Вы находитесь на странице: 1из 300

Perspectives of Stem Cells

Henning Ulrich
Editor

Perspectives
of Stem Cells
From Tools for Studying
Mechanisms of Neuronal
Differentiation towards Therapy

123
Editor
Prof. Henning Ulrich
Universidade de Sao Paulo
Instituto de Quimica
Departamento de Bioquimica
Av. Prof. Lineu Prestes 748
Sao Paulo-SP
Brazil
henning@iq.usp.br

ISBN 978-90-481-3374-1 e-ISBN 978-90-481-3375-8


DOI 10.1007/978-90-481-3375-8
Springer Dordrecht Heidelberg London New York
Library of Congress Control Number: 2009940404

Springer Science+Business Media B.V. 2010


No part of this work may be reproduced, stored in a retrieval system, or transmitted in any form or by
any means, electronic, mechanical, photocopying, microfilming, recording or otherwise, without written
permission from the Publisher, with the exception of any material supplied specifically for the purpose of
being entered and executed on a computer system, for exclusive use by the purchaser of the work.

Printed on acid-free paper

Springer is part of Springer Science+Business Media (www.springer.com)


Preface

Stem cells are fascinating cell types. They can replicate themselves forever while
retaining the potential to generate progeny with specific functions. Because of these
special properties, stem cells have been subjects of intensive investigation, from
understanding basic mechanisms underlying tissue generation, to modeling human
diseases, to application for cell replacement therapy. Stem cells come in different
forms. For example, mouse embryonic stem cells can general all cell types in a body,
either in a dish or when put back into mouse embryos. On the other hand, neural
stem cells in the adult brain generate neurons and glia cells that contribute to the
brains plasticity. Rapid progress has been made in the stem cell field with discover-
ies published in a record speed. A quick Pubmed search has returned 2789 hits for
embryonic stem cells and 815 hits for adult neural stem cells/neurogenesis in the
year 2008 alone. It remains a taunting task for all who are interested in stem cells to
keep up with rapidly accumulating literatures. The Perspectives of Stem Cells by a
truly international team of experts provides a timely and invaluable highlight of the
stem cell field gearing toward future therapeutic applications in the nervous system.
Stem cells with neural potentials have attracted a lot of attention because of their
promise for cell replacement therapy, ranging from degenerative neurological disor-
ders to spinal cord injuries. Before such potentials to be realized, however, we need
to understand the basic biology of these stem cells. For example, understanding how
stem cell behaviors are controlled by intrinsic and extrinsic factors will help to direct
stem cells into a specific fate while avoiding undesired tumorigenesis. Equally impor-
tant, we need to understand adult nervous system milieu where substitute neurons
need to integrate into proper circuitry for maximal recovery. In this regard, the recent
discovery of functional neurogenesis in discrete regions of the adult mammalian
brain, including humans, has a major impact on regenerative medicine. Not only
there exist residual adult neural stem cells as endogenous cellular sources for neu-
rogenesis, the adult nervous system itself exhibits surprising plasticity. They provide
proper signals to support normal neurogenesis and, furthermore, additional signals
upon injuries to activate neural stem cells and guide their neuronal progeny to the
right location. The dogma, In the adult centers, the nerve paths are something fixed,
ended, and immutable. Everything may die, nothing may be regenerated, is now long
gone.
The Prospective covers a broad spectrum of a fast evolving field of stem cells:
from different model systems and stem cell types, to their cell biology and molecu-
lar signaling mechanisms; from overview of protocols for directed neuronal subtype
differentiation from embryonic stem cells, and even the latest induced pluripotent

v
vi Preface

stem cells, to specific considerations for the therapeutic application. The historical
view of neurogenesis since the time of Cajal was a delight retreat; the discussion of
retrotransposons in generating neuronal diversity through neurogenesis was fascinat-
ing. The Prospective provides a much needed overview of the state of the art in the
field and a rich resource of updated information. More importantly, it sets up a stage
for flourishing of new ideas in stem cell biology and for fostering novel therapeutic
applications in the nervous system for years to come.

Clarksville, Maryland Hongjun Song


August 2009
Editor Preface

The field of stem cell biology is geared towards translation into clinical practice
through in vitro tissue production and regeneration therapy. The discovery of neu-
rogenesis in selected areas of the adult brain has revolutionized neuroscience. This
discovery has overturned the central assumption that no new neurons were origi-
nated in the brain after birth, and provided the basis for understanding the molecular
mechanisms of neural differentiation. Several in vitro models have been developed
to investigate signalling pathway of neurogenesis regulation and cell fate specifica-
tion. Massive propagation of embryonic cells into just the right type of phenotype
of a neural progenitor cell or strategies for mobilizing endogenous neural stem or
progenitor cells provide replacement therapies for brain injury resulting from stroke
or neurodegenerative diseases. Such strategies are getting more important, since the
development of ex vivo cultures of stem cells allows collection of multipotent cells
from patients, their differentiation and transplantation into diseased areas.
This book includes a chapter on mechanisms of neural induction in early embryos
as well as a detailed discussion of changes in paradigms in view of the discov-
ery of adult neurogenesis. Neuronal differentiation is detailed using the olfactory
epithelium, one of the tissues bearing neurogenesis along life. In addition to chapters
on therapeutic applicability of embryonic, very small-embryonic like, mesenchymal
stem and neural progenitor cells, this book covers signalling mechanisms guiding
induction to differentiation and selective achievement of specific phenotypes. Cell
diversification of the neuronal system is explained using the example of neural crest
cell differentiation. Alterations in genetic material, such as loss of chromosomes
and retrotransposition, are discussed as possible mechanisms for cell diversification.
Furthermore, fundamental aspects of stem cell biology and neurogenesis, such as the
importance of proliferation induction, programmed cell death, as well as the func-
tion of glia in differentiation of stem cells and development of neuronal circuits, are
also highlighted. The participation of cytoskeletal elements in cell polarization as pre-
requisites of asymmetric division and differentiation induction is discussed. Further
topics include the analysis of extracellular signals, such as neurotrophic factors and
neurotransmitters, their receptor molecules and the propagation of these signals by
intracellular signal transduction leading to activation of selective gene expression dur-
ing differentiation. Rhythmic gene expression for activation and inhibition of Notch
signalling is discussed as a mechanism for regulating the progress of neurogenesis. In
vitro cultures of embryonic, mesenchymal and neural stem cells as well as mobiliza-
tion of endogenous stem and precursor cells for brain repair and replacement therapy
in neurological disorders are important issues of this book.

vii
viii Editor Preface

Each chapter provides an invaluable resource for information on the most current
advances in the field and possible therapeutic applications, with discussions of con-
troversial issues and areas of emerging importance. By providing an up-to-date and
critical view of the state of Science, we hope that this book shall be a base for excit-
ing scientific ideas regarding functions and therapeutic applications of stem cells in
the adult brain. The book is directed to neuroscientists, physicians, students and all
who are engaged and interested in the exciting and rapidly expanding field of modern
neuroscience and stem cell biology.

So Paulo Henning Ulrich


June 2009
Contents

1 Neural Induction . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Karla Loureiro Almeida, Jos Abreu, and C. Y. Irene Yan
2 Neurogenesis: A Change of Paradigms . . . . . . . . . . . . . . . . 11
Luiz E. Mello and Beatriz M. Longo
3 Neurogenesis in the Olfactory Epithelium . . . . . . . . . . . . . . . 35
Bettina Malnic and Lucia Armelin-Correa
4 Cell Diversification During Neural Crest Ontogeny:
The Neural Crest Stem Cells . . . . . . . . . . . . . . . . . . . . . . 47
Elisabeth Dupin, Giordano W. Calloni, and Nicole M. Le
Douarin
5 Intermediate Filament Expression in Mouse Embryonic
Stem Cells and Early Embryos . . . . . . . . . . . . . . . . . . . . . 59
Zhigang Xue, Vivaldo Moura-Neto, Araksya Izmiryan,
Sheila Cristina de Souza Martins, Jean Christophe Larcher,
Denise Paulin, and Zhenlin Li
6 Aneuploidy in Embryonic Stem Cells . . . . . . . . . . . . . . . . . 73
Rafaela C. Sartore, Priscila B. Campos,
Michael J. McConnell, and Stevens K. Rehen
7 Retrotransposition and Neuronal Diversity . . . . . . . . . . . . . . 87
Maria C. N. Marchetto, Fred H. Gage, and Alysson R. Muotri
8 Directing Differentiation of Embryonic Stem Cells
into Distinct Neuronal Subtypes . . . . . . . . . . . . . . . . . . . . 97
Noelle Ammon, Nathaniel Hartman, and Laura Grabel
9 Neurotransmitters as Main Players in the Neural
Differentiation and Fate Determination Game . . . . . . . . . . . . 115
Katia K. Yuahasi, Katia N. Gomes, Marcelo Campos, Arthur
A. Nery, Ariane Nunes-Alves, Cleber A. Trujillo, and Henning Ulrich
10 Rhythmic Expression of Notch Signaling in Neural
Progenitor Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Hiromi Shimojo, Toshiyuki Ohtsuka, and Ryoichiro Kageyama

ix
x Contents

11 Neuron-Astroglial Interactions in Cell Fate Commitment


in the Central Nervous System . . . . . . . . . . . . . . . . . . . . . 145
Joice Stipursky, Tnia Cristina Leite de Sampaio e Spohr,
Luciana Ferreira Romo, and Flvia Carvalho Alcantara Gomes
12 The Origin of Microglia and the Development of the Brain . . . . . 171
Flavia R. S. Lima, Anna Carolina C. da Fonseca,
Giselle P. Faria, Luiz Gustavo F. Dubois, Trcia R. Alves,
Jane Faria, and Vivaldo Moura Neto
13 Tissue Biology of Proliferation and Cell Death Among
Retinal Progenitor Cells . . . . . . . . . . . . . . . . . . . . . . . . 191
Rafael Linden, Rodrigo A.P. Martins, Mariana S. Silveira,
Helena L. Borges, Alfred Sholl-Franco, Lucianne
Fragel-Madeira, and Ana Carolina Dudenhoeffer-Carneiro
14 Potential Application of Very Small Embryonic Like
(VSEL) Stem Cells in Neural Regeneration . . . . . . . . . . . . . . 231
Mariusz Z. Ratajczak, Ewa Zuba-Surma, Magda Kucia,
Przemyslaw Nowacki, and Bogdan Machalinski
15 Embryonic Stem Cell Transplantation for the Treatment
of Parkinsons Disease . . . . . . . . . . . . . . . . . . . . . . . . . 245
Asuka Morizane and Jun Takahashi
16 Functional Multipotency of Neural Stem Cells
and Its Therapeutic Implications . . . . . . . . . . . . . . . . . . . 255
Yang D. Teng, Serdar Kabatas, Jianxue Li,
Dustin R. Wakeman, Evan Y. Snyder, and Richard L. Sidman
17 Dual Roles of Mesenchymal Stem Cells in Spinal Cord
Injury: Cell Replacement Therapy and as a Model System
to Understand Axonal Repair . . . . . . . . . . . . . . . . . . . . . 271
Cecile King, Shyam Patel, Treena Livingston Arinzeh,
and Pranela Rameshwar
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285
Contributors

Jos Abreu Cellular and Developmental Biology Program, Institute of Biomedical


Sciences, Universidade Federal do Rio de Janeiro, Centro de Cincias da Sade
bloco F, Cidade Universitria, Rio de Janeiro, RJ 21949-590, Brazil,
garciajr@anato.ufrj.br
Karla Loureiro Almeida Departamento de Morfologia, Centro de Cincias da
Sade, Universidade Federal do Esprito Santo - Vitria/ES - Brazil,
karlaloureiro25@hotmail.com
Ariane Nunes-Alves Departamento de Bioqumica, Instituto de Qumica,
Universidade de So Paulo, So Paulo, Brazil, anunesalves@usp.br
Trcia R. Alves Programa de Anatomia, Instituto de Cincias Biomdicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,
terciabio@yahoo.com.br
Noelle Ammon Hall-Atwater Laboratories, Biology Department, Wesleyan
University, Middletown, CT, USA, nammon@wesleyan.edu
Lucia Armelin-Correa Departamento de Bioqumica, Instituto de Qumica,
Universidade de So Paulo, So Paulo, Brazil, armelin@iq.usp.br
Helena L. Borges Instituto de Cincias Biomdicas, Universidade Federal do Rio
de Janeiro, Cidade Universitaria, Rio de Janeiro, Brazil, hborges@anato.ufrj.br
Giordano W. Calloni CNRS UPR2197 Laboratoire Dveloppement, Evolution et
Plasticit du Systme Nerveux, Institut de Neurobiologie Alfred Fessard, 91198
Gif-sur-Yvette, France; Departamento de Biologia Celular, Embriologia e Gentica,
Centro de Cincias Biolgicas, Universidade Federal de Santa Catarina,
Florianpolis, Brazil, giordano@ccb.ufsc.br
Priscila B. Campos Instituto de Cincias Biomdicas, Universidade Federal do Rio
de Janeiro, Rio de Janeiro, Brazil, brittopris@anato.ufrj.br
Marcelo Campos Departamento de Bioqumica, Instituto de Qumica,
Universidade de So Paulo, So Paulo, Brazil, camposm@iq.usp.br
Anna Carolina C. da Fonseca Programa de Anatomia, Instituto de Cincias
Biomdicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,
annafonseca@anato.ufrj.br
Luiz Gustavo F. Dubois Programa de Anatomia, Instituto de Cincias Biomdicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil, dubois@anato.ufrj.br

xi
xii Contributors

Ana Carolina Dudenhoeffer-Carneiro Instituto de Biofsica, Universidade


Federal do Rio de Janeiro, Cidade Universitaria, Rio de Janeiro, Brazil,
anacarol@biof.ufrj.br
Elisabeth Dupin CNRS UPR2197 Laboratoire Dveloppement, Evolution et
Plasticit du Systme Nerveux, Institut de Neurobiologie Alfred Fessard, 91198
Gif-sur-Yvette, France, dupin@inaf.cnrs-gif.fr
Giselle P. Faria Programa de Anatomia, Instituto de Cincias Biomdicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,
giselle.faria@gmail.com
Jane Faria Programa de Anatomia, Instituto de Cincias Biomdicas, Universidade
Federal do Rio de Janeiro, Rio de Janeiro, Brazil, jane@anato.ufrj.br
Lucianne Fragel-Madeira Instituto de Biofsica, Universidade Federal do Rio de
Janeiro, Cidade Universitaria, Rio de Janeiro, Brazil, lufragel@biof.ufrj.br
Fred H. Gage Laboratory of Genetics, The Salk Institute for Biological Studies, La
Jolla, CA, USA, gage@salk.edu
Katia N. Gomes Departamento de Bioqumica, Instituto de Qumica, Universidade
de So Paulo, So Paulo, Brazil, knevesgomes@yahoo.com.br
Flvia Carvalho Alcantara Gomes Laboratrio de Neurobiologia Celular,
Programa de Biologia Celular e do Desenvolvimento, Instituto de Cincias
Biomdicas, Centro de Cincias da Sade, Universidade Federal do Rio de Janeiro,
Rio de Janeiro, Brazil, fgomes@anato.ufrj.br
Laura Grabel Hall-Atwater Laboratories, Biology Department, Wesleyan
University, Middletown, CT, USA, lgrabel@wesleyan.edu
Nathaniel Hartman Hall-Atwater Laboratories, Biology Department, Wesleyan
University, Middletown, CT, USA, nhartman@wesleyan.edu
Araksya Izmiryan UPMC Univ Paris 6, UMR 7079, Paris, France,
araksya.izmiryan@inserm.fr
Serdar Kabatas Department of Neurosurgery, Baskent University Istanbul
Hospital, Istanbul, Turkey, kabatasserdar@hotmail.com
Ryoichiro Kageyama Institute for Virus Research, Kyoto University, Kyoto, Japan;
Japan Science and Technology Agency, CREST, Kyoto, Japan,
rkageyam@virus.kyoto-u.ac.jp
Cecile King UMDNJ-New Jersey Medical School, Department of Medicine,
Newark, NJ, USA, kingc4@umdnj.edu
Magda Kucia Stem Cell Institute at James Graham Brown Cancer Center,
University of Louisville, Louisville, KY, USA, mjkuci01@louisville.edu
Jean Christophe Larcher Laboratoire de Biologie du Dveloppement, Paris,
France, jclarche@snv.jussieu.fr
Nicole M. Le Douarin CNRS UPR2197 Laboratoire Dveloppement, Evolution et
Plasticit du Systme Nerveux, Institut de Neurobiologie Alfred Fessard, 91198
Gif-sur-Yvette, France, nicole.ledouarin@academie-sciences.fr
Contributors xiii

Jianxue Li Department of Neurology, Beth Israel Deaconess Medical Center,


Harvard Medical School, Boston, MA, USA, jli7@caregroup.harvard.edu
Zhenlin Li UPMC Univ Paris 6, UMR 7079, Paris, France, zhenlin.li@upmc.fr
Flavia R.S. Lima Programa de Anatomia, Instituto de Cincias Biomdicas,
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil, flima@anato.ufrj.br
Rafael Linden Instituto de Biofsica, Universidade Federal do Rio de Janeiro,
Cidade Universitaria, Rio de Janeiro, Brazil, rlinden@biof.ufrj.br
Treena Livingston Arinzeh New Jersey Institute of Technology, Department of
Biomedical Engineering, Newark, NJ, USA, arinzeh@adm.njit.edu
Beatriz M. Longo Departamento de Fisiologia, Universidade Federal de So Paulo,
So Paulo, Brazil, beatriz.longo@unifesp.br
Bogdan Machalinski Department of Physiopathology Pomeranian Medical
University, Szczecin, Poland, machalin@pam.szczecin.pl
Bettina Malnic Departamento de Bioqumica, Instituto de Qumica, Universidade
de So Paulo, So Paulo, Brazil, bmalnic@iq.usp.br
Maria C. N. Marchetto Laboratory of Genetics, The Salk Institute for Biological
Studies, La Jolla, CA, USA, marchetto@salk.edu
Sheila Cristina de Souza Martins Instituto de Cincias Biomdicas-Universidade
Federal de Rio de Janeiro, Rio de Janeiro, Brazil, sheila@anato.ufrj.br
Rodrigo A.P. Martins Instituto de Biofsica, Universidade Federal do Rio de
Janeiro, Cidade Universitaria, Rio de Janeiro, Brazil, Rodrigo.Martins@biof.ufrj.br
Michael J. McConnell Crick-Jacobs Center for Theoretical and Computational
Biology, Salk Institute for Biological Studies, La Jolla, California, USA,
mikemc@salk.edu
Luiz E. Mello Departamento de Fisiologia, Universidade Federal de So Paulo, So
Paulo, Brazil, lemello@unifesp.br
Asuka Morizane Department of Biological Repair, Institute for Frontier Medical
Sciences, Kyoto University, Kyoto, Japan, asuka.morizane@frontier.kyoto-u.ac.jp
Vivaldo Moura-Neto Instituto de Cincias Biomdicas-Universidade Federal de
Rio de Janeiro, Rio de Janeiro, Brazil, vivaldo@anato.ufrj.br
Alysson Muotri University of California at San Diego, School of Medicine,
Department of Pediatrics/Rady Childrens Hospital San Diego and Department of
Pediatrics/Cellular and Molecular Medicine, La Jolla, CA, USA, muotri@ucsd.edu
Arthur A. Nery Departamento de Bioqumica, Instituto de Qumica, Universidade
de So Paulo, So Paulo, Brazil, arthur.nery@gmail.com
Przemyslaw Nowacki Department of Physiopathology Pomeranian Medical
University, Szczecin, Poland, rektor@szczecin.pl
Toshiyuki Ohtsuka Institute for Virus Research, Kyoto University, Kyoto, Japan;
Japan Science and Technology Agency, CREST, Kyoto, Japan,
tohtsuka@virus.kyoto-u.ac.jp
xiv Contributors

Shyam Patel UMDNJ-New Jersey Medical School, Department of Medicine,


Newark, NJ, USA, patel120@umdnj.edu
Denise Paulin UPMC Univ Paris 6, UMR 7079, Paris, France,
denise.paulin@upmc.fr
Pranela Rameshwar UMDNJ-New Jersey Medical School, Department of
Medicine, Newark, NJ, USA, rameshwa@umdnj.edu
Mariusz Z. Ratajczak Stem Cell Institute at James Graham Brown Cancer Center,
University of Louisville, Louisville, KY, USA; Department of Physiopathology
Pomeranian Medical University, Szczecin, Poland, mzrata01@louisville.edu
Stevens K. Rehen Instituto de Cincias Biomdicas, Universidade Federal do Rio
de Janeiro, Rio de Janeiro, Brazil, srehen@anato.ufrj.br
Luciana Ferreira Romo Laboratrio de Neurobiologia Celular, Programa de
Biologia Celular e do Desenvolvimento, Instituto de Cincias Biomdicas, Centro de
Cincias da Sade, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,
romao@anato.ufrj.br
Rafaela C. Sartore Instituto de Cincias Biomdicas, Universidade Federal do Rio
de Janeiro, Rio de Janeiro, Brazil, fa_sartore@yahoo.com.br
Hiromi Shimojo Institute for Virus Research, Kyoto University, Kyoto, Japan;
Japan Science and Technology Agency, CREST, Kyoto, Japan,
hshimojo@virus.kyoto-u.ac.jp
Alfred Sholl-Franco Instituto de Biofsica, Universidade Federal do Rio de
Janeiro, Cidade Universitaria, Rio de Janeiro, Brazil, asholl@biof.ufrj.br
Richard L. Sidman Department of Neurology, Beth Israel Deaconess Medical
Center, Harvard Medical School, Boston, MA, USA,
Richard_Sidman@hms.harvard.edu
Mariana S. Silveira Instituto de Biofsica, Universidade Federal do Rio de Janeiro,
Cidade Universitaria, Rio de Janeiro, Brazil, silveira@biof.ufrj.br
Evan Y. Snyder Stem Cell and Regeneration Program, The Burnham Institute, La
Jolla, CA, USA, esnyder@burnham.org
Tnia Cristina Leite de Sampaio e Spohr Laboratrio de Neurobiologia Celular,
Programa de Biologia Celular e do Desenvolvimento, Instituto de Cincias
Biomdicas, Centro de Cincias da Sade, Universidade Federal do Rio de Janeiro,
Rio de Janeiro, Brazil, spohr@anato.ufrj.br
Joice Stipursky Laboratrio de Neurobiologia Celular, Programa de Biologia
Celular e do Desenvolvimento, Instituto de Cincias Biomdicas, Centro de Cincias
da Sade, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil,
joice@anato.ufrj.br
Jun Takahashi Department of Biological Repair, Institute for Frontier Medical
Sciences, Kyoto University, Kyoto, Japan, jbtaka@frontier.kyoto-u.ac.jp
Contributors xv

C.Y. Irene Yan Department of Cell and Developmental Biology, Institute of


Biomedical Sciences, Universidade de So Paulo, Av. Prof Lineu Prestes 1524, So
Paulo, SP 05508-900, Brazil, ireneyan@usp.br
Yang D. Teng Department of Neurosurgery, Harvard Medical School, Brigham and
Womens Hospital, and Childrens Hospital Boston, Boston, MA, USA; Department
of Physical Medicine and Rehabilitation, Harvard Medical School, Spaulding
Rehabilitation Hospital, Boston, MA, USA; Division of SCI Research, Veterans
Affairs Boston Healthcare System, Boston, MA, USA, yang_teng@hms.harvard.edu
Cleber A. Trujillo Departamento de Bioqumica, Instituto de Qumica,
Universidade de So Paulo, So Paulo, Brazil, clebertrujillo@yahoo.com.br
Henning Ulrich Departamento de Bioqumica, Instituto de Qumica, Universidade
de So Paulo, So Paulo, Brazil, henning@iq.usp.br
Dustin R. Wakeman Stem Cell and Regeneration Program, The Burnham Institute,
La Jolla, CA, USA; University of California at San Diego: Graduate Program in
Biomedical Sciences, La Jolla, CA 92093, USA, dwakeman@burnham.org
Zhigang Xue UPMC Univ Paris 6, UMR 7079, Paris, France,
zhigang.xue@upmc.fr
Katia K. Yuahasi Programa de Ps-graduao em Neurologia, Departamento de
Neurologia e Neurocirurgia, Universidade Federal de So Paulo, So Paulo, Brazil,
yuahasi@usp.br
Ewa Zuba-Surma Stem Cell Institute at James Graham Brown Cancer Center,
University of Louisville, Louisville, KY, USA, ewa.zuba-surma@uj.edu.pl
Chapter 1

Neural Induction

Karla Loureiro Almeida, Jos Abreu, and C. Y. Irene Yan

Contents layer of complexity to the interpretation of the results


obtained from the amphibian model. Here, we will
1.1 Introduction . . . . . . . . . . . . . . . . . 1 focus on the landmark experiments that address the
1.2 Neural Induction in the Xenopus Embryo earliest step of neural induction in these two models.
The Early Experiments . . . . . . . . . . . 2 Specifically, we will discuss the Neural Default model
1.3 Neural Default Model . . . . . . . . . . . . 3
that was generated from experiments in the amphib-
1.4 BMP and the Neural Inducers . . . . . . . . 4
1.5 Challenges to the Neural Default Model . . . . 4 ian embryo to explain the choice between epidermal
1.6 Neural Induction and the Avian Node . . . . . 4 and neural precursor fate and the modifications on this
1.7 Epiblast The Responsive Tissue . . . . . . . 5 model based on conclusions derived from the chick
1.8 Inhibition of BMP in the Avian Context . . . . 6
embryo.
1.9 FGF Signaling and Neural Induction . . . . . 7
References . . . . . . . . . . . . . . . . . . . . 8
Keywords BMP signaling Ectoderm FGF Neural
induction Smad Xenopus Chick
Abstract Neural induction, i.e. definition of the
Abbreviations
neural domain from the ectoderm, is a fundamental
topic that has fascinated developmental biologists for
years. The concept was first proposed by Spemman BMP bone morphogenetic protein
TGF- transforming growth factor
and Mangold after their classic experiment in the
FGF fibroblast growth factor
amphibian Xenopus laevis where transplantation of
MAPK mitogen activated protein kinase
the embryos dorsal blastopore lip induced a com-
plete neural axis from the acceptor embryos ectoderm.
Since then, much effort has been applied into iden-
tifying the signals that bias the ectoderm into neural
1.1 Introduction
fate and the resulting picture clearly indicates that
neural induction is a multi-step process that requires
the interplay of various pathways. A major part of The induction of neural tissue is a fundamental ques-
our current understanding of neural induction orig- tion that has fascinated developmental biologists since
inates from the original amphibian model Xenopus the classic experiment by Spemmann and Mangold.
laevis. Recently, the chick embryo has added another In 1924, based on their results from grafting experi-
ments performed in amphibian embryos, the authors
proposed for the first time the concept of neural induc-
tion. At the time, it was known that the blastopore
C.Y.I. Yan () lip initial involution site during gastrulation marked
Department of Cell and Developmental Biology, Universidade
the dorsal region of the embryo, and that the future
de So Paulo, Av Prof. Lineu Prestes, 1524, So Paulo, SP,
05508-900, Brazil neural plate arose from the dorsal ectoderm the ven-
e-mail: ireneyan@usp.br tral ectoderm forms mainly epidermal tissue. Spemann

H. Ulrich (ed.), Perspectives of Stem Cells, 1


DOI 10.1007/978-90-481-3375-8_1, Springer Science+Business Media B.V. 2010
2 K.L. Almeida et al.

