis and the effect on circulating IgA immune
randomized, double-blind, placebo-controlled trial. complexes. Clin Nephrol. 1981;16(3):155–60.
J Pediatr. 2006;149(2):241–7. 445. Camerone G, Garelli S, Valbonesi M, Mosconi L.
432. Dudley J, Smith G, Llewelyn-Edwards A, Bayliss K, Plasma exchange treatment in a patient with severe
Pike K, Tizard J. Randomised, double-blind, placebo- Schoenlein-Henoch purpura. Minerva Med.
controlled trial to determine whether steroids 1982;73(18):1185–7.
reduce the incidence and severity of nephropathy in 446. Coppo R, Basolo B, Roccatello D, Giachino O,
Henoch-Schonlein Purpura (HSP). Arch Dis Child. Lajolo D, Martina G, et al. Plasma exchange in pro-
2013;98(10):756–63. gressive primary IgA nephropathy. Int J Artif Organs.
433. Rosenblum ND, Winter HS. Steroid effects on 1985;8 Suppl 2:55–8.
the course of abdominal pain in children with 447. Hattori M, Ito K, Konomoto T, Kawaguchi H,
Henoch-Schonlein purpura. Pediatrics. 1987;79(6): Yoshioka T, Khono M. Plasmapheresis as the sole
1018–21. therapy for rapidly progressive Henoch-Schonlein
32 Immunoglobulin A Nephropathies in Children (Includes HSP) 1033

purpura nephritis in children. Am J Kidney Dis. 456. Butani L, Morgenstern BZ. Long-term outcome in
1999;33(3):427–33. children after Henoch-Schonlein purpura nephritis.
448. Ninchoji T, Kaito H, Nozu K, Hashimura Y, Kanda K, Clin Pediatr (Phila). 2007;46(6):505–11.
Kamioka I, et al. Treatment strategies for 457. Edstrom Halling S, Soderberg MP, Berg UB. Predic-
Henoch-Schonlein purpura nephritis by histological tors of outcome in Henoch-Schonlein nephritis.
and clinical severity. Pediatr Nephrol. 2011;26(4): Pediatr Nephrol. 2010;25(6):1101–8.
563–9. 458. Ronkainen J, Nuutinen M, Koskimies O. The adult
449. Chartapisak W, Opastirakul S, Hodson EM, kidney 24 years after childhood Henoch-Schonlein
Willis NS, Craig JC. Interventions for preventing purpura: a retrospective cohort study. Lancet.
and treating kidney disease in Henoch-Schonlein Pur- 2002;360(9334):666–70.
pura (HSP). Cochrane Database Syst Rev. 2009;3: 459. Halling SF, Soderberg MP, Berg UB. Henoch
CD005128. Schonlein nephritis: clinical findings related to renal
450. Chartapisak W, Opastiraku S, Willis NS, Craig JC, function and morphology. Pediatr Nephrol.
Hodson EM. Prevention and treatment of renal dis- 2005;20(1):46–51.
ease in Henoch-Schonlein purpura: a systematic 460. Shin JI, Park JM, Kim JH, Lee JS, Jeong HJ. Factors
review. Arch Dis Child. 2009;94(2):132–7. affecting histological regression of crescentic
451. Davin JC, Coppo R. Pitfalls in recommending Henoch-Schonlein nephritis in children. Pediatr
evidence-based guidelines for a protean disease like Nephrol. 2006;21(1):54–9.
Henoch-Schonlein purpura nephritis. Pediatr 461. Brenner BM. Hemodynamically mediated glomerular
Nephrol. 2013;28(10):1897–903. injury and the progressive nature of kidney disease.
452. Davin JC, Coppo R. Henoch-Schonlein purpura Kidney Int. 1983;23(4):647–55.
nephritis in children. Nat Rev Nephrol. 2014;10(10): 462. Meulders Q, Pirson Y, Cosyns JP, Squifflet JP, van
563–73. Ypersele de Strihou C. Course of Henoch-Schonlein
453. Farine M, Poucell S, Geary DL, Baumal R. nephritis after renal transplantation. Report on ten
Prognostic significance of urinary findings and patients and review of the literature. Transplantation.
renal biopsies in children with Henoch- 1994;58(11):1179–86.
Schonlein nephritis. Clin Pediatr (Phila). 1986; 463. White RHR, Yoshikawa N. Henoch-Schönlein
25(5):257–9. nephritis. In: Holliday MA, Barratt TM, Avner ED,
454. Schärer K, Krmar R, Querfeld U, Ruder H, editors. Pediatric nephrology. Baltimore: Williams &
Waldherr R, Schaefer F. Clinical outcome of Wilkins; 1993. p. 729–38.
Schonlein-Henoch purpura nephritis in children. 464. White RHR. Henoch-Schönlein purpura. In: Churg A,
Pediatr Nephrol. 1999;13(9):816–23. Churg J, editors. Systemic vasculitides. New York:
455. Coppo R, Andrulli S, Amore A, Gianoglio B, Igaku-Shoin; 1991:203–217.
Conti G, Peruzzi L, et al. Predictors of outcome in 465. KDIGO guidelines on glomerulonephritis Chapter 11:
Henoch-Schonlein nephritis in children and adults. Henoch–Schönlein purpura nephritis. Kidney Int
Am J Kidney Dis. 2006;47(6):993–1003. Suppl. 2012;2:218–220; doi:10.1038/kisup.2012.24.
Membranoproliferative and
C3-Mediated GN in Children 33
Christoph Licht, Magdalena Riedl, Matthew C. Pickering,
and Michael Braun

Contents Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1045


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1036 Differential Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1045
Histopathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1037 Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1046
Supportive Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1046
Complement System . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1037 Immunosuppressive Therapy . . . . . . . . . . . . . . . . . . . . . . . 1046
Lessons Learned from Animal Models . . . . . . . . . . 1040 Complement-Targeting Therapy . . . . . . . . . . . . . . . . . . . 1047

Lessons Learned from Patients . . . . . . . . . . . . . . . . . . 1041 Clinical Outcomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1047


Autoimmune Forms of C3G . . . . . . . . . . . . . . . . . . . . . . . . 1041 Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1048
Genetic Forms of C3G . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1042
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1049
Clinical Manifestation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1043

C. Licht (*)
Division of Nephrology, The Hospital for Sick Children,
University of Toronto, Toronto, ON, Canada
Research Institute, Cell Biology Program, The Hospital for
Sick Children, Toronto, ON, Canada
Department of Paediatrics, University of Toronto, Toronto,
ON, Canada
e-mail: christoph.licht@sickkids.ca
M. Riedl
Research Institute, Cell Biology Program, The Hospital for
Sick Children, Toronto, ON, Canada
Department of Paediatrics, Innsbruck Medical University,
Innsbruck, Tyrol, Austria
M.C. Pickering
Centre for Complement and Inflammation Research,
Imperial College, London, UK
M. Braun
Renal Section, Department of Pediatrics, Texas Children’s
Hospital, Balyor College of Medicine, Houston, TX, USA

# Springer-Verlag Berlin Heidelberg 2016 1035


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_29
1036 C. Licht et al.

and capillary wall remodeling with formation of


List of Abbreviations
double contours. This injury pattern is a result of
aHUS Atypical hemolytic uremic
deposition of immunoglobulins (Ig)/immune
syndrome
complexes (IC) and/or complement proteins in
AMD Age-related macular degeneration
the mesangium and/or along the capillary wall of
AP Alternative pathway
the glomerulus [1, 2]. The increasing understand-
(of complement)
ing of the underlying pathophysiology has
aPL Acquired partial lipodystrophy
recently resulted in a reclassification of MPGN
C Complement
into (i) a primary complement-mediated C3
C3G C3 glomerulopathy
glomerulopathy (C3G) with predominant C3
C3GN C3 glomerulonephritis
deposition and (ii) a secondary Ig-/IC-mediated
C3NeF C3 nephritic factor
glomerulonephritis with mainly IgG staining on
CFB Complement factor B
immunohistochemistry [2, 3]. C3 glomerulopathy
CFH Complement factor H
enables the inclusion of cases with C3 deposition
CFHR1-5 Complement factor H-related
without a membranoproliferative pattern on light
protein 1–5
microscopy [3] and encompasses all glomerular
CFI Complement factor I
lesions in which there is predominant C3 staining
CFP Properdin
as defined as an immunohistochemistry C3c
CP Classical pathway
intensity 2 orders of magnitude more than any
(of complement)
other immune reactant (e.g., IgG/IgA, C1q) using
DDD Dense deposit disease
a scale ranging from 0 to 3 [3]. MPGN, in distinc-
DEAP-HUS Deficiency of CFHR plasma
tion, may result from Ig/IC deposition in the kid-
proteins and CFH autoantibody
ney and may be either idiopathic or secondary to
positive HUS
infections, autoimmune diseases, malignancies,
ESRD End-stage renal disease
or monoclonal gammopathy (Table 1) [4–6].
FFP Fresh frozen plasma
C3G is considered a primary complement disease,
GN Glomerulonephritis
where complement deposition is a result of defec-
IC Immune complexes
tive control of the complement AP [7, 8]. Of note,
Ig Immunoglobulins
complement activation – primarily of the classical
IVCP Intravenous cyclophosphamide
pathway (CP) – also occurs in Ig-/IC-mediated
LP Lectin pathway
glomerulonephritis [9]. The diagnosis of C3
(of complement)
glomerulopathy is based on the result of light
MCP Membrane cofactor protein
and electron microscopy including immunohisto-
MPGN Membranoproliferative
chemistry [3]. Treatment approaches include
glomerulonephritis
immunosuppressant therapy and treatment of the
NeF Nephritic factor
underlying condition in secondary MPGN and
PE Plasma exchange
targeting the complement system for primary
PI Plasma infusion
C3G. Some patients may even recover spontane-
PIGN Postinfectious glomerulonephritis
ously without specific treatment. Evidence for
SCR Short consensus repeat
treatment-specific benefits is still lacking, and
TMA Thrombotic microangiopathy
formal treatment trials will be required to establish
treatment guidelines and define long-term treat-
ment benefits.
Introduction While focusing on C3G, references used
in this chapter are mainly based on the
Membranoproliferative glomerulonephritis (MPGN) traditional nomenclature but are interpreted as
describes a histopathological pattern character- best as possible in light of the new consensus
ized by mesangial and endocapillary proliferation guidelines.
33 Membranoproliferative and C3-Mediated GN in Children 1037

Table 1 Causes of secondary MPGN proliferative glomerulonephritis, or even a necro-


Infectious diseases: bacterial/viral/protozoal tizing and crescentic glomerulonephritis [3, 10].
Hepatitis B, C, EBV, HIV Electron microscopy (EM) is needed to differen-
Endocarditis/visceral abcesses tiate C3GN and DDD. In C3GN, discrete C3
Infected ventriculoatrial shunts/empyema deposits are located in the mesangium and along
Malaria, schistosomiasis, mycoplasma the capillary walls, whereas in DDD, C3 deposits
Tuberculosis, leprosy are more intense in the mesangium and within the
Epstein-Barr virus infection glomerular basement membrane (GBM) forming
Brucellosis a unique ribbon-shaped band [3, 11].
Systemic immune diseases Mesangial C3 with at least codominant IgG
Cryoglobulinemia deposition compared to C3G with mesangial,
Systemic lupus erythematosus
subendothelial, and subepithelial humps by EM
Sjögren’s syndrome
are characteristic for postinfectious GN (PIGN), a
Rheumatoid arthritis
major differential diagnosis of C3G [3]. Some
Hereditary deficiencies of complement components
patients showed only the combination of C3 depo-
X-linked agammaglobulinemia
Neoplasms/dysproteinemias
sition without IgG and humps. As clinical signs
Plasma cell dyscrasia such as low C3, hematuria/proteinuria, or deterio-
Fibrillary and immunotactoid glomerulonephritis ration of renal function were persistent and defects
Light-chain deposition disease in the AP were detected, these patients received the
Heavy-chain deposition disease diagnosis of “atypical” PIGN, very likely being
Light- and heavy-chain deposition disease patients with C3G [12]. As infections – including
Leukemias and lymphomas (with cryoglobulinemia) streptococcal – are a common trigger for C3G and
Waldenstrom macroglobulinemia humps also occur in biopsies of patients with C3G,
Carcinomas, Wilms’ tumor, malignant melanoma the differential diagnosis is difficult and mostly
Chronic liver disease relies on clinical observation.
Chronic active hepatitis (B, C)
Cirrhosis
Alpha-1-antitrypsin deficiency Complement System
Miscellaneous
Thrombotic microangiopathy
The complement system (Fig. 2) is an integral part
Sickle-cell disease
of the innate immune system for antimicrobial
Partial lipodystrophy
defense and immune complex clearing
Transplant glomerulopathy
[13–15]. However, defective regulation or
Niemann-Pick disease (Type C)
overactivation caused by mutations and autoanti-
bodies has been linked with a variety of renal
diseases, including thrombotic microangiopathy
Histopathology (TMA) especially atypical hemolytic uremic syn-
drome (aHUS), systemic lupus erythematosus
C3G presently includes two entities: C3 glomer- (SLE), antibody-mediated rejection (AMR),
ulonephritis (C3GN e.g., CFHR5 nephropathy) ANCA-associated vasculitis (AAV), and membra-
and dense deposit disease (DDD), former classi- nous nephropathy (MN) [16, 17]. The classical
fied as MPGN Type II (Fig. 1) [3]. On light pathway (CP) is activated by immunoglobulins
microscopy, C3GN and DDD have an or immune complexes and thus especially
overlapping spectrum of features including involved in autoimmune diseases and the Ig-/IC-
membranoproliferative glomerulonephritis, dif- mediated form of MPGN. The lectin pathway
fuse proliferative glomerulonephritis, mesangial (LP) is activated by repetitive carbohydrate
1038 C. Licht et al.

Fig. 1 Histological features of C3G. Panel highlighting the scanty (as illustrated here) but can include large subepithelial
microscopic features of MPGN Type I (a–c), C3G (d–f) and and/or subendothelial deposits, with or without mesangial
DDD (g–i) by light microscopy (top row), immunofluores- interposition (also seen here). (g–i) DDD imparts a uniform
cence (middle row) and electron microscopy (bottom row). ribbonlike appearance to the glomerular basement mem-
(a–c) Typical MPGN results in a cellular glomerulus with brane, highlighted here by PAS staining. Mesangial regions
expanded mesangial regions resulting from increased matrix show increased matrix and cellularity, but mesangial interpo-
and cellularity, as well as immune complex deposition. Cap- sition along capillary loops is an inconsistent feature. By
illary loops are thickened with narrowed lumina from immunofluorescence, C3 deposition can be detected,
enlarged endothelial cells, mesangial interposition, and although usually less than seen in C3G. Electron microscopy
immune complex deposition. There is strong staining for is diagnostic with the presence of an intramembranous
both IgG and C3 by immunofluorescence (only IgG is electron-dense alteration to the basement membranes along
shown). By electron microscopy, capillary loops show prom- capillary loops and sometimes in mesangial regions. Early in
inent subendothelial deposits and mesangial interposition. the disease, this change involved loops partially but becomes
(d–f) C3G can have a wide spectrum of appearances ranging confluent as the disease progresses. (a, d, f) PAS staining,
from mesangial proliferative, membranoproliferate, to a nec- original magnification 400, (b) IgG, original magnification
rotizing crescentic glomerulonephritis (illustrated here). 400, (e) and (h) and B3: C3, original magnification 400,
C3 deposition predominates by immunofluorescence. By (c, f, i) original magnification 10,000)
electron microscopy, immune complex deposition may be
33 Membranoproliferative and C3-Mediated GN in Children 1039

Fig. 2 (a) Activation and a C3(H2O)


regulation of the complement
alternative pathway (AP): CFH
Physiological balance DAF
between spontaneous C3(H2O)Bb
activation and regulation of AP initation C3 convertase
the AP. Mutations in CFH
and C3 or autoantibodies to C3bBb
CFH, C3bBb (i.e., C3NeF) C3 C3b AP amplification
or CFB cause excessive C3 convertase
fluid-phase C3 activation CFH
resulting in the glomerular CFI
accumulation of C3 MCP
split products giving rise C3bBbC3b
iC3b C5 convertase
to the specific
immunofluorescence
patterns found in DDD and
C3GN. (b) Complement C3d, C3e, C3f, C3g C5b-9
dysregulation in C3G:
Mutations and
autoantibodies (red) b C3(H2O)
involved in C3G
CFH
DAF
C3(H2O)Bb C3NeF
AP initation C3 convertase CFB
C3b
C3bBb
C3 C3b AP amplification
CFH C3 convertase
(mut. + abs.)
CFI
MCP C3bBbC3b
iC3b
C5 convertase

Deposition of Injury / cell death


C3 split products (C5b-9)
(C3d, C3e, C3f, C3g)

structures found, e.g., on the surface of bacteria deposited on target surfaces (“opsonization”) and
[13–15]. Different from the CP and LP, the AP is allows for the formation of the C5 convertase
constitutively active and requires tight (negative) (C3bBbC3b), activation of C5, and induction of
regulation to maintain the balance between phys- the terminal complement cascade (TCC), ulti-
iological activation of complement when and mately resulting in the formation of the membrane
where needed and to prevent harmful attack complex (MAC) C5b-9 (Fig. 2a) [13–15].
overactivation. Initiation of the CP and LP results A number of soluble and membrane-anchored
in the activation of C3 to C3b, which – in the regulators limit complement activation via inacti-
AP – occurs spontaneously via a process called vation of C3b or acceleration of the physiological
“tick over.” Together with complement factor B decay of the C3 convertase, C3bBb. The most
(CFB) – activated by complement factor D important complement regulator, fluid-phase
(CFD) – and properdin (CFP), C3b forms the AP complement factor H (CFH), provides comple-
C3 convertase (C3bBb), which drastically ment regulation via three different mechanisms:
enhances C3 activation (“amplification”). C3b is it limits C3b binding to the endothelial surface and
1040 C. Licht et al.

formation of the C3 convertase by competition, CFH (through either plasma or purified protein)
C3b cleavage (cofactor activity), and acceleration delayed disease progression [26, 27]. Similarly,
of the natural decay of the C3 convertase (decay- C3G developed spontaneously in mice with com-
accelerating activity) (Fig. 2a). plete CFH deficiency (cfh/), generated
While the C-terminal region of CFH is respon- through gene targeting [25]. In both the
sible for surface recognition and binding to C3b, the CFH-deficient pig and mouse models, C3 accu-
N-terminal region of CFH provides cofactor and mulates along the GBM in the setting of systemic
decay-accelerating function [18]. It is not surprising hypocomplementemia. In both animal models,
that aHUS – a disease characterized by uncontrolled immune electron microscopy identified comple-
complement activation on endothelial cells – is ment components (i.e., C3, C5, C5b-9) within
associated with mutations in the C-terminal region the electron-dense deposits [25, 26]. An essential
of CFH, while in C3G – a disease associated with role for the AP was demonstrated by the observa-
enhanced C3 conversion – mutations are rather tion that cfh/ mice did not develop renal dis-
detected in the N-terminal region [19]. ease if they were also genetically deficient in the
Besides CFH, there are five proteins AP activation protein, factor B [25]. The develop-
sharing sequence and structural homology with ment of C3 along the GBM in complete CFH
CFH – the complement factor H-related proteins deficiency is dependent of CFI since cfh/
(CFHR) 1–5. CFHR 1 contributes to complement mice lacking CFI (cfh/; cfi/) developed
regulation on the level of the C5 convertase [20]. mesangial and not GBM-associated C3
CFHR1, CFHR2, and CFHR5 share a dimeriza- deposition [28].
tion motif, which allows them to form dimers, Importantly, the phenotype associated with
which can antagonize CFH at physiological con- complete CFH deficiency in humans included
centrations [21]. By contrast, recent studies have not only C3 glomerulopathy but also atypical
shown that CFHR1 [22], CFHR5 [21], and hemolytic uremic syndrome (aHUS) indicating
CFHR4 [23] unlike CFH do not have the ability that defects in the complement AP can give rise
to negatively regulate C3 at physiological concen- to at least two distinct renal pathologies. The
trations. In fact, by competing for surface ligands majority of aHUS-associated CFH mutations do
with CFH, they can prevent the ability of CFH to not result in complete deficiency of CFH but
negatively regulate C3 activation. This process selectively impair the surface recognition
has been termed CFH deregulation [21]. domains of the protein. These mutations were
In addition, cellular surfaces are protected via modeled experimentally by creating a
complement receptor 1 (CR1) and membrane CFH-deficient mouse strain that expressed
cofactor protein (MCP; CD46), both acting as transgenically a mutant CFH protein that func-
cofactors to CFI; via decay-accelerating factor tionally mimicked the human aHUS-associated
(DAF; CD55); and via CD59 which prevents CFH mutations. The mutant protein lacked the
assembly of the C5b-9 complex (Fig. 2a) [13–15]. terminal 5 surface recognition domains (denoted
C3 inactivation results in the formation of CFHΔ16-20). When this protein was expressed in
iC3b, C3dg, and C3d, split products which can CFH-deficient mice (cfh/. CFHΔ16-20), sys-
be used to detect complement activation but are temic hypocomplementemia was ameliorated
also of biological relevance, as they interact with since the mutant protein had intact complement
the adaptive immune system (Fig. 2a) [24]. regulatory domains. However, the animals spon-
taneously developed aHUS and not C3G, and the
aHUS phenotype was dependent on C5 activation
Lessons Learned from Animal Models [29, 30]. The conclusions of these studies were
that for aHUS to develop, defective complement
A lesion consistent with MPGN type 2 developed regulation along the renal endothelium was
spontaneously in pigs with complete genetic defi- required but so too was an intact (or partially
ciency of CFH [25]. Notably, administration of intact) complement system that enabled sufficient
33 Membranoproliferative and C3-Mediated GN in Children 1041

C3 and C5 to be available to mediate the enhanced levels of TCC activation were also
complement-induced endothelial damage. In the found in C3G patients [19].
cfh/. CFHΔ16-20 animals both were present. The pathogenetic role of complement was
In contrast in the cfh/ animals, while there further supported by recent work by Sethi
was clearly no CFH-mediated regulation of et al., who was able to confirm the occurrence
complement along endothelium since the mice of proteins of the alternative and terminal path-
lacked CFH, the deficiency also resulted in way in the glomerulus of patients with C3G
secondary depletion of C3 and C5. The auto- using a combination of laser microdissection
depletion of these components, and consequent and mass spectrometry [7, 8]. Of note, since
lack of an intact complement system, meant CFB was not detected, the authors concluded
that these mice were protected from the develop- that AP activation occurred in the fluid phase
ment of spontaneous aHUS. Conversely, the but not locally.
observation that cfh/. CFHΔ16-20 animals
did not develop C3G suggested that the
systemic hypocomplementemia due to
complete fluid-phase dysregulation of the com- Autoimmune Forms of C3G
plement AP was driving the C3G seen in the
cfh/ mice. There are examples of human In 1965, the first association of low C3 and
C3G where there is and is not systemic MPGN was reported, followed by the hypothesis,
hypocomplementemia, so fluid-phase comple- that a factor in serum of patients with GN leads to
ment AP dysregulation is relevant for some but increased cleavage of C3 [31]. Subsequently, an
not all C3G. antibody binding and stabilizing the AP C3
convertase (C3bBb), thus enhancing C3 activa-
tion and C3b generation, was detected and
named C3 nephritic factor (C3NeF) [32]. C3NeF
Lessons Learned from Patients was detected in 86 % of DDD and 46 % of C3GN
patients, respectively, and is associated with
There has been a long recognized role for com- decreased C3 levels [19]. C3NeF levels can be
plement in patients with MPGN, as low C3 is a fluctuant during the clinical course of a patient
hallmark feature of the clinical presentation. and do not necessarily reflect disease activity or
Mutations or autoantibodies result in the loss of treatment status [33]. Of note, in 1 case, C3NeF
control of AP C3 convertase via one or more of was reported to disappear after renal
the following scenarios (Fig. 2b): transplantation [34].
As C3NeF are heterogeneous, reliable detec-
– Antibodies stabilizing the AP C3 tion is challenging. Current assays involve mea-
convertase thus prolonging the natural decay surement of IgG binding to the C3 convertase or
of the AP C3 convertase and rendering it the detection of the ability to stabilize the AP C3
overactive convertase [35]. The latter was reported as
– Mutations or antibodies to CFH that result in the (currently) most sensitive test [36]. Efforts
the absence or loss of function of CFH creating an international standard for the detection
resulting in the loss of control of the AP C3 of C3NeF are currently under way (Michael
convertase and its enhanced activity Kirschfink – personal communication). C3NeF
– Mutations in C3 or CFB that result in an was also reported in other renal diseases, such
exceedingly stable C3 convertase with as SLE [37–39], PIGN [12], patients with
prolonged decay and enhanced function meningococcal meningitis [40], and healthy
individuals [35]. The finding of C3NeF does
In all scenarios, the functional consequence is not exclude the coexistence of complement
an enhanced activation rate of C3. Of note mutations, and despite C3NeF positivity,
1042 C. Licht et al.