and Mangold transplanted the blastopore lip of donor a bona fide candidate for direct neural inducer had to
embryos to the ventral region of host embryos in gas- fulfill certain criteria: it should cause axis duplication
trula stage. The host embryos went on to develop in whole embryos, it should be expressed in the dor-
a second, ventral neuraxis and anterior nervous sys- sal blastopore (Organizer) region and elimination of its
tem. More strikingly, the duplicate nervous system activity should interfere with normal neural develop-
was fully composed of host tissue, while the trans- ment. The experimental paradigm used to screen for
plant gave rise to a second notochord (dorsal meso- candidate neural inducers was based on the fact that,
derm) underlying it. This result suggested strongly that by definition, induction involves a signaling source
the grafted tissues determinative influences on its and a responsive target. Based on Spemmans exper-
surroundings converted the surrounding ventral ecto- iment, the endogenous source of neural inducers is
derm into the second nervous system (Spemann and the Organizer and the responsive tissue the ectoderm.
Mangold, 1924). The authors named the dorsal blasto- Thus, ectoderm explants assays were used as an initial
pore lip the Organizer, and hypothesized that during screen for candidates. Ectoderm explants (also known
normal development this region determined the choice as animal caps) are cultured from a piece of ecto-
of a neural fate for the dorsal ectoderm. They also derm excised from the animal pole of late blastulas,
proposed that the effect of the Organizer on the respon- the lower part of which constitutes the blastocoele
sive ectoderm necessarily would involve cell-to-cell roof (Fig. 1.1a). At this stage, the ectoderm is not
communication. yet committed to an epidermal or neural fate and
In the ensuing years, much effort has been applied responds to growth factors in the media or overex-
for identifying the exact signals that emanate from the pression of relevant mRNAs by adopting different cell
Organizer and activate the signaling pathways that bias fates, which are verified through the expression of
the ectoderm into neural fate in vertebrates. The result- marker genes. When cultured as an intact tissue in
ing picture, derived from data obtained by various saline solution, ectoderm explants express genes char-
groups, indicates that neural induction is a multi-step acteristic of epidermal tissue (Kintner and Melton,
process. The amphibian model, Xenopus laevis, has 1987). However, if the explant is co-cultured with a
continued to be of major importance to our understand- dorsal blastopore lip, neural markers are expressed
ing of neural induction due to the ease of experimental instead (Kintner and Melton, 1987). Thus, a genes
readout of neural induction in ectoderm explants. In neural-inducing activity is identified if there is upregu-
recent years, the chick embryo has added another lation of the expression of neural markers and decrease
layer of complexity to the interpretation of the results in the expression of epidermal genes. Importantly,
obtained from the amphibian model. In the following because the Organizer is part of the dorsal meso-
sections we will present the major results derived from derm, genes that increased neural marker expression
both model systems and the model that is emerging but also induced mesoderm markers, were not con-
from those results. For the purposes of this chapter, we sidered direct neural inducers, as their effect could
will focus our discussion on the earliest step of neural be indirect, through additional factors secreted by the
induction, which is the choice between epidermal and mesoderm.
neural precursor fate. The first molecule to fulfill all of the above-
mentioned criteria for direct neural induction was
Noggin, a secreted polypeptide first identified by
Smith and Harland (1992) in the Xenopus. Afterwards,
1.2 Neural Induction in the Xenopus Follistatin (Hemmati-Brivanlou et al., 1994) and
Chordin (Sasai et al., 1994), were also isolated from
Embryo The Early Experiments
Xenopus embryos on the basis of their neuraliz-
ing activity. All of these factors fulfilled the above-
In the decade of 19902000, the search for the mentioned conditions, including expression at the
Organizers neural inducing factors intensified and was Organizer. At the time, these molecules were thought
mainly performed in the Xenopus embryo. Based on to act by directly stimulating neural fate, albeit through
the characteristic of the Organizer, it was agreed that an as yet unidentified mechanism.
1 Neural Induction 3

Fig. 1.1 Epidermal default


model versus neural default
Model. (a) In the epidermal
default model the normal fate
of an ectodermic tissue would
be epidermal, unless this
ectoderm is stimulated by
external factors (such as those
provided by addition of the
dorsal blastopore lip). (b) In
the neural default model,
the intact ectoderm secretes
anti-neural/pro-epidermal
factors. Induction of neural
fate occurs either by
co-culture with the dorsal
blastopore lip, which secretes
neuralizing factors or
dissociation of the ectoderm.
In the former, neuralizing
factors counteract the effect of
endogenous pro-epidermal
factors. In the latter case,
dissociation of the ectoderm
dilutes these factors and
generates neural fate.
Addition of ectoderm extract
restores epidermal fate

1.3 Neural Default Model of neural fate (Godsave and Slack, 1989; Grunz and
Tacke, 1989; Sato and Sargent, 1989). Thus, it was
proposed that the ectoderm has neural default fate,
Insight on the mode of action of these molecules came
which is revealed in the absence of exogenous sig-
from a second series of experiments that explored
naling (reviewed by Muoz-Sanjun and Brivanlou,
the effect of cell dissociation on ectoderm cell fate.
2002).
When ectoderm explants are dissociated into individ-
The addition of concentrated ectodermal super-
ual cells and cultured as such for a set period of time,
natant to dissociated cell cultures prevented the expres-
they express neural markers, instead of epidermal ones
sion of neural markers after ectodermal dissociation
(Fig. 1.1b). Remarkably, this occurs in the absence of
(Grunz and Tacke, 1990). Thereafter, candidate pro-
the dorsal blastopore lip and without the addition of
teins for the role of epidermal factor were added
exogenous factors (Godsave and Slack, 1989; Grunz
onto dissociated cultures and tested for their ability to
and Tacke, 1989; Sato and Sargent, 1989; Wilson et al.,
restore epidermal fate while suppressing neuralization.
1997). These data led to the hypothesis that neural-
These screens identified Bone Morphogenetic Protein
ization is the default fate for ectodermal cells, and
4 (BMP4), a member of the Transforming Growth
that the cellcell interactions that occur in an intact
Factor beta (TGF-) superfamily as a potent epidermal
ectodermic tissue somehow inhibit this developmen-
inducer. When BMP4 is added to a culture of cells dis-
tal path, resulting in an epidermal fate (Fig. 1.1b).
sociated from the ectoderm it induces the expression of
Once the tissue is dissociated, these epidermal fac-
epidermal markers (Wilson and Hemmati-Brivanlou,
tors are sufficiently diluted so as to allow development
1995). Moreover, the expression pattern of BMPs in
4 K.L. Almeida et al.

the Xenopus gastrula is consistent with the role of of BMP-inhibiting neural inducers. Accordingly, it
epidermal factor: BMP4 is found throughout the has relatively low levels of BMP signaling. Likewise,
ectoderm prior to gastrulation but, afterwards it is this model explains the double-neural axis phenotype
excluded from the neural plate (Fainsod et al., 1994; in Spemann and Mangolds original Organizer graft
Hemmati-Brivanlou and Thomsen, 1995). Finally, experiment: the grafting of an additional Organizer in
inhibition of BMP signaling in ectodermal cells with the ventral region provided a source of neural inducers
dominant-negative receptors or antisense BMP4 RNA that inhibited BMP signaling in that region, allow-
neuralizes ectodermal cells (Sasai et al., 1995). This ing the ventral ectodermal cells to follow their default
last set of data was consistent with the model that neural fate.
inhibition of endogenous BMP signaling, through
dilution, directs dissociated ectodermal cells towards
neural fate.
1.5 Challenges to the Neural Default
Model

1.4 BMP and the Neural Inducers The model of neural induction based on the sim-
ple inhibition of BMP signaling by its antagonists
The discovery of the neuralization-suppressing effect expressed at the Organizer has been challenged, how-
of BMP4 suggested a new hypothesis for the mode ever, by results which suggest that neural induction
of action of the direct neuralizers (Noggin, Chordin is a more complex process, involving additional fac-
and Follistatin), that is through the inhibition of BMP4 tors. One of these might be Fibroblast Growth Factor
action. Further experiments showed that, indeed, (FGF; Kengaku and Okamoto, 1993). FGF treatment
Noggin and Chordin directly bind to BMP4 protein and increases expression of neural markers and decreases
interfere with its ligation to its receptor (Zimmerman that of epidermal markers, (Kengaku and Okamoto,
et al., 1996; Piccolo et al., 1996). Follistatin also 1993, 1995; Lamb and Harland, 1995; Uzgare et al.,
binds to BMPs and, while still allowing ligation to its 1998). Furthermore, dominant-negative FGF receptor
receptor, forms a trimeric complex that inhibits sig- inhibits the neuralizing effects of ectoderm dissocia-
naling (Nakamura et al., 1990; Fainsod et al., 1997; tion and of noggin overexpression in whole embryos
Iemura et al., 1998). Interestingly, molecular studies (Hongo et al., 1999; Launay et al., 1996). Together,
have shown that different from Noggin and Follistatin these data suggested that FGF might also be nec-
the inhibitory activity of Chordin on BMP resides in essary to promote neural induction. This was just
specific cysteine-rich (CR) domains and is phylogenet- the beginning of a series of questions regarding the
ically conserved (Abreu et al., 2002). sufficiency of BMP inhibition in the neural induc-
The model that emerged was one in which the deci- tion model, which was primarily based on amphibian
sion on the neural or epidermal fate of the ectoderm embryos. The strongest evidence against the neural
depends on the level of BMP signaling. When BMP default model of BMP inhibition, however, came from
signaling is decreased, either through dilution in disso- experiments conducted in chick embryos.
ciated cultures or inhibition by neural inducers, ecto-
derm will progress towards a neural fate. Conversely,
when BMP signaling prevails, the ectoderm will form
epidermis. 1.6 Neural Induction and the Avian Node
This model is consistent with the conditions occur-
ring during normal Xenopus development: On the ven- Unlike the Xenopus embryos, whose development is
tral ectoderm of the gastrulating embryo, which is dia- completely external, the avian embryo initiates its
metrically opposite to the Organizer and which devel- development in the oviduct (reviewed in Wittler and
ops into the epidermis, high levels of BMP are detected Kessel, 2004). The initial cleavage cycles that occur
(Jones et al., 1996; Reem-Kalma et al., 1995). In con- there generate a flat blastoderm disc overlying the yolk.
trast, the dorsal ectoderm, where neurulation occurs, is When the egg is laid, the avian embryo is a translu-
in close proximity to the Organizer, which is the source cent disc composed of an epithelial monolayer the
1 Neural Induction 5

epiblast , which is subdivided into a central area pel- expanding anteriorly and bisects the embryo into left
lucida and an yolk-rich, extra-embryonic area opaca. and right regions (Fig. 1.2).
The circumference where the pellucida and the opaca The node is considered the avian homologue of the
meet is known as the Marginal Zone. After a few hours, amphibian dorsal blastopore lip. Its neural inductive
a half-moon-shaped thickened region appears at the abilities and gene expression pattern are reminiscent
Marginal Zone. This structure is known as Kohlers of the Organizer: transplantation of the node to the
sickle and is the morphological landmark for the pos- extraembryonic area opaca induces a secondary neu-
terior end of the embryo and the site for initiation of raxis (Waddington, 1932; Storey et al., 1992), with
gastrulation. At the stage of its appearance, the epiblast minimal participation of donor node cells (Storey
cells migrate posteriorly in a bilaterally symmetric et al., 1992). Furthermore, the node expresses the avian
movement and anteriorly at the midline, forming the homologues of Goosecoid (Izpisua-belmonte et al.,
primitive streak through which epiblast cell ingress and 1993), Goosecoid-like gene (Gsx, Lemaire et al., 1997)
form the definitive endoderm and mesoderm (Hatada and Chordin (Streit et al., 1998), which are found in the
and Stern, 1994; Voiculescu et al., 2007; Joubin and Xenopus Organizer.
Stern, 1999 ). When sickle cells and the central epiblast
cells meet at the anteriormost edge of the primi-
tive streak, they form a thickened structure known as 1.7 Epiblast The Responsive Tissue
Hensens Node, or simply the node (Fig. 1.2; Lawson
and Schoenwolf, 2001; Bachvarova et al., 1998 ). As The induction and patterning of the avian nervous
gastrulation continues, the primitive streak continues system is a stepwise process that can be subdivided

Fig. 1.2 The expression pattern of BMP, Chordin and FGF during different stages of early chick development. The first row
represents a simplified dorsal view of the pre-gastrula and gastrulating embryo
6 K.L. Almeida et al.

into the ability of the epiblast to respond to neu- 1.8 Inhibition of BMP in the Avian
ralizing signals (competence), the progressive sta- Context
bilization of this response (specification) and the
subsequent patterning of the neural region in its
diverse axis. The initial experiments by Waddington The search for avian neural inducers that compartmen-
(1932, 1993) showed that the avian blastulas epi- talize the epiblast into neural or epidermal fate was
blast layer is competent to respond to neuralizing initially based on a parallelism between the inductive
signals derived from the node. Indeed, fate mapping abilities of Hensens Node and Spemanns Organizer.
experiments show that neural structures arise from In support of this idea was the expression pattern of
a widespread region of the epiblast prior to gas- BMPs and its inhibitors in late Primitive Streak stages:
trulation (Hatada and Stern, 1994; Garca-Martnez Prior to egg-laying, BMP is present throughout the
et al., 1993). Waddingtons conclusions were fur- epiblast but, when neuro-epidermal compartmentaliza-
ther refined by Storey et al. who transplanted ectopic tion occurs, it becomes excluded from the prospective
nodes to progressively older host embryos and deter- neural tissue (Wilson et al., 2000; Streit et al., 1998,
mined that the epiblast can generate a full antero- Watanabe and Le Douarin, 1996; Streit et al., 1998).
posterior neural axis up to early gastrula stages (Storey Likewise, the TGF-beta inhibitors chordin and noggin,
et al., 1992; Streit et al., 1997). Thereafter, the which are expressed anterior to Kohlers Sickle prior to
epiblast cannot be induced to form anterior neural gastrulation, are found at the anterior tip of the prim-
structures. itive streak in early gastrulas and are restricted to the
The precise stage at which the epiblast first demon- notochord and the node in late gastrulas (Streit et al.,
strates that it is competent to follow neural fate has 1998; Streit and Stern, 1999; Connolly et al., 1997).
been progressively pushed back as more molecular Altogether, these data suggested that in chick, simi-
markers have become available. For instance, the early lar to Xenopus, BMP and its inhibitors are present in
neural marker Sox3 and late marker Sox2 have been complementary regions and that definition of a BMP-
used as standard indicators of chick neural specifi- activity-free neural domain plays a crucial role in
cation (Rex et al., 1997; Streit et al., 2000, 1997; neural induction.
Uchikawa et al., 2003). Sox3 is detected throughout However, contrary to the results obtained in
the epiblast before neural induction in pre-gastrula amphibian embryos, application of ectopic chordin
embryos and becomes restricted to the future neu- onto early gastrula embryos cannot induce neural fate
roectoderm as development progresses. Sox2 is first in non-neural ectoderm (Streit et al., 1998). Moreover,
detected around the time when neural induction is it would be expected, from the results in the frog
believed to occur and its expression is limited to model, that exposure to ectopic BMP would con-
the neuroectoderm (Rex et al., 1997; Muhr et al., vert the presumptive neural domain into epidermal.
1999). Surprisingly, application of BMP onto early gastru-
Accordingly, immediately prior to gastrulation, the las neural domains does not inhibit Sox3 or Sox2
potential of different regions of the epiblast differ. expression (Streit et al., 1998). Inhibition of BMP
Cultures of explants derived from central epiblast gen- signaling through overexpression of Smad6 or dom-
erated Sox2 and Sox3-positive cells whereas cultures inant negative BMP receptor is also not sufficient
derived from explants removed from regions closer for neural induction (Linker and Stern, 2004). These
to the marginal zone did not. Rather, these periph- results, together with the findings that central epiblast
eral explants express genes indicative of epidermal is specified as neural prior to egg-laying (see previ-
fate (Fig. 1.2, Wilson et al., 2000). Thus, by fol- ous section), indicated that at early gastrula stages
lowing the expression of Sox3 and Sox2 in cultured the neuro-ectodermal regions are already specified and
epiblast explants, the earliest stage in which epiblast is that the search for the initial neuralizing step should
compartmentalized into neural and epidermal domains include earlier developmental stages.
was identified to be immediately prior to egg-laying Thus, Wilson and collaborators investigated the
(Wilson et al., 2000). At this stage, neural fate is identity of the signals that compartmentalized the cen-
restricted to the central epiblast and epidermal fate to tral and peripheral epiblast into their respective neural
the peripheral epiblast. and epidermal fates in pre-gastrula embryos. At this
1 Neural Induction 7

stage, the central epiblast is still susceptible to BMP epiblast can be compensated for by the addition of
and will respond to its presence by converting from BMP inhibitors (Streit et al., 2000; Wilson et al., 2000,
neural to epidermal fate (Streit et al., 1998; Wilson 2001). Together, these data suggest that FGF is a puta-
et al. 2000). Thus, in early chick epiblasts, the Xenopus tive early neural inducer that acts by counteracting
neural induction model holds true, in that BMP sig- BMP signaling in the central epiblast.
naling confers an epidermal bias and that its absence These results agree with the previously mentioned
is necessary for neural fate. The dynamics of BMP effects of FGF on Xenopus embryos. However, at the
expression at this stage is consistent with its role as the time that those reports appeared, FGF was considered
endogenous epidermalizing signal BMP is downreg- mainly a posteriorizing signal that acted secondarily on
ulated in central epiblast and maintained in peripheral the neural domain generated by inhibition of BMP sig-
epiblast (Streit et al., 1998; Wilson et al., 2000). This naling. In light of the compelling data obtained from
plasticity ends with the onset of gastrulation (HH4) chick embryos, the role of FGF as a primary neural-
(Fig. 1.2; Wilson et al., 2000). The neural domains izing signal was revisited in the amphibian embryo as
progressive resistance to BMP reflects the gradual well. This reassessment was done with ex vivo ectoder-
commitment to neural fate that occurs during normal mal explants and in vivo analysis of ventral ectoderm
embryonic development. fate in whole embryos. The results derived from in
vivo experiments differed somewhat from the clas-
sical ex vivo experiments. While overexpression of
truncated TGF-beta receptor was sufficient to induce
1.9 FGF Signaling and Neural Induction Sox2 expression in amphibian ectodermal explants
(Wilson and Hemmati-Brivanlou, 1995), it did not
The question that remains is: what is the identity of induce a similar response in whole embryo ventral
the endogenous factor(s) that inhibit BMP signaling in ectoderm (Linker and Stern, 2004; Delaune et al.,
the pre-gastrula central epiblast? Contrary to expecta- 2005). In this experimental paradigm, ectopic expres-
tions, BMP signaling cannot be directly antagonized sion of neural markers was achieved when there was
by secreted BMP-inhibitors in pre-gastrula embryo. concomitant inhibition of BMP and stimulation of FGF
Although Chordin is expressed at the gastrulas node, signaling (Linker and Stern, 2004). Moreover, in the
neither Chordin, Noggin, Follistatin or Caronte were absence of FGF signaling, the ectoderm cannot be neu-
detected in central or peripheral epiblast in pre-gastrula ralized by inhibition of BMP (Delaune et al., 2005).
embryos (Levin, 1998; Wilson et al., 2000). Moreover, These results strongly suggest that, similar to the
these inhibitors cannot induce neural markers by them- avian embryo, neuralization in the amphibian embryo
selves (Streit et al., 1998, 2000). In other words, an requires interaction of the FGF and BMP pathways.
alternative signaling mechanism must maintain the The interaction between both pathways has been
central epiblast BMP-free for the initial step in neural mapped to Smad1, a downstream nuclear effector of
induction to occur. the BMP pathway. Smad1 nuclear translocation and
The answer came from a series of elegant experi- transcriptional activity are increased when it is phos-
ments that provided strong evidence that FGF meets all phorylated at the carboxy-terminal upon activation of
the requirements for a role as an endogenous inhibitor the BMP receptor serine/threonine kinase (Massagu
of BMP in avian blastulas. Firstly, FGF3 is expressed and Chen, 2000). This activity is required for BMP-
in pre-gastrula central epiblast (Mahmood et al., 1995; induced epidermal fate (Wilson et al., 1997; Nakayama
Wilson et al., 2000, 2001). Furthermore, exogenously et al., 1998). In contrast, when Smad1 is phospho-
applied FGF can induce the expression of early neural rylated by MAPK in the central linker region, both
markers (Streit et al., 2000). Blockade of endogenous nuclear translocation and transcription are inhibited
FGF signaling inhibits expression of Sox3. Inhibition (Kretzschmar et al., 1997).
of FGF signaling blocks neuralization and induction FGF signals through receptor tyrosine kinases that
of ectopic neural plate by a grafted organizer (Streit ultimately activate MAPK, which in turn phospho-
et al., 2000). Lastly, the FGF pathway is required rylates Smad1 (Pera et al., 2003). Underscoring the
for downregulation of BMP levels in the central epi- importance of the MAPK pathway during Xenopus
blast, and absence of FGF signaling in the central neural development, MAPK activity is required for
8 K.L. Almeida et al.

Fig. 1.3 Neural and Chordin


epidermal fate are Noggin
determined by Smad1 FGF
activity, which in turn is
regulated by
phosphorylation of its BMP
MAPK
serine/threonine residues.
FGF-induced phosphorylation
of the linker region retains
Smad1 in the cytoplasm and
P P
results in neural fate, whereas
BMP-induced N- MH1 LINKER MH2 -C
phosphorylation of the
carboxy terminal promotes
translocation of Smad1 to the
nucleus and results in
NEURAL EPIDERMAL
epidermal fate

neural induction by FGF and cell dissociation in ecto- the FGF pathway itself is modulated by other signals
derm explants (Uzgare et al., 1998; Kuroda et al., that are present during acquisition of neural compe-
2005). Thus, Smad1 integrates signals from the FGF tence. For instance, in the chick embryo, the Wnt
and BMP pathway. Its activity results from the oppos- pathway suppresses FGF signaling in the lateral epi-
ing effects between FGF-induced linker region phos- blast (Wilson et al., 2001). Lastly, as mentioned above,
phorylation versus the BMP-driven phosphorylation of cell fate induction occurs in a continuous and progres-
the carboxy-region. Consistent with this idea, overex- sive fashion. Therefore, the response of a target tissue
pression of a MAPK-kinase insensitive Smad1 inhib- to neuralizing or epidermalizing signals depends on its
ited neural development in whole embryos, whereas differentiation state at the time of exposure. An exam-
mutation of both MAPK and BMP-sensitive regions ple of this is neuralization through BMP inhibition in
resulted in very mild phenotype (Pera et al., 2003). Xenopus embryos. The response to BMP inhibition
Thus, the final model that emerges places Smad1 is lost prior to the onset of gastrulation (Wawersik
in the centre of the choice between neural and epi- et al., 2005). Likewise, neural induction in Xenopus
dermal fate. In the presence of high levels of BMP embryos is most sensitive to removal of FGF signaling
signaling, Smad1 is phosphorylated in the carboxy during mid-blastula transition (Delaune et al., 2005).
terminal, which activates its nuclear activity and cul- Although these results are still under discussion (de
minates in epidermal fate. This epidermalizing effect Almeida et al., 2008) and the exact period when each
can be counteracted by FGF, which phosphorylates the identified player is required for normal progression of
Smad1 linker, inhibits its nuclear functions, resulting neural development is still unclear, it is the general
in adoption of neural fate (Fig. 1.3). consensus that the plasticity of the ectoderm decreases
Although this model accounts for most of the results with time due to stabilization of cell fate (Streit et al.,
in the field, there are some points that must be consid- 1998; Wawersik et al., 2005; Linker and Stern, 2004;
ered: firstly, besides FGF there are other growth factors reviewed in Stern, 2005).
that can activate MAPK activity, which raises the pos- In conclusion, since the molecular identification
sibility that additional secreted proteins can modulate of direct neural inducers the development field has
neural induction (Linker and Stern, 2004). Second, proposed and refined models for the signaling that
MAPK has other target proteins, amongst them Smad2 underlies the choice between epidermal and neural fate
and Smad3, components of another TGF-beta pathway. from the ectoderm. Even though the current model
Therefore, it is possible that FGF modulates additional does not account for all the complexity that occurs in
pathways for its neuralizing effect. Indeed, there is this process, the speed with which new findings are col-
evidence that suppression of both Smad1 and Smad2 lected and incorporated into the most recent hypothesis
activity are necessary for neural induction in ventral has increased, and a more comprehensive panorama
ectoderm (Chang and Harland, 2007). Furthermore, should emerge in the next few years.
1 Neural Induction 9

References Izpisua-Belmonte JC, De Robertis EM, Storey KG, Stern CD


(1993) The homeobox gene goosecoid and the origin of orga-
nizer cells in the early chick blastoderm. Cell 74:645659.
Abreu JG, Coffinier C, Larrain J, Oelgeschlager M, De Robertis Jones CM, Dale L, Hogan BL, Wright CV, Smith JC (1996)
EM (2002) Chordin-like CR domains and the regulation Bone morphogenetic protein-4 (BMP-4) acts during gas-
of evolutionarily conserved extracellular signaling systems. trula stages to cause ventralization of Xenopus embryos.
Gene 287:3947. Development 122:15451554.
Bachvarova RF, Skromne I, Stern CD (1998) Induction of primi- Joubin K, Stern CD (1999) Molecular interactions continu-
tive streak and Hensens node by the posterior marginal zone ously define the organizer during the cell movements of
in the early chick embryo. Development 125:35213534. gastrulation. Cell 98:559571.
Chang C, Harland RM (2007) Neural induction requires contin- Kengaku M, Okamoto H (1993) Basic fibroblast growth fac-
ued suppression of both Smad1 and Smad2 signals during tor induces differentiation of neural tube and neural crest
gastrulation. Development 134:38613872. lineages of cultured ectoderm cells from Xenopus gastrula.
Connolly DJ, Patel K, Cooke J (1997) Chick noggin is expressed Development 119:10671078.
in the organizer and neural plate during axial develop- Kengaku M, Okamoto H (1995) bFGF as a possible morphogen
ment, but offers no evidence of involvement in primary axis for the anteroposterior axis of the cental nervous system in
formation. Int J Dev Biol 41:389396. Xenopus. Development 121:31213130.
de Almeida I, Rolo A, Batut J, Hill C, Stern CD, Linker C (2008) Kintner CR, Melton DA (1987) Expression of Xenopus N-CAM
Unexpected activities of Smad7 in Xenopus mesodermal and RNA in ectoderm is an early response to neural induction.
neural induction. Mech Dev 125:421431. Development 99:311325.
Delaune E, Lemaire P, Kodjabachian L (2005) Neural induction Kretzschmar M, Doody J, Massague J (1997) Opposing BMP
in Xenopus requires early FGF signaling in addition to BMP and EGF signalling pathways converge on the TGF-beta
inhibition. Development 132:299310. family mediator Smad1. Nature 389:618622.
Fainsod A, Deissler K, Yelin R, Marom K, Epstein M, Pillemer Kuroda H, Fuentealba L, Ikeda A, Reversade B, De Robertis EM
G, Steinbeisser H, Blum M (1997) The dorsalizing and neu- (2005) Default neural induction: neuralization of dissociated
ral inducing gene follistatin is an antagonist of BMP-4. Mech Xenopus cells is mediated by Ras/MAPK activation. Genes
Dev 63:3950. Dev 19:10221027.
Fainsod A, Steinbeisser H, De Robertis EM (1994) On the Lamb TM, Harland RM (1995) Fibroblast growth factor is a
function of BMP-4 in patterning the marginal zone of the direct neural inducer, which combined with noggin gen-
Xenopus embryo. EMBO J 13:50155025. erates anterior-posterior neural pattern. Development 121:
Garcia-Martinez V, Alvarez IS, Schoenwolf GC (1993) 36273636.
Locations of the ectodermal and nonectodermal subdivisions Launay C, Fromentoux V, Shi DL, Boucaut JC (1996) A trun-
of the epiblast at stages 3 and 4 of avian gastrulation and cated FGF receptor blocks neural induction by endogenous
neurulation. J Exp Zool 267:431446. Xenopus inducers. Development 122:869880.
Godsave SF, Slack JM (1989) Clonal analysis of mesoderm Lawson A, Schoenwolf GC (2001) Cell populations and mor-
induction in Xenopus laevis. Dev Biol 134:486490. phogenetic movements underlying formation of the avian
Grunz H, Tacke L (1989) Neural differentiation of Xenopus primitive streak and organizer. Genesis 29:188195.
laevis ectoderm takes place after disaggregation and Lemaire L, Roeser T, Izpisua-Belmonte JC, Kessel M (1997)
delayed reaggregation without inducer. Cell Differ Dev 28: Segregating expression domains of two goosecoid genes dur-
211217. ing the transition from gastrulation to neurulation in chick
Grunz H, Tacke L (1990) Extracellular matrix components pre- embryos. Development 124:14431452.
vent neural differentiation of disaggregated Xenopus ecto- Levin M (1998) The roles of activin and follistatin signaling in
derm cells. Cell Differ Dev 32:117123. chick gastrulation. Int J Dev Biol 42:553559.
Hatada Y, Stern CD (1994) A fate map of the epiblast of the early Linker C, Stern CD (2004) Neural induction requires BMP inhi-
chick embryo. Development 120:28792889. bition only as a late step, and involves signals other than FGF
Hemmati-Brivanlou A, Kelly OG, Melton DA (1994) Follistatin, and Wnt antagonists. Development 131:56715681.
an antagonist of activin, is expressed in the Spemann Mahmood R, Kiefer P, Guthrie S, Dickson C, Mason I (1995)
organizer and displays direct neuralizing activity. Cell 77: Multiple roles for FGF-3 during cranial neural development
283295. in the chicken. Development 121:13991410.
Hemmati-Brivanlou A, Thomsen GH (1995) Ventral mesoder- Massague J, Chen YG (2000) Controlling TGF-beta signaling.
mal patterning in Xenopus embryos: expression patterns and Genes Dev 14:627644.
activities of BMP-2 and BMP-4. Dev Genet 17:7889. Muhr J, Graziano E, Wilson S, Jessell TM, Edlund T (1999)
Hongo I, Kengaku M, Okamoto H (1999) FGF signaling and Convergent inductive signals specify midbrain, hindbrain,
the anterior neural induction in Xenopus. Dev Biol 216: and spinal cord identity in gastrula stage chick embryos.
561581. Neuron 23:689702.
Iemura S, Yamamoto TS, Takagi C, Uchiyama H, Natsume Muoz-Sanjuan I, Brivanlou AH (2002) Neural induction, the
T, Shimasaki S, Sugino H, Ueno N (1998) Direct binding default model and embryonic stem cells. Nat Rev Neurosci
of follistatin to a complex of bone-morphogenetic protein 3:271280.
and its receptor inhibits ventral and epidermal cell fates Nakamura T, Takio K, Eto Y, Shibai H, Titani K, Sugino H
in early Xenopus embryo. Proc Natl Acad Sci USA 95: (1990) Activin-binding protein from rat ovary is follistatin.
93379342. Science 247:836838.
10 K.L. Almeida et al.