Table 2 Diagnostic workup for C3G MPGN 1, C3G, and DDD, it is not
Global complement CH50, APH50 possible to detect a mutation in CFH, CFI, or
function CD46. To date, complement mutations have
Specific complement C3, C4, C3d been reported in the following complement
activation genes: CFH, CFHR5, CFI, MCP/CD46, C3, and
Terminal pathway SC5b-9
CFB [19, 21, 22, 47–59]. Additional mutations or
activation
Complement protein CFH, CFI, CFB
internal duplications in complement factor
levels H-related proteins (CFHRs), or the formation of
Autoantibody screening C3 Nephritic Factor hybrid genes, were associated with familial C3G
CFH/CFB/C3b [60, 61]. Risk haplotypes were identified in CFH,
Autoantibodies C3, and MCP/CD46, with the CFH Y402H hap-
Mutation screening CFH, CFI, MCP/CD46, C3 lotype more frequently reported in DDD and the
CFHR1-5 (MLPA) MCP -652A4G polymorphism in C3G and
Adapted from Pickering et al., Kidney Int 2013 (Ref. [3]) MPGN1 [19, 47]. The presence of two or more
MLPA multiplex ligation-dependent probe amplification
complement haplotypes significantly increased
Italic: rare conditions
The authors acknowledge that not all tests may be available the risk of disease manifestation [19, 47].
to all clinicians and that local/national clinical practice Table 3 gives an overview of mutations, rare
recommendations for the diagnostic workup of C3G variants, and polymorphisms associated with
may apply
C3G. Interestingly, several mutations have
already been described in patients with aHUS
[19]. Compared to aHUS, mutations in CFH asso-
ciated with C3G are preferentially located in the
comprehensive complement diagnostics (Table 2) N-terminal region of CFH or affect Cys
should be performed [41]. residues throughout the CFH protein. In either
Other autoantibodies including anti-CFB and case, these mutations result in the loss of function
anti-C3b antibodies have been reported, all of of CFH complement regulatory activity similar to
which result in the loss of AP C3 convertase a CFH null genotype [19, 55, 62]. Deletions of
control [42]. Strobel et al. reported in a patient CFHR3-CFHR1, unlike in (DEAP-) aHUS
with DDD an antibody binding to factors B and patients, have not been associated with CFH
Bb, which decreased terminal pathway activation antibodies in patients with C3GN [42]. Most CFI
by inhibiting the C5 convertase [43]. Factor H mutations associated with MPGN were
antibodies, frequently associated with aHUS already reported in patients with aHUS, and the
[44], are reported but rare in MPGN. A CFH functional consequences in fluid phase are not
mini-autoantibody (i.e., antibody fragment-like λ clear [19].
light-chain dimer) was reported to block C3b In 2009, Gale et al. described in 26 of 84 people
binding of CFH thus abolishing CFH regulatory of Cypriotic ancestry with unexplained renal dis-
capacity [45, 46]. ease, a mutation in CFHR5 comprising a duplica-
tion in the dimerization domain of CFHR5
[51]. Several more mutations and hybrid variants
in CFHR genes have been detected and are asso-
Genetic Forms of C3G ciated with C3G (Table 3). The role of CFHRs in
C3G was unclear until Goicoechea de Jorge
In one series of 134 patients in which the CFH, et al. reported that CFHR1, CFHR2, and CFHR5
CFI, and CD46 genes were screened, mutations form homo- and heterodimers among themselves
were detected in 8/48 (16.6 %), 5/29 (17.2 %), and [21]. In the dimer form, these proteins are able to
11/56 (19.6 %) patients with MPGN 1, C3G, and compete with CFH for C3b binding and protect
DDD, respectively [19]. Note that this C3b from inactivation and the AP C3 convertase
indicated that in the majority of patients with from its decay. This process, termed deregulation,
33 Membranoproliferative and C3-Mediated GN in Children 1043

Table 3 Overview of mutations, rare variants, common normal variants (CNV), and polymorphism associated with C3G
Gene/ Pheno-
protein Mutation/variant Function type References
CFH Mutations • Intact surface binding DDD/ Levy et al. [57], Vogt et al. [54], Ault
• Homo-/compound • Reduced C3b binding C3G et al. [48], Dragon-Durey et al. [50],
heterozygous • Loss of CFH cofactor and Licht et al. [55], Habbig et al. [52]
• SCRs 1–4 decay-accelerating
(regulatory activity
domain)
• Cys residues
(tertiary structure)
CFH Polymorphisms • Impaired C3b/heparin DDD Hageman et al. [53], Abrera-Abeleda
• Y402H (SCR 7) binding et al. [47], Abrera-Abeleda et al. [58]
• Impaired CFH cofactor
activity
CFI Type I and II • Decreased activity on cell DDD/ Servais et al. [19]
surface C3GN
MCP/ Rare SNP • Defective control on cell C3GN Servais et al. [19]
CD46 surface
CFHR1 Internal duplication • Alters FHR DDD Tortajada et al. [22]
oligomerization
• Greater degree of CFHR-
mediated deregulation
CFHR3-1 CNV • Greater degree of CFHR- C3G Malik et al. [61], Goicoechea de Jorge
• CFHR3-1 hybrid mediated deregulation et al. [21]
gene
CFHR5 CNV • Greater degree of CFHR- C3G Gale et al. [51], Goicoechea de Jorge
• Duplication in mediated deregulation et al. [21]
CFHR5 exons 2/3
CFHR5 Polymorphisms • Not tested DDD Abrera-Abeleda et al. [47], Abrera-
Abeleda et al. [58]
CFHR2-5 CFHR2-CFHR5 • Stabilizes C3 convertase, DDD Chen et al. [49]
hybrid gene reduced CFH-mediated
decay
C3 Mutations • C3mut – resistant to DDD Martinez-Barricarte et al. [56]
• Heterozygous cleavage by C3bBb
deletion • 3mut convertase – resistant
to CFH inactivation,
normal MCP/CD46
regulation
C3 Polymorphisms • Not tested DDD Smith et al. [59], Abrera-Abeleda
et al. [47]

was reported to be increased in patients with a and adult) [63, 64]. Once thought to be primarily
CFHR1-3 hybrid genes or CFHR5 mutations pediatric diseases, it is clear that they also com-
[21, 51, 61]. monly present in the adult population [19].
The recent French series noted that only 39 % of
patients were less than 16 years at the time of
Clinical Manifestation diagnosis, with the mean age at diagnosis for
MPGN, DDD, and C3GN reported to be 20.7
MGPN, C3GN, and DDD are rare diseases with years  16.8, 18.9 years  17.7, and 30.3 years
an individual annual incidence estimated at 1–2  19.3, respectively [19]. Estimates range from
per million per total population (both pediatric 43 % to 58 % of patients with DDD and 25–54 %
1044 C. Licht et al.

of patients with C3GN being under the age of versus C3GN is limited by the small number of
16 at diagnosis. Within the pediatric population, case series that have been reported. Doubtless,
DDD tends to present at a younger age with 70 % there will be refinements regarding variations in
of patients diagnosed prior to age 15 [65]. With presentation; however, it is unlikely that there will
respect to gender, there may be a slight bias with be unique features that clearly distinguish MPGN,
males predominating in MPGN, DDD, and C3GN DDD, and C3GN clinically. The exception to this
[19, 63]. As the large case series that discriminate appears to CFHR5-associated C3GN which pre-
MPGN, DDD, and C3GN are predominately from sents in childhood with persistent microscopic
Northern European populations, it is not clear at hematuria, synpharyngitic gross hematuria, and a
present if there are ethnic or racial differences in strong family history of ESRD. At present, this
disease frequency. form of G3GN has been reported primarily in the
The clinical presentations of MPGN, DDD, Cypriot population but also in two patients with
and C3GN are nonspecific and require a high no cypriotic heritage [51, 68–70].
index of suspicion. There is significant overlap Historically hypocomplementemia has been
with regard to clinical features at presentation. regarding as a distinguishing feature of MPGN I
Depending on the case series, nephrotic syndrome (low C3 and low C4) and other types of MPGN
has been reported as the presenting feature in (low C3 and normal C4) [71]. In the current clas-
40–65 % of patients with MPGN, 30–40 % with sification system, MPGN is regarded as a form of
DDD, and 25–40 % with C3GN. Regardless of immune complex disease, and depressed C4
disease type, microscopic hematuria has been levels are commonly held as distinguishing fea-
reported in 40–75 % of patients, gross hematuria ture; C3GN is thought primarily to be due to
in 10–20 %, and non-nephrotic proteinuria in dysregulation of the complement alternative path-
30–40 %. Presentations with acute glomerulone- way, and therefore C4 levels are presumed to be
phritis are common in MPGN, DDD, and C3GN, normal. Recent retrospective case series compar-
with hypertension noted in 30–60 % of patients ing complement levels in patients with MGPN,
and renal insufficiency in 20–50 % of patients [19, DDD, and C3GN generally confirm the utility of
63, 66, 67]. Although rare, rapidly progressive C3 and C4 levels in distinguishing these disease
glomerulonephritis has been reported in MPGN, types [19, 63]. There were, however, important
DDD, and C3GN (Table 4). caveats noted in these studies. In the French series
As the classification system for MPGN has (115 patients), at diagnosis, only 46 % of patient
recently evolved, our understanding of the differ- with MPGN had C3 levels less than 66 mg/dl and
ences in the clinical presentations of MPGN only 2.4 % had C4 levels less than 9.3 mg/dl; low
C3 levels were noted in 59 % of DDD patients and
39.6 % of C3GN patients, with 1 DDD patient
Table 4 Clinical manifestation and outcome of DDD
and C3G
noted to have a low C4 level and none of the
C3GN patients [19]. The series from England
DDD C3G
(80 patients) reported low C3 levels, <65 mg/dl,
Pediatric onset (<16 years) 43–58 % 25–54 %
in 79 % of DDD and 48 % of C3GN patients, with
Mean age at onset (years) 19  18 30  19
suppressed C4 levels, <16 mg/dl, in 15 % of
Clinical presentation
DDD and 36 % of C3GN patients [63]. It appears
Nephrotic syndrome 38–43 % 27–44 %
Microhematuria 76 % 65 %
based on these and other reports that while assess-
Arterial hypertension 21–60 % 40 % ment of C3 and C4 levels is suggestive of a diag-
Impaired renal function (>1.5 29 % 50 % nosis of MPGN, DDD, or C3GN, suppressed C3
mg/dL creatinine) or C4 levels are neither sufficient nor essential in
Low C3 (<75 mg/dL) 59–79 % 40–48 % the presentation of MPGN, DDD, or C3GN
Long-term outcome [19, 63]. It is important to recognize that of the
Duration to ESRD (years) 10  11 11  10 combined patients reported in these case series
Adapted from Refs. [19, 63] with biopsy proven MPGN, DDD, or C3GN,
33 Membranoproliferative and C3-Mediated GN in Children 1045

43 % had normal C3 levels at the time of diagnosis Differential Diagnosis


[19, 63]. Furthermore, the patients with CFHR5
associated C3GN are also reported to have normal In the setting of hypocomplementemia, low C3
serum C3 concentrations [72]. levels, the differential diagnosis is narrow, and
Extrarenal manifestations are uncommon depressed C4 levels can refine the differential
in C3G. These include acquired partial further. In the pediatric population, low C3 and
lipodystrophy (aPL) and ocular C3 deposits, sim- C4 levels are seen primarily in patients with either
ilar to soft drusen seen in age-related macular MPGN or SLE and in some patients with PIGN.
degeneration (AMD) [53]. Interestingly, polymor- A throughout history and physical examination
phisms in complement genes are also associated with attention to extrarenal disease manifestations
with the risk of developing AMD [73]. These such as arthralgias, rashes, and neurologic, hepatic,
drusen consist of C3 split products and CFH lung, or cardiac involvement combined with labo-
[74, 75]. The long-term risk for visual problems is ratory evidence of immune dysregulation including
about 10 % [76]. aPL usually precedes the diagno- antibody-mediated hemolysis, leukopenia,
sis of MPGN and may present with low C3 levels coagulopathy, myositis, or hepatitis strongly sup-
and C3NeF. It is presumably caused by comple- port a diagnosis of SLE as opposed to MPGN.
ment dysregulation on adipocytes [77, 78]. Although exceedingly rare in the pediatric popula-
tion, essential cryoglobulinemia may also present
with multi-organ involvement and systemic find-
Diagnosis ings. Of note, secondary forms of MPGN may
present with low C3 and C4 levels [79]. The dis-
Physicians must maintain a high index of suspi- eases presenting with low C3 and normal C4 levels
cion when considering a diagnosis of MPGN, have traditionally been limited to PIGN, the nephri-
DDD, or C3GN. The clinical presentations of tis of chronic bacteremia, shunt nephritis, and
MPGN, DDD, and C3GN are variable, while DDD. The presence of persistent fevers or V-A
most commonly seen in the setting of acute shunts should alert the physician to the possibility
nephritis or nephrotic syndrome, more subtle pre- of the nephritis of chronic bacteremia or shunt
sentations are also seen, and there is considerable nephritis. The finding of isolated low C3 levels
clinical overlap with the presentations of many with clinical features of acute GN presents a par-
other forms of renal disease. Analysis of the com- ticular challenge to the clinician. Historically, the
plement system can be useful particularly C3 and history of a recent episode of pharyngitis, elevated
C4 levels. However, as noted above, normal com- ASO or anti-DNase titers, and low C3 levels has
plement levels do not exclude the diagnosis of defined the clinical features of postinfectious
MPGN, DDD, or C3GN [19]. The measurement GN. DDD or C3GN is considered only if the clin-
of nephritic factor is also supportive of a diagnosis ical course is atypical or if the C3 level does not
of C3GN or DDD; however, these assays are return to normal within 8–10 weeks of presentation
neither standardized, widely available, nor have [12]. The report by Medjeral-Thomas et al. noted
diagnostic specificity. Genetic testing of comple- elevated ASO titers in 43 % of patients with DDD
ment components may provide insight into under- and 57 % of patients with C3GN; further compli-
lying pathogenic mechanisms; however, cating matters was the finding that of the
mutations in the complement system that are spe- DDD/C3GN patients with elevated ASO titers,
cific from a diagnostic standpoint have yet to be 21 % had a documented antecedent upper respira-
identified. The exception to this is the CFHR5 tory tract infection [63]. Normalization of C3 levels
mutation seen in a specific subset of patients has been used to differentiate PIGN from other
with C3GN [51]. Ultimately, a diagnosis of forms of nephritis; however, current data suggests
MPGN, DDD, or C3GN is made by renal biopsy that a significant percentage of patients with C3GN
in conjunction with complement diagnostics and DDD have normal C3 levels at the time of
(Table 2). diagnosis [19]; it is not clear what percentage of
1046 C. Licht et al.

the C3GN patients with normal C3 levels at the may only be a partial response [80, 81]. Predni-
time of diagnosis had previously been sone is considered the first-line agent in patients
hypocomplementemic [63]. As the vast majority with Ig-/IC-mediated GN who present with
of patients with presumed PIGN are never nephrotic-range proteinuria with or without renal
biopsied, the clinician should have a low threshold failure. A reasonable approach would be
for biopsy if the patient has a clinical course that is 60 mg/m2 [2] (maximal 60 mg/d)  4 weeks
atypical for PIGN or there is a family history of (induction) followed by 40 mg/m2 [2] on
unexplained ESRD. [12] Because MPGN, DDD, alternate-day for 6–12 (24) months (mainte-
and C3GN may also present with normal comple- nance). No beneficial effect was shown in patients
ment levels, they should also be considered in any with DDD.
differential diagnosis for glomerular disease even if Mycophenolate mofetil (MMF) was adminis-
the patient’s C3 and C4 levels are normal. Until it is tered alone or in combination with prednisone in
clear what the frequency of CHFR5 mutations are idiopathic MPGN and generated encouraging
in the population outside of Cyprus, C3GN should results. In 13 patients with steroid-resistant pri-
be considered in the differential diagnosis of mary MPGN, the addition of MMF resulted in
patients with a family history of ESRD, persistent sustained improvement (1-year follow-up) of pro-
microscopic hematuria, and synpharyngitic gross teinuria and renal function [82]. In 51 patients
hematuria (Table 1). with primary GN, including 15 patients with
MPGN, a partial or complete remission was
reported to be 70 % after 1 year [83]. In a small
Treatment case series of nine children with MPGN I, treat-
ment with MMF and prednisone for a mean time
No treatment standard for patients with MPGN or of 40 months resulted in complete or partial
C3G exists, and current treatment recommenda- response in 5. However, all patients with low C3
tions are mainly based on small-size single-center levels were treatment failure [84]. No reports on
studies and expert opinions. the effectiveness of MMF in patients with DDD
are available.
Calcineurin inhibitors (e.g., cyclosporine A
Supportive Therapy and tacrolimus) were used in prednisone-resistant
MPGN patients. One study demonstrated its effi-
Angiotensin-converting-enzyme inhibitors (ACEi) cacy in a small trial including 18 patients with
or angiotensin II receptor antagonists (ARB) are refractory MPGN and additional low-dose pred-
used in many patients with MPGN due to their anti- nisone therapy, by inducing long-term (2 years)
proteinuric and antihypertensive effect. The use of reduction of proteinuria and stable renal function
ACEi or ARB was associated with better renal in 94 %. Only one patient showed a recurrence
survival in MPGN patients [19]. The treatment of after treatment discontinuation [85]. In 11 adult
arterial hypertension and CKD/ESRD follows patients with steroid resistant, in five also
established standards. cyclophosphamide-resistant MPGN, tacrolimus
plus prednisone in six resulted in partial/complete
remission in nine [86, 87]. In two children with
Immunosuppressive Therapy MPGN and suboptimal response to long-term
prednisone, a rapid and complete remission was
In children, there are multiple studies reporting on achieved with tacrolimus [88]. In two patients
the use of steroids in MPGN. A beneficial effect of with DDD, low-dose prednisone and cyclosporine
steroids for proteinuria and long-term renal func- A was able to induce remission [89, 90]. On the
tion was detected by using long-term low-dose other hand, Appel et al. were not able to see a
prednisone. However, only a subgroup of patients benefit of calcineurin inhibitors in the treatment of
formerly classified as MPGN I will respond, and it DDD [76].
33 Membranoproliferative and C3-Mediated GN in Children 1047

The detection of C3NeF has prompted the use progression and stabilized kidney function
of B-cell depleting agents like rituximab, a chi- [52, 55, 100]. However, plasma infusion might
meric monoclonal CD20 antibody. Several case not be enough as induction therapy [111].
reports in patients with MPGN I, especially with In 2012, several reports of the successful use of
IC-mediated disease, indicate partial or complete eculizumab in patients with MPGN were
remission after administration of rituximab published [97, 100, 112, 113]. Eculizumab is a
(in half of the cases in addition to steroids) in monoclonal antibody binding C5 and therefore
11 of 13 patients [91–95]. Despite a decrease in preventing the assembly of the terminal comple-
C3NeF levels, two patients with DDD did not ment complex (TCC, C5b-9) [114]. This complex
show any change in proteinuria or renal function is integrated in cell membranes and can lead to
[96, 97]. Of note, both patients were rescued by cell lysis. Its role in the pathogenesis in C3G is not
eculizumab therapy (below). known. However, elevated soluble C5b-9 levels
were detected in patients with C3G, and in mice
deficient in C5, anti-murine C5 antibody amelio-
Complement-Targeting Therapy rated the disease [115].
At present, 13 patients with different forms of
With the emerging role of complement in C3G C3G treated with eculizumab have been reported
pathogenesis, complement-targeting treatment in the literature. Six are part of a small clinical
strategies are increasingly considered for patients trial, while the others are single case reports
with C3G. Several case reports exist on the use of [96, 97, 100, 112, 113, 116–118]. A significant
plasma exchange (PE) in patients with MPGN I response was reported in 10, a partial response in
and DDD for disease manifestation and recur- one and no response in two of these patients, as
rence in native and graft kidneys. A partial or reviewed by Vivarelli et al. [119]. These patients
complete remission was observed in 17 out of include cases of C3G and DDD, in native and
21 patients (81 %) reported in the literature transplanted kidney, and both cases with C3NeF
[93, 98–109]. In one patient with Ig-mediated and genetic mutations. Although numbers are
GN (MPGN I), despite a transient improvement small, a better outcome seems to be associated
of renal function and proteinuria, her clinical con- with elevated SC5b-9 levels and shorter disease
dition worsened including seizures, respiratory duration [119]. Of note, treatment with
distress, and sustained anuria. Due to a CFHR1 eculizumab in C3G is not always successful
deletion (without CFH antibodies), low C3 and [112] (personal unpublished observation), and
elevated SC5b-9, indicating alternative and termi- criteria clearly identifying individuals with the
nal complement pathway activation, and potential to respond to eculizumab treatment are
TMA-related symptoms, the patient was switched currently lacking.
from PE to eculizumab. This resulted in a dra-
matic improvement of her clinical condition, pro-
teinuria and renal function [100]. One case report Clinical Outcomes
also shows the successful treatment of MPGN
recurrence during pregnancy and enabled the Long-term renal outcome data with the new clas-
delivery of a healthy child [110]. sification system is limited for C3GN and DDD.
Three case reports, including two siblings with Sethi, in a small case series of 12 of primarily
DDD on the background of a CFH mutation in adult C3GN patients, reported stable short-term
SCR4 and one patient with MPGN I and renal function with a mean follow-up of 26.4
MCP/CD46 mutation, were treated for >3 years months [67]. Medjeral-Thomas, comparing out-
with chronic infusion of fresh frozen plasma comes of DDD and C3GN in a cohort of
(FFP). FFP as maintenance therapy, individual- 80 patients, reported progression to ESRD in
ized according to the patient needs (e.g., intercur- 47 % of DDD patients and 23 % of C3GN patients
rent infection), was able to prevent disease with a median follow-up of 28 months; there was
1048 C. Licht et al.