Nakayama T, Gardner H, Berg LK, Christian JL (1998) Smad6 Streit A, Sockanathan S, Perez L, Rex M, Scotting PJ, Sharpe
functions as an intracellular antagonist of some TGF-beta PT, Lovell-Badge R, Stern CD (1997) Preventing the loss
family members during Xenopus embryogenesis. Genes of competence for neural induction: HGF/SF, L5 and Sox-2.
Cells 3:387394. Development 124:11911202.
Pera E, Ikeda A, Eivers E, de Robertis EM (2003) Integration of Streit A, Stern CD (1999) Establishment and maintenance of the
IGF, FGF and anti-BMP signals via Smad1 phosphorylation border of the neural plate in the chick: involvement of FGF
in neural induction. Genes Dev 17:30233028. and BMP activity. Mech Dev 82:5166.
Piccolo S, Sasai Y, Lu B, De Robertis EM (1996) Dorsoventral Uchikawa M, Ishida Y, Takemoto T, Kamachi Y, Kondoh
patterning in Xenopus: inhibition of ventral signals by direct H (2003) Functional analysis of chicken Sox2 enhancers
binding of chordin to BMP-4. Cell 86:589598. highlights an array of diverse regulatory elements that are
Reem-Kalma Y, Lamb T, Frank D (1995) Competition between conserved in mammals. Dev Cell 4:509519.
noggin and bone morphogenetic protein 4 activities may reg- Uzgare AR, Uzman JA, El-Hodiri HM, Sater AK (1998)
ulate dorsalization during Xenopus development. Proc Natl Mitogen-activated protein kinase and neural specification in
Acad Sci USA 92:1214112145. Xenopus. Proc Natl Acad Sci USA 95:1483314838.
Rex M, Orme A, Uwanogho D, Tointon K, Wigmore PM, Sharpe Voiculescu O, Bertocchini F, Wolpert L, Keller RE, Stern
PT, Scotting PJ (1997) Dynamic expression of chicken Sox2 CD (2007) The amniote primitive streak is defined by
and Sox3 genes in ectoderm induced to form neural tissue. epithelial cell intercalation before gastrulation. Nature 449:
Dev Dyn 209:323332. 10491052.
Sasai Y, Lu B, Steinbeisser H, Geissert D, Gont LK, De Robertis Waddington CH (1932) Experiments on the development of
EM (1994) Xenopus chordin: a novel dorsalizing factor chick and duck embryos cultivated in vitro. Philos Trans R
activated by orgaizerspecific homeobox genes. Cell 79: Soc Lond B Biol Sci 221:179230.
779790. Waddington CH (1933) Induction by the primitive streak and its
Sasai Y, Lu B, Steinbeisser H, de Robertis EM (1995) Regulation derivatives in the chick. J Exp Biol 10:3848.
of neural induction by the Chd and Bmp-4 antagonistic Watanabe Y, Le Douarin NM (1996) A role for BMP-4 in the
patterning signals in Xenopus. Nature 376:333336. development of subcutaneous cartilage. Mech Dev 57:6978.
Sato SM, Sargent TD (1989) Development of neural induc- Wawersik S, Evola C, Whitman M (2005) Conditional BMP
ing capacity in dissociated Xenopus embryos. Dev Biol inhibition in Xenopus reveals stage-specific roles for BMPs
134:263266. in neural and neural crest induction. Dev Biol 277:
Smith WC, Harland RM (1992) Expression cloning of noggin, a 425442.
new dorsalizing factor localized to the Spemann organizer in Wilson SI, Graziano E, Harland R, Jessell TM, Edlund T (2000)
Xenopus embryos. Cell 70:829840. An early requirement for FGF signalling in the acquisition of
Spemann H, Mangold H (1924) Induction of embryonic primor- neural cell fate in the chick embryo. Curr Biol 10:421429.
dia by implantation of organizers from a different species. Wilson PA, Hemmati-Brivanlou A (1995) Induction of epi-
Rouxs Arch Entw Mech 100:599638. Re-published in Int J dermis and inhibition of neural fate by Bmp-4. Nature
Dev Biol 45:1338. 376:331333.
Stern CD (2005) Neural induction: old problem, new findings, Wilson PA, Lagna G, Suzuki A, Hemmati-Brivanlou A
yet more questions. Development 132:20072021. (1997) Concentration-dependent patterning of the Xenopus
Storey KG, Crossley JM, De Robertis EM, Norris WE, Stern ectoderm by BMP4 and its signal transducer Smad1.
CD (1992) Neural induction and regionalisation in the chick Development 124:31773184.
embryo. Development 114:729741. Wilson SI, Rydstrom A, Trimborn T, Willert K, Nusse R, Jessell
Streit A, Berliner AJ, Papanayotou C, Sirulnik A, Stern CD TM, Edlund T (2001) The status of Wnt signalling regu-
(2000) Initiation of neural induction by FGF signalling lates neural and epidermal fates in the chick embryo. Nature
before gastrulation. Nature 406:7478. 411:325330.
Streit A, Lee KJ, Woo I, Roberts C, Jessell TM, Stern CD Wittler L, Kessel M (2004) The acquisition of neural fate in the
(1998) Chordin regulates primitive streak development and chick. Mech Dev 121:10311042.
the stability of induced neural cells, but is not sufficient Zimmerman LB, De Jesus-Escobar JM, Harland RM (1996) The
for neural induction in the chick embryo. Development 125: Spemann organizer signal noggin binds and inactivates bone
507519. morphogenetic protein 4. Cell 86:599606.
Chapter 2

Neurogenesis: A Change of Paradigms

Luiz E. Mello and Beatriz M. Longo

Contents under physiological and pathological conditions from


the self repair and medical perspective. We discuss fac-
2.1 Historical Overview . . . . . . . . . . . . . 12 tors that influence adult neurogenesis and stem cell
2.2 Neurogenesis and Neurogenic Regions . . . . 14 behavior and their potential therapeutic implications.
2.3 Cell Death and Neurogenesis . . . . . . . . . 17
2.4 Neurogenesis and Inflammation . . . . . . . 20 Keywords Adult neurogenesis Cell death
2.5 Stem Cell Therapies for CNS Disorders . . . . 23
2.6 Concluding Remarks . . . . . . . . . . . . 25 Epilepsy Inflammation Stem cell
References . . . . . . . . . . . . . . . . . . . . 26
Abbreviations

Abstract Neurogenesis and neural stem cells in the AD Alzheimers disease


adult brain are relatively recent discoveries in neu- BDNF brain-derived neurotrophic factor
roscience. Since Cajals statement in 1928 it was bFGF basically fibroblast growth factor
conceived that in the adult brain no new neurons BrdU bromodeoxyuridine
were formed. However, along the history of neuro- CXCR4 chemokine receptor 4
science new findings and observations did not fit to the DG dentate gyrus
established paradigm and novel concepts were formed. EGF epidermal growth factor
Thereafter, neurogenesis, a process of generation of EGFP enhanced green fluorescent protein
new neurons was proved to occur in the adult brain. GABA -aminobutyric acid
Later, the existence of stem cells made adult neuroge- GFAP glial fibrillary acidic protein
nesis more plausible, and thus dividing cells leading GFP green fluorescent protein
GDNF glial-derived neurotrophic factor
to new neurons in the adult brains became widely
HD Huntingtons disease
accepted. These two concepts, adult neurogenesis and
hNSC human neural stem cell(s)
neural stem cells, together overturned the so-called
IFN- interferon-gamma
no-new-neurons dogma and shifted the old paradigm.
IGF-1 insulin-like growth factor-1
Currently, adult neurogenesis is largely accepted by IL-1 interleukin-1 alpha
the scientific community, and is a classic example of IL-1 interleukin-1 beta
a long-held scientific theory being brought down. Here IL-6 interleukin-6
we review the most recent findings in neurogenesis and LPS lipopolysaccharide
neural stem cell and their implication in brain function NeuN neuronal nuclear antigen
NPC neuroprogenitor cell(s)
NPY neuropeptide Y
PD Parkinsons disease
L.E. Mello ()
Departamento de Fisiologia, Universidade Federal de So Pilo pilocarpine
Paulo, So Paulo, Brazil POMC pro-opiomelanocortin
e-mail: lemello@unifesp.br RMS rostral migratory stream

H. Ulrich (ed.), Perspectives of Stem Cells, 11


DOI 10.1007/978-90-481-3375-8_2, Springer Science+Business Media B.V. 2010
12 L.E. Mello and B.M. Longo

SDF1 stromal cell-derived factor-1 overturned the so-called no-new-neurons dogma and
SE status epilepticus shifted the old paradigm.
SGZ subgranular zone With respect to Cajals studies in 1928, with the
SRMS spontaneous recurrent motor seizures technology available at that time, he could not distin-
SVZ subventricular zone guish newly formed neurons from neurons that were
TLE temporal lobe epilepsy already there since birth in adult brain. Cajal never
TNF- tumor necrosis factor-alpha found evidence whether neurons could divide, and he
was right. Even today it remains established (at least
for the moment) that neurons do not divide.
The first evidence that the adult brain contained
2.1 Historical Overview dividing and proliferating cells with neural phenotypes
was reported by Joseph Altman in the 1960s (Altman,
Thomas Kuhn, in his book The Structure of Scientific 1962, 1963; Altman and Das, 1965). Although histori-
Revolutions (1962) stated that a scientific paradigm is a cally the first suggestion of the existence of dividing
set of practices that define a scientific discipline during cells in the adult CNS was raised by Hamilton (see
a particular period of time. Kuhn described a paradigm Trujillo et al., 2009 for review), no credits were given
shift as a change in basic assumptions within the rul- to him, and the nonexistence of new postnatal neurons
ing theory of science, and wrote Successive transition became a central dogma in neuroscience for almost
from one paradigm to another via revolution is the a century. Autoradiography with 3 H-thymidine con-
usual developmental pattern of mature science. tributed for the advances in this area. 3 H-thymidine
By the end of the nineteenth century, the great incorporates into the DNA during the S phase of
anatomist Ramon y Cajal, together with other main fig- the cellular cycle, thus enabling cell division to be
ures of that time, Koelliker and His, proposed that the visualized by autoradiography, and demonstrating that
structure of the adult mammalian CNS remains fixed progenitor cells divide and their progeny shows neural
and constant (Gross, 2000). phenotypes. Altman combined intracranial injection of
In his 1928 paper, Cajal wrote: Once development tritiated thymidine with brain lesions in rats, and found
was ended, the fonts of growth and regeneration of labeled neurons and neuroblasts suggesting the possi-
the axons and dendrites dried up irrevocably. In the bility of proliferation of cells and differentiation onto
adult centers, the nerve paths are something fixed and neurons in adult rats. In his paper he wrote: It is
immutable: everything may die, nothing may be regen- commonly stated that in higher vertebrates neogene-
erated. (Ramon y Cajal, 1928). This suggestion was sis of nerve cells is restricted to the early stages of
well accepted and since then, it was thus created a embryonic development. This belief is based on the
paradigm that in the adult brain no new neurons were observation that neurons with mitotic figures are absent
formed. in the central nervous system of higher vertebrates.
However, along the history of neuroscience new However, this does not definitely rule out the neogene-
findings and observations did not fit to the estab- sis of neurons in the adult, for new neurons might arise
lished paradigm and new concepts were formed. from nondifferentiated precursors, such as ependymal
Neurogenesis and neural stem cells in the adult brain cells. . . . If the general observation is valid that mitotic
are relatively recent discoveries in neuroscience. The figures are absent in the brain of adult mammals,
former is a process of generation of new neurons in these findings might suggest that the labeled neurons
the CNS of adult brain (Kempermann, 2006), and the were formed from undifferentiated cells which divided
latter refers to the presence of undifferentiated cells in mitotically during the period at which the adminis-
the adult brain that show the properties of undergoing tered thymidine-H3 was available. The presence of
unlimited self-renewal and the potency to generate at labeled neuroblasts, mostly in fiber tracts, would sup-
least two different cell types (McKay, 1997; van der port such a process of neurogenesis (Altman, 1962).
Kooy and Weiss, 2000; Weissman et al., 2001). Adult Altman was very careful in interpreting his findings
neurogenesis originates from undifferentiated (precur- and was aware of the false impression of neuronal
sor) cells in the adult brain. In this logic, these two con- labeling (Nottebohm, 1985). At that time, the concept
cepts, adult neurogenesis and neural stem cell, together of stem cell in the adult brain did not yet exist, causing
2 Neurogenesis: A Change of Paradigms 13

adult neurogenesis to remain an issue that was hard to synthetic analog bromodeoxyuridine (BrdU). BrdU is
sustain. a halopyrimidine used therapeutically as antiviral and
Fifteen years later, a series of electron microscopy antineoplastic agent (Begg et al., 2000; Freese et al.,
studies by Michael Kaplan demonstrated the neuronal 1994). BrdU labeling was developed as an alterna-
nature of the cells labeled with tritiated thymidine in tive approach for determining the proliferative index
the hippocampus, the olfactory bulb, and the visual of tumors (Hoshino et al., 1989; Struikmans et al.,
cortex (Kaplan and Hinds, 1977). In these labeled cells 1997), and was introduced for studying cell prolif-
they identified somatic synapses and small neurites eration in the developing brain by Nowakowski and
which are typical of neural cells (Kaplan, 1981, 1985; colleagues (1989). George Kuhn, working with Fred
Kaplan and Bell, 1984). Gage (1996), replaced tritiated thymidine by BrdU and
Although well documented in these previous stud- double-labeled cells to investigate the newly born cells
ies, neurogenesis could not be confirmed in rhesus in the adult hippocampus.
monkeys. Pasko Rakic in 1984, did not find radiola- Two areas of the adult mammalian brain are con-
beled neurons in the adult brain of rhesus monkeys, sidered as neurogenic regions, the subgranular zone
and concluded that all neurons of the rhesus monkey of hippocampal dentate gyrus and subventricular zone
brain are generated during prenatal and early postna- (see details about these neurogenic regions in the
tal life (Eckenhoff and Rakic, 1984). Based on this, next section). With these techniques (3 H-thymidine
Rakic speculated that, differently of rodents, a stable and BrdU), several laboratories confirmed the pres-
population of neurons in primates may be important ence of new neurons in these regions of the adult
for the continuity of learning and memory over a life- brain. They reported adult neurogenesis to be modu-
time (Eckenhoff and Rakic, 1988). In 1999, however, lated by a broad range of physiological, experimental
also Rakic published evidence for neurogenesis in the and pathophysiological conditions, in both rodents and
dentate gyrus of adult primates (Kornack and Rakic, primates.
1999). Factors that increase neurogenesis include dietary
In 1981, Fernando Nottebohm published an elegant restriction (Lee et al., 2000), enriched environments
paper named A brain for all seasons: cyclical anatom- (Barnea and Nottebohm, 1994; Kempermann et al.,
ical changes in song control nuclei of the canary 1998; Nilsson et al., 1999; van Praag et al., 1999),
brain (Nottebohm, 1981), followed by a series of hippocampal-dependent learning-tasks (Gould et al.,
studies on adult neurogenesis in songbirds (Goldman 1999a; Lemaire et al., 2000), and voluntary physical
and Nottebohm, 1983; Paton and Nottebohm, 1984; activity (van Praag et al., 1999; Kronenberg et al.,
Nottebohm, 1985; Burd and Nottebohm, 1985; 2003). However, other studies showed that cell pro-
Nottebohm, 1996). Interestingly, Nottebohms studies liferation decreases with age (Altman and Das, 1965;
on the canary system of singing demonstrated that the Seki and Arai, 1995; Kempermann et al., 1998) and
production of new neurons was related to the song can also be reduced by stressful experiences (Gould
learning. These contributions corroborated with the et al., 1997, 1998; Tanapat et al., 2001) related to
idea that new functional neurons with a clear behav- glucocorticoid levels, which are released in response
ioral function could be generated in the adult brain in to stress, suggesting that stress-induced inhibition of
the learning context. cell proliferation is mediated by glucocorticoids (see
During the 1990s, with the advances of new Gould and Gross, 2002, for an overview). There is also
techniques, a considerable number of experiments some evidence that the decrease in the number of new
demonstrated the presence of newborn neurons in the neurons affects performance in some hippocampal-
adult mammal brain. Confirmation of adult neuro- dependent learning-tasks (Shors et al., 2001, 2002) or
genesis came with important studies combining the the acquisition in the water maze task (Kempermann
3 H-thymidine labeling method with cell type-specific and Gage, 2002). Reduced neurogenesis might con-
markers for immunohistochemical identification of the tribute to major depression (Santarelli et al., 2003;
phenotype of newly generated cells (Cameron et al., Kempermann and Kronenberg, 2003).
1993; Okano et al., 1993; Seki and Arai, 1993). The Similar studies showed that various trophic factors,
other important contribution in this area was the use neurotransmitters and drugs modulate neurogenesis
of the immunohistochemicaly detectable thymidine in the dentate gyrus and subventricular zone (Craig
14 L.E. Mello and B.M. Longo

et al., 1996; Kuhn et al., 1997; Brezun and Daszuta, potential of generating neurospheres in the presence of
1999; Malberg et al., 2000; Jin et al., 2006). Using growth factors.
immunohistochemical detection of markers of the cell The existence of stem cells, thus, made adult neuro-
cycle or BrdU incorporation, it has been demonstrated genesis more plausible and thus dividing cells leading
that neurogenesis increased in the subventricular zone to new neurons in the adult brains became widely
of Huntingtons disease (HD), and in the hippocam- accepted. Finally, adult neurogenesis has been largely
pus of Alzheimers disease (AD) brains (Curtis et al., accepted by the scientific community, and is a classic
2003; Jin et al., 2004; Tattersfield et al., 2004). Newly example of a long-held scientific theory being brought
formed neurons are also increasingly found in the hip- down.
pocampus and subventricular zone of epileptic animals
(Parent et al., 1997), in strokes (Liu et al., 1998) and
traumatic brain injuries (Dash et al., 2001).
By the end of 1990s, some authors started to sug- 2.2 Neurogenesis and Neurogenic
gest that adult neurogenesis could also occur in brain Regions
areas other than hippocampus and subventricular zone
(Shankle et al., 1999; Gould et al., 1999b; Zhao et al., Adult neurogenesis and stem cells are firmly asso-
2003), although others, including Rakic (2002), have ciated to one another. To better understand the phe-
questioned the scientific evidence of these findings. nomena of neurogenesis in the adult brain, the stem
Neurogenesis had also been described in the dentate cell concept should be considered. A stem cell is an
gyrus of human aged subjects (Ericksson et al., 1998). undifferentiated (unspecialized) cell that has two main
Newborn neuronal cells immunopositive for BrdU and properties: the ability to divide (self-renewal) infinitely
neuronal nuclear antigen (NeuN), a marker of mature (or seemingly so), and the potential to give rise to
neurons, were identified in the human adult dentate specialized cell types (multipotency). Conceptually,
gyrus of postmortem tissue samples obtained from the immediate progeny of stem cells are the pro-
cancer patients who had been administered with the genitor cells that in turn generate differentiated cells
drug as part of their treatment. In 2002, van Praag (Kempermann, 2006). To ensure self-renewal, stem
and coworkers used retrovirus labeled with the green cells undergo two types of cell division: symmetric
fluorescent protein (GFP) to visualize newly gener- division, that gives rise to two identical daughter cells
ated cells in living tissue slices and were thus able to with the same stem cell properties, and asymmet-
confirm that newborn neurons in the adult rodent hip- ric division (Fig. 2.1a), that produces only one stem
pocampus become functional granule cells (van Praag cell and a progenitor cell with limited self-renewal
et al., 2002). The occurrence of neurogenesis in adult potential. Progenitors can go through several rounds
rodents was also confirmed by BrdU and retroviral of cell of asymmetric division leading to progenitor
labeling in other studies (Seki and Arai, 1993; Corotto cell and a specialized cell (Fig. 2.1b) before terminally
et al., 1993; Kuhn et al., 1996; Yamada et al., 2004). differentiating into a mature cell.
Neural stem cells in turn were characterized in the The adult stem cell is an undifferentiated cell that is
adult brain by Brent Reynolds and Sam Weiss, by found in a differentiated (specialized) tissue, which can
Perry Barlett and colleagues and by Fred Gage and col- renew itself for a lifetime. Adult stem cells are asso-
leagues in the early 1990s (Reynolds and Weiss, 1992; ciated with most tissues of the body probably as part
Richards et al., 1992; Kilpatrick et al., 1993; Kilpatrick of a tissue/cell renewal mechanism. Sources of adult
and Barlett, 1995; Palmer et al., 1995; Ray et al., stem cells have been found in almost all areas of the
1995). Weiss and Reynolds observed the formation of body including bone marrow, blood stream, cornea and
spheres in cell cultures from adult mice striatum grown retina of the eye, the dental pulp of the tooth, liver,
in suspension (Reynolds and Weiss, 1992). The authors skin, gastrointestinal tract, and the pancreas. Indeed, it
called these structures neurospheres, and suggested seems that the discovery of more sources of adult stem
they derived from the adult brain cell population able cells in the body is only a question of time.
to divide dynamically. The potential of self-renewal Traditionally, adult stem cells are primarily multi-
and the differentiation of stem cells could be observed potent, meaning that they appear to be more limited
in neurospheres in vitro, which in turn, reflected the in their differentiation potential and to be committed
2 Neurogenesis: A Change of Paradigms 15

Fig. 2.1 True neural stem


cells provide slow occurring
unlimited self-replication in
asymmetric divisions that
produces progenitor cells
with limited self-renewal
potential and another stem
cell (a). Neural progenitor
cells are already committed to
a neural lineage
(compromised cells) leading
to specialized cells terminally
differentiating into mature
cells

to their tissue of origin, as compared to embryonic with even a smaller number assuming neuronal char-
stem cells, which are able to give rise to all cell types acteristics suggesting the presence of a specific genetic
of the body. This view was dropped into question program of these cells of non-neural origin. In view
by a number of studies that seemed to suggest that of that their progeny and, as a consequence, their
stem or progenitor cells from one organ could cross cellular identity is somewhat limited (Avots et al.,
limits of tissue specificity and generate differentiated 2002).
cells in other organs, implying that adult stem cells Stem cells can be found in the adult brain. These
have a wider potential for differentiation (transdiffer- neural stem cells (NCS) are self-renewing, multipotent
entiation or plasticity) than previously assumed. For cells that generate the main phenotypes of the nervous
example, studies have shown the generation of blood system, including neurons and glial cells (astrocytes,
cells from neural stem cells and of neurons and glia oligodendrocytes) (McKay, 1997; van der Kooy and
from hematopoietic stem cells (Brazelton et al., 2000; Weiss, 2000; Weissman et al., 2001).
Mezey et al., 2000). Moreover, Purkinje neurons origi- Adult neurogenesis is defined as the process of gen-
nated from transplanted bone marrow cells were found erating new neurons in the adult brain, after fetal and
in the cerebellum one year after bone marrow trans- early postnatal development has ceased. The process
plantation (Priller et al., 2001; Weimann et al., 2003). of adult neurogenesis comprises many steps, from the
Likewise, in the brains of female patients who had division of a stem cell, called precursor cell, to the
received a bone marrow graft from male donors, brain full integration of the new neurons into the working
autopsy results revealed neurons carrying the Y chro- brain. It involves decisions at the precursor level, such
mosome (Mezey et al., 2000; Cogle et al., 2004). In as whether symmetric or asymmetric divisions occur
our hands, cells originating from the bone marrow have before become a neuron.
the capacity to migrate proliferate and populate brain A brain region that can generate neurons is called
regions in epileptic chimeric animals (Longo et al., neurogenic region, a term that implies the pres-
2005). However, in all of these studies the experimen- ence of immature precursor cells and more importantly
tally observed plasticity of adult stem cells occurs in a microenviroment that is permissive for neuroge-
a very low frequency in terms of numbers of cells, nesis to occur. New neurons are continually born
16 L.E. Mello and B.M. Longo

throughout adulthood in predominantly two regions of The SVZ, also known as ependymal region, con-
the mammalian brain: tains at least four different cell types defined by their
morphology, ultrastructure, and molecular markers.
The subventricular zone (SVZ), where the new Glial fibrillary acidic protein (GFAP)-expressing astro-
cells migrate to the olfactory bulb via the rostral cytic cells act as the primary stem cells in the adult
migratory stream (Fig. 2.2a,b). SVZ (Doetsch et al., 1997). The authors of this work
The subgranular zone (SGZ), part of the dentate named these astrocytes as B cells. Another type, a
gyrus of the hippocampal complex, where new transiently amplifying progenitor cell (C cell), origi-
granule cells are produced (Fig. 2.2c,d). nates from B cells and, in turn, give rise to immature
neurons or neuroblasts (A cells). A cells are migra-
For these two regions several types of progenitor tory cells passing through the rostral migratory stream
cells were identified. (RMS) and reaching the olfactory bulb, where they dif-
In the adult SVZ, new neurons are continuously ferentiate into interneurons. Type C cells are highly
produced suggesting that neural stem cells persist proliferative precursors and form clusters next to the
within this germinal layer (Alvarez-Buylla and Garca- chains of migrating A cells. The SVZ is largely sepa-
Verdugo, 2002). The SVZ is defined as a one- or two- rated from the ventricle cavity by a layer of ependymal
cell body wide zone below the ependyma, lining the cells (type E cells). B cells interact closely with E cells
lateral ventricles, adjacent to striatum (Kempermann, and occasionally contact the ventricle lumen. Ongoing
2006). In the adult olfactory system, the population replication of B and C cells suggests that one or both
of precursor cells is found in the SVZ in the tempo- of these cell types could be involved in the generation
ral walls of the lateral ventricles. Progenies migrate of the new neurons, the A cells (Garca-Verdugo et al.,
over long distances along the rostral migratory stream 1998; Lois et al., 1996; Doetsch and Alvarez-Buylla,
to the olfactory bulb, where they differentiate into 1996). Because A cells, themselves, divide (Lois and
interneurons in the granule and periglomerular lay- Alvarez-Buylla, 1994; Menezes et al., 1995; Thomas
ers. The olfactory system also comprises the olfactory et al., 1996), it was also formally possible that they
epithelium, which is part of the peripheral nervous sys- simply generate more A cells. As many cell types in
tem and in which massive adult neurogenesis occurs. the subventricular zone incorporate 3 H-thymidine in

Fig. 2.2 The two


neurogenic regions in the
brain, where the presence of
immature precursor cells
and a microenviroment
permissive allow for
neurogenesis.
Immunofluorescence for
BrdU stained cells in (a) and
(b) subventricular zone
(SVZ), where the new cells
migrate to the olfactory bulb
via the rostral migratory
stream; and in (c) and (d) the
subgranular zone (SGZ), part
of the dentate gyrus of the
hippocampal complex, where
new granule cells are
produced
2 Neurogenesis: A Change of Paradigms 17

adult mice, it is believed that more than one cell type vivo neurogenesis observed in the adult dentate gyrus
is dividing in this region. There is some controversy, is thus currently considered as derived from a neu-
however, related to the identity of the stem cells that ronal progenitor population and not from true stem cell
could be E cells (Johansson et al., 1999) or type B population.
astrocytes (Doetsch et al., 1999).
For several decades, evidence identifying the hip-
pocampus as a region with a large degree of structural
plasticity has been accumulated. Investigations were 2.3 Cell Death and Neurogenesis
initially focused on exploring the organization of the
hippocampal formation, proposing that the hippocam- Similarly to developmental neurogenesis, adult neuro-
pus is constantly changing under normal conditions. genesis is also followed by significant apoptotic cell
According to these studies, the hippocampal forma- death. During adult neurogenesis, neuronal progenitor
tion is a dynamic area whose synapses and dendrites cells present in neurogenic areas divide and migrate to
are undergoing continuous rearrangement (see Gould, their appropriate destination, while differentiating into
2007). their mature phenotypes and connecting to their target
In the adult hippocampus, neural precursor cells cells. These new neurons need support from neighbor-
reside in a narrow band of tissue in the border zone ing glial cells and nutrients from blood to survive and
between the granule cell layer and the hilus, the sub- become part of the working brain. Importantly, as a
granular zone (SGZ). SGZ is defined as a layer about vital condition, they need to make connections with
three cell nuclei wide, including the basal cell band of other neurons. This process also includes migration
the granule cell layer and a two nucleus-wide zone in and differentiation of the progeny, and proliferation
the hilus (Kempermann, 2006). The progeny of these processes (dendrites and axons establishing connec-
continuously dividing cells migrate varying distances tions and forming synapses), and finally the presence
into the granule cell layer where they differentiate of sodium currents and the ability to generate action
(Altman and Das, 1965; Cameron et al., 1993; Kuhn potentials. Without these connections, the new neu-
et al., 1996; Palmer et al., 2000). They then extend rons shrink and die. In fact, many of these newborn
their dendrites, as all other granule cells do, into the cells die shortly after their birth (being not much differ-
molecular layer and send axons along the mossy fiber ent from the process taking place during development,
tract to the CA3, forming together with the hilus the except for the massive scale of the latter), while the
projection area of granule cells. surviving cells become functionally integrated into the
As in the SVZ, the putative stem cells of the surrounding brain tissue.
adult SGZ reveal a characteristic morphology resem- It has been suggested that adult neurogenesis reca-
bling radial glia and astrocytic properties (Seri et al., pitulates events occurring during embryonic develop-
2001). Seri and colleagues demonstrated that BrdU ment (Kintner, 2002). In this view, newly produced
labeled astrocytes after antimytotic treatment in the neurons in adult brain may die for the same reasons as
hippocampus generated neurons, astrocytes and oligo- embryonic neurons do during development, resulting
dendrocytes. Thus, astrocytes from the SGZ were able from loss of trophic support and lack of connections.
to generate the three main types of cells in the brain, Neurons that are not integrated in the neuronal network
exhibiting characteristics of multipotentiality of stem are not functional and thus are removed by apopto-
cell. sis. Since the generation and integration of these new
Seaberg and Van der Kooy (2002), however, ques- neurons takes time, it seems reasonable and a good
tioned whether the hippocampus could indeed contain strategy to have some of them in stock all the time. In
stem cells. They speculated that the detection of stem case of necessity they can be integrated fast. If they
cell in the primary precursor cell cultures derived from are not anymore needed, cells die and are replaced
the hippocampus could in fact be due to stem cell by new ones (Kempermann et al., 2004). Although
contamination from the ventricular wall. It may also in the adult brain the large amount of the dying cells
be possible that the discrepancy resulted from differ- are immature cells, the mature neurons generated dur-
ences in cell culture and isolation protocols, leading ing development also undergo apoptosis (Buss et al.,
to the enrichment of different cell population. The in 2006; Kempermann, 2006). Coincidently, of all adult
18 L.E. Mello and B.M. Longo

brain regions the neurogenic zones show the high- patients with PD (Madrazo et al., 1987, 1988). The
est incidence of apoptotic cell death, being about a studies by these and various other groups indicated that
hundred times higher than in the rest of the brain transplanted cells from a number of different sources
(Blaschke et al., 1996; Biebl et al., 2000), suggesting can replace dopaminergic neurons damaged in PD,
that these two processes, apoptosis and neurogene- thereby restoring and regenerating brain function.
sis are strongly linked. Moreover, in these neuro- In the hippocampus, the link between neurogen-
genic zones the location of apoptotic profiles coincides esis and cell death has been well documented (see
exactly with the area where dividing progenitor cells Gould in Andersen, 2007) as observed in studies
reside, as observed in cells double-immunostained for with adrenalectomy (Gould et al., 1991; Cameron
apoptotic profiles and immature neuronal cells (Kuhn and Gould, 1994), mechanical or excitotoxic lesions
et al., 2005). In the rodent brain, it was demonstrated (Gould and Tanapat, 1997), ischemia and seizures
that approximately 70% of newly produced DG cells (Parent et al., 1997; Scott et al., 2000; Kee et al.,
and approximately 50% of olfactory bulb cells undergo 2001; Kokaia and Lindvall, 2003; Scharfman, 2004),
apoptosis within 30 days of their birth (Biebl et al., all inducing cell death and proliferation of granule
2000; Kempermann et al., 1997). cell precursors. Hastings and Gould (2003) suggested
Different views have been proposed for the rela- that cell proliferation is enhanced with neuronal dam-
tionship between these two events (adult neurogenesis age by two alternative mechanisms: dying neurons
and apoptosis). In fact there are at least two possi- can release molecules that stimulate proliferation of
bilities for this interconnection. One is that already precursor cells; or neurons in the intact brain release
mentioned whereby apoptosis allows the selection of molecules that suppress cell proliferation. This inhi-
cells generated in excess (neurogenesis followed by bition is removed after damage. This hypothesis is
apoptosis). The other is the condition in which apopto- reinforced by other lines of arguments based on the
sis is followed by neurogenesis as a sort of replacement balance between the birth and death of granule cells
strategy. Magavi and colleagues (2000) showed that (Gould et al., 1991), by the fact that proliferation in
degeneration-induced apoptosis in layer VI of the ante- the SVZ is upregulated under inflammatory condi-
rior cortex induces proliferation of endogenous neural tions (Calza et al., 1998), and by the observation that
precursors. These precursors showed the ability of dif- the neurogenic structures do not grow in size. There
ferentiation into neurons from the damaged region, and is however evidence for the contrary indicating that
these new neurons were able to reestablish appropri- between 3 and 12 months of age (a life period dur-
ate cortical-thalamic connections. As proposed by the ing which a mouse is considered an adult) the mouse
authors, the induction of highly selective apoptotic cell dentate granule cell layer incorporates some additional
death in individual cells could trigger neurogenesis. 300,000 new neurons (Bayer, 1982; Bayer et al., 1982;
A number of different combinations of both strategies Kaplan and Hinds, 1977) and that young adult rats
could take place given that apoptosis and neurogenesis have 9,400 dividing cells in the dentate proliferating
may in addition to the above represent different strate- with a cell cycle time of 25 h (Cameron and Mckay,
gies adapt to changing conditions. Furthermore it must 2001). Therefore the balance between loss and incor-
be considered that no process occurs in isolation and poration of new neurons is not always maintained both
one may affect the other. in physiological and pathological conditions.
The fact that neurogenesis might ensue after nat- Indeed, it has been shown that hippocampi of
urally occurring or more obvious and widespread patients with epilepsy show remarkable structural
lesional phenomena has clear implications for cell changes of the dentate gyrus that have been initially
replacement therapies. Nowhere has this been devel- labeled granule cell dispersion and more recently tec-
oped as much and as long as in treatment of tonic layer displacements (Houser, 1990; Sloviter
Parkinsons disease (PD) by Sweden scientists et al., 2004). This issue itself has been the subject of
(Lindvall et al., 1990; Bjrklund and Lindvall, 2000). a certain degree of debate regarding whether seizures
Yet the pioneering studies on the subject came from the are cause or consequence of the cell derangement and
efforts by Madrazo in Mexico, who showed improve- cell proliferation or the reverse (or both). The fact that
ment in PD symptoms after transplanting adrenal and adult rodents (Mello et al., 1992; Gray and Sundstrom,
fetal substantia nigra cell extracts into the striatum of 1998; Rougier et al., 2005) and adult primates (our own
2 Neurogenesis: A Change of Paradigms 19

unpublished observations) subject to status epilepticus areas such as the dentate gyrus, there is a sixfold
(SE) subsequently develop these altered hippocampal increase in proliferation rate (Bengzon et al., 1997)
profiles speaks for the possibility that these may also and thus neurogenesis has been shown to be induced
be a consequence of seizures. It may thus be that the independent of cell death (Ferland et al., 2002). In
adequate balance between loss and birth of neurons the kainate-induced SE mouse model, neurogenesis
in the hippocampus is not only valid for all species largely exceeds dentate cell loss as evidenced by the
but in addition is perturbed in neurological conditions marked increase in the area occupied by the dentate
and that the excess of neurons contributes to disease cell layer (Suzuki et al., 1995; Gray and Sundstrom,
progression. 1998; Bouilleret et al., 1999). Similarly, a comparative
A good example of compensatory neurogenesis study using both the kainate and pilocarpine mod-
replacing older neurons by new ones, renewing neu- els of SE, Covolan and Mello (2000) suggested that
ral circuits, is the seasonal programmed cell death and increased proliferation in seizures and epilepsy is a
neurogenesis of high vocal center neurons in songbirds function of stimulating available progenitors by means
(Nottebohm, 2004). As pondered by Nottebohm, there of seizures without providing too much excitation and
is little evidence that adult neurogenesis contributes ending up killing such cells. Therefore, similarly to
to neuronal turnover in neurogenic regions. It seems a host of naturally occurring conditions that stim-
unlikely that animals in nature should have time to ulate neural proliferation and neurogenesis, seizures
recover from brain dysfunction by means of neuroge- and epilepsy constitute powerful means to stimulate
nesis process. Nottebohm suggested that neurogenesis this phenomenon (Fig. 2.3). To this end, experimen-
acts as source of neuronal replacement to keep neu- talmodels of epilepsy became widely used as a strategy
ronal circuits functionally young (Nottebohm, 2002). for studying the genes and relevant molecules that
Accordingly, Kempermann proposed the hypothesis regulate adult neural proliferation and neurogenesis
that brain development never ends and that plasticity (Elliott et al., 2003; Altar et al., 2004; Hagihara et al.,
can be taken as continued development (Kempermann 2005; Pirtilla et al., 2005; Wang and Baraban, 2007;
et al., 2004). In the hippocampus, new neurons are Choi et al., 2008 Steiner et al., 2008). Using pro-
added to the existing networks and to the dentate gyrus opiomelanocortin-enhanced green fluorescent protein
throughout life. (POMC-EGFP) transgenic mice to identify newborn
Epilepsy, of all the neurological disorders, is the granule cells, the group of Linda Overstreet Wadiche
only condition where there is evidence that more neu- found that the seizure activity not only increased the
rogenesis may act as to increase rather than merely number of newborn granule cells, but it also dramati-
replace dying neurons. Indeed, dentate granule cell cally altered their dendritic morphology (see Zhao and
dispersion shown in humans (Houser, 1990; Houser Overstreet-Wadiche, 2008). Labeling mitotic cells with
et al., 1990), initially suggested as a predisposing BrdU 2 days before SE and quantifying 2 weeks later,
developmental event, was later shown to be induced
in adult rodents by status epilepticus (SE) (Mello
et al., 1992) and to result from increased proliferation
rate (Parent et al., 1997). The relative contribution of
this event to the ensuing mossy fiber sprouting that
later develops in the rodent models of SE was subse-
quently discarded given that X-ray irradiated rodents,
in which dentate neurogenesis is impaired, still have
robust mossy fiber sprouting (Parent et al., 1999). The
issue of whether SE-induced neurogenesis is finely
adjusted as a compensatory phenomenon or generates
an excess of cells has been addressed in several stud-
ies (Mohapel et al., 2004; Covolan and Mello, 2000;
Fig. 2.3 Schematic view of neural proliferation and cell loss
Zhao and Overstreet-Wadiche, 2008). Indeed, in the
in epilepsy as a function of seizure intensity. The dashed line
kindling model of seizures where cell loss is mini- represents the resultant outcome (net effect) of proliferation and
mal, and may even be non existent in some resistant cell loss
20 L.E. Mello and B.M. Longo

they also suggested that preexisting newborn granule neuropeptide Y (NPY) and brain-derived neurotrophic
cells as well as seizure-induced granule cells undergo factor (BDNF) (Kuhn et al., 1997; berg et al., 2003;
abnormal dendrite development during epileptogene- Benraiss et al., 2001; Pencea et al., 2001; Scharfman
sis. The authors concluded that seizures accelerated et al., 2002; Hansel et al., 2001; Howell et al., 2003).
the dendritic development of newborn granule cells Neurotransmitters, hormones, and psychotropic agents
altering the wiring of individual cells into the existing have been also shown to influence neurogenesis in the
circuit. adult brain (see Parent, 2003 for review).
Not all developmentally triggered protein expres- An interesting aspect is the correlation between
sion is associated with seizure-induced proliferation neurogenesis and inflammation caused by insults like
and migration process in the dentate gyrus (Mello ischemia, trauma and seizures (Makowiak et al.,
and Mendez-Otero, 1996), and it has even been sug- 2004; Bernardino et al., 2005; Vezzani, 2005; Ekdahl
gested that granule cell dispersion is not accompanied et al., 2009). There is a growing body of evidence
by enhanced neurogenesis in temporal lobe epilepsy indicating that inflammation in the CNS can either pro-
(TLE) patients (Fahrner et al., 2007). It has been shown mote or inhibit adult neurogenesis (Simard and Rivest,
in animal models of TLE that migration is disturbed, 2004; Ekdahl et al., 2009; Bernardino et al., 2005).
leading to ectopic granule cells near the CA3 region A possible molecular explanation for these findings is
(Scharfman et al., 2000). Thus, careful understanding that the nervous and immune systems are integrated
of how much of the seizure-induced neurogenic pro- in an intense crosstalk (Kerschensteiner et al., 2009).
cesses recapitulate ontogeny is important for the poten- Both systems produce a range of overlapping factors
tial of using electrical stimulation (or even seizures) as that modulate cell growth and differentiation, such as
part of therapeutic strategies. cytokines and chemokines released by the immune sys-
It has been considered that epileptic seizures repre- tem and neurotrophic and growth factors by the CNS,
sent one of the most severe stimulatory stresses that the although neither cytokines nor neurotrophic factors are
brain is exposed to, and that generalized status epilep- completely exclusive to either system (Kerschensteiner
ticus represents a very severe form of seizure (Kulkarni et al., 2003). Expression in the nervous system has
and George, 1995). The participation of the newly born now been described for cytokines of many families
cells in network reorganization and in the chronicity including interleukins, interferons and members of the
of epileptic seizure is still unclear. The presence of tumor necrosis (TNF) family (Neumann et al., 1997;
newly born cells (in normotypic and ectopic locations) Knoblach et al., 1999; Krumbholz et al., 2005; Liu
is systematically observed in the different models of et al., 2007).
epilepsy. The mechanisms by which seizure activity The immune-competent cells of the CNS are the
stimulates neurogenesis are still unknown. It remains microglia, also considered as part of a very elabo-
unclear to what degree the neurogenic response to rate innate immune system and one of the cell types
seizures is in attempt at regeneration or part of the that constitute the stem cell niche (Simard and Rivest,
pathology itself. 2004; Kempermann, 2006). Microglia are recruited
by many pathological stimuli in the defense of the
neural parenchyma against infection, inflammation,
ischemia, trauma, brain tumors and neurodegenera-
2.4 Neurogenesis and Inflammation tion. They contribute to the immune response by acting
as antigen presenting cells, cleaning, unwanted debris
Adult neurogenesis can be profoundly modulated by phagocytosis, as well as secreting factors associ-
by several environmental stimuli, trophic factors, ated with inflammation, such as cytokines and other
cytokines, drug treatments, and various physio- and signaling molecules. Over the last few years the role
pathological conditions. It is well established that of microglia in adult neurogenesis in the intact and
specific growth or neurotrophic factors influence neu- injured brain has been discussed. A growing body of
ral precursor proliferation in the adult hippocampus evidence shows that microglia, depending on their state
and SVZ. This group of molecules includes basically of activation, can be either detrimental or supportive
fibroblast growth factor (bFGF), insulin-like growth for neurogenesis acting on different steps in the for-
factor-1 (IGF-1), epidermal growth factor (EGF), mation, maturation and functional integration of the
2 Neurogenesis: A Change of Paradigms 21

new neurons (see Ekdhal et al., 2009; for review). To circumstances can be beneficial and supportive of the
elucidate their neurotoxic and supportive role in adult different steps in adult neurogenesis and gliogenesis
neurogenesis, it is necessary first to understand the (Selmaj et al., 1990; Wu et al., 2000; Arnett et al.,
microglial function in different activation profiles and 2001; Butovsky et al., 2006; Battista et al., 2006)
their associated cytokine. An interesting view is that (see Table 2.1). By the use of TNF--receptor defi-
microglia activation, as an indicator of inflammation, is cient mice, Arnett and colleagues found that the lack
not pro- or antineurogenic per se but the net outcome is of TNF- leads to a delay in remyelination, which is
dependent on the balance between secreted molecules associated with a reduction in the pool of proliferating
with pro- and anti-inflammatory action (Ekdhal et al., oligodendrocyte progenitors followed by a reduction in
2009). the number of mature oligodendrocytes (Arnett et al.,
Cytokines IL-1, IL-1, IL-6, IFN- and TNF- 2001). Moreover, TNF- has been shown to promote
in the CNS have been shown to have a suppres- astrocyte proliferation (Selmaj et al., 1990).
sive effect over neurogenesis that depends on the A beneficial role of microglia in adult neurogen-
degree of activation of the brain resident microglia esis is also supported by in vitro studies on neural
on inflammation (Vallieres et al., 2002; Ekdahl et al., stem cells co-cultured with microglia or grown in con-
2003; Monje et al., 2003; Ben-Hur et al., 2003; ditioned media from microglia (Aarum et al., 2003;
Wong et al., 2004; Heldmann et al., 2005; Cacci Morgan et al., 2004; Walton et al., 2006; Nakanishi
et al., 2005; Iosif et al., 2006; Koo and Duman, et al., 2007). Microglia and microglia-conditioned
2008) (Table 2.1). Vallieres and colleagues have shown medium rescued the in vitro formation of neurob-
that interleukin-6 (IL-6) when expressed over the lasts from SVZ neural stem cells, which otherwise
long term in astroglia of young adult transgenic mice was lost during continued culture (Aarum et al., 2003;
is able to decrease dentate gyrus neurogenesis by Walton et al., 2006). In a recent paper, Cacci and
63% (Vallieres et al., 2002). Other studies demon- colleagues (Cacci et al., 2008), showed that pro-
strated that inflammation caused by lipopolysaccha- inflammatory cytokines interleukin (IL-1, IL-1 beta,
ride (LPS), known to induce several inflammatory IL-6), and TNF- were strongly reduced after chronic
responses, inhibits neurogenesis in the hippocampus stimulation of microglia, as compared to acute stim-
(Ekdahl et al., 2003; Monje et al., 2003). Neurogenesis ulation. The authors suggested that, in a chronically
can be restored by indomethacin, a common nons- altered environment, persistently activated microglia
teroidal anti-inflammatory drug (Monje et al., 2003). can display protective functions that favor rather
Monje and colleagues concluded that cell death under than hinder brain repair processes. They suggested
pathological conditions can be associated with an that acutely activated microglia, or their conditioned
antineurogenic inflammatory response. Interestingly, medium, reduced neural progenitor cell survival, pre-
Wong and colleagues have shown that adult neural vented neuronal differentiation and strongly increased
stem cells are able to respond to the inflammatory glial differentiation, likely through the release of proin-
cytokines interferon-gamma (IFN-) and tumor necro- flammatory cytokines, whereas chronically activated
sis factor-alpha (TNF-), with significant effects on microglia were permissive to neuronal differentiation
neural stem cell function. While TNF- was toxic and cell survival, and still supported glial differenti-
to proliferating neural stem cells, it did not appear ation. Recently, Ziv and colleagues showed that hip-
to affect differentiation. IFN-, on the other hand, pocampal neurogenesis could be restored in mice by
inhibited neural stem cell proliferation and promoted the transfer of CNS-reactive T cells (Ziv et al., 2006).
neuronal differentiation and neurite outgrowth (Wong These regulatory T-lymphocytes were also necessary
et al., 2004). Other studies also showed the inhibitory for the completion of spatial learning and memory
effect on neurogenesis by IL-6, as well as by other tasks and, notably, for BDNF expression in the den-
inflammation-related cytokine products such as IFN- tate gyrus. Another relevant molecule associated to
, interleukin-1beta (IL-1), and TNF- (Heldmann et microglial activation under conditions of inflamma-
al., 2005; Cacci et al., 2005; Iosif et al., 2006; Ben-Hur tory reaction is stromal derived cell factor (SDF1)
et al., 2003; Koo and Duman, 2008). that induces microglia migration by inducing CXC
Interestingly, recent experimental evidence indi- chemokine receptor 4 (CXCR4) expression (Wang
cates that inflammation or microglia under certain et al., 2008). It was furthermore shown that glutamate
22 L.E. Mello and B.M. Longo

Table 2.1 Examples of studies showing the effect of cytokines and other factors on neurogenesis and gliogenesis
Effect on
Cytokine/factor neurogenesis/gliogenesis Animal model Study
IL-6 Decrease SE (pilo) in transgenic mice Vallieres et al. (2002)
expressing IL-6
IL-1 Decrease LPS Monje et al. (2003)
IL-1 /IL-6 Decrease SE and LPS Ekdall et al. (2003)
IL-1 Decrease Stress and NPC culture Koo and Duman
(2008)
TNF-/IFN- Decrease NPC culture Ben-Hur et al. (2003)
TNF- Decrease Middle cerebral artery Heldmann et al. (2005)
occlusion
TNF- Decrease Hippocampal progenitor cell Cacci et al. (2005)
culture
TNF- Decrease TNF-R1/R2-deficient Iosif et al. (2006)
mice/SE
TNF-/ Decrease Neural stem cell (SVZ) Wong et al. (2004)
IFN- Increase proliferation of culture
BIII-tub+ cells
TNF-/IL-6 Proliferation of astrocyte Cell culture Selmaj et al. (1990)
TNF- Increase Exogenous TNF- Wu et al. (2000)
administration in SVZ/VZ
TNF- Oligodendrocyte TNF-deficient Arnett et al. (2001)
progenitor proliferation mice/cuprizone
IL-4/ Increase NPC co-cultured with IL-4, Butovsky et al. (2006)
IFN- oligodendrogenesis IFN-, LPS
Increase neurogenesis
TGF- Increase Adrenalectomy Battista et al. (2006)
IGF-1 Increase Status Epilepticus Choi et al. (2008)
IGF-1 Increase Igf-1 / mice Beck et al. (1995)
IGF-1 Increase Overexpressed Igf OKusky et al. (2000)
IGF-1 Increase transgenic mice Trejo et al. (2001)
IGF-1 Increase Runner rats and IGF-1 Aberg et al. (2003)
infusion
Hippocampal NPC culture
treated with IGF-1
Microglia Increase Microglia culture from Aarum et al. (2003)
embrionic cortical tissue
Microglia Increase Microglial conditioned Morgan et al. (2004)
medium on cerebellar
cultures
Microglia Increase Neural stem cell adherent Walton et al. (2006)
culture

also increased the CXCR4-mediated T cell chemotac- In epilepsy studies, the presence of immunologi-
tic migration in the direction of the localization of the cal dysfunctions after seizures has been extensively
key chemokine stromal cell-derived factor-1 (SDF-1) demonstrated (Bhatt et al., 2003; Cook and Persinger,
(Ganor et al., 2003). SDF1-mediated CXCR4 signal- 1999; Vezzani, 2005). Inflammatory cells have been
ing was also shown to influence rat and human cortical shown to infiltrate the brain parenchyma within
neural progenitor cells (Peng et al., 2004) and to regu- limbic structures after lithium-pilocarpine-induced
late the migration of dentate granule cells (Bagri et al., seizures (Cook and Persinger, 1999). Moreover, after
2002) placing it at a pivotal step as a candidate for amygdala-induced kindling seizures bone marrow pro-
the trophic effects of seizures over neurogenesis and genitor cells were described to proliferate in vitro, sug-
associated phenomena. gesting alterations in immunological functions after
2 Neurogenesis: A Change of Paradigms 23