no difference in cumulative renal survival by has also been reported in the NAPRTCS database
Kaplan-Meier analysis [63]. The French cohort [123]. The etiology of this apparent increased risk
of 134 patients reported a 10-year renal survival in the pediatric population has not been identified.
rate of 63.5 % with no differences in renal survival As it is not possible to reclassify patients in the
between groups (MPGN, DDD, C3GN) existing databases, it is not clear how the new
[19]. These data are in agreement with previous classification system will impact the relative risk
reports indicating that progression to ESRD of disease recurrence and allograft survival. There
occurs in 40–50 % of patients with MPGN regard- are, however, a number of limited case series that
less of type within 10 years of diagnosis. Analysis confirm increased risk of disease recurrence for
by both the French and English groups suggested C3GN and the potential negative impact on allo-
that both eGFR and age at presentation were graft survival. Zand et al. reported outcomes on
strongly negatively correlated with long-term 21 patients with C3GN; 14 (66.7 %) had disease
renal survival [19, 63]. Medjeral-Thomas also recurrence with a median time to graft failure of
reported that the presence of glomerular crescents 77 months (6.4 years) [124]. Using the new clas-
or DDD by biopsy was a strong independent sification system, the French group reported recur-
predictor of disease progression [63] (Table 4). rence rates of 43 %, 55 %, and 60 % for MPGN,
While disease recurrence of MPGN and DDD DDD, and C3GN, respectively [19]. The impact on
in renal transplants has been well described, with allograft survival was not reported and the total
recurrence rates ranging from 10 % to 100 % number of transplants was small, 35 [19]. The
depending on the case series, the impact of disease English group reported data on 13 transplants,
recurrence on allograft survival has been contro- 6 with DDD and 7 with C3GN. All of the DDD
versial, as a number of small case series report patients had recurrence of disease and 50 % failed
accelerated rates of graft loss from disease recur- due to disease recurrence. Of the 7 C3GN patients,
rence, while larger studies suggest a more limited 57 % had disease recurrence; three of the four were
impact. Analysis of the UNOS database noted to have recurrent disease contributing to
(811 patients with MPGN I and 179 patients allograft failure [63]. Overall, the allograft survival
with DDD) reported a modest reduction in at 5 years was 69 % (95 % CI: 65–99 %). CFHR5-
10-year graft survival for patients with MPGN I, associated C3GN has also been reported to recur in
56.2 % versus 60 %, compared to the dataset as renal transplants [125].
whole with allograft failures from disease recur-
rence in 14.5 % patients [120]. The 10-year allo-
graft survival for DDD was not significantly Summary
different from the general population, 57.5 %,
despite almost 30 % of graft failures reported to Lessons learned from animals and humans have
be from disease recurrence; however, the authors helped to significantly advance our understanding
noted significantly worse allograft survival in of the pathogenesis of the disease spectrum
pediatric DDD patients compared to patients previously labeled as MPGN. While so far classi-
transplanted as adults, with age at transplantation fied based on morphology, today the identification
being positively associated with graft survival of a central role of the complement AP has
[120]. Data supporting a modest impact of allowed for a pathomechanism-based new classifi-
MPGN I disease recurrence has also been reported cation system introducing the term C3
from the ANZDATA registry [121]. A more recent glomerulopathy (C3G), which reflects the predom-
report from the ESPN/ERA-EDTA registry indi- inant finding of C3 in kidney biopsy specimens –
cated that there was significant increase risk of C3 glomerulonephritis (C3GN) or dense deposit
allograft loss, 32.4 %, at 5 years posttransplant disease (DDD) – or in combination with immuno-
in pediatric patients with MPGN [122]. Data globulins – secondary MPGN. Complement
suggesting that pediatric patients with DDD are dysregulation resulting in C3G seems – different
at greater risk of graft loss from disease recurrence from aHUS with uncontrolled complement
33 Membranoproliferative and C3-Mediated GN in Children 1049

activation on the vascular endothelium – to be 12. Sethi S, Fervenza FC, Zhang Y, et al. Atypical
characterized by the overshooting activation of postinfectious glomerulonephritis is associated with
abnormalities in the alternative pathway of comple-
C3 and generation of C3 split products. Identifica- ment. Kidney Int. 2013;83:293–9.
tion of a central role of complement dysregulation 13. Walport MJ. Complement. Second of two parts.
in C3G pathogenesis has for the first time identified N Engl J Med. 2001;344:1140–4.
a specific treatment strategy for C3G, a break- 14. Walport MJ. Complement. First of two parts. N Engl J
Med. 2001;344:1058–66.
through in the management of this disease spec- 15. Zipfel PF, Skerka C. Complement regulators and
trum historically characterized by its progressive inhibitory proteins. Nat Rev Immunol. 2009;9:
nature and poor outcomes. 729–40.
16. Riedl M, Fakhouri F, Lequintrec M. The spectrum of
complement-mediated thrombotic microangiopathies
Acknowledgments The authors wish to thank Dr. Paul – pathogenetic insights identifying novel treatment
Thorner, The Hospital for Sick Children and Department of approaches. Semin Thromb Hemost. 2014;
Laboratory Medicine and Pathobiology, University of 40(4):444–64.
Toronto, Toronto, ON, Canada for preparing Fig. 1 and 17. Noone D, Al-Matrafi J, Tinckam K, et al. Antibody
discussing pathohistological aspects of C3G. mediated rejection associated with complement
factor H-related protein 3/1 deficiency successfully
treated with Eculizumab. Am J Transplant. 2012;12:
2546–53.
References 18. Noris M, Remuzzi G. Overview of complement acti-
vation and regulation. Semin Nephrol. 2013;33:
1. Sethi S, Fervenza FC. Membranoproliferative 479–92.
glomerulonephritis–a new look at an old entity. 19. Servais A, Noel L-H, Roumenina LT, et al. Acquired
N Engl J Med. 2012;366:1119–31. and genetic complement abnormalities play a critical
2. Sethi S, Fervenza FC. Membranoproliferative glo- role in dense deposit disease and other C3
merulonephritis: pathogenetic heterogeneity and pro- glomerulopathies. Kidney Int. 2012;82:454–64.
posal for a new classification. Semin Nephrol. 20. Heinen S, Hartmann A, Lauer N, et al. Factor
2011;31:341–8. H-related protein 1 (CFHR-1) inhibits complement
3. Pickering MC, D’Agati VD, Nester CM, et al. C3 C5 convertase activity and terminal complex forma-
glomerulopathy: consensus report. Kidney Int. tion. Blood. 2009;114:2439–47.
2013;84:1079–89. 21. Goicoechea de Jorge E, Caesar JJ, Malik TH, et al.
4. Zand L, Fervenza FC, Nasr SH, Sethi S. Dimerization of complement factor H-related proteins
Membranoproliferative glomerulonephritis associ- modulates complement activation in vivo. Proc Natl
ated with autoimmune diseases. J Nephrol. 2014;27: Acad Sci U S A. 2013;110:4685–90.
165–71. 22. Tortajada A, Yebenes H, Abarrategui-Garrido C,
5. Zand L, Kattah A, Fervenza FC, et al. C3 glomerulo- et al. C3 glomerulopathy-associated CFHR1 mutation
nephritis associated with monoclonal gammopathy: a alters FHR oligomerization and complement regula-
case series. Am J Kidney Dis. 2013;62:506–14. tion. J Clin Invest. 2013;123:2434–46.
6. Rennke HG. Secondary membranoproliferative glo- 23. Hebecker M, Jozsi M. Factor H-related protein 4 acti-
merulonephritis. Kidney Int. 1995;47:643–56. vates complement by serving as a platform for the
7. Sethi S, Gamez JD, Vrana JA, et al. Glomeruli of assembly of alternative pathway C3 convertase via its
Dense Deposit Disease contain components of the interaction with C3b protein. J Biol Chem.
alternative and terminal complement pathway. Kid- 2012;287:19528–36.
ney Int. 2009;75:952–60. 24. Holers VM. Complement and its receptors: new
8. Sethi S, Vrana JA, Theis JD, Dogan A. Mass spec- insights into human disease. Annu Rev Immunol.
trometry based proteomics in the diagnosis of kidney 2014;32:433–59.
disease. Curr Opin Nephrol Hypertens. 2013;22: 25. Pickering MC, Cook HT, Warren J, et al.
273–80. Uncontrolled C3 activation causes membranoproli-
9. Fervenza FC, Smith RJH, Sethi S. Association of a ferative glomerulonephritis in mice deficient in com-
novel complement factor H mutation with severe plement factor H. Nat Genet. 2002;31:424–8.
crescentic and necrotizing glomerulonephritis. Am J 26. Jansen JH, Hogasen K, Harboe M, Hovig T. In situ
Kidney Dis. 2012;60:126–32. complement activation in porcine membranoproli-
10. Walker PD. Dense deposit disease: new insights. Curr ferative glomerulonephritis type II. Kidney Int.
Opin Nephrol Hypertens. 2007;16:204–12. 1998;53:331–49.
11. Cook HT, Pickering MC. Histopathology of MPGN 27. Hegasy GA, Manuelian T, Hogasen K, Jansen JH,
and C3 glomerulopathies. Nat Rev Nephrol. Zipfel PF. The molecular basis for hereditary porcine
2015;11:14–22. membranoproliferative glomerulonephritis type II:
1050 C. Licht et al.

point mutations in the factor H coding sequence block 41. Leroy V, Fremeaux-Bacchi V, Peuchmaur M, et al.
protein secretion. Am J Pathol. 2002;161:2027–34. Membranoproliferative glomerulonephritis with
28. Rose KL, Paixao-Cavalcante D, Fish J, et al. Factor I C3NeF and genetic complement dysregulation.
is required for the development of membranoproli- Pediatr Nephrol. 2011;26:419–24.
ferative glomerulonephritis in factor H-deficient 42. Chen Q, Muller D, Rudolph B, et al. Combined C3b
mice. J Clin Invest. 2008;118:608–18. and factor B autoantibodies and MPGN type II.
29. de Jorge EG, Macor P, Paixao-Cavalcante D, et al. N Engl J Med. 2011;365:2340–2.
The development of atypical hemolytic uremic syn- 43. Strobel S, Zimmering M, Papp K, Prechl J, Jozsi
drome depends on complement C5. J Am Soc M. Anti-factor B autoantibody in dense deposit dis-
Nephrol. 2011;22:137–45. ease. Mol Immunol. 2010;47:1476–83.
30. Pickering MC, de Jorge EG, Martinez-Barricarte R, 44. Hofer J, Janecke AR, Zimmerhackl LB, et al. Com-
et al. Spontaneous hemolytic uremic syndrome trig- plement factor H-related protein 1 deficiency and
gered by complement factor H lacking surface recog- factor H antibodies in pediatric patients with atypical
nition domains. J Exp Med. 2007;204:1249–56. hemolytic uremic syndrome. Clin J Am Soc Nephrol.
31. Spitzer RE, Stitzel AE. On the origin and control of 2013;8:407–15.
C3NeF. In Vivo. 1988;2:79–81. 45. Jokiranta TS, Solomon A, Pangburn MK, Zipfel PF,
32. Berthoux FC, Carpenter CB, Traeger J, Merrill JP. Meri S. Nephritogenic lambda light chain dimer: a
[C3 nephritic factor and heat labile complement unique human miniautoantibody against complement
inactivator in chronic hypocomplementemic factor H. J Immunol. 1999;163:4590–6.
mesangioproliferative glomerulonephritis, French]. 46. Meri S, Koistinen V, Miettinen A, Tornroth T,
Le Facteur Nephritique (C3 Nephritic Factor) Et Seppala IJ. Activation of the alternative pathway of
L’inactivateur Thermolabile Du Complement (Heat complement by monoclonal lambda light chains in
Labile Complement Inactivator) Dans Les membranoproliferative glomerulonephritis. J Exp
Glomerulonephrites Mesangioproliferatives Med. 1992;175:939–50.
Hypocomplementaires Chroniques. Actualites 47. Abrera-Abeleda MA, Nishimura C, Frees K, et al.
Nephrologiques de l’Hopital Necker. 1974;1974: Allelic variants of complement genes associated
141–56. with dense deposit disease. J Am Soc Nephrol.
33. Nicolas C, Vuiblet V, Baudouin V, et al. C3 nephritic 2011;22:1551–9.
factor associated with C3 glomerulopathy in children. 48. Ault BH, Schmidt BZ, Fowler NL, et al. Human fac-
Pediatr Nephrol. 2014;29:85–94. tor H deficiency. Mutations in framework cysteine
34. Fremeaux-Bacchi V, Weiss L, Brun P, Kazatchkine residues and block in H protein secretion and intra-
MD. Selective disappearance of C3NeF IgG autoan- cellular catabolism. J Biol Chem. 1997;272:
tibody in the plasma of a patient with membranopro- 25168–75.
liferative glomerulonephritis following renal 49. Chen Q, Wiesener M, Eberhardt HU, et al. Comple-
transplantation. Nephrol Dial Transplant. 1994;9: ment factor H-related hybrid protein deregulates com-
811–4. plement in dense deposit disease. J Clin Invest.
35. Paixao-Cavalcante D, Lopez-Trascasa M, Skattum L, 2014;124:145–55.
et al. Sensitive and specific assays for C3 nephritic 50. Dragon-Durey MA, Fremeaux-Bacchi V, Loirat C,
factors clarify mechanisms underlying complement et al. Heterozygous and homozygous factor h defi-
dysregulation. Kidney Int. 2012;82:1084–92. ciencies associated with hemolytic uremic syndrome
36. Zhang Y, Meyer NC, Wang K, et al. Causes of or membranoproliferative glomerulonephritis: report
alternative pathway dysregulation in dense and genetic analysis of 16 cases. J Am Soc Nephrol.
deposit disease. Clin J Am Soc Nephrol. 2012;7: 2004;15:787–95.
265–74. 51. Gale DP, de Jorge EG, Cook HT, et al. Identification
37. Waldo FB, Forristal J, Beischel L, West CD. A circu- of a mutation in complement factor H-related protein
lating inhibitor of fluid-phase amplification. C3 5 in patients of Cypriot origin with glomerulonephri-
convertase formation in systemic lupus tis. Lancet. 2010;376:794–801.
erythematosus. J Clin Invest. 1985;75:1786–95. 52. Habbig S, Mihatsch MJ, Heinen S, et al. C3 deposi-
38. Arroyave CM, Wilson MR, Tan EM. Serum factors tion glomerulopathy due to a functional factor H
activating the alternative complement pathway in defect. Kidney Int. 2009;75:1230–4.
autoimmune disease: description of two different fac- 53. Hageman GS, Anderson DH, Johnson LV, et al.
tors from patients with systemic lupus erythematosus. A common haplotype in the complement regulatory
J Immunol. 1976;116:821–6. gene factor H (HF1/CFH) predisposes individuals to
39. Wilson MR, Arroyave CM, Nakamura RM, age-related macular degeneration. Proc Natl Acad Sci
Vaughan JH, Tan EM. Activation of the alternative U S A. 2005;102:7227–32.
complement pathway in systemic lupus 54. Vogt BA, Wyatt RJ, Burke BA, Simonton SC,
erythematosus. Clin Exp Immunol. 1976;26:11–20. Kashtan CE. Inherited factor H deficiency and colla-
40. Lewis LA, Ram S. Meningococcal disease and the gen type III glomerulopathy. Pediatr Nephrol.
complement system. Virulence. 2014;5:98–126. 1995;9:11–5.
33 Membranoproliferative and C3-Mediated GN in Children 1051

55. Licht C, Heinen S, Jozsi M, et al. Deletion of Lys224 mutations: clinical characteristics of 91 patients in
in regulatory domain 4 of Factor H reveals a 16 pedigrees. Clin J Am Soc Nephrol. 2011;6:
novel pathomechanism for dense deposit disease 1436–46.
(MPGN II). Kidney Int. 2006;70:42–50. 69. Besbas N, Gulhan B, Gucer S, Korkmaz E, Ozaltin F.
56. Martinez-Barricarte R, Heurich M, Valdes-Canedo F, A novel CFHR5 mutation associated with C3 glomer-
et al. Human C3 mutation reveals a mechanism of ulonephritis in a Turkish girl. J Nephrol. 2014;27:
dense deposit disease pathogenesis and provides 457–60.
insights into complement activation and regulation. 70. Medjeral-Thomas N, Malik TH, Patel MP, et al. A
J Clin Invest. 2010;120:3702–12. novel CFHR5 fusion protein causes C3
57. Levy M, Halbwachs-Mecarelli L, Gubler MC. H defi- glomerulopathy in a family without Cypriot ancestry.
ciency in two brothers with atypical dense Kidney Int. 2014;85:933–7.
intramembranous deposit disease. Kidney Int. 71. Jackson EC, McAdams AJ, Strife CF. Differences
1986;30:949–56. between membranoproliferative glomerulonephritis
58. Abrera-Abeleda MA, Nishimura C, Smith JLH, et al. types I and III in clinical presentation, glomerular
Variations in the complement regulatory genes factor morphology, and complement perturbation. Am J
H (CFH) and factor H related 5 (CFHR5) are associ- Kidney Dis. 1987;9:115–20.
ated with membranoproliferative glomerulonephritis 72. Deltas C, Gale D, Cook T, Voskarides K,
type II (dense deposit disease). J Med Genet. Athanasiou Y, Pierides A. C3 glomerulonephritis/
2006;43:582–9. CFHR5 nephropathy is an endemic disease in Cyprus:
59. Smith RJH, Alexander J, Barlow PN, et al. New clinical and molecular findings in 21 families. Adv
approaches to the treatment of dense deposit disease. Exp Med Biol. 2013;735:189–96.
J Am Soc Nephrol. 2007;18:2447–56. 73. Zhan X, Larson DE, Wang C, et al. Identification of a
60. Xiao X, Pickering MC, Smith RJ. C3 rare coding variant in complement 3 associated with
glomerulopathy: the genetic and clinical findings in age-related macular degeneration. Nat Genet.
dense deposit disease and c3 glomerulonephritis. 2013;45:1375–81.
Semin Thromb Hemost. 2014;40:465–71. 74. Mullins RF, Aptsiauri N, Hageman GS. Structure and
61. Malik TH, Lavin PJ, De Jorge EG, et al. A hybrid composition of drusen associated with glomerulone-
CFHR3-1 gene causes familial C3 glomerulopathy. phritis: implications for the role of complement acti-
J Am Soc Nephrol. 2012;23:1155–60. vation in drusen biogenesis. Eye (Lond). 2001;15:
62. Fremeaux-Bacchi V, Fakhouri F, Garnier A, et al. 390–5.
Genetics and outcome of atypical hemolytic uremic 75. Mullins RF, Russell SR, Anderson DH, Hageman GS.
syndrome: a nationwide french series comparing chil- Drusen associated with aging and age-related macular
dren and adults. Clin J Am Soc Nephrol. 2013;8: degeneration contain proteins common to extracellu-
554–62. lar deposits associated with atherosclerosis, elastosis,
63. Medjeral-Thomas NR, O’Shaughnessy MM, amyloidosis, and dense deposit disease. FASEB
O’Regan JA, et al. C3 glomerulopathy: clinicopatho- J. 2000;14:835–46.
logic features and predictors of outcome. Clin J Am 76. Appel GB, Cook HT, Hageman G, et al. Membrano-
Soc Nephrol. 2014;9:46–53. proliferative glomerulonephritis type II (dense
64. Coppo R, Gianoglio B, Porcellini MG, Maringhini S. deposit disease): an update. J Am Soc Nephrol.
Frequency of renal diseases and clinical indications 2005;16:1392–403.
for renal biopsy in children (report of the Italian 77. Mathieson PW, Wurzner R, Oliveria DB,
National Registry of Renal Biopsies in Children). Lachmann PJ, Peters DK. Complement-mediated adi-
Group of Renal Immunopathology of the Italian Soci- pocyte lysis by nephritic factor sera. J Exp Med.
ety of Pediatric Nephrology and Group of Renal 1993;177:1827–31.
Immunopathology of the Italian Society of Nephrol- 78. Misra A, Peethambaram A, Garg A. Clinical features
ogy. Nephrol Dial Transplant. 1998;13:293–7. and metabolic and autoimmune derangements in
65. Lu D-F, Moon M, Lanning LD, McCarthy AM, acquired partial lipodystrophy: report of 35 cases
Smith RJH. Clinical features and outcomes of 98 chil- and review of the literature. Medicine (Baltimore).
dren and adults with dense deposit disease. Pediatr 2004;83:18–34.
Nephrol. 2012;27:773–81. 79. Alchi B, Jayne D. Membranoproliferative
66. Nasr SH, Valeri AM, Appel GB, et al. Dense deposit glomerulonephritis. Pediatr Nephrol. 2010;25:
disease: clinicopathologic study of 32 pediatric and 1409–18.
adult patients. Clin J Am Soc Nephrol. 2009;4:22–32. 80. Braun MC, West CD, Strife CF. Differences between
67. Sethi S, Fervenza FC, Zhang Y, et al. C3 glomerulo- membranoproliferative glomerulonephritis types I
nephritis: clinicopathological findings, complement and III in long-term response to an alternate-day pred-
abnormalities, glomerular proteomic profile, treat- nisone regimen. Am J Kidney Dis. 1999;34:1022–32.
ment, and follow-up. Kidney Int. 2012;82:465–73. 81. McEnery PT, McAdams AJ, West CD. The effect
68. Athanasiou Y, Voskarides K, Gale DP, et al. Familial of prednisone in a high-dose, alternate-day
C3 glomerulopathy associated with CFHR5 regimen on the natural history of idiopathic
1052 C. Licht et al.

membranoproliferative glomerulonephritis. Medi- 96. McCaughan JA, O’Rourke DM, Courtney


cine. 1985;64:401–24. AE. Recurrent dense deposit disease after renal trans-
82. Yuan M, Zou J, Zhang X, et al. Combination therapy plantation: an emerging role for complementary ther-
with mycophenolate mofetil and prednisone in apies. Am J Transplant. 2012;12:1046–51.
steroid-resistant idiopathic membranoproliferative 97. Daina E, Noris M, Remuzzi G. Eculizumab in a
glomerulonephritis. Clin Nephrol. 2010;73:354–9. patient with dense-deposit disease. N Engl J Med.
83. Dimkovic N, Jovanovic D, Kovacevic Z, et al. 2012;366:1161–3. [Erratum appears in N Engl J
Mycophenolate mofetil in high-risk patients with Med. 2012 Apr 12;366(15):1454].
primary glomerulonephritis: results of a 1-year pro- 98. Nord AT, Nord BL, Schmidt AE, Smith DS. Manage-
spective study. Nephron Clin Pract. 2009;111: ment of dense deposit disease with plasmapheresis
c189–96. and eculizumab. J Clin Apher. 2014;29(1):28–9.
84. Mazo A, Margieva T, Vashurina T, Zrobok O, 99. McGinley E, Watkins R, McLay A, Boulton-Jones
Tsygin A. The treatment of membranoproliferative JM. Plasma exchange in the treatment of mesangio-
glomerulonephritis in children with mycophenolate capillary glomerulonephritis. Nephron. 1985;40:
mofetil. Pediatr Nephrol. 2013;28(8):1607–8. 385–90.
85. Bagheri N, Nemati E, Rahbar K, Nobakht A, 100. Radhakrishnan S, Lunn A, Kirschfink M, et al.
Einollahi B, Taheri S. Cyclosporine in the treatment Eculizumab and refractory membranoproliferative
of membranoproliferative glomerulonephritis. Arch glomerulonephritis. N Engl J Med. 2012;366:
Iran Med. 2008;11:26–9. 1165–6.
86. Fan L, Liu Q, Liao Y, et al. Tacrolimus is an alterna- 101. Oberkircher OR, Enama M, West JC, Campbell P,
tive therapy option for the treatment of adult steroid- Moran J. Regression of recurrent membranoproli-
resistant nephrotic syndrome: a prospective, multi- ferative glomerulonephritis type II in a transplanted
center clinical trial. Int Urol Nephrol. 2013;45: kidney after plasmapheresis therapy. Transplant Proc.
459–68. 1988;20:418–23.
87. Li X, Li H, Ye H, et al. Tacrolimus therapy in adults 102. Kurtz KA, Schlueter AJ. Management of membrano-
with steroid- and cyclophosphamide-resistant proliferative glomerulonephritis type II with plasma-
nephrotic syndrome and normal or mildly reduced pheresis. J Clin Apher. 2002;17:135–7.
GFR. Am J Kidney Dis. 2009;54:51–8. 103. Banks RA, May S, Wallington T. Acute renal failure
88. Haddad M, Lau K, Butani L. Remission of in dense deposit disease: recovery after plasmaphere-
membranoproliferative glomerulonephritis type I sis. Br Med J (Clin Res Ed). 1982;284:1874–5.
with the use of tacrolimus. Pediatr Nephrol. 104. Masutani K, Katafuchi R, Ikeda H, et al. Recurrent
2007;22:1787–91. nephrotic syndrome after living-related renal
89. Kiyomasu T, Shibata M, Kurosu H, et al. Cyclosporin transplantation resistant to plasma exchange: report
A treatment for membranoproliferative glomerulone- of two cases. Clin Transplant. 2005;19 Suppl
phritis type II. Nephron. 2002;91:509–11. 14:59–64.
90. Hiramatsu M. Cyclosporin a treatment for dense 105. Montoliu J, Bergada E, Botey A, et al. Plasmapheresis
deposit disease with steroid resistant nephrotic syn- induced recovery from renal failure in mesangio-
drome. Pediatr Nephrol. 2010;25(9):1804. capillary glomerulonephritis of acute onset. Proc Eur
91. Dillon JJ, Hladunewich M, Haley WE, Reich HN, Dial Transplant Assoc. 1983;19:794–9.
Cattran DC, Fervenza FC. Rituximab therapy for 106. Muczynski KA. Plasmapheresis maintained renal
type I membranoproliferative glomerulonephritis. function in an allograft with recurrent membranopro-
Clin Nephrol. 2012;77:290–5. liferative glomerulonephritis type I. Am J Nephrol.
92. Kong WY, Swaminathan R, Irish A. Our experience 1995;15:446–9.
with rituximab therapy for adult-onset primary glo- 107. Pipeleers L, Sennesael J, Massart A, et al. Successful
merulonephritis and review of literature. Int Urol use of plasma exchange to prevent recurrence of C3
Nephrol. 2013;45:795–802. glomerulonephritis after kidney transplantation:
93. Perez-Saez MJ, Toledo K, Navarro MD, A case report. Transplantation. 2012;94:1050.
et al. Recurrent membranoproliferative glomerulone- 108. Saxena R, Frankel WL, Sedmak DD, Falkenhain ME,
phritis after second renal graft treated with plasma- Cosio FG. Recurrent type I membranoproliferative
pheresis and rituximab. Transplant Proc. 2011;43: glomerulonephritis in a renal allograft: successful
4005–9. treatment with plasmapheresis. Am J Kidney Dis.
94. Sugiura H, Takei T, Itabashi M, et al. Effect of single- 2000;35:749–52.
dose rituximab on primary glomerular diseases. 109. Yadav P, Ognjanovic M, Coulthard M, Moghal N,
Nephron Clin Pract. 2011;117:c98–105. Lambert H, Tse Y. Recurrent (MPGN) membranopro-
95. Guiard E, Karras A, Plaisier E, et al. Patterns of liferative glomerulonephritis type 1 successfully
noncryoglobulinemic glomerulonephritis with mono- treated with plasma exchange (PE). Pediatr Nephrol.
clonal Ig deposits: correlation with IgG subclass and 2011;26(9):1665–6.
response to rituximab. Clin J Am Soc Nephrol. 110. Morton MR, Bannister KM. Renal failure due
2011;6:1609–16. to mesangiocapillary glomerulonephritis in
33 Membranoproliferative and C3-Mediated GN in Children 1053