Fig. 2.4 Expression of


GFP+ /BrdU+ double stained
cells in the hippocampus of
epileptic animals at 24 h, 7
and 24 days after status
epilepticus of chimeric
animals transplanted with
GFP+ bone marrow cells
(kindly provided by S.
Romariz)

chronic seizures (Bhatt et al., 2003). The continuous producing and releasing the cytokines, the functional
traffic of inflammatory cells into the cerebral tissue is state of neurons, local cytokine concentration and
accelerated after injury (Schilling et al., 2003; Vallieres period of tissue exposure, the presence of specific
and Sawchenko, 2003), and is involved in cytokine receptors on target cells, and the presence or absence
response to seizures (Voutsinos-Porche et al., 2004; of modulators of cytokine activity. Indeed, a sus-
Bernardino et al., 2005; Vezzani, 2005). Our own stud- tained expression of specific cytokines has been asso-
ies, using chimeric animals transplanted with bone ciated to chronic TLE. Many authors argued that the
marrow cells, characterized the temporal pattern of cytokines, up-regulated following seizures, may act
migration and distribution of bone marrow cells after through specific receptors in both protective (Albensi,
epileptic seizures (Fig. 2.4). The majority of these 2001; Hamano et al., 2002) and toxic ways (Vezzani
cells could be labeled with BrdU as well as expressed et al., 1999; Eriksson et al., 2000; Vezzani et al., 2000)
microglial markers meaning that the bone marrow to control the pattern of neuronal loss and replacement
cells proliferated within the brain parenchyma. Our associated with seizures. The beneficial or detrimental
evidence is consistent with the hypothesis that these roles of the innate immune response in the epilep-
cells participate in inflammatory reaction that follows tic tissue and its positive or negative modulation on
epileptic seizures (Romariz et al., 2008). seizure-induced neurogenesis still needs to be clarified
As speculated by Bernardino and colleagues, neu- (Vezzani, 2005).
rogenesis that occurs in TLE is under the control
of two opposite driving forces. On one hand, acute
seizures induce differentiation of new neurons. On
the other hand, chronic TLE, inflammation and IL-6 2.5 Stem Cell Therapies for CNS
release contribute to inhibit neurogenesis (Bernardino Disorders
et al., 2005). The authors suggested that pathological
alterations that occurs in chronic TLE can be influ- The CNS, differently from other organs, has a limited
enced by the inflammatory response, and the functional capacity of self repair and self regeneration. Obviously,
role of cytokines in inflammation can shift from ben- these mechanisms are not sufficient for a complete
eficial to detrimental depending on the type of cells recovery facing the intense damage to the CNS in some
24 L.E. Mello and B.M. Longo

cases. From a clinical perspective, it is a consensus injured areas in the brain. Indeed, freshly injected bone
that there is an incredible need for strategies and thera- marrow cells might even be able to block seizures.
pies developed for preventing or reversing neurological In our hands, transplantation of precursor cells only
disturbances. Over the last few years, the therapeutic 30 min before a convulsive stimulation in mice and
potential of stem cells has been suggested as a pos- rats, altered seizure susceptibility, making it more dif-
sible treatment in various pathological conditions of ficult for a seizure to be triggered in these animals.
the CNS. A growing body of evidence shows that stem Given the narrow temporal window between the trans-
cells from different sources, when transplanted into the plant and seizure induction in those experiments, it is
brain, can migrate to the damaged tissue improving likely that this protection is due to a paracrine action
neurological function (Azizi et al., 1998; Kopen et al., of these recently injected marrow cells (Ferrazoli et al.,
1999; Lee et al., 2003), and even differentiate into 2008).
glial (Wernig et al., 2008) and neuronal cells (Wernig This paracrine effect opens the possibility of
et al., 2008; Mezey, 2000)!!! Here we address some neuroregenerative therapies, but important points
of the questions regarding the strategies of therapies should be emphasized before proposing a therapy
based on stem cell to treat neurological diseases. In based on stem cell transplantation. As suggested by
general, stem cell therapy in the CNS can be divided Pluchino and Martino (2005), it is important to be
in two strategies, the transplantation of precursors or aware that relevant issues concerning the transplan-
stem cells, and the stimulation of endogenous neural tation strategies, number of cells, timing and route
progenitor cells. The transplanted cell strategy is based of administration should be elucidated before any
on the replacement potential expecting that these cells therapeutic application.
could migrate, differentiate and integrate, and may be Not less important, questions about the best source
transplanted locally or administered intravenously. The of stem cell to be transplanted and whether trans-
stimulation of endogenous neural progenitor or stem planted cells are functional and integrated to the neu-
cells locally would represent a strategy to promote ronal circuitry should be also considered. Obviously,
regeneration of injured brain (Taupin, 2008). the degree of neural damage and finally, the exper-
Neuroprotection can be also provided by the imental model elected for therapeutic studies should
paracrine effect of stem cell in the lesion site by releas- be adequate to answer these questions. In this regard,
ing cytokine and trophic factors, irrespective of the embryonic stem cells, neural stem cells and bone
chosen strategy, whether transplantation or stimula- marrow cells may constitute the ideal candidates for
tion of progenitor cells (Ruschenschmidt et al., 2005; transplantation therapy in many neurological diseases
Gttinger et al., 2005; Zhao et al., 2004; Borlongan in both clinical and experimental studies (Bossolasco
et al., 2004). Besides triggering differentiation and sur- et al., 2005; Cogle et al., 2004; Chu et al., 2003, 2004;
vival of transplanted cells by autocrine and paracrine Silani et al., 2004; Zhang et al., 2004). Hypothetically,
mechanisms, genetically modified neural stem cells the ideal precursor cell for CNS therapy is of autolo-
secreting trophic and differentiation-inducing factors gous origin and multipotent in terms of its capacity to
can also recruit endogenous neural progenitor cells for give rise to the three cell lineages comprising the CNS
tissue repair (Trujillo et al., 2009). Endogenous cells (neurons, astrocytes and oligodendrocytes), as well as
that proliferate and migrate to the lesioned areas pos- their restricted capacity to differentiate into other cell
sibly exert paracrine effects, rescuing damaged cells in types and their lack of tumorigenic potential (Ziv and
the brain or stimulating neural stem cells and neuro- Schwartz, 2008).
genesis. Obviously for the contribution of these stem One of the criticisms on stem cell therapy and
cells or progenitors from other tissues to brain repair transplantation is based on the suggestion that these
makes it hard to unambiguously demonstrate the con- strategies are less efficacious in chronic neurological
tribution of true neural stem or progenitor cells. Recent diseases, in neurodegenerative processes, as compared
papers have shown that the paracrine action of these to the acute period, during disease onset, indicating
foreign migrated cells into the brain from transplanted a direct correlation with the immune system response
bone marrow (Zhao et al., 2004), umbilical cord blood (Glezer et al., 2007). However, recent findings in stud-
(Borlongan et al., 2004), and embryonic stem cells ies with Alzheimers disease revealed positive results
(Gttinger et al., 2005) can have positive effect on in this chronic neurodegenerative disease. The group
2 Neurogenesis: A Change of Paradigms 25

of Serge Rivest from Laval University of Quebec brain regions into the hippocampus (Bortolotto et al.,
have shown that newly differentiated blood-derived 1990; Buzski et al., 1991) and correlated to epilep-
microglia are able to remove -amyloid plaque and tic activity. Buzski and coworkers had concluded
consequently decelerate the progress of Alzheimers that hippocampal grafts into the intact brain induce
disease, whereas microglia cells resident in CNS are epileptic patterns. On the other hand, Bortolotto and
inefficient in phagocyting the amyloid deposit (Simard colleagues showed that noradrenergic neurons pre-
et al., 2006; Napoli and Neumann, 2009). These pared from the locus coeruleus region from E13 or
authors suggested that therapeutic strategies aiming E14 and injected bilaterally into the hippocampus of
to improve microglia recruitment from bone marrow epileptic rats suppressed spontaneous recurrent epilep-
could potentially lead to a new powerful tool for the tic seizures in intra-hipocampal grafted rats.
elimination of toxic senile plaques. More recently, studies on epilepsy models dealt with
In Parkinson disease, as already mentioned here, embryonic (Gttinger et al., 2005; Ruschenschmidt
much of the present studies about transplantation have et al., 2005) and human fetal neural stem cells (Chu
been developed by Bjrklund, Lindvall and their col- et al., 2004) to treat seizures or to study their behav-
leagues in Sweden (Lindvall et al., 1990; Bjrklund ior in epileptic condition. Grafting of human fetal
and Lindvall, 2000). These studies indicated that these neural stem cells into rat brains one day after sta-
transplanted cells can replace dopaminergic neurons tus epilepticus induction led to marked suppression of
damaged in PD and restore and regenerate brain func- spontaneous recurrent motor seizures (SRMS). Indeed,
tion. Replacement therapies for neurodevelopmental approximately one month after SE onset, 87% of
and neurodegenerative diseases have revealed success the untreated animals showed spontaneous recurrence
when stem cells were placed into a microenviron- of seizure activity in contrast to only 13% of the
ment favoring differentiation and survival of newborn hNSC-treated animals. Phenotypic analysis indicated
cells. Such environmental conditions can be generated that whereas only a small fraction of the injected
by transplantation of genetically engineered neural hNSC expressed markers of mature neurons and
stem cells, which produce and secrete growth factors yet transplanted cells co-expressed phenotypic mark-
and neurotransmitters with neuroprotective and trophic ers of interneurons (GABA, parvalbumin), suggesting
actions (Trujillo et al., 2009). In PD, an alternative that hNSC can differentiate into GABA-synthesizing
method for mechanical pumps delivering glial-derived cells after transplantation into the lesioned hippocam-
neurotrophic factor (GDNF) can be the use of bio- pus. The authors hypothesize these new GABA-
logical minipumps, such as genetically modified neu- synthesizing cells may decrease neuronal excitability
rospheres. Neurospheres modified to produce GDNF and thus suppress spontaneous seizures.
can be transplanted into the brain for delivery of the
trophic factor. Using an inducible viral system, such as
that described by Capowski and coworkers (2007), it
is possible to control the amounts of GDNF delivery 2.6 Concluding Remarks
as well as to stop the therapy at any time. This type of
cell therapy associated with gene therapy is still under It is clear that functionally diverse roles expected
evaluation in animal models (Trujillo et al., 2009). of cellular replacement cannot be fulfilled by the
Concerning the functionality of transplanted stem transplantation of any single cell type; rather, spe-
cells in neurological diseases, electrophysiological cialized cells or tittered mixtures of cells may be
recordings after transplantation in a model of PD important in replacement strategies. As claimed by
have shown that grafted neurons were functionally Kempermann (2006), neural stem cells have some-
integrated in the host brain (Wernig et al., 2008; times been greeted as the last missing link in an
lvares-Dolado et al., 2006). Morphological analy- otherwise obviously coherent picture of what would
sis of the recorded cells confirmed their differenti- constitute successful neuroregeneration. As with the
ation into dopaminergic (Wernig et al., 2008) and discovery of precursor cells and adult neurogenesis,
GABAergic neurons (lvares-Dolado et al., 2006). everything should fall into place and neuroregener-
In the beginning of 1990s two different groups had ation become suddenly possible. However, the adult
published interesting papers about grafting embryonic brain still regenerates poorly. It does so despite the
26 L.E. Mello and B.M. Longo

presence of neural stem cells and not, as was previ- MG (2004) Electroconvulsive seizures regulate gene expres-
ously thought, because of their absence. To this end, sion of distinct neurotrophic signaling pathways. J Neurosci
24:26672677.
circumventing this limitation by means foreign to the
Altman J (1962) Are new neurons formed in the brains of adult
brain is not only a difficult task but might in addition mammals? Science 135:11271128.
produce mixed results. Altman J (1963) Differences in the utilization of tritiated
If stem cells are mobilized in the case of danger, leucine by single neurones in normal and exercised rats:
an autoradiographic investigation with microdensitometry.
than we are facing another type of defense system!
Nature 199:777780.
These cells are reacting somehow similar to immuno- Altman J, Das GD (1965) Autoradiographic and histological evi-
competent cells, which are responsible for the so called dence of postnatal hippocampal neurogenesis in rats. J Comp
body immune surveillance. Maybe the stem cells, Neurol 124:319336.
Alvarez-Buylla A, Garcia-Verdugo J (2002) Neurogenesis in
besides their task to generate all other kind of differen- adult subventricular zone. J Neurosci 22:629634.
tiated cells, are not working on surveillance but rather Alvarez-Dolado M, Calcagnotto ME, Karkar KM, Southwell
are mobilized by specific biochemical signals emitted DG, Jones-Davis DM, Estrada RC, Rubenstein JL, Alvarez-
by the injured organ in order to regenerate it. Their Buylla A, Baraban SC (2006) Cortical inhibition modified by
embryonic neural precursors grafted into the postnatal brain.
tropism for such an organ justifies this hypothesis of J Neurosci 26:73807389.
such a signal. This could be universal or specific to Arnett HA, Mason J, Marino M, Suzuki K, Matsushima GK,
the tissue or more likely specific to the given brain Ting JP (2001) TNF alpha promotes proliferation of oligo-
structure or cell population within a brain structure. dendrocyte progenitors and remyelination. Nat Neurosci
4:11161122.
We conclude with Karl Popper statement in his 1934 Avots A, Harder F, Schmittwolf C, Petrovic S, Mller AM
book The Logic of Scientific Discovery (1959, first (2002) Plasticity of hematopoietic stem cells and cellular
English edition): Science is not a system of certain, or memory. Immunol Rev 187:921.
well-established, statements; nor is it a system which Azizi SA, Stokes D, Augelli BJ, DiGirolamo C, Prockop DJ
(1998) Engraftment and migration of human bone mar-
steadily advances towards a state of finality. Our sci- row stromal cells implanted in the brains of albino rats
ence is not knowledge (episteme): it can never claim to similarities to astrocyte grafts. Proc Natl Acad Sci USA
have attained truth, or even a substitute for it, such as 95:39083913.
probability. Bagri A, Gurney T, He X, Zou YR, Littman DR, Tessier-Lavigne
M, Pleasure SJ (2002) The chemokine SDF1 regulates migra-
tion of dentate granule cells. Development 129:42494260.
Acknowledgments We gratefully acknowledge Dr Aurel Barnea A, Nottebohm F (1994) Seasonal recruitment of hip-
Popescu for suggestions and fertile discussion on immunoin- pocampal neurons in adult free-ranging black-capped chick-
flammation, Simone Romariz for the preparation of immunoflu- adees. Proc Natl Acad Sci USA 91:1121711221.
orescence assays and images, and Thomas Perlaky for the Battista D, Ferrari CC, Gage FH, Pitossi FJ (2006) Neurogenic
assistance with all the references. This work was supported by niche modulation by activated microglia: transforming
FAPESP and CNPq. growth factor beta increases neurogenesis in the adult dentate
gyrus. Eur J Neurosci 23:8393.
Bayer S (1982) Changes in the total number of dentate gran-
ule cells in juvenile and adult rats: A correlated volumetric
and 3H-thymidine autoradiographic study. Exp Brain Res
References 46:315323.
Bayer S, Yackel JW, Puri PS (1982) Neurons in the rat dentate
gyrus granular layer substantially increase during juvenile
Aarum J, Sandberg K, Haeberlein SL, Persson MA (2003) and adult life. Science 216:890892.
Migration and differentiation of neural precursor cells can be Beck D, Gross N, Beretta-Brognara C (1995) Effects of stem cell
directed by microglia. Proc Natl Acad Sci USA 100:15983 factor and other bone marrow-derived growth factors on the
15988. expression of adhesion molecules and proliferation of human
Aberg MA, Aberg ND, Palmer TD, Alborn AM, Carlsson- neuroblastoma cells. Eur J Cancer 31A:467470.
Skwirut C, Bang P, Rosengren LE, Olsson T, Gage FH, Begg AC, Hofland I, Van Der Pavert I, Van Der Schueren B,
Eriksson PS (2003) IGF-I has a direct proliferative effect Austermans K (2000) Use of thymidine analogues to indicate
in adult hippocampal progenitor cells. Mol Cell Neurosci vascular perfusion in tumours. Br J Cancer 83:899905.
24:2340. Ben-Hur T, Ben-Menachem O, Furer V, Einstein O, Mizrachi-
Albensi BC (2001) Potential roles for tumor necrosis factor and Kol R, Grigoriadis N (2003) Effects of proinflammatory
nuclear factor-kappaB in seizure activity. J Neurosci Res cytokines on the growth, fate, and motility of multipotential
66:151154. neural precursor cells. Mol Cell Neurosci 24:623631.
Altar CA, Laeng P, Jurata LW, Brockman JA, Lemire A, Bengzon J, Kokaia Z, Elmr E, Nanobashvili A, Kokaia M,
Bullard J, Bukhman YV, Young TA, Charles V, Palfreyman Lindvall O (1997) Apoptosis and proliferation of dentate
2 Neurogenesis: A Change of Paradigms 27

gyrus neurons after single and intermittent limbic seizures. Cacci E, Ajmone-Cat MA, Anelli T, Biagioni S, Minghetti
Proc Natl Acad Sci USA 94:1043210437. L (2008) In vitro neuronal and glial differentiation from
Benraiss A, Chmielnicki E, Lerner K, Roh D, Goldman embryonic or adult neural precursor cells are differently
SA (2001) Adenoviral brain-derived neurotrophic factor affected by chronic or acute activation of microglia. Glia
induces both neostriatal and olfactory neuronal recruitment 56:412425.
from endogenous progenitor cells in the adult forebrain. J Cacci E, Claasen JH, Kokaia Z (2005) Microglia-derived tumor
Neurosci 21:67186731. necrosis factor-alpha exaggerates death of newborn hip-
Bernardino L, Ferreira R, Cristovao AJ, Sales F, Malva pocampal progenitor cells in vitro. J Neurosci 80:789797.
JO (2005) Inflammation and neurogenesis in temporal Calza L, Giardino L, Pozza M, Bettelli C, Micera A, Aloe L
lobe epilepsy. Curr Drug Targets CNS Neurol Disord 4: (1998) Proliferation and phenotype regulation in the subven-
349360. tricular zone during experimental allergic encephalomyelitis:
Bhatt R, Rameshwar P, Goldstein K, Siegel A (2003) Effects in vivo evidence of a role for nerve growth factor. Proc Natl
of kindled seizures upon hematopoiesis in rats. Epilepsy Res Acad Sci USA 95:32093214.
54:209219. Cameron HA, Gould E (1994) Adult neurogenesis is regu-
Biebl M, Cooper C, Winkler J, Kuhn HG (2000) Analysis of neu- lated by adrenal steroids in the dentate gyrus. Neuroscience
rogenesis and programmed cell death reveals a self-renewing 61:203209.
capacity in the adult rat brain. Neurosci Lett 291:1720. Cameron HA, Mckay R (2001) Adult neurogenesis produces a
Bjrklund A, Lindvall O (2000) Cell replacement therapies for large pool of new granule cells in the dentate gyrus. J Comp
central nervous system disorders. Nat Neurosci 3:537544. Neurol 435:406417.
Blaschke AJ, Staley K, Chun J (1996) Widespread programmed Cameron HA, Wooley CS, McEwen BS, Gould E (1993)
cell death in proliferative and postmitotic regions of the fetal Differentiation of newly born neurons and glia in the dentate
cerebral cortex. Development 122:11651174. gyrus if the adult rat. Neuroscience 56:337344.
Borlongan CV, Hadman M, Sanberg CD, Sanberg PR (2004) Capowski EE, Schneider BL, Ebert AD, Seehus CR, Szulc J,
Central nervous system entry of peripherally injected umbil- Zufferey R, Aebischer P, Svendsen CN (2007) Lentiviral
ical cord blood cells is not required for neuroprotection in vector-mediated genetic modification of human neural pro-
stroke. Stroke 35:23852389. genitor cells for ex vivo gene therapy. J Neurosci Methods
Bortolotto ZA, Calderazzo L, Cavalheiro EA (1990) Some evi- 163:338349.
dence that intrahippocampal grafting of noradrenergic neu- Choi YS, Cho HY, Hoyt KR, Naegele JR, Obrietan K (2008)
rons suppresses spontaneous seizures in epileptic rats. Braz J IGF-1 receptor-mediated ERK/MAPK signaling couples sta-
Med Biol Res 23:12671269. tus epilepticus to progenitor cell proliferation in the subgran-
Bossolasco P, Cova L, Calzarossa C, Rimoldi SG, Borsotti C, ular layer of the dentate gyrus. Glia 56:791800.
Deliliers GL, Silani V, Soligo D, Polli E (2005) Neuro-glial Chu K, Kim M, Jeong SW, Kim SU, Yoon BW (2003) Human
differentiation of human bone marrow stem cells in vitro. neural stem cells can migrate, differentiate, and integrate
Exp Neurol 193:312325. after intravenous transplantation in adult rats with transient
Bouilleret V, Ridoux V, Depaulis A, Marescaux C, Nehlig A, forebrain ischemia. Neurosci Lett 343:129133.
Le Gal La Salle G (1999) Recurrent seizures and hippocam- Chu K, Kim M, Jung KH, Jeon D, Lee ST, Kim J, Jeong SW,
pal sclerosis following intrahippocampal kainate injection Kim SU, Lee SK, Shin HS, Roh JK (2004) Human neu-
in adult mice: electroencephalography, histopathology and ral stem cell transplantation reduces spontaneous recurrent
synaptic reorganization similar to mesial temporal lobe seizures following pilocarpine-induced status epilepticus in
epilepsy. Neuroscience 89:717729. adult rats. Brain Res 1023:213221.
Brazelton TR, Rossi FM, Keshet GI, Blau HM (2000) From Cogle CR, Yachnis AT, Laywell ED, Zander DS, Wingard JR,
marrow to brain: expression of neuronal phenotypes in adult Steindler DA, Scott EW (2004) Bone marrow transdifferen-
mice. Science 290:17751779. tiation in brain after transplantation: a retrospective study.
Brezun JM, Daszuta A (1999) Depletion in serotonin decreases Lancet 363:14321437.
neurogenesis in the dentate gyrus and the subventricular zone Cook LL, Persinger MA (1999) Infiltration of lymphocytes in
of adult rats. Neuroscience 89:9991002. the limbic brain following stimulation of subclinical cellular
Burd GD, Nottebohm F (1985) Ultrastructural characterization immunity and low dosages of lithium and a cholinergic agent.
of synaptic terminals formed on newly generated neurons in Toxicol Lett 109:7785.
a song control nucleus of the adult canary forebrain. J Comp Corotto FS, Henegar JA, Maruniak JA (1993) Neurogenesis per-
Neurol 240:143152. sists in the subependymal layer of the adult mouse brain.
Buss RR, Sun W, Oppenheim RW (2006) Adaptive roles of Neurosci Lett 149:111114.
programmed cell death during nervous system development. Covolan L, Mello LE (2000) Temporal profile of neuronal
Annu Rev Neurosci 29:135. injury following pilocarpine or kainic acid-induced status
Butovsky O, Ziv Y, Schwartz A, Landa G, Talpalar AE, Pluchino epilepticus. Epilepsy Res 39:133152.
S, Martino G, Schwartz M (2006) Microglia activated by Craig CG, Tropepe V, Morshead CM, Reynolds BA, Weiss S,
IL-4 or IFN-gamma differentially induce neurogenesis and van der Kooy D (1996) In vivo growth factor expansion of
oligodendrogenesis from adult stem/progenitor cells. Mol endogenous subependymal neural precursor cell populations
Cell Neurosci 31:149160. in the adult mouse brain. J Neurosci 16:26492658.
Buzski G, Masliah E, Chen LS, Horvth Z, Terry R, Gage Curtis MA, Connor B, Faull RL (2003) Neurogenesis in the
FH (1991) Hippocampal grafts into the intact brain induce diseased adult human brain new therapeutic strategies for
epileptic patterns. Brain Res 554:12. neurodegenerative diseases. Cell Cycle 2:428430.
28 L.E. Mello and B.M. Longo

Dash PK, Mach SA, Moore AN (2001) Enhanced neurogenesis subventricular zone: in search of the stem cells. J Neurobiol
in the rodent hippocampus following traumatic brain injury. 36:234248.
J Neurosci Res 63:313319. Glezer I, Simard AR, Rivest S (2007) Neuroprotective role of
Doetsch F, Alvarez-Buylla A (1996) Network of tangential path- the innate immune system by microglia. Neuroscience 147:
ways for neuronal migration in the adult mammalian brain. 867883.
Proc Natl Acad Sci USA 93:1489514900. Goldman SA, Nottebohm F (1983) Neuronal production, migra-
Doetsch F, Caille I, Lim DA (1999) Subventricular zone astro- tion, and differentiation in a vocal control nucleus of the adult
cytes are neural stem cells in the adult mammalian brain. Cell female canary brain. Proc Natl Acad Sci USA 80:23902394.
97:120. Gould E (2007) Structural plasticity. In: The Hippocampus Book
Doetsch F, Garca-Verdugo JM, Alvarez-Buylla A (1997) (Andersen P, ed), pp. 321335. Oxford University Press,
Cellular composition and three-dimensional organization of Oxford.
the subventricular germinal zone in the adult mammalian Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ (1999a)
brain. J Neurosci 17:50465061. Learning enhances adult neurogenesis in the hippocampal
Eckenhoff MF, Rakic P (1984) Radial organization of the formation. Nat Neurosci 2:260265.
hippocampal dentate gyrus: a Golgi, ultrastructural, and Gould E, Gross CG (2002) Neurogenesis in adult mammals:
immunocytochemical analysis in the developing rhesus mon- some progress and problems. J Neurosci 22:619623.
key. J Comp Neurol 223:121. Gould E, McEwen BS, Tanapat P, Galea LA, Fuchs E (1997)
Eckenhoff MF, Rakic P (1988) Nature and fate of proliferative Neurogenesis in the dentate gyrus of the adult tree shrew
cells in the hippocampal dentate gyrus during the life span of is regulated by psychosocial stress and NMDA receptor
the rhesus monkey. J Neurosci 8:27292747. activation. J Neurosci 17:24922498.
Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O (2003) Gould E, Reeves AJ, Graziano MS, Gross CG (1999b)
Inflammation is detrimental for neurogenesis in adult brain. Neurogenesis in the neocortex of adult primates. Science
Proc Natl Acad Sci USA 100:1363213637. 286:548552.
Ekdahl CT, Kokaia Z, Lindvall O (2009) Brain inflammation and Gould E, Tanapat P (1997) Lesion-induced proliferation of
adult neurogenesis: the dual role of microglia. Neuroscience neuronal progenitors in the dentate gyrus of the adult rat.
158:10211029. Neuroscience 80:427436.
Elliott RC, Miles MF, Lowenstein DH (2003) Overlapping Gould E, Tanapat P, McEwen BS, Flugge G, Fuchs E (1998)
microarray profiles of dentate gyrus gene expression dur- Proliferation of granule cell precursors in the dentate gyrus
ing development- and epilepsy-associated neurogenesis and of adult of monkeys is diminished by stress. Proc Natl Acad
axon outgrowth. J Neurosci 23:22182227. Sci USA 95:31683171.
Eriksson PS, Perfilieva E, Bjrk-Eriksson T, Alborn AM, Gould E, Woolley C, McEwen BS (1991) Adrenal steroids
Nordborg C, Peterson DA, Gage FH (1998) Neurogenesis in regulate postnatal development of the rat dentate gyrus. I.
the adult human hippocampus. Nat Med 4:13131317. Effects of glucocorticoids on cell death. J Comp Neurol
Eriksson C, Zou LP, Ahlenius S, Winblad B, Schultzberg 313:479485.
M (2000) Inhibition of kainic acid induced expression Gray W, Sundstrom LE (1998) Kainic acid increases the prolif-
of interleukin-1 beta and interleukin-1 receptor antagonist eration of granule cell progenitors in the dentate gyrus of the
mRNA in the rat brain by NMDA receptor antagonists. Brain adult rat. Brain Res 790:5259.
Res Mol Brain Res 85:103113. Gross CG (2000) Neurogenesis in the adult brain: death of a
Fahrner A, Kann G, Flubacher A, Heinrich C, Freiman TM, dogma. Nature Rev Neurosci 1:6773.
Zentner J, Frotscher M, Haas CA (2007) Granule cell dis- Gttinger M, Fedele D, Koch P, Padrun V, Pralong WF, Brustle
persion is not accompanied by enhanced neurogenesis in O, Boison D (2005) Suppression of kindled seizures by
temporal lobe epilepsy patients. Exp Neurol 203:320332. paracrine adenosine release from stem cell-derived brain
Ferland RJ, Gross RA, Applegate CD (2002) Increased mitotic implants. Epilepsia 46:11621169.
activity in the dentate gyrus of the hippocampus of adult Hagihara H, Hara M, Tsunekawa K, Nakagawa Y, Sawada
C57bl/6j mice exposed to the flurothyl kindling model of M, Nakano K (2005) Tonic-clonic seizures induce divi-
epileptogenesis. Neuroscience 115:669683. sion of neuronal progenitor cells with concomitant changes
Ferrazoli E, Senedese A, Romariz S, Bahia L, Santos RR, in expression of neurotrophic factors in the brain of
Blanco M, Mello LE, Longo B (2008) Avaliao curso- pilocarpine-treated mice. Brain Res Mol Brain Res 139:
temporal do efeito protetor do transplante de clulas de 258266.
medula ssea em um modelo agudo de crise convulsiva. Hamano H, Noguchi M, Fukui H, Issiki A, Watanabe Y (2002)
J Epil Clin Neurophysiol 14:5455. Regulation of brain cell environment on neuronal protection:
Freese A, ORourke D, Judy K, OConnor MJ (1994) The appli- role of TNFalpha in glia cells. Life Sci 72:565574.
cation of 5-bromodeoxyuridine in the management of CNS Hansel DE, Eipper BA, Ronnett GV (2001) Neuropeptide
tumors. J Neurooncol 20:8195. Y functions as a neuroproliferative factor. Nature 410:
Ganor Y, Besser M, Ben-Zakay N, Unger T, Levite M (2003) 940944.
Human T cells express a functional ionotropic glutamate Hastings NB, Gould E (2003) Neurons inhibit neurogenesis. Nat
receptor GluR3, and glutamate by itself triggers integrin- Med 9:264266.
mediated adhesion to laminin and fibronectin and chemotac- Heldmann U, Thored P, Claasen JH, Arvidsson A, Kokaia Z,
tic migration. J Immunol 170:43624372. Lindvall O (2005) TNF-alpha antibody infusion impairs sur-
Garca-Verdugo JM, Doetsch F, Wichterle H, Lim D, Alvarez- vival of stroke-generated neuroblasts in adult rat brain. Exp
Buylla A (1998) Architecture and cell types of the adult Neurol 196:204208.
2 Neurogenesis: A Change of Paradigms 29