pregnancy: use of plasma exchange therapy. Clin eculizumab restores renal function in C3nef-related
Nephrol. 1993;40:74–8. DDD. Pediatr Nephrol. 2014;29:1107–11.
111. Roord JJ, van Diemen-van Steenvoorde RA, 119. Vivarelli M, Emma F. Treatment of C3G with com-
Schuurman HJ, et al. Membranoproliferative glomer- plement blockers. Semin Thromb Hemost. 2014;
ulonephritis in a patient with congenital deficiency of 40(4):472–7.
the third component of complement: effect of treat- 120. Angelo JR, Bell CS, Braun MC. Allograft failure in
ment with plasma. Am J Kidney Dis. 1989;13:413–7. kidney transplant recipients with membranoproli-
112. Bomback AS, Smith RJ, Barile GR, et al. Eculizumab ferative glomerulonephritis. Am J Kidney Dis.
for dense deposit disease and C3 glomerulonephritis. 2011;57:291–9.
Clin J Am Soc Nephrol. 2012;7:748–56. 121. Briganti EM, Russ GR, McNeil JJ, Atkins RC,
113. Vivarelli M, Pasini A, Emma F. Eculizumab for the Chadban SJ. Risk of renal allograft loss from recur-
treatment of dense-deposit disease. N Engl J Med. rent glomerulonephritis. N Engl J Med. 2002;347:
2012;366:1163–5. 103–9.
114. Rother RP, Rollins SA, Mojcik CF, Brodsky RA, 122. Van Stralen KJ, Verrina E, Belingheri M, et al.
Bell L. Discovery and development of the comple- Impact of graft loss among kidney diseases with a
ment inhibitor eculizumab for the treatment of parox- high risk of post-transplant recurrence in the paediat-
ysmal nocturnal hemoglobinuria. Nat Biotechnol. ric population. Nephrol Dial Transplant. 2013;28:
2007;25:1256–64. 1031–8.
115. Pickering MC, Warren J, Rose KL, et al. Prevention of 123. Braun MC, Stablein DM, Hamiwka LA, Bell L,
C5 activation ameliorates spontaneous and experi- Bartosh SM, Strife CF. Recurrence of membranopro-
mental glomerulonephritis in factor H-deficient liferative glomerulonephritis type II in renal allo-
mice. Proc Natl Acad Sci U S A. 2006;103:9649–54. grafts: the North American Pediatric Renal
116. Gurkan S, Fyfe B, Weiss L, Xiao X, Zhang Y, Smith Transplant Cooperative Study experience. J Am Soc
RJ. Eculizumab and recurrent C3 glomerulonephritis. Nephrol. 2005;16:2225–33.
Pediatr Nephrol. 2013;28:1975–81. 124. Zand L, Lorenz EC, Cosio FG, et al. Clinical findings,
117. Ozkaya O, Nalcacioglu H, Tekcan D, et al. pathology, and outcomes of C3GN after kidney trans-
Eculizumab therapy in a patient with dense-deposit plantation. J Am Soc Nephrol. 2014;25:1110–7.
disease associated with partial lipodystrophy. Pediatr 125. Vernon KA, Gale DP, De Jorge EG, et al. Recurrence
Nephrol. 2014;29:1283–7. of complement factor H-related protein 5 nephropathy
118. Rousset-Rouviere C, Cailliez M, Garaix F, Bruno D, in a renal transplant. Am J Transplant. 2011 Jan;
Laurent D, Tsimaratos M. Rituximab fails where 11(1):152–5. Doi 10.1111/j. 1600-6143. 2010. 03333.x.
Membranous Nephropathy in Children
34
Rudolph P. Valentini

Contents Other Secondary Causes of MN . . . . . . . . . . . . . . . . . . . . 1063


Background . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1056 Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1064
Treatment of Idiopathic Membranous
Incidence . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1056 Nephropathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1064
Histopathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1056 Corticosteroid Monotherapy . . . . . . . . . . . . . . . . . . . . . . . 1065
Idiopathic Membranous Nephropathy . . . . . . . . . . . . . 1056 Alkylating Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1066
Secondary Membranous Nephropathy . . . . . . . . . . . . . 1059 Alkylating Agents + Corticosteroids . . . . . . . . . . . . . . . 1066
Calcineurin Inhibitors + Corticosteroids . . . . . . . . . . . 1067
Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1060 Mycophenolate Mofetil (MMF) . . . . . . . . . . . . . . . . . . . . 1068
Rituximab (RTX) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1070
Pathogenesis of Idiopathic MN . . . . . . . . . . . . . . . . . . . 1060
ACTH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1070
Historical Animal Models . . . . . . . . . . . . . . . . . . . . . . . . . . 1060
M-Type Phospholipase A2 Receptor Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1071
Antibodies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1061
Neutral Endopeptidase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1061 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1071
Other Causes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1062
Pathogenesis of Secondary MN . . . . . . . . . . . . . . . . . . 1062
Hepatitis B Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1062
Systemic Lupus Erythematosus (SLE) . . . . . . . . . . . . . 1062

R.P. Valentini (*)


Pediatric Nephrology, Children’s Hospital of Michigan,
Detroit, MI, USA
Wayne State University School of Medicine, Detroit, MI,
USA
e-mail: rvalenti@dmc.org

# Springer-Verlag Berlin Heidelberg 2016 1055


E.D. Avner et al. (eds.), Pediatric Nephrology,
DOI 10.1007/978-3-662-43596-0_30
1056 R.P. Valentini

Background nephrotic syndrome, MN appears to increase


markedly. Moxey-Mims reported the incidence
Membranous nephropathy (MN), also known as of MN in the setting of nephrotic syndrome
membranous glomerulonephritis, is a pathologic increases from 1 % in children 1–12 years of age
condition associated with an immune complex to 22 % in children 13–19 years of age [14]. Sim-
deposition within the glomerulus resulting in glo- ilarly, an 18.5 % incidence of MN was reported
merular dysfunction. Clinical manifestations of from the Southwest Pediatric Nephrology Study
MN include proteinuria, hematuria, overt Group in 65 adolescents having undergone a renal
nephrotic syndrome, and in some cases renal dys- biopsy in the setting of nephrotic syndrome [15].
function. In adults, MN is the most common pri- Further data in support of a rising incidence of
mary glomerulopathy associated with the MN in adolescents comes from a recent study
nephrotic syndrome, accounting for approxi- from Pakistan involving 538 patients with
mately 25–35 % of adults diagnosed with this nephrotic syndrome including 173 adolescents
condition [1–3]. In children, MN may present as (13–18 years of age); MN was found in 3 %
the nephrotic syndrome or as asymptomatic pro- of children 12 and under, but 18.5 % of adoles-
teinuria and is considered to a much less common cents [16]. The United States Renal Data Systems
condition than that seen in adults [4, 5]. MN may (USRDS) 2013 report indicates that 36 pediatric
be idiopathic or secondary to infections, autoim- patients (0.6 %) had MN as an underlying cause of
mune diseases, or medications. Recent break- their end-stage renal disease over the 5-year
through discoveries regarding the pathogenesis period (2007–2011). These patients had a median
of idiopathic MN have reenergized the field and age of 16 years and were nearly equally split
should aid clinicians going forward with regard to between Caucasians and African Americans at
developing novel means to monitor and treat nearly 40 % each without any gender bias [17].
patients with membranous nephropathy [6–8].

Histopathology

Incidence Membranous nephropathy is a pathologic entity


with common histologic features, but does not
A comprehensive review of 40 studies estimated represent one disease state [7, 18]. The histologic
the incidence of primary glomerulonephritis feature characteristic of MN is a normocellular
worldwide; it was determined that MN occurs in glomerulus with prominent capillary loops sec-
1.2/100,000/year in adults, and approximately ondary to subepithelial immune complex
0.05–0.1/100,00/year in children and adolescents deposits. To best delineate the subtle histologic
[9–11]. With regard to idiopathic MN in the set- features that aid in the distinction between idio-
ting of nephrotic syndrome, despite being respon- pathic and secondary forms of MN, they will be
sible for 25–35 % of adults with new onset described separately.
disease, MN accounts for <5 % of children with
nephrotic syndrome. A multicenter, prospective
study conducted by International Study of Kidney Idiopathic Membranous Nephropathy
Disease in Children analyzed 521 children
presenting with the nephrotic syndrome from The pathologic features of membranous nephrop-
1967 to 1974; in that era, pretreatment biopsies athy were initially described in 1968 by
revealed MN in only 1.5 % of children [12]. Other Ehrenreich and Churg [19]. This staging system
more recent reports still show the incidence of consisted of four stages seen as the evolution of
MN to be <5 % of children with nephrotic syn- immune complex formation in relation to the glo-
drome [9, 13]. However, when one focuses on a merular basement membrane (GBM):
more select demographic, the adolescent with subepithelial deposits and normal GBM usually
34 Membranous Nephropathy in Children 1057

Fig. 1 Idiopathic
membranous
nephropathy: hematoxylin
and eosin,  200; arrow
points out thickened
capillary loop, without
cellular proliferation

Fig. 2 Idiopathic
membranous nephropathy:
Periodic Acid Schiff stain,
 200; glomerulus with
widely patent capillary
loops, without cellular
proliferation

only seen on EM (Stage 1), global subepithelial 9 children [18]. Exquisite details of the histopa-
deposits with projections of newly formed matrix thology allowed one to see the characteristic light
material (Stage 2), incorporation of the deposits and electron microscopic features [18, 23]. Light
into the GBM seen as an envelopment of the microscopy is characterized by widely patent cap-
subepithelial deposits by the projections of the illary loops with normal mesangial matrix and
GBM (Stage 3), and a remodeling phase with normal mesangial cellularity, and almost “exag-
thickening of the GBM and electron-lucent or gerated appearance of normality” [20] (Figs. 1
absent deposits (Stage 4) [7, 19, 20]. Adult and and 2). The capillary wall thickening can be seen
pediatric studies have failed to provide convinc- by various light stains: hematoxylin and eosin,
ing evidence that the Ehrenreich-Churg classifica- methenamine silver-PAS (Jones), and trichrome
tion system for MN correlates with clinical stain. However, the methenamine silver-PAS
presentation or is helpful in predicting renal stain demonstrates the characteristic spikes from
outcomes [7, 21, 22]. Work by Olbing et al. projections of the glomerular basement membrane
enlightened the pediatric community on idio- (GBM) between the nonargyrophilic subepithelial
pathic MN as it was described in a cohort of deposits.
1058 R.P. Valentini

Fig. 3 Idiopathic
membranous nephropathy:
electron microscopy, 
5492; arrows point out
subepithelial electron-dense
deposits