Hoshino T, Prados M, Wilson CB, Cho KG, Lee KS, Davis RL Kerschensteiner M, Meinl E, Hohlfeld R (2009) Neuro-
(1989) Prognostic implications of the bromodeoxyuridine immune crosstalk in CNS diseases. Neuroscience 158:
labeling index of human gliomas. J Neurosurg 71:335341. 11221132.
Houser C (1990) Granule cell dispersion in the dentate gyrus Kerschensteiner M, Stadelmann C, Dechant G, Wekerle H,
of humans with temporal lobe epilepsy. Brain Res 535: Hohlfeld R (2003) Neurotrophic cross-talk between the ner-
195204. vous and immune systems: implications for neurological
Houser CR, Miyashiro JE, Swartz BE, Walsh GO, Rich JR, diseases. Ann Neurol 53:292304.
Delgado-Escueta AV (1990) Altered patterns of dynor- Kilpatrick TJ, Bartlett PF (1995) Cloned multipotential precur-
phin immunoreactivity suggest mossy fiber reorganization in sors from the mouse cerebrum require FGF-2, whereas glial
human hippocampal epilepsy. J Neurosci 10:267282. restricted precursors are stimulated with either FGF-2 or
Howell OW, Scharfman HE, Herzog H, Sundstrom LE, Beck- EGF. J Neurosci 15:36533661.
Sickinger A, Gray WP (2003) Neuropeptide Y is neuro- Kilpatrick TJ, Talman PS, Bartlett PF (1993) The differentiation
proliferative for post-natal hippocampal precursor cells. J and survival of murine neurons in vitro is promoted by sol-
Neurochem 86:646659. uble factors produced by an astrocytic cell line. J Neurosci
Iosif RE, Ekdahl CT, Ahlenius H, Pronk CJ, Bonde S, Kokaia Z, Res 35:147161.
Jacobsen SE, Lindvall O (2006) Tumor necrosis factor recep- Kintner C (2002) Neurogenesis in embryos and in adult neural
tor 1 is a negative regulator of progenitor proliferation in stem cells. J Neurosci 22:639643.
adult hippocampal neurogenesis. J Neurosci 26:97039712. Knoblach SM, Fan L, Faden AI (1999) Early neuronal
Jin K, Peel AL, Mao XO, Xie L, Cottrell BA, Henshall DC, expression of tumor necrosis factor-alpha after experimen-
Greenberg DA (2004) Increased hippocampal neurogene- tal brain injury contributes to neurological impairment.
sis in Alzheimers disease. Proc Natl Acad Sci USA 101: J Neuroimmunol 95:115125.
343347. Kokaia Z, Lindvall O (2003) Neurogenesis after ischaemic brain
Jin K, Wang X, Xie L, Mao XO, Zhu W, Wang Y, Shen J, Mao insults. Curr Opin Neurobiol 13:127132.
Y, Banwait S, Greenberg DA (2006) Evidence for stroke- Komack DR, Rakic P (1999) Continuation of neurogenesis in the
induced neurogenesis in the human brain. Proc Natl Acad hippocampus of the adult macaque monkey. Proc Natl Acad
Sci USA 103:1319813202. Sci USA 96:57685773.
Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U, Koo JW, Duman RS (2008) IL-1beta is an essential mediator of
Frisn J (1999) Identification of a neural stem cell in the adult the antineurogenic and anhedonic effects of stress. Proc Natl
mammalian central nervous system. Cell 96:2534. Acad Sci USA 105:751756.
Kaplan MS (1981) Neurogenesis in the 3-month-old rat visual Kopen GC, Prockop DJ, Phinney DG (1999) Marrow stro-
cortex. J Comp Neurol 195:323338. mal cells migrate throughout forebrain and cerebellum,
Kaplan MS (1985) Formation and turnover of neurons in young and they differentiate into astrocytes after injection into
and senescent animals: an electron microscopic and morpho- neonatal mouse brains. Proc Natl Acad Sci USA 96:
metric analysis. Ann NY Acad Sci 457:173192. 1071110716.
Kaplan MS, Bell DH (1984) Mitotic neuroblasts in the 9-day- Kronenberg G, Reuter K, Steiner B, Brandt MD, Jessberger
old and 11-month-old rodent hippocampus. J Neurosci 4: S, Yamaguchi M, Kempermann G (2003) Subpopulations
14291441. of proliferating cells of the adult hippocampus respond dif-
Kaplan MS, Hinds J (1977) Neurogenesis in the adult rat: elec- ferently to physiologic neurogenic stimuli. J Comp Neurol
tron microscopic analysis of light radioautographs. Science 467:455463.
197:10921094. Krumbholz M, Theil D, Derfuss T, Rosenwald A, Schrader F,
Kee NJ, Preston E, Wojtowicz JM (2001) Enhanced neurogene- Monoranu CM, Kalled SL, Hess DM, Serafini B, Aloisi F,
sis after transient global ischemia in the dentate gyrus of the Wekerle H, Hohlfeld R, Meinl E (2005) BAFF is produced
rat. Exp Brain Res 136:313320. by astrocytes and up-regulated in multiple sclerosis lesions
Kempermann G (2006) Adult Neurogenesis: Stem Cell and and primary central nervous system lymphoma. J Exp Med
Neuronal Development in the Adult Brain. Oxford University 201:195200.
Press, Oxford. Kuhn T (1962) The Structure of Scientific Revolutions.
Kempermann G, Gage FH (2002) Genetic determinants of adult University of Chicago Press, Chicago, IL.
hippocampal neurogenesis correlate with acquisition, but Kuhn HG, Biebl M, Wilhelm D, Li M, Friedlander RM, Winkler
not probe trial performance, in the water maze task. Eur J (2005) Increased generation of granule cells in adult Bcl-2-
J Neurosci 16:129136. overexpressing mice: a role for cell death during continued
Kempermann G, Jessberger S, Steiner B, Kronenberg G (2004) hippocampal neurogenesis. J Neurosci 22:19071915.
Milestones of neuronal development in the adult hippocam- Kuhn HG, Dickison-Anson H, Gage FH (1996) Neurogenesis
pus. Trends Neurosci 27:447452. in the dentate gyrus of the adult rat: age-related decrease
Kempermann G, Kronenberg G (2003) Depressed new neurons of neuronal progenitor proliferation. J Neurosci 16:
adult hippocampal neurogenesis and a cellular plasticity 20272033.
hypothesis of major depression. Biol Psychiatry 54:499503. Kuhn HG, Winkler J, Kempermann G, Thal LJ, Gage FH (1997)
Kempermann G, Kuhn H, Gage FH (1997) More hippocampal Epidermal growth factor and fibroblast growth factor-2 have
neurons in adult mice living in an enriched environment. different effects on neural progenitors in the adult rat brain. J
Nature 386:493495. Neurosci 17:58205829.
Kempermann G, Kuhn HG, Gage FH (1998) Experience- Kulkarni SK, George B (1995) Lithium-pilocarpine neurotoxi-
induced neurogenesis in the senescent dentate gyrus. J city: a potential model of status epilepticus. Methods Find
Neurosci 18:32063212. Exp Clin Pharmacol 17:551567.
30 L.E. Mello and B.M. Longo

Lee J, Duan W, Long JM, Ingram DK, Mattson MP (2000) the neonatal mammalian forebrain. Mol Cell Neurosci 6:
Dietary restriction increases the number of newly generated 496508.
neural cells, and induces BDNF expression, in the dentate Mezey E, Chandross KJ, Harta G, Maki RA, McKercher SR
gyrus of rats. J Mol Neurosci 15:99108. (2000) Turning blood into brain: cells bearing neuronal
Lee J, Kuroda S, Shichinohe H, Ikeda J, Seki T, Hida K, Tada M, antigens generated in vivo from bone marrow. Science
Sawada K, Iwasaki Y (2003) Migration and differentiation of 290:17791782.
nuclear fluorescence-labeled bone marrow stromal cells after Mohapel P, Ekdahl C, Lindvall O (2004) Status epilepti-
transplantation into cerebral infarct and spinal cord injury in cus severity influences the long-term outcome of neu-
mice. Neuropathology 23:169180. rogenesis in the adult dentate gyrus. Neurobiol Dis 15:
Lemaire V, Koehl M, Le Moal M, Abrous DN (2000) Prenatal 196205.
stress produces learning deficits associated with an inhibi- Monje ML, Toda H, Palmer TD (2003) Inflammatory blockade
tion of neurogenesis in the hippocampus. Proc Natl Acad Sci restores adult hippocampal neurogenesis. Science 302:1760
USA 97:1102311027. 1765.
Lindvall O, Rehncrona S, Brundin P, Gustavii B, Astedt B, Morgan SC, Taylor DL, Pocock JM (2004) Microglia release
Widner H, Lindholm T, Bjrklund A, Leenders KL, Rothwell activators of neuronal proliferation mediated by activation of
JC (1990) Neural transplantation in Parkinsons disease: the mitogen-activated protein kinase, phosphatidylinositol-
Swedish experience. Prog Brain Res 82:729734. 3-kinase/Akt and delta-Notch signalling cascades.
Liu Z, Fan Y, Won SJ, Neumann M, Hu D, Zhou L, Weinstein J Neurochem 90:89101.
PR, Liu J (2007) Chronic treatment with minocycline pre- Nakanishi M, Niidome T, Matsuda S, Akaike A, Kihara T,
serves adult new neurons and reduces functional impairment Sugimoto H (2007) Microglia-derived interleukin-6 and
after focal cerebral ischemia. Stroke 38:146152. leukaemia inhibitory factor promote astrocytic differentia-
Liu J, Solway K, Messing RO, Sharp FR (1998) Increased neu- tion of neural stem/progenitor cells. Eur J Neurosci 25:
rogenesis in the dentate gyrus after transient global ischemia 649658.
in gerbils. J Neurosci 18:77687778. Napoli I, Neumann H (2009) Microglial clearance function in
Lois C, Alvarez-Buylla A (1994) Long-distance neuronal migra- health and disease. Neuroscience 158:10301038.
tion in the adult mammalian brain. Science 264:11451148. Neumann H, Schmidt H, Wilharm E, Behrens L, Wekerle H
Lois C, Garca-Verdugo JM, Alvarez-Buylla A (1996) Chain (1997) Interferon gamma gene expression in sensory neu-
migration of neuronal precursors. Science 271:978981. rons: evidence for autocrine gene regulation. J Exp Med
Longo B, Blanco M, Senra J, Brustolim D, Bahia L, Soares M, 186:20232031.
Mello LE, dos Santos RR (2005) Migration of bone mar- Nilsson SK, Dooner MS, Weier HU, Frenkel B, Lian JB, Stein
row stem cell into the brain after status epilepticus or acute GS, Quesenberry PJ (1999) Cells capable of bone production
seizures. Epilepsia 46:360361 Suppl. 366. engraft from whole bone marrow transplants in nonablated
Madrazo I, Drucker-Colin R, Daz V, Martnez-Mata J, Torres C, mice. J Exp Med 189:729734.
Becerril JJ (1987) Open microsurgical autograft of adrenal Nottebohm F (1981) A brain for all seasons: cyclical anatomical
medulla to the right caudate nucleus in two patients with changes in song control nuclei of the canary brain. Science
intractable Parkinsons disease. N Engl J Med 316:831834. 214:13681370.
Madrazo I, Leon V, Torres C, Aguilera MC, Varela G, Alvarez F, Nottebohm F (1985) Neuronal replacement in adulthood. Ann
Fraga A, Drucker-Coln R, Ostrosky F, Skurovich M (1988) NY Acad Sci 457:143161.
Transplantation of fetal substantia nigra and adrenal medulla Nottebohm F (1996) The King Solomon lectures in neuroethol-
to the caudate nucleus in two patients with Parkinsons ogy. A white canary on Mount Acropolis. J Comp Physiol A
disease. N Engl J Med 319:370371. 179:149156.
Magavi SS, Leavitt B, Macklis JD (2000) Induction of neuroge- Nottebohm F (2002) Neuronal replacement in the adult brain.
nesis in the neocortex of adult mice. Nature 405:951955. Brain Res Bull 57:737749.
Malberg JE, Eisch AJ, Nestler EJ, Duman RS (2000) Chronic Nottebohm F (2004) The road we travelled: discovery, choreog-
antidepressant treatment increases neurogenesis in adult rat raphy, and significance of brain replaceable neurons. Ann N
hippocampus. J Neurosci 20:91049110. Y Acad Sci 1016:628658.
Makowiak M, Chocyk A, Markowicz-Kula K, Wedzony K Nowakowski RS, Lewin SB, Miller MW (1989)
(2004) Neurogenesis in the adult brain. Pol J Pharmacol Bromodeoxyuridine immunohistochemical determina-
56:673687. tion of the legths of the cell cycle and the DNA-synthetic
McKay R (1997) Stem cells in the central nervous system. phase for an anatomically defined population. J Neurocytol
Science 276:6671. 18:311318.
Mello LE, Cavalheiro E, Tan AM, Pretorius JK, Babb TL, Finch Okano HJ, Pfaff DW, Gibbs RB (1993) RB and Cdc2 expression
DM (1992) Granule cell dispersion in relation to mossy fiber in brain: correlations with 3H-thymidine incorporation and
sprouting, hippocampal cell loss, silent period and seizure neurogenesis. J Neurosci 13:29302938.
frequency in the pilocarpine model of epilepsy. Epilepsy Res OKusky JR, Ye P, DErcole AJ (2000) Insulin-like growth
Suppl 9:5159. factor-I promotes neurogenesis and synaptogenesis in the
Mello LE, Mendez-Otero R (1996) Expression of 9-O-acetylated hippocampal dentate gyrus during postnatal development. J
gangliosides in the rat hippocampus. Neurosci Lett 213: Neurosci 20:84358442.
1720. Palmer TD, Ray J, Gage FH (1995) FGF-2-responsive neuronal
Menezes JR, Smith CM, Nelson KC, Luskin MB (1995) The progenitors reside in proliferative and quiescent regions of
division of neuronal progenitor cells during migration in the adult rodent brain. Mol Cell Neurosci 6:474486.
2 Neurogenesis: A Change of Paradigms 31

Palmer TD, Willhoite AR, Gage FH (2000) Vascular niche Rougier A, Arthaud S, Zombre N, La Salle L (2005) Patterns of
for adult hippocampal neurogenesis. J Comp Neurol 425: dentate granule cell responses to perforant path stimulation
479494. in epileptic mice with granule cell dispersion. Epilepsy Res
Parent J (2003) Injury-induced neurogenesis in the adult mam- 63:119129.
malian brain. Neuroscientist 9:261272. Ruschenschmidt C, Koch PG, Brustle O, Beck H (2005)
Parent JM, Tada E, Fike JR, Lowenstein DH (1999) Inhibition of Functional properties of ES cell-derived neurons engrafted
dentate granule cell neurogenesis with brain irradiation does into the hippocampus of adult normal and chronically epilep-
not prevent seizure-induced mossy fiber synaptic reorganiza- tic rats. Epilepsia 46:174183 Suppl 5.
tion in the rat. J Neurosci 19:45084519. Santarelli L, Saxe M, Gross C, Surget A, Battaglia F, Dulawa S,
Parent JM, Yu TW, Leibowitz RT, Gerschwind DH, Sloviter RS, Weisstaub N, Lee J, Duman R, Arancio O, Belzung C, Hen
Lowenstein DH (1997) Dentate granule cell neurogenesis R (2003) Requirement of hippocampal neurogenesis for the
is increased by seizures and contributes to aberrant net- behavioral effects of antidepressants. Science 301:805809.
work reorganization in the adult rat hippocampus. J Neurosci Scharfman HE (2004) Functional implications of seizure-
17:37273738. induced neurogenesis. Adv Exp Med Biol 548:192212.
Paton JA, Nottebohm FN (1984) Neurons generated in the Scharfman HE, Goodman JH, Sollas AL (2000) Granule-like
adult brain are recruited into functional circuits. Science neurons at the hilar/CA3 border after status epilepticus and
225:10461048. their synchrony with area CA3 pyramidal cells: functional
Pencea V, Bingaman KD, Wiegand SJ, Luskin MB (2001) implications of seizure-induced neurogenesis. J Neurosci
Infusion of brain-derived neurotrophic factor into the lat- 20:61446158.
eral ventricle of the adult rat leads to new neurons in the Scharfman HE, Sollas AL, Smith KL, Jackson MB,
parenchyma of the striatum, septum, thalamus, and hypotha- Goodman JH (2002) Structural and functional asym-
lamus. J Neurosci 21:67066717. metry in the normal and epileptic rat dentate gyrus. J Comp
Peng H, Huang Y, Rose J, Erichsen D, Herek S, Fujii N, Neurol 454:424439.
Tamamura H, Zheng J (2004) Stromal cell-derived factor 1- Schilling M, Besselmann M, Leonhard C, Mueller M,
mediated CXCR4 signaling in rat and human cortical neural Ringelstein EB, Kiefer R (2003) Microglial activation pre-
progenitor cells. J Neurosci Res 76:3550. cedes and predominates over macrophage infiltration in tran-
Pirttil TJ, Manninen A, Jutila L, Nissinen J, Klviinen R, sient focal cerebral ischemia: a study in green fluorescent
Vapalahti M, Immonen A, Paljrvi L, Karkola K, Alafuzoff I, protein transgenic bone marrow chimeric mice. Exp Neurol
Mervaala E, Pitknen A (2005) Cystatin C expression is asso- 183:2533.
ciated with granule cell dispersion in epilepsy. Ann Neurol Scott BW, Wojtowicz JM, Burnham WM (2000) Neurogenesis
58:211223. in the dentate gyrus of the rat following electroconvulsive
Pluchino S, Martino G (2005) The therapeutic use of stem cells shock seizures. Exp Neurol 165:231236.
for myelin repair in autoimmune demyelinating disorders. J Seaberg RM, Van der Kooy D (2002) Adult rodent neurogenic
Neurol Sci 233:117119. regions: the ventricular subependyma contains neural stem
Popper K (2002) The Logic of Scientific Discovery. Routledge cells, but the dentate gyrus contains restricted progenitors.
Classics Routledge, London. J Neurosci 22:17841793.
Priller J, Flugel A, Wehner T, Boentert M, Haas CA, Prinz Seki T, Arai Y (1993) Highly polysialylated neural cell adhe-
M, Fernandez-Klett F, Prass K, Bechmann I, de Boer BA, sion molecule (NCAM-H) is expressed by newly generated
Frotscher M, Kreutzberg GW, Persons DA, Dirnagl U (2001) granule cells in the dentate gyrus of the adult rat. J Neurosci
Targeting gene-modified hematopoietic cells to the central 13:23512358.
nervous system: use of green fluorescent protein uncovers Seki T, Arai Y (1995) Age-related production of new granule
microglial engraftment. Nat Med 7:13561361. cells in the adult dentate gyrus. Neuroreport 6:24792482.
Rakic P (2002) Neurogenesis in adult primates. Prog Brain Res Selmaj KW, Farooq M, Norton WT, Raine CS, Brosnan CF
138:314. (1990) Proliferation of astrocytes in vitro in response to
Ramn y Cajal S (1928) Degeneration and Regeneration of the cytokines. A primary role for tumor necrosis factor. J
Nervous System. Oxford University Press, Oxford. Immunol 144:129135.
Ray J, Raymon HK, Gage FH (1995) Generation and cul- Seri B, Garca-Verdugo JM, McEwen BS, Alvarez-Buylla A
turing of precursor cells and neuroblasts from embryonic (2001) Astrocytes give rise to new neurons in the adult
and adult central nervous system. Methods Enzymol 254: mammalian hippocampus. J Neurosci 21:71537160.
2037. Shankle WR, Rafii MS, Landing BH, Fallon JH (1999)
Reynolds B, Weiss S (1992) Generation of neurons and astro- Approximate doubling of numbers of neurons in postnatal
cytes from isolated cells of the adult mammalian central human cerebral cortex and in 35 specific cytoarchitectural
nervous system. Science 255:17071710. areas from birth to 72 months. Pediatr Dev Pathol 2:244259.
Richards LJ, Kilpatrick TJ, Bartlett PF (1992) De novo genera- Shors TJ, Miesegaes G, Beylin A, Zhao M, Rydel T, Gould E
tion of neuronal cells from the adult mouse brain. Proc Natl (2001) Neurogenesis in the adult is involved in the formation
Acad Sci USA 89:85918595. of trace memories. Nature 410:372376.
Romariz S, Blanco M, Senra J, Ferrazoli E, Soares M, Santos Silani V, Cova L, Corbo M, Ciammola A, Polli E (2004)
RR, Mello LE, Longo B (2008) Avaliao da migrao Stem-cell therapy for amyotrophic lateral sclerosis. Lancet
e proliferao no hipocampo de camundongos epilpticos 364:200202.
transplantados com clulas de medula ssea GFP. J Epil Clin Simard AR, Rivest S (2004) Role of inflammation in the neuro-
Neurophysiol 14:6263. biology of stem cells. Neuroreport 15:23052310.
32 L.E. Mello and B.M. Longo

Simard AR, Soulet D, Gowing G, Julien JP, Rivest S (2006) Vezzani A (2005) Inflammation and epilepsy. Epilepsy Curr 5:
Bone marrow-derived microglia play a critical role in 16.
restricting senile plaque formation in Alzheimers disease. Vezzani A, Conti M, De Luigi A, Ravizza T, Moneta
Neuron 49:489502. D, Marchesi F, De Simoni MG (1999) Interleukin-1beta
Sloviter RS, Kudrimoti HS, Laxer KD, Barbaro NM, Chan immunoreactivity and microglia are enhanced in the rat
S, Hirsch LJ, Goodman RR, Pedley TA (2004) Tectonic hippocampus by focal kainate application: functional evi-
hippocampal malformations in patients with temporal lobe dence for enhancement of electrographic seizures. J Neurosci
epilepsy. Epilepsy Res 59:123153. 19:50545065.
Steiner B, Zurbog S, Hrster H, Fabel K, Kempermann G (2008) Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi
Differential 24 h responsiveness of Prox1-expressing pre- A, De Simoni MG, Sperk G, Andell-Jonsson S, Lundkvist J,
cursor cells in adult hippocampal neurogenesis to physical Iverfeldt K, Bartfai T (2000) Powerful anticonvulsant action
activity, environmental enrichment, and kainic acid-induced of IL-1 receptor antagonist on intracerebral injection and
seizures. Neuroscience 154:521529. astrocytic overexpression in mice. Proc Natl Acad Sci USA
Struikmans H, Rutgers DH, Jansen GH, Tulleken CA, van der 21:1153411539.
Tweel I, Battermann JJ (1997) S-phase fraction, 5-bromo-2 - Voutsinos-Porche B, Koning E, Kaplan H, Ferrandon A,
deoxy-uridine labelling index, duration of S-phase, potential Guenounou M, Nehlig A, Motte J (2004) Temporal patterns
doubling time, and DNA index in benign and malignant brain of the cerebral inflammatory response in the rat lithium-
tumors. Radiat Oncol Investig 5:170179. pilocarpine model of temporal lobe epilepsy. Neurobiol Dis
Suzuki F, Junier MP, Guilhem D, Srensen JC, Onteniente 17:385402.
B (1995) Morphogenetic effect of kainate on adult hip- Walton NM, Sutter BM, Laywell ED, Levkoff LH, Kearns
pocampal neurons associated with a prolonged expres- SM, Marshall GP 2nd, Scheffler B, Steindler DA (2006)
sion of brain-derived neurotrophic factor. Neuroscience 64: Microglia instruct subventricular zone neurogenesis. Glia
665674. 54:815825.
Tanapat P, Hastings NB, Rydel TA, Galea LA, Gould E (2001) Wang Y, Baraban SC (2007) Granule cell dispersion and aberrant
Exposure to fox odor inhibits cell proliferation in the hip- neurogenesis in the adult hippocampus of an LIS1 mutant
pocampus of adult rats via an adrenal hormone-dependent mouse. Dev Neurosci 29:9198.
mechanism. J Comp Neurol 437:496504. Wang Y, Deng Y, Zhou GQ (2008) SDF-1alpha/CXCR4-
Tattersfield AS, Croon RJ, Liu YW, Kells AP, Faull RL, Connor mediated migration of systemically transplanted bone mar-
B (2004) Neurogenesis in the striatum of the quinolinic row stromal cells towards ischemic brain lesion in a rat
acid lesion model of Huntingtons disease. Neuroscience model. Brain Res 1195:104112.
127:319332. Weimann JM, Charlton CA, Brazelton TR, Hackman RC, Blau
Taupin P (2008) Adult neurogenesis, neuroinflammation ther- HM (2003) Contribution of transplanted bone marrow cells
apeutic potential of adult neural stem cells. Int J Med Sci to Purkinje neurons in human adult brains. Proc Natl Acad
5:127132. Sci USA 100:20882093.
Thomas LB, Gates MA, Steindler DA (1996) Young neurons Weissman IL, Anderson DJ, Gage F (2001) Stem and pro-
from the adult subependymal zone proliferate and migrate genitor cells: origins, phenotypes, lineage commitments,
along an astrocyte, extracellular matrix-rich pathway. Glia and transdifferentiations. Annu Rev Cell Dev Biol 17:
17:114. 387403.
Trejo JL, Carro E, Torres-Aleman I (2001) Circulating insulin- Wernig M, Zhao JP, Pruszak J, Hedlund E, Fu D, Soldner
like growth factor I mediates exercise-induced increases in F, Broccoli V, Constantine-Paton M, Isacson O, Jaenisch
the number of new neurons in the adult hippocampus. J R (2008) Neurons derived from reprogrammed fibroblasts
Neurosci 21:16281634. functionally integrate into the fetal brain and improve symp-
Trujillo CA, Schwindt TT, Martins AH, Alves JM, Mello LE, toms of rats with Parkinsons disease. Proc Natl Acad Sci
Ulrich H (2009) Novel perspectives of neural stem cell differ- USA 105:58565861.
entiation: from neurotransmitters to therapeutics. Cytometry Wong G, Goldshmit Y, Turnley AM (2004) Interferon-gamma
A 75:3853. but not TNF alpha promotes neuronal differentiation and
Vallieres L, Sawchenko PE (2003) Bone marrow-derived cells neurite outgrowth of murine adult neural stem cells. Exp
that populate the adult mouse brain preserve their hematopoi- Neurol 187:171177.
etic identity. J Neurosci 23:51975207. Wu JP, Kuo JS, Liu YL, Tzeng SF (2000) Tumor necrosis factor-
Vallires L, Campbell II, Gage FH, Sawchenko PE (2002) alpha modulates the proliferation of neural progenitors in the
Reduced hippocampal neurogenesis in adult transgenic subventricular/ventricular zone of adult rat brain. Neurosci
mice with chronic astrocytic production of interleukin-6. J Lett 292:203206.
Neurosci 22:468492. Yamada M, Onodera M, Mizuno Y, Mochizuki H (2004)
Van Praag H, Kempermann G, Gage FH (1999) Running Neurogenesis in olfactory bulb identified by retroviral
increases cell proliferation and neurogenesis in the adult labeling in normal and 1-methyl-4-phenyl-1,2,3,6-
mouse dentate gyrus. Nat Neurosci 2:266270. tetrahydropyridine-treated adult mice. Neuroscience
Van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD, 124:173181.
Gage FH (2002) Functional neurogenesis in the adult hip- Zhang J, Li Y, Chen J, Yang M, Katakowski M, Lu M, Chopp M
pocampus. Nature 415:10301034. (2004) Expression of insulin-like growth factor 1 and recep-
Van der Kooy D, Weiss S (2000) Why stem cells? Science tor in ischemic rats treated with human marrow stromal cells.
287:14391441. Brain Res 1030:1927.
2 Neurogenesis: A Change of Paradigms 33

Zhao M, Momma S, Delfani K, Carln M, Cassidy RM, to transform mesenchymal stem cells into advantageous
Johansson CB, Brismar H, Shupliakov O, Frisn J, Janson engineering cells for neuroregeneration and neuroprotection.
M (2003) Evidence for neurogenesis in the adult mammalian Exp Neurol 190:396406.
substantia nigra. Proc Natl Acad USA 100:79257930. Ziv Y, Ron N, Butovsky O, Landa G, Sudai E, Greenberg
Zhao CS, Overstreet-Wadiche L (2008) Integration of adult gen- N, Cohen H, Kipnis J, Schwartz M (2006) Immune cells
erated neurons during epileptogenesis. Epilepsia 49(Suppl contribute to the maintenance of neurogenesis and spatial
5):312. learning abilities in adulthood. Nat Neurosci 9:268275.
Zhao LX, Zhang J, Cao F, Meng L, Wang DM, Li YH, Nan X, Ziv Y, Schwartz M (2008) Orchestrating brain-cell renewal: the
Jiao WC, Zheng M, Xu XH, Pei XT (2004) Modification role of immune cells in adult neurogenesis in health and
of the brain-derived neurotrophic factor gene: a portal disease. Trends Mol Med 14:471478.
Chapter 3

Neurogenesis in the Olfactory Epithelium

Bettina Malnic and Lucia Armelin-Correa

Contents neurodegenerative diseases or to induce regeneration


of injured axons.
3.1 Organization of the Mammalian Olfactory
System . . . . . . . . . . . . . . . . . . . 35 Keywords Globose basal cells Horizontal basal
3.2 The Olfactory Epithelium . . . . . . . . . . 36 cells Olfactory ensheating cells Olfactory
3.3 Neurogenesis in the Olfactory Epithelium . . . 38 epithelium Olfactory sensory neurons
3.4 The Olfactory Ensheating Cells . . . . . . . . 41
References . . . . . . . . . . . . . . . . . . . . 42
Abbreviations

GAP-43 growth associated protein of 43-kDa molec-


Abstract The olfactory sensory neurons in the olfac- ular mass
tory epithelium generally live for only about 3060 BG Bowmans glands
days. In addition, they are constantly killed by environ- GBC globose basal cell(s)
mental insults such as pathogens and toxic substances HBC horizontal basal cell(s)
and therefore need to be replaced throughout adult OE olfactory epithelium
life. This unique capability of neuronal regeneration is OEC olfactory ensheating cell(s)
due to the presence of olfactory stem cells localized OMP olfactory marker protein
in the basal layer of the olfactory epithelium which ONF olfactory nerve fibroblast(s)
proliferate and differentiate into new olfactory sen- OSN olfactory sensory neuron(s)
sory neurons. The newly generated neurons are able OST olfactory sulfotransferase
to extend their axons to the olfactory bulb, where NCAM neural cell adhesion molecule
they form synapses with the mitral cells. The olfac- Sus supporting cells
tory ensheating cells, which are specialized olfactory
glial cells, enclose the newly formed axons and help to
direct them through the cribriform plate to the olfac- 3.1 Organization of the Mammalian
tory bulb. Because the olfactory epithelium is readily
Olfactory System
accessible from its location in the nasal cavity, it has
been considered to be a potential source of basal stem
cells and olfactory ensheating cells which could be Mammals can discriminate a vast number of diverse
used in therapeutic applications for the treatment of odorants (Buck, 2000, 2004). Odorants, which are usu-
ally small organic and volatile molecules, are initially
detected by olfactory sensory neurons, located in a spe-
cialized olfactory epithelium lining the nasal cavity.
Each olfactory sensory neuron extends a single den-
B. Malnic ()
drite towards the apical region of the epithelium. The
Departamento de Bioqumica, Instituto de Qumica,
Universidade de So Paulo, So Paulo, Brazil tip of the dendrite carries cilia which are embedded in
e-mail: bmalnic@iq.usp.br the mucus layer of the epithelium, and are therefore

H. Ulrich (ed.), Perspectives of Stem Cells, 35


DOI 10.1007/978-90-481-3375-8_3, Springer Science+Business Media B.V. 2010
36 B. Malnic and L. Armelin-Correa

in direct contact with the environment. Each neuron locations in different individuals. These results indi-
also projects an axon to the olfactory bulb, where it cate that the information provided by different odorant
synapses with the dendrites of mitral cells, in spherical receptors in the nose is organized into a stereotyped
structures called glomeruli. From the olfactory bulb, sensory map in the olfactory bulb.
sensory information is transmitted to various higher It is not known yet how the sensory information pro-
brain regions via the axons of mitral cells, that project vided by the different odorant receptors is organized
to the olfactory cortex and other regions. beyond the olfactory bulb. That is, it is not known how
Odorants enter the nasal cavity and bind to odorant mitral cells that innervate one given glomerulus con-
receptors that are localized in the cilia of the olfactory nect to the olfactory cortex. Genetic tracing techniques
sensory neurons (Buck and Axel, 1991). The odor- (Horowitz et al., 1999; Boehm et al., 2005), where one
ant receptors belong to the super-family of G-protein transneuronal tracer is co-expressed with one given
coupled receptors and are extremely diverse in their type of odorant receptor should allow for the visual-
amino acid sequences, consistent with the ability to ization of the cortical neurons that receive input from
recognize a large variety of odorants. The odorant a particular odorant receptor. Analysis of the neuronal
receptor genes constitute the largest gene families in circuits of different odorant receptors should contribute
mammalian genomes, with around 1,000 functional to the understanding of how odorant perceptions are
receptors in rodents (Young et al., 2002; Godfrey et al., generated by the brain.
2004; Zhang et al., 2004; Niimura and Nei, 2005)
and 400 functional receptors in humans (Glusman
et al., 2001; Niimura and Nei, 2003; Malnic et al.,
2004). 3.2 The Olfactory Epithelium
A series of in situ hybridization experiments using
odorant receptors as molecular probes determined The olfactory epithelium (OE) is found on the nasal
how sensory information is organized in the olfactory septum and on a series of turbinates in the posterior
epithelium and olfactory bulb. These experiments sug- region of the nasal cavity (Fig. 3.1a). The OE can be
gested that each olfactory sensory neuron expresses divided into three major layers: an apical layer, which
only one out of the 1,000 odorant receptor genes contains the supporting cells, a medium layer, which
(Ressler et al., 1993; Vassar et al., 1993). This was later contains the mature and immature olfactory sensory
verified by examining gene expression in single neu- neurons, and a basal cell layer, which contains the
rons using single cell RT-PCR (Malnic et al., 1999). horizontal and globose basal cells (Fig. 3.1b).
The presence of small amounts of odorant receptor The supporting cells make up 1518% of the olfac-
mRNAs in the axon terminals of the olfactory neu- tory cells in the OE (Carr et al., 1991). They are
rons allowed for a visualization of the glomeruli that non-neuronal glial-like cells that extend processes
receive input from olfactory neurons expressing differ- from the surface to the basal lamina of the epithe-
ent odorant receptors. The results indicated that neu- lium. The supporting cells are capped by microvilli
rons expressing one same given odorant receptor type and contain abundant endoplasmic reticulum, but their
converge into one or few glomeruli at two specific sites functions are not entirely clear. They express mul-
in the olfactory bulb (Ressler et al., 1994; Vassar et al., tiple cytochrome P450s, and other biotransformation
1994). Since each mitral cell in the bulb is connected enzymes, that are involved in metabolizing foreign
to a single glomerulus, it receives incoming signals compounds, suggesting a role in detoxifying toxic sub-
from only one type of odorant receptor. These results stances present in the air, to which the OE is exposed
were confirmed by experiments that used molecular- (Yu et al., 2005). The supporting cells also phagocytose
genetic techniques to produce transgenic mice were the dead olfactory sensory neurons (OSN) and probably
olfactory sensory neurons expressing one same type provide support to the OSN. There is also a second
of odorant receptor co-express tau-lacZ in their axons, type of supporting cells, denominated microvillar cells,
allowing for the visualization of their axonal projec- which lack the abundant endoplasmic reticulum of
tion to the olfactory bulb (Mombaerts et al., 1996). supporting cells (Carr et al., 1991).
Interestingly, glomeruli that receive input from each Mature OSN are bipolar cells that extend a single
odorant receptor type show approximately the same ciliated dendrite to the epithelial surface and project
3 Neurogenesis in the Olfactory Epithelium 37

both, the olfactory epithelium and the vomeronasal


epithelium (Berghard et al., 1996). The expression of
the olfactory specific adenyl cyclase III (ACIII), and
other proteins involved in odorant signal transduc-
tion, also characterizes mature OSN (Menco et al.,
1992). Immature OSN also express growth associ-
ated protein of 43-kDa molecular weight (GAP-43)
(Verhaagen et al., 1989). The axons of the mature OSN
and their associated (glial) ensheating cells form bun-
dles that run anterior to posterior in the lamina propria
of the epithelium and project to the olfactory bulb,
where they synapse with the mitral cells, forming the
glomeruli.
The basal cells of the olfactory epithelium can
be subdivided into two different cell types, based
on cell morphology (Graziadei and Monti-Graziadei,
1979) and on the expression of specific cell mark-
ers (Holbrook et al., 1995). The horizontal basal cells
(HBC) are flat cells, with elongated nuclei, that stain
darkly with toluidine blue and proliferate at a low rate
(Holbrook et al., 1995). They lie deepest in the olfac-
tory epithelium, in a single-cell layer that is in direct
contact with the basal lamina (Holbrook et al., 1995).
The horizontal cells are the only cells in the olfactory
epithelium that express cytokeratins 5 and 14 (K5 and
Fig. 3.1 The olfactory epithelium. (a) Schematic diagram of
a coronal section through the mouse nasal cavity showing the K14) (Calof and Chikaraishi, 1989; Holbrook et al.,
septum and the turbinates. (b) Representation of the olfactory 1995).
epithelium which is populated by supporting cells (Sus), mature The globose basal cells (GBC) have round nuclei
and immature olfactory sensory neurons (OSNm and OSNim,
and show light staining with toluidine blue (Graziadei
respectively), two types of globose basal cells (GBC), the imme-
diate neuronal precursor and the progenitor cells (GBCinp and and Monti-Graziadei, 1979). GBC do not express
GBCprog, respectively), the horizontal basal cells (HBC), the cytokeratins, but they express NCAM, like the OSN.
olfactory ensheating cells (OEC) and the Bowmans gland cells They lie above the HBC and are the major proliferat-
(BG). Some of the cell specific markers are shown in paren-
ing population in the olfactory epithelium (Caggiano
thesis: OMP (olfactory marker protein), OST (olfactory sul-
fotransferase), GAP-43 (growth associated protein of 43-kDa et al., 1994). This population of cells contains the pro-
molecular weight), Ngn1 (neurogenin-1), Mash1 (mammalian genitors that divide and give rise to the OSN. The
achaete-scute homolog 1), K5/K14 (keratin 5 and keratin 14) progenitors sequentially express the proneuronal genes
Mash1 (these are denominated GBC progenitor cells
or GBCprog) and Ngn1 (denominated GBC immedi-
a single unmyelinated axon to the olfactory bulb of ate neuronal precurssor cells or GBCinp) (Fig. 3.1b)
the brain. The ciliated dendrites are the primary sites (Calof et al., 2002; Cau et al., 2002).
of odorant transduction. OSN can be identified by In addition, Bowmans glands are present in the
their expression of different proteins. For example, OE. They consist of acini and ducts that extend from
both mature and immature OSN express the neural cell the lamina propria through the epithelium to discharge
adhesion molecule (NCAM) (Key and Akeson, 1990), mucus secretions at the apical surface (Getchell and
but only the mature OSN express the olfactory marker Getchell, 1992). The mucus layer covering the olfac-
protein (OMP) (Margolis, 1972) (Fig. 3.1b). OMP is a tory epithelium, which is produced by the Bowmans
cytoplasmic protein that is abundantly and exclusively glands and by the supporting cells, is believed to cre-
expressed in the mature olfactory sensory neurons of ate an optimal environment for OSN function (Getchell
38 B. Malnic and L. Armelin-Correa

et al., 1984). These secretions are lipid rich and also that the right amount of cells will populate the OE stem
contain odorant binding proteins, which are small cell niche, while in the established tissue these factors
proteins that belong to a large family of ligand carrier play a role in maintaining a tight regulation of the
proteins, the lipocalins (Bignetti et al., 1985; Pevsner total number of OE cells (Beites et al., 2005). Indeed,
and Snyder, 1990). These proteins may enhance odor- after olfactory bulbectomy, which induces OSN apop-
ant detection by assisting in the solubilization and tosis, increased neuronal progenitor cell mitoses are
transport of the odorants in the mucus. observed (Leung et al., 2007), supporting the idea that
OSN death removes an inhibitory feedback signal that
normally acts to block progenitor cell proliferation.
Commitment and differentiation of the cells that
3.3 Neurogenesis in the Olfactory
originate the OSN is controlled by a series of genes
Epithelium that encode transcription factors of the basic helix-
loop-helix (bHLH) class which are homologues of the
The OE provides one of the few examples of contin- Drosophila proneural genes of the achaete-scute (asc)
uous neurogenesis in the adult nervous system. The and atonal (ato) complex, such as Mash1, neurogenin1
OSN are directly exposed to pathogens and toxic sub- (Ngn1) and NeuroD (Cau et al., 1997, 2002). The
stances present in the environment and need to be fre- commitment of basal cells to the OSN lineage is ini-
quently replaced. These neurons generally live for only tiated with the expression of Mash1. Accordingly, in
about 3060 days and are continuously replaced from Mash1/ embryos neuronal differentiation in the OE
stem cells localized in the basal region of the epithe- is blocked. There is increased cell death and cells do
lium (Graziadei and Monti-Graziadei, 1979; Calof and not express Ngn1 and NeuroD (Guillemot et al., 1993;
Chikaraishi, 1989; Caggiano et al., 1994). Cau et al., 1997). The Ngn1 mutant phenotype has
OE neurogenesis can be subdivided in two phases: revealed that this transcription factor is required for
the primary and the established neurogenesis (Beites NeuroD expression, which in turn promotes neuronal
et al., 2005). During the primary neurogenesis the differentiation of OSN (Cau et al., 2002).
olfactory placode, a multipotent group of cells that will A combination of single-cell RT-PCR and microar-
give rise to all cell types in the OE, is determined, ray was used to identify transcription factors that are
and the establishment of the OE neuronal lineage is downstream of Mash1 and are involved in the genera-
initiated. The interaction between the olfactory pla- tion of further cellular diversity of the OSN (Tietjen
codal epithelium and its associated mesenchyme is et al., 2003). In these experiments, cDNA libraries
essential for the generation of cellular and molecular were prepared from isolated single cell progenitors
diversity, as well as for the development of appropriate (expressing Mash1 and the cell division markers cdc2
axon trajectories in the peripheral portion of the olfac- and Ki67) or mature OSN (expressing OMP and odor-
tory pathway (Balmer and Lamantia, 2005). Signaling ant receptor). Probes prepared from these single-cell
molecules such as retinoic acid (RA), fibroblast growth libraries were hybridized to an Affymetrix microar-
factor 8 (FGF8), sonic hedgehog (Shh) and bone mor- ray containing a large number of mouse probe sets.
phogenetic protein 4 (BMP4) are involved in this initial Transcripts enriched in the progenitor cells versus
events (Balmer and Lamantia, 2005). mature OSN were identified. Some of these genes
Established neurogenesis is initiated in mice around (such as Notch1, Lhx2, Hes6) are involved in cell
E13.5E14.5, when the OE becomes organized into proliferation and were shown to be dependent on
its mature pattern. The nuclei of the supporting cells Mash1 expression, since their expression is abolished
become organized in a single apical layer and there in Mash1/ mice.
is a decrease in the overall number of mitotic nuclei, There are several transcription factors expressed in
which are localized primarily to the basal compartment the OE that are not involved in OSN differentiation,
of the OE. Signaling molecules such as BMP2, 4 and but are required for OSN maturation. Mature OSN
7; and growth and differentiation factor 11 (GDF11) express one single type of odorant receptor (Chess
exert important roles in this stage (Shou et al., 2000; et al., 1994; Malnic et al., 1999), and olfactory sig-
Wu et al., 2003). Factors that predominate during the nal transduction proteins that are required for proper
primary olfactory neurogenesis are likely to determine neuronal function. The expression of odorant receptor
3 Neurogenesis in the Olfactory Epithelium 39

genes is segregated in the dorso-ventral axis of the OE,


so that OSN expressing one given odorant receptor are
contained within one of four distinct zones of the OE
(Ressler et al., 1993; Vassar et al., 1993). Interestingly,
some transcripts enriched in the progenitor cells, such
as Pax6, Sox11 and Eya2, were found to be expressed
in specific zones in E15 OE, and this pattern of expres-
sion was maintained in the Mash1/ mice (Tietjen
et al., 2003). These transcripts may be involved in the
zonal organization of odorant receptor expression in
the OE.
The O/E like proteins are transcription factors that
bind to promoter regions of several genes expressed
in mature OSN, such as OMP, ACIII, Golf and odor-
ant receptors (Kudrycki et al., 1993; Wang and Reed,
1993; Michaloski et al., 2006), and therefore must be
involved in the regulation of the expression of these
genes. Another characterized gene that is expressed
downstream of Mash1 is Zfp423/OAZ (Cheng and
Reed, 2007). This transcription factor is transiently
expressed in newly differentiating neurons and is
required for maturation of the OSN. Zfp423/OAZ binds
to the O/E like proteins and inhibits their binding to
the promoters of the olfactory specific genes (Tsai
and Reed, 1997), thereby preventing the premature
differentiation of the immature OSN.
The newly-generated neurons are result of mitoses
that occur near the basal lamina of the OE, and the pro-
gression of neuronal maturation occurs from the basal
to the apical portion of the tissue (Fig. 3.2). The basal
region of the epithelium contains cells with distinct
proliferating rates: stem cells thought to undergo slow,
asymmetric cell divisions (the HBC), and a popula-
Fig. 3.2 Neuronal regeneration in the olfactory epithelium.
tion of rapidly-dividing transit amplifying progenitors (a) In physiological OSN turnover or under mild injury condi-
which are committed to a neuronal cell fate (the GBC). tions, where only the OSN are ablated, GBC have the capacity
It has been difficult to define exactly which one of to regenerate the missing neurons. (b) Under severe olfactory
epithelium lesions, that ablate the OSN, supporting cells and the
these two distinct cell types represents the stem cell
GBC, HBC are able to regenerate GBC, OSN, Bowmans gland
population of the olfactory epithelium (Calof et al., and supporting cells. Model based on Beites et al. (2005) and
1998). HBC, which undergo slow cell division, a hall- Leung et al. (2007)
mark of other well-characterized adult tissue stem
cell (Weissman, 2000), express cytokeratins (Holbrook progenitor cells which sequentially express the proneu-
et al., 1995), integrins (Carter et al., 2004) and also ral genes Mash1 and Ngn1 (Cau et al., 1997). Studies
respond to epidermal growth factor (EGF), a factor of both developmental and regeneration models sug-
shown to be mitogenic for stem cells in the CNS. gest that the Mash1 expressing cells are daughters of
Besides, when in culture, HBC have demonstrated the the OE stem cell (Gordon et al., 1995). Some retroviral
capacity to generate neurons as well as non-neuronal lineage analyses, including models of injury induced
cells (Sicard et al., 1998; Carter et al., 2004). GBC, OE regeneration as well as cell transplantation experi-
on the other hand, even though cytokeratin-negative, ments, supported the idea that a subpopulation of GBC
contain the two populations of transit amplifying was a multipotent progenitor giving rise not only to
40 B. Malnic and L. Armelin-Correa

OSN, but also to supporting cells (Caggiano et al., mice, bearing the GFP coding sequence replacing
1994; Huard et al., 1998; Chen et al., 2004). either Mash1 or Ngn1 genes, were crossed with K5-
Recently, the technology of homologous recombi- CreERT2 /R26R animals. These triple heterozygous
nation was used to clarify the controversy that exists animals, once subjected to severe OE lesions, showed
concerning the identity of the OE stem cells. Leung GBC co-expressing GFP and LacZ. These findings are
and cols (2007) performed an in vivo fate mapping consistent with in vitro studies (Carter et al., 2004),
of HBC exploiting the restricted expression of cytok- and provide in vivo evidence that HBC can give rise to
eratin K5 gene in post natal HBC. A transgenic both neuronal and non-neuronal cell types in the OE.
mouse expressing a tamoxifen-inducible Cre recombi- HBC undergo a regulated transient proliferative burst
nase under the control of the cytokeratin K5 promoter to regenerate OE when an extensive cellular depletion
(K5-CreER T2 ) was crossed to a strain containing a occurs, while under bulbectomy no change is detected
lox-STOP-lox LacZ reporter gene at the Rosa26 locus in their proliferation rate (Leung et al., 2007).
(R26R), a locus from which genes can be ubiqui- The study of OE regeneration in K5-CreERT2 /R26R
tously expressed. Injection of tamoxifen into double- animals provides a new model for neural stem cell
heterozygous K5-CreERT2 /R26R animals led to the dynamics in which distinct cell populations mediate
induction of Cre recombinase in HBC, and resulted normal neuronal turnover and neuronal replacement
in CreER mediated excision of the lox-flanked STOP in traumatic injuries. It also opens the possibility to
sequence at the LacZ reporter locus. This permanent perform other cell specific lineage tracing in the OE.
activation of the LacZ reporter gene in HBC and all of Lineage tracing of GBC at specific commitment points
their descendents, allows for a HBC-specific lineage would help to elucidate if these progenitor cells also
tracing strategy. originate supporting cells and Bowmans glands, and
Analysis of HBC contribution to the natural OSN at what point they do so, or if HBC have another cell
turnover in these double-heterozygous animals indi- intermediate to originate these non-neuronal cell types.
cated that these cells, under normal conditions, remain Also it is still a mystery which cells are the true pro-
quiescent in the OE. Since specific lesions in the OE genitors of OEC. There are studies which suggest that
lead to different dynamics of cell proliferation and OEC may originate from stem cells in the OE (Beites
differentiation (Murray and Calof, 1999), three dif- et al., 2005), and it remains to be determined whether
ferent OE regeneration models were tested in these HBC are these progenitors. The complete understand-
transgenic animals: bulbectomy, exposure to methyl ing of the differentiation pathway of neuronal and
bromide and injection of methimazole. Animals which non-neuronal cells in the OE should therefore aid in the
suffered bulbectomy did not show a different HBC potential application of OE stem and progenitor cells
dynamic from that of control animals, suggesting that in the treatment of CNS lesions and neurodegenerative
HBC remain quiescent under specific ablation of OSN diseases.
in OE. In this condition, only mature OSN are lost, and The mechanisms of OE regeneration in humans are
new neurons are probably regenerated from another however not as well understood as in rodents. Although
pool of progenitor cells, presumably the GBC (Gordon the human OE is similar to the rodent OE regard-
et al., 1995) (Fig. 3.2a). But under extensive OE ing its cellular and molecular composition, there are
lesions, as those caused by exposure to methyl bro- a few differences, which suggest that neurogenesis
mide or methimazole, which damage all cell types of OSN in humans may differ from that in rodents.
in the OE including the neuronal progenitors GBC, First, the molecular and morphological differences that
transgenic animals showed clusters of marked cells exist between GBC and HBC in rodents are not found
throughout the OE including GBC, OSN, Bowmans in humans (Hahn et al., 2005). In humans, the cells
glands and supporting cells (Fig. 3.2b). OSN and adjacent to the basal lamina show no morphological
supporting cells were respectively identified by co- differences from the cells located above them. All cells
immunostaining with antibodies anti-OMP and anti- in the basal layer are round and resemble the rodent
olfactory sulfotransferase (OST). Bowmans gland GBC, with no cells showing the morphological char-
cells were identified based on their peculiar localiza- acteristics of the rodent HBC. Also, differently from
tion regarding the neuroepithelium basal lamina. To what is observed in rodents, all of these GBC-like basal
determine whether GBC lie in the HBC-initiated path- cells express cytokeratin-5. Second, in humans, the
way of differentiation into mature neurons, knock-in laminar organization of immature and mature neurons
3 Neurogenesis in the Olfactory Epithelium 41

observed in rodents, does not exist. In rodents, imma- closely associated with the axons of the OSN. In this
ture and mature OSN exist in two layers, with the case, hundreds of axons are enclosed by only a thin
immature OSN being localized closer to the basal lam- OEC cytoplasmatic process (Field et al., 2003). When
ina. In humans, the immature and mature OSN were the olfactory nerve reaches the olfactory bulb, the basal
found to be dispersed throughout the OE (Hahn et al., lamina and the ONF are reflected off and the OEC
2005). processes open up to interdigitate with the astrocytic
The fact that human OE biopsies can be obtained processes which cover the brain, enabling the olfac-
from living human subjects should contribute to the tory axons to enter the olfactory bulbs (Raisman, 1985;
study of human neuronal differentiation. Because of Valverde et al., 1992). Microscopic investigations of
its regenerative capacity and the fact that it is read- the tubular arrangement of the OEC/ONF during OSN
ily accessible from its location in the nasal cavity, the degeneration and regeneration revealed that it remains
human OE represents a source of mitotically active stable, with no indications of cell death, division or
neuronal progenitors, which once differentiated into migration, differing completely from Schwann cells
neurons, could be used in potential therapies of autol- in pheripheral nerve lesions (Williams et al., 2004; Li
ogous transplantation for the treatment of neurodegen- et al., 2005). The persistence of the OEC tunnels con-
erative diseases (Marshall et al., 2006). Neurospheres tributes to the effectiveness of the regeneration of the
obtained from cultured human OE have already been olfactory axons.
shown to differentiate into mature neurons, emphasiz- During development the OEC, like the other cells in
ing the potential use of adult human OE as a source of the OE, originate from the olfactory placode (Farbman
progenitors for future therapies (Othman et al., 2005; and Squinto, 1985). Even though the OEC morpho-
Zhang et al., 2006). logically resemble Schwann cells, they physiologically
do not produce myelin and they demonstrate a highly
plastic and heterogeneous morphology, being capa-
ble of acquiring in vitro, an astrocyte-like morphology
3.4 The Olfactory Ensheating Cells (Richter and Roskams, 2008). Microarray analysis of
the transcriptional profiling of OEC, Schwann cells
Continuous neurogenesis in the OE requires that not and astrocytes revealed that these cells have overlap-
only OSN are replaced, but also that the axons of the ping but distinct gene expression profiles, and that
newly formed OSN target the correct glomerulus in OEC are transcriptionally closer to Schwann cells than
the olfactory bulb. The ability of OSN to regenerate to astrocytes (Vincent et al., 2005). However, OEC
their axonal connections with the mitral cells in the have a transcriptome more similar to astrocytes than
bulb throughout life is believed to be in part because Schwann cells do (Vincent et al., 2005), and while
the olfactory axons are ensheated by specialized glial the co-culture of randomly dispersed Schwann cells
cells, the olfactory ensheating cells (OEC). and astrocytes shows cell type-specific territories, co-
The axons of OSN form loose bundles at the OE cultured OEC and astrocytes intermingle, indicating
basal lamina. Once these axons have entered the lam- that OEC integrate more effectively than Schwann
ina propria they are enwrapped by OEC, which guide cells into astrocytic environments (Lakatos et al.,
them through the cribriform plate to the olfactory bulb 2000).
(Boyd et al., 2003; Franssen et al., 2007). OEC consist Nerve fibers usually require an aligned glial path-
of a thin cytoplasmatic sheet that encloses a tunnel- way to establish connections between neurons. During
like space through which hundreds of unmyelinated a spinal cord or dorsal root lesion this pathway is dis-
olfactory axons run (Field et al., 2003). Therefore, rupted, and astrocytes immediately seal off the breach,
the olfactory nerves are constituted by olfactory axons to recover the external wall of the nervous system,
enclosed by a series of end-to-end OEC and overlaid which avoids invasion of non-neural cells and other
by the basal lamina and by olfactory nerve fibrob- damaging materials (Raisman and Li, 2007). However,
lasts (ONF) (Li et al., 2005). While in the peripheral the formation of a fibrous scar or cavity at the site of
nerves Schwann cells enwrap around 20 axons that are injury abrogates the repair of the nerve connections,
separated one from another by folds of Schwann cells because the glial pathway required for the growth of
cytoplasm, in the olfactory nerve, the OEC are not so the nerve fibers are disrupted.
42 B. Malnic and L. Armelin-Correa