Fig. 4 Idiopathic
membranous nephropathy:
electron microscopy, 
12,960; arrows point out
electron-dense deposits
situated beneath podocyte
foot process

Electron microscopy has helped best Nephrology Study Group in the United States
characterize this disease. Subepithelial or revealed that 31 % of patients classified as idio-
epimembranous electron-dense deposits resting pathic MN had mesangial deposits; while this was
upon the GBM with projections of cytoplasm far lower than the 100 % seen in those with MN
between them are characteristic features (Figs. 3 associated with systemic lupus erythematosus
and 4). These deposits are typically diffusely dis- (SLE), the presence or absence of mesangial
tributed, but can be segmental in some cases. deposits appear to lack specificity in the
Intramembranous deposits are also seen in pediatric patient with MN [24]. Subendothelial
idiopathic MN. Mesangial deposits, on the other deposits are even less commonly seen in idio-
hand, are uncommon in idiopathic MN in pathic MN; in the same study, subendothelial
adults, and when seen, secondary MN should deposits occurred in only 13 % of idiopathic MN
be entertained [20]. However, a pediatric versus 78 % of lupus associated MN in this pedi-
study performed by the Southwest Pediatric atric cohort [24].
34 Membranous Nephropathy in Children 1059

Fig. 5 Membranous
nephropathy secondary to
lupus nephritis:
hematoxylin and eosin,
 400. Note: thickened
capillary loops and focal
minimal increase in
mesangial cellularity at
11–12 o’clock position

Fig. 6 Membranous
nephropathy secondary to
lupus nephritis: electron
microscopy,  12,500; long
thin arrows point out
subepithelial electron-dense
deposits; short thick arrow
(on right) shows
tubuloreticular inclusion
within glomerular
endothelial cell

Immunofluorescence is characterized by a Secondary Membranous Nephropathy


granular, glomerular capillary wall distribution
of immune deposits, comprised of immunoglobu- Light microscopic features more commonly seen
lin G (IgG) and to a lesser extent complement with secondary MN include glomerular lobula-
component 3 (C3). Further characterization of tion, mesangial hypercellularity, segmental
IgG reveals a predominance of subclass involvement (scars or inflammation), and necrosis
IgG4 and IgG1 within the deposits, at 81 % [20] (Fig. 5). By electron microscopic, mesangial
and 75 %, respectively, in idiopathic MN deposits and subendothelial deposits are much
[25]. Subclass IgG3, which is not seen in idio- more commonly seen than in idiopathic
pathic MN, is present in 50 % of patients with MN. Some findings speak to the underlying
lupus associated MN [26]. A large pediatric cause of secondary MN such as SLE where
pathology study revealed granular, capillary wall tubuloreticular inclusions (TRI) may be seen
staining for IgG in 100 % and C3 staining in within the glomerular endothelial cells [4]
77 % [27]. (Figs. 6 and 7). The formation of these subcellular
1060 R.P. Valentini

children [33]. Hypertension occurs in <10 % of


children with MN, and thromboembolic events in
<5 % of cases [4, 7, 35].

Pathogenesis of Idiopathic MN

An animal model was developed in the 1950s by


Heymann et al. that replicates the autoimmune
disease which results in nephrotic syndrome
[36]. A rat autoantibody model which is triggered
by the injection of a rat proximal tubular antigenic
preparation. This results in a subepithelial,
immune complex deposition, the activation of
complement, and sublethal injury to the
podocytes. This topic is reviewed in greater detail
elsewhere [7, 37].

Historical Animal Models

Fig. 7 Membranous nephropathy secondary to lupus


The Heymann nephritis animal model, while
nephritis: electron microscopy,  40,000; short thick developed many decades ago, has been instru-
arrow shows tubuloreticular inclusion within glomerular mental in gaining an understanding to the evolu-
endothelial cell tion and pathogenesis of human, idiopathic
membranous nephropathy. This mammalian
organelles within the endoplasmic reticulum is model of immune complex nephritis consists of
induced by alpha interferon (IFN-α) [28]. While immunization of rats with an antigenic fraction of
a “full house” immunostaining pattern of featur- rat proximal tubular brush border (Fx1A). One
ing glomerular deposition of IgG, IgM, IgA, C3, key pathophysiologic aspect of this immune com-
and C1q can be seen in proliferative forms of SLE, plex disease was determining whether circulating
this is uncommon in membranous lupus nephritis. immune complexes were deposited into the
The subclass IgG3 predominance, noted above, subepithelial space. It had been hypothesized
aids in the distinction from idiopathic MN, where that unique physicochemical properties of these
IgG3 is not typically seen [26]. immune complexes allowed them to move across
the endothelial and glomerular basement mem-
brane to allow access beneath the podocytes.
Clinical Features Alternatively, circulating antibodies could react
to in situ antigens in the subepithelial region
Most children with MN present with some degree resulting in immune complex formation in that
of proteinuria. This can be asymptomatic protein- location. Subsequent studies would later validate
uria picked up incidentally on a urinalysis speci- this latter theory that antigen-antibody immune
men. This moderate proteinuria is accompanied by complexes were formed locally and could occur
microscopic hematuria in the majority (70–90 %) in the absence of circulating antigen thereby prov-
of patients [13, 29]. Nephrotic syndrome has been ing the in situ (“fixed”) antigen theory in this rat
the clinical presentation in 25–100 % of pediatric model [38, 39].
studies [13, 18, 21, 22, 29–34]. A study from South Later studies would reveal that megalin, a large
Korea noted gross hematuria in up to 40 % of glycoprotein involved in protein reabsorption in
34 Membranous Nephropathy in Children 1061

the proximal tubule, is the target antigen in the rat immunochemistry, even in patients with no or
(Heymann nephritis) model. This same protein is low levels of circulating anti-PLA2 R antibodies
also contained on the rat podocytes foot process – [43], which demonstrate that staining for PLA2 R
thereby explaining the immune complex location is more sensitive than circulating antibodies
in the subepithelial space. Subsequent to immune [44, 45]. However, PLA2 R have been detected
complex deposition, complement fixation occurs, in the immune deposits of a few patients with
leading to C5-C9 insertion into the podocyte cell membranous nephropathy-associated NSAIDS
membrane resulting in sublethal injury and pro- (naproxen and piroxicam), hepatitis B, and sar-
teinuria. While this model led to a more advanced coidosis, but was not detected in MN secondary to
understanding of human disease, it was not SLE [46, 47].
completely applicable. While humans have a PLA2R staining has been reported in 45 % of
megalin analogue expressed in the brush border children with membranous nephropathy, the
of the proximal tubule, unlike the rat, humans do youngest of which was 10 years old. None of the
not express this megalin like protein on their negative patients went on to show signs of sec-
podocytes. Moreover, this megalin like protein ondary MN at >3 years of follow-up, which sug-
was absent from immune deposits in patients gest that PLA2R staining sensitivity is much
with membranous nephropathy [40, 41]. lower in the pediatric than the adult primary mem-
The search for the antigen in human membra- branous glomerulopathy population [27].
nous nephropathy continued. Anti-PLA2 R antibody titers have been shown
to be an excellent marker of disease activity being
elevated in a relapse state and absent in a state of
M-Type Phospholipase A2 Receptor remission [41]. The antibody titer following
Antibodies rituximab predicts the clinical response [48].

In 2009, a major breakthrough in the understand-


ing of the pathogenesis of idiopathic membranous Neutral Endopeptidase
nephropathy occurred with the identification of
phospholipase A2 receptor (PLA2 R) antibodies. Neutral endopeptidase (NEP), which is expressed
These PLA2 R antibodies are directed at this on human podocytes, has been shown to be the
transmembrane glycoprotein expressed on the target antigen in some rare cases of antenatal
podocyte and have been shown to be present in MN. This is the case when a mother, genetically
70 % of adults with idiopathic MN but not sec- deficient for neutral endopeptidase, has been
ondary MN [6]. These anti-PLA(2)R autoanti- alloimmunized against NEP present on the fetal
bodies were mainly IgG4, the predominant IgG podocyte during a prior pregnancy. The transpla-
subclass found in the glomerular deposits in idio- cental passage of the anti-NEP antibodies is
pathic MN but not in the secondary forms of responsible for the subepithelial immune deposits
MN. It should be noted that the antibodies eluted in the fetus and the neonate. The severity of the
from biopsy samples from patients with idiopathic nephrotic syndrome correlates to the anti-NEP
MN, but not from secondary forms, recognized antibody titer in the mother. The nephrotic syn-
PLA2 R(6). A Chinese study of patients with drome is present at birth but proteinuria decreases
idiopathic MN, membranous lupus nephritis, and with time with a progressive decrease of the
MN associated with hepatitis B virus (HBV) maternal antibodies and a disappearance of the
infection found that anti-PLA2 R antibodies were immune deposits [49]. However, residual protein-
present in 82 % of adults with idiopathic MN, and uria and chronic renal insufficiency may occur in
only 5 % and 6 % of those with MN secondary to relation to nephron loss and renal scarring, per-
lupus and HBV, respectively [42]. haps in relation to renal arterial lesions, which are
PLA2 R antigen may be identified in the observed in addition to the subepithelial immune
immune deposits by immunofluorescence or deposits. Interestingly, the IgG fraction from the
1062 R.P. Valentini

mother serum produces proteinuria and immune Taiwan [52]. Both the causes of nephrotic syn-
deposits when injected into the rabbit drome and the presence of hepatitis B carrier state
were assessed in this study. Remarkably, the fre-
quency of HBV-associated membranous nephrop-
Other Causes athy (HBVMN) decreased steadily over the
25-year study period: 11.6 % (1974–1984), 4.5
Another antigen has been identified in young % (1984–1994), 2.1 % (1994–2004), and 0 %
pediatric patients with MN – cationic bovine (2004–2009). At the same time, this Taiwanese
serum albumin (cBSA). In young infants with study showed a marked decline in the prevalence
MN, immune complexes were identified with of HBV surface Ag (+) carrier state, which was
subepithelial deposits, which colocalized both consistent with earlier studies [52, 53]. Others
cBSA protein and cBSA IgG antibodies. In this have shown the consequences of HBV infection
study, the BSA antibodies were specific to bovine with hepatitis B e Antigen (HBeAg) detected in
serum albumin, in that they did not react to human 88 % of the glomerular deposits in children diag-
serum albumin. It was hypothesized that cBSA nosed with HBV MN [54].
was a modified, food-derived antigen from dietary Liao’s study also revealed some interesting
cow milk protein which is absorbed through the epidemiologic information as it relates to HBV
relatively immature alimentary tract, and the cat- MN in Taiwan – as they studied 471 children
ionic nature of BSA allows it to cross the nega- with nephrotic syndrome from 1974 to 2009. Of
tively charged glomerular basement membrane the 35 HBV seropositive patients, 25 had biopsy-
and allows it to be “planted” into the subepithelial proven HBV MN [no evidence of SLE or other
region for in situ formation of immune secondary causes of MN, characteristic histologic
complexes [50]. features of MN on light, immunofluorescence,
and electron microscopy and positive serum for
hepatitis B surface antigen (HBsAg)]. These
Pathogenesis of Secondary MN patients were predominantly male (84 %), and
96 % were infected via horizontal transmission
The 2012 clinical practice guidelines from the in that their mothers were HBsAg seronegative.
Kidney Diseases Improving Global Outcomes Also, it was shown that horizontal and not vertical
(KDIGO) estimated that secondary MN is more transmission of HBV is important in the develop-
common in children, accounting for 75 % of chil- ment of HBV MN, raising the question of whether
dren with MN versus 25 % of adults [51]. It is or not the response to HBV and perhaps HBeAg is
generally accepted that systemic lupus different in the neonatal and infant kidney as
erythematosus (SLE) and chronic infection with opposed to the older child [52].
hepatitis B virus (HBV) are the most common Children with HBV MN usually present with
secondary causes of MN in both children and nephritic syndrome, hematuria, and normal renal
adults [7]. Both disease entities will be discussed function. Blood tests show the presence of
along with the less common causes of secondary HBsAg, HBeAg, and anti-HBc antibodies. Com-
MN as it relates to children. plement C3 level is often low. The presence of
anti-HBe antibodies correlates with the disappear-
ance of HBeAg and clinical remission of
Hepatitis B Virus

Вам также может понравиться