It has been shown that axons are capable of sprout- OEC transplantation in animals that suffered spinal
ing after spinal cord injury, and that transplantation cord injuries demonstrated that OEC minimize cavity
of Schwann cells amplifies this response, but in these and scar formation, induce local angiogenesis and dif-
experiments the axons show minimal capacity to reen- ferential axon sprouting and regeneration (Ramer et al.,
ter the host CNS (Li and Raisman, 1994; Keyvan- 2004), and improved animals behavioral, breathing
Fouladi et al., 2005). The fact that there is a continuous and climbing capacities (Keyvan-Fouladi et al., 2003;
neurogenesis in the olfactory system and that OEC Li et al., 2003). These findings suggest that OEC are
play a crucial role in this process, maintaining the either capable of promoting sprouting or facilitating
olfactory nerve structure and interacting with astro- plasticity and neuroprotection in injured spinal cord,
cytes in the olfactory bulb, prompted researchers to which may lead to improved behavioral outcomes, but
study their value as theraupeutic agents for treatment the capacity of OEC to promote spinal cord axonal
of CNS injuries (reviewed in Franssen et al., 2007; regeneration is still questioned (Richter and Roskams,
Raisman and Li, 2007; Richter and Roskams, 2008). 2008).
It was shown that OEC are capable of producing a Several important points must be taken into con-
series of factors which support axonal growth, such as sideration when trying to understand the potential
nerve growth factor (NGF), brain derived neurotrophic effect of OEC in promoting axonal regeneration: the
factor (BDNF), neurotrophin 4/5 (NT-4/5) and also size and type of lesion inflicted to the nervous tissue,
other growth factors, such as fibroblast growth factor 2 the type of nervous tract involved and collateral axon
(FGF-2) (reviewed in Richter and Roskams, 2008). A branching and differences in the preparations of OEC
different number of in vitro assays have shown that co- that will be transplanted. There are at least two sources
culture of not only OSN, but also CNS neurons, with of OEC for transplantation: the lamina propria of the
OEC have led to increased neurite growth (reviewed in OE and the olfactory bulb. OEC may be transplanted
Richter and Roskams, 2008). Besides, OEC produce purified or associated with the basal lamina and ONF
several components of the olfactory system extracellu- which physiologically enclose the olfactory nerve.
lar matrix which may also contribute to neuronal axon These cells maybe cultured or directly transplanted,
growth. and the delay between injury and cell application
OEC transplantation consistently induces a more also impacts the OEC functions and their ability to
permissive scar boundary for the extension of growing promote regeneration.
axons than that induced by Schwann cell transplan- Despite some conflicting results the current experi-
tation (Raisman and Li, 2007; Richter and Roskams, mental observations raise hope that transplantation of
2008). When a lesion occurs, astrocytes and their pro- OEC will contribute to the treatment of spinal cord and
cesses form a dense mass around it, and transplantation dorsal root injuries. With these perspectives in mind
of OEC leads to reorganization of the astrocytic scar to some neurosurgical teams have established clinical tri-
form a newly aligned pathway (Raisman and Li, 2007; als involving autologous transplantation of olfactory
Richter and Roskams, 2008). OEC are also capable mucosa or OEC in patient with spinal cord injuries
of producing myelin in vitro and in vivo (reviewed in (Lima et al., 2006; Mackay-Sim et al., 2008). Future
Richter and Roskams, 2008). investigations should reveal the potential of using OEC
It has been shown that transplanted OEC can inter- for therapeutic applications in axonal repair.
act with host astrocytes to produce a bridge through
the dorsal root entry zone (DREZ) along which axons
are able to regenerate into the dorsal columns (Li
et al., 2004). But these results have been challenged by References
studies using labeled OEC and more complex axonal
tracing techniques which suggested that apparent long- Balmer C, Lamantia A-S (2005) Noses and neurons: induction,
distance afferent in-growth could be the result of morphogenesis, and neuronal differentiation in the peripheral
olfactory pathway. Dev Dyn 234:464481.
spared fibers, leaking label, or growth fibers only along
Beites C, Kawauchi S, Crocker C, Calof A (2005) Identification
physically disrupted injection tracts (Gomez et al., and molecular regulation of neural stem cells in the olfactory
2003; Ramer et al., 2004b; Riddell et al., 2004). epithellium. Exp Cell Res 306:309316.
3 Neurogenesis in the Olfactory Epithelium 43

Berghard A, Buck LB, Liman ER (1996) Evidence for distinct Franssen E, de Bree F, Verhaagen J (2007) Olfactory ensheathing
signaling mechanisms in two mammalian olfactory sense glia: their contribution to primary olfactory nervous sys-
organs. Proc Natl Acad Sci USA 93:23652369. tem regeneration and their regenerative potential following
Bignetti E, Cavaggioni A, Pelosi P, Persaud K, Sorbi R, transplantation into the injured spinal cord. Brain Res Rev
Tirindelli R (1985) Purification and characterisation of 56:236258.
an odorant-binding protein from cow nasal tissue. Eur J Getchell M, Getchell T (1992) Fine structural aspects of secre-
Biochem 149:227231. tion and extrinsic innervation in the olfactory mucosa.
Boehm U, Zou Z, Buck L (2005) Feedback loops link odor Microsc Res Tech 23:111127.
and pheromone signaling with reproduction. Cell 123: Getchell T, Margolis F, Getchell M (1984) Perireceptor and
683695. receptor events in vertebrage olfaction. Prog Neurobiol
Boyd J, Skihar V, Kawaja M, Doucette R (2003) Olfactory 23:317345.
ensheating cells: historical perspective and therapeutic Glusman G, Yanai I, Rubin I, Lancet D (2001) The complete
potential. Anat Rec B New Anat 271:4960. human olfactory subgenome. Genome Res 11:685702.
Buck LB (2000) The molecular architecture of odor and Godfrey PA, Malnic B, Buck LB (2004) The mouse olfac-
pheromone sensing in mammals [comment]. Cell 100: tory receptor gene family. Proc Natl Acad Sci USA 101:
611618. 21562161.
Buck LB (2004) Olfactory receptors and odor coding in mam- Gomez V, Averill S, King V, Yang Q, Doncel Perez E, Chacon
mals. Nutr Rev 62:S184S188. S, Ward R, Nieto-Sampedro M, Priestley J, Taylor J (2003)
Buck L, Axel R (1991) A novel multigene family may encode Transplantation of olfactory ensheathing cells fails to pro-
odorant receptors: a molecular basis for odor recognition. mote significant axonal regeneration from dorsal roots into
Cell 65:175187. the rat cervical cord. J Neurocytol 32:5370.
Caggiano M, Kauer J, Hunter D (1994) Globose basal cells are Gordon M, Mumm J, Davis R, Holcomb J, Calof A (1995)
neuronal progenitors in the olfactory epithelium: a lineage Dynamics of MASH1 expression in vitro and in vivo sug-
analysis using a replication-incompetent retrovirus. Neuron gest a non-stem cell site of MASH1 action in the olfactory
13:339352. receptor neuron lineage. Mol Cell Neurosci 6:363379.
Calof A, Bonnin A, Crocker C, Kawauchi S, Murray R, Shou Graziadei PPC, Monti-Graziadei GA (1979) Neurogenesis and
J, Wu H-H (2002) Progenitor cells of the olfactory receptor neuron regeneration in the olfactory system of mammals.
neuron lineage. Microsc Res Tech 58:176188. I. Morphological aspects of differentiation and structural
Calof AL, Chikaraishi DM (1989) Analysis of neurogenesis organization of the olfactory sensory neurons. J Neurocytol
in a mammalian neuroepithelium: proliferation and differ- 8:118.
entiation of an olfactory neuron precursor in vitro. Neuron Guillemot F, Lo L, Johnson J, Auerbach A, Anderson D, Joyner
3:115127. A (1993) Mammalian achaete-scute homolog 1 is required
Calof A, Mumm J, Rim P, Shou J (1998) The neuronal stem cell for the early development of olfactory and autonomic neu-
of the olfactory epithelium. J Neurobiol 36:190205. rons. Cell 124:463476.
Carr V, Farbman A, Colletti L, Morgan J (1991) Identification Hahn C-G, Han L-Y, Rawson N, Mirza N, Borgmann-Winter
of a new non-neuronal cell type in rat olfactory epithelium. K, Lenox R, Arnold S (2005) In vivo and in vitro neuroge-
Neuroscience 45:433449. nesis in human olfactory epithelium. J Comp Neurol 483:
Carter L, MacDonald J, Roskams A (2004) Olfactory horizontal 154163.
basal cells demonstrate a conserved multipotent progenitor Holbrook E, Szumowski K, Schwob J (1995) An immunochem-
phenotype. J Neurosci 24:56705683. ical, ultrastructural, and developmental characterization of
Cau E, Casarosa S, Guillemot F (2002) Mash1 and Ngn1 con- the horizontal basal cells of rat olfactory epithelium. J Comp
trol distinct steps of determination and differentiation in Neurol 363:129146.
the olfactory sensory neuron lineage. Development 129: Horowitz LF, Montmayeur J-P, Echelard Y, Buck LB (1999) A
18711880. genetic approach to trace neural circuits. Proc Natl Acad Sci
Cau E, Gradwohl G, Fode C, Guillemot F (1997) Mash1 acti- USA 96:31943199.
vates a cascade of bHLH regulators in olfactory neuron Huard J, Youngentob S, Goldstein B, Luskin M, Schwob J
progenitors. Development 124:16111621. (1998) Adult olfactory epithelium contains multipotent pro-
Chen X, Fang H, Schwob J (2004) Multipotency of purified, genitors that give rise to neurons and non-neural cells. J
transplanted globose basal cells in olfactory epithelium. Comp Neurol 400:469486.
J Comp Neurol 469:457474. Key B, Akeson R (1990) Olfactory neurons express a unique
Cheng L, Reed R (2007) Zfp423/OAZ participates in a devel- glycosylated form of the neural cell adhesion molecule
opmental switch during olfactory neurogenesis. Neuron (N-CAM). J Cell Biol 110:17291743.
54:547557. Keyvan-Fouladi N, Raisman G, Li Y (2003) Functional repair of
Chess A, Simon I, Cedar H, Axel R (1994) Allelic inactiva- the corticospinal tract by delayed transplantation of olfactory
tion regulates olfactory receptor gene expression. Cell 78: ensheathing cells in adult rats. J Neurosci 23:94289434.
823834. Keyvan-Fouladi N, Raisman G, Li Y (2005) Delayed repair of
Farbman A, Squinto L (1985) Early development of olfactory corticospinal tract lesions as an assay for the effectiveness of
receptor cell axons. Brain Res 351:205213. transplantation of Schwann cells. Glia 51:306311.
Field P, Li Y, Raisman G (2003) Ensheatment of the olfactory Kudrycki K, Stein-Izsak C, Behn C, Grillo M, Akeson R,
nerves in the adult rat. J Neurocytol 32:317324. Margolis F (1993) Olf-1 binding site: characterization of
44 B. Malnic and L. Armelin-Correa

an olfactory neuron-specific promoter motif. Mol Cell Biol Othman M, Lu C, Klueber K, Winstead W, Roisen F (2005)
13:30023014. Clonal analysis of adult human olfactory neurosphere form-
Lakatos A, Franklin R, Barnett S (2000) Olfactory ensheathing ing cells. Biotech Histochem 80:189200.
cells and Schwann cells differ in their in vitro interactions Pevsner J, Snyder S (1990) Odorant binding protein: odorant
with astrocytes. Glia 32:214225. transport function in the nasal epithelium. Chemical Senses
Leung C, Coulombe P, Reed R (2007) Contribution of olfactory 15:217222.
neural stem cells to tissue maintenance and regeneration. Nat Raisman G (1985) Specializedneuroglial arrangement may
Neurosci 6:720726. explain the capacity of vomeronasal axons to reinnervate
Li Y, Carlstedt T, Berthold CH, Raisman G (2004) Interaction of central neurons. Neuroscience 14:237254.
transplanted olfactory-ensheathing cells and host astrocytic Raisman G, Li Y (2007) Repair of neural pathways by olfactory
processes provides a bridge for axons to regenerate across ensheating cells. Nat Rev Neurosci 8:312319.
the dorsal root entry zone. Exp Neurol 188:300308. Ramer L, Au E, Richter M, Liu J, Tetzlaff W, Roskams A
Li Y, Field P, Raisman G (2005) Olfactory ensheathing cells (2004a) Peripheral olfactory ensheathing cells reduce scar
and olfactory nerve fibroblasts maintain continuous open and cavity formation and promote regeneration after spinal
channels for regrowth of olfactory nerve fibres. Glia 52: cord injury. J Comp Neurol 473:115.
245251. Ramer L, Richter M, Roskams A, Tetzlaff W, Ramer M (2004b)
Li Y, Raisman G (1994) Schwann cells induce sprouting in Peripherally-derived olfactory ensheathing cells do not pro-
motor and sensory axons in the adult rat spinal cord. J mote primary afferent regeneration following dorsal root
Neurosci 14:40504063. injury. Glia 47:189206.
Li Y, Sauve Y, Li D, Lund R, Raisman G (2003) Transplanted Ressler KJ, Sullivan SL, Buck LB (1993) A zonal organiza-
olfactory ensheathing cells promote regeneration of cut adult tion of odorant receptor gene expression in the olfactory
rat optic nerve axons. J Neurosci 23:77837788. epithelium. Cell 73:597609.
Lima C, Pratas-Vital J, Escada P, Hasse-Ferreira A, Capucho Ressler KJ, Sullivan SL, Buck LB (1994) Information cod-
C, Peduzzi J (2006) Olfactory mucosa autografts in human ing in the olfactory system: evidence for a stereotyped and
spinal cord injury: a pilot clinical study. J Spinal Cord Med highly organized epitope map in the olfactory bulb. Cell 79:
29:191203. 12451255.
Mackay-Sim A, Feron F, Cochrane J, Bassingthwaighte L, Richter M, Roskams A (2008) Olfactory ensheathing cell trans-
Bayliss C, Davies W, Fronek P, Gray C, Kerr G, Licina P, plantation following spinal cord injury: hype or hope? Exp
Nowitzke A, Perry C, Silburn P, Urquhart S, Geraghty T Neurol 209:353367.
(2008) Autologous olfactory ensheathing cell transplanta- Riddell J, Enriquez-Denton M, Toft A, Fairless R, Barnett S
tion in human paraplegia: a 3-year clinical trial. Brain 131: (2004) Olfactory ensheathing cell grafts have minimal influ-
23762386. ence on regeneration at the dorsal root entry zone following
Malnic B, Godfrey PA, Buck LB (2004) The human olfac- rhizotomy. Glia 47:150167.
tory receptor gene family. Proc Natl Acad Sci USA 101: Shou J, Murray R, Rim P, Calof A (2000) Opposing effects of
25842589. bone morphogenetic proteins on neuron production and sur-
Malnic B, Hirono J, Sato T, Buck LB (1999) Combinatorial vival in the olfactory receptor neuron lineage. Development
receptor codes for odors. Cell 96:713723. 127:54035413.
Margolis F (1972) A brain protein unique to the olfactory bulb. Sicard G, Feron F, Andrieu J, Holley A, Mackay-Sim A (1998)
Proc Natl Acad Sci USA 69:12211224. Generation of neurons from a nonneuronal precursor in
Marshall C, Lu C, Winstead W, Zhang X, Xiao M, Harding adult olfactory epithelium in vitro. Ann N Y Acad Sci 855:
G, Klueber K, Roisen F (2006) The therapeutic potential 223225.
of human olfactory-derived stem cells. Histol Histopatol Tietjen I, Rihel J, Cao Y, Koentges G, Zakhary L, Dulac C (2003)
21:633643. Single-cell transcriptional analysis of neuronal progenitors.
Menco BPM, Bruch RC, Dau B, Danho W (1992) Ultrastructural Neuron 38:161175.
localization of olfactory transduction components: the G Tsai R, Reed R (1997) Cloning and functional characterization
protein subunit Golf and type III adenylyl cyclase. Neuron of Roaz, a zinc finger protein that interacts with O/E-1 to
8:441453. regulate gene expression: implications for olfactory neuronal
Michaloski J, Galante P, Malnic B (2006) Identification of poten- development. J Neurosci 17:41594169.
tial regulatory motifs in odorant receptor genes by analysis of Valverde F, Santacana M, Heredia M (1992) Formation
promoter sequences. Genome Res 16:10911098. of an Olfactory Glomerulus: Morphological Aspects of
Mombaerts P, Wang F, Dulac C, Chao S, Nemes A, Mendelsohn Development and Organization. Neuroscience 49:255275.
M, Edmondson J, Axel R (1996) Visualizing an olfactory Vassar R, Chao S, Sitcheran R, Nunez J, Vosshall L, Axel R
sensory map. Cell 87:675686. (1994) Topographic organization of sensory projections to
Murray R, Calof A (1999) Neuronal regeneration: lessons from the olfactory bulb. Cell 79:981991.
the olfactory system. Cell Dev Biol 10:421431. Vassar R, Ngai J, Axel R (1993) Spatial segregation of odorant
Niimura Y, Nei M (2003) Evolution of olfactory receptor genes receptor expression in the mammalian olfactory epithelium.
in the human genome. Proc Natl Acad Sci USA 100: Cell 74:309318.
1223512240. Verhaagen J, Oestreicher A, Gispen W, Margolis FL (1989) The
Niimura Y, Nei M (2005) Comparative evolutionary analysis of expression of the growth associated protein B50/GAP43 in
olfactory receptor genes clusters between humans and mice. the olfactory system of neonatal and adult rats. J Neurosci
Gene 346:1321. 9:683691.
3 Neurogenesis in the Olfactory Epithelium 45

Vincent A, Taylor J, Choi-Lundberg D, West A, Chuah M (2005) Young JM, Friedman C, Williams EM, Ross JA, Tonnes-Priddy
Genetic expression profile of olfactory ensheathing cells is L, Trask BJ (2002) Different evolutionary processes shaped
distinct from that of Schwann cells and astrocytes. Glia the mouse and human olfactory receptor gene families. Hum
51:132147. Mol Genet 11:535546.
Wang M, Reed R (1993) Molecular cloning of the olfactory neu- Yu T, McIntyre J, Bose S, Hardin D, Owen M, McClintock T
ronal transcription factor Olf-1 by genetic selection in yeast. (2005) Differentially expressed transcripts from phenotypi-
Nature 364:121126. cally identified olfactory sensory neurons. J Comp Neurol
Weissman I (2000) Stem cells: units of development, units of 483:251261.
regeneration, and units in evolution. Cell 100:157168. Zhang X, Klueber K, Guo Z, Cai J, Lu C, Winstead W, Qiu
Williams S, Franklin R, Barnett S (2004) Response of olfac- M, Roisen F (2006) Induction of neuronal differentiation of
tory ensheathing cells to the degeneration and regeneration adult human olfactory neuroepithelial-derived progenitors.
of the peripheral olfactory system and the involvement of the Brain Res 10731074:109119.
neuregulins. J Comp Neurol 470:5062. Zhang X, Rodriguez I, Mombaerts P, Firestein S (2004) Odorant
Wu H-H, Ivkovic S, Murray R, Jaramillo S, Lyons K, Johnson J, and vomeronasal receptor genes in two mouse genome
Calof A (2003) Autoregulation of neurogenesis by GDF11. assemblies. Genomics 83:802811.
Neuron 37:197207.
Chapter 4

Cell Diversification During Neural Crest Ontogeny:


The Neural Crest Stem Cells

Elisabeth Dupin, Giordano W. Calloni, and Nicole M. Le Douarin

Contents and progenitors in the NC cells (NCC) that undergo


migration during embryogenesis. We focus on one
4.1 Introduction . . . . . . . . . . . . . . . . . 47 striking property of the cephalic NCC, i.e., the capac-
4.2 Formation of the Neural Crest, a Structure ity to yield chondrocytes and bone-forming cells in
Between CNS and Epidermis in Vertebrate addition to skin melanocytes and nerve cells of the
Embryos . . . . . . . . . . . . . . . . . . 49
peripheral nervous system (PNS). We also emphasize
4.3 Identification of Neural Crest Progenitors and
Stem Cells by In Vitro Single Cell Cultures . . 49 the role of environmental cues in ensuring the survival
4.4 Pluripotent Neural Crest Stem Cells in and directing the differentiation of these progenitors in
Tissues and Organs; Developmental Remnant their various sites of homing. Finally we also include
and Potential Source of Stem Cells for
recent advances that uncover stem cell properties of
Regenerative Medicine . . . . . . . . . . . . 51
4.5 In Vivo and In Vitro Demonstration of the NC-derived cells in the adult body. On one hand,
Influence of Environmental Cues on the differentiated cell types of NC origin are prone to ded-
Differentiation of Neural Crest Derivatives . . 53 ifferentiate, as shown by in vitro experiments; on the
4.5.1 In Vivo Studies . . . . . . . . . . . . 53
other hand, undifferentiated multipotent NCC persist
4.5.2 In Vitro Studies . . . . . . . . . . . . 53
4.6 Plasticity and Dedifferentiation Ability of Neural in many tissues and organs. These findings suggest that
Crest-Derived Differentiated Cells . . . . . . 54 a diversity of NC-derived cells could be mobilized to
4.7 Concluding Remarks . . . . . . . . . . . . 55 function as stem cells in adult tissue repair.
References . . . . . . . . . . . . . . . . . . . . 55
Keywords Chondrocyte Clonal culture
Multipotentiality Mesenchymal Neural
Abstract The neural crest (NC) is a transitory ecto- Osteoblast Quail embryo
dermal structure that forms in the vertebrate embryo
at the junction between the presumptive CNS and epi- Abbreviations
dermis. As it is at the origin of very diverse (neural
and non-neural) cell types in adult tissues, the NC has ET3 endothelin-3
attracted for long the interest of developmental biolo- NC neural crest
gists and is a valuable model to investigate stem cell NCC neural crest cell(s)
biology. Here we review a number of data mainly NF neural fold
provided by in vitro single cell culture experiments, PNS peripheral nervous system
which led to characterizing multipotent stem cells Shh Sonic Hedgehog

4.1 Introduction
E. Dupin ()
CNRS UPR2197 Laboratoire Dveloppement, Evolution et
The neural crest (NC) is a transitory structure of
Plasticit du Systme Nerveux, Institut de Neurobiologie Alfred
Fessard, 91198 Gif-sur-Yvette, France the vertebrate embryo. The cells that enter in the
e-mail: dupin@inaf.cnrs-gif.fr constitution of the crest are pluripotent and endowed

H. Ulrich (ed.), Perspectives of Stem Cells, 47


DOI 10.1007/978-90-481-3375-8_4, Springer Science+Business Media B.V. 2010
48 E. Dupin et al.

with migratory properties. After the NC is formed at


the border of the neural plate, forming an ectoder-
mal area intermediate between the presumptive neural
epithelium and the epidermis (and designated as the
neural fold NF), the NC cells (NCC) loose their
epithelial arrangement and colonize the embryonic tis-
sues to finally settle in elected points in the embryo
where they differentiate in a variety of cell types (see
Le Douarin, 1982; Le Douarin and Kalcheim, 1999 for
reviews).
The invasive properties of the NCC are impres-
sive since there is virtually no tissue in the body
devoid of cells derived from the NC. The NC is
at the origin of the peripheral nervous system (PNS
i.e., neurones of the sympathetic, parasympathetic and
enteric ganglia and plexuses, ganglionic glial cells
and Schwann cells lining peripheral nerves) and glan-
dular cells (carotid-body cells, calcitonin-producing
cells of the ultimobranchial bodies in birds and of the
thyroid gland in mammals, adrenal medullary cells).
The NC is also the source of mesenchymal cells
(designated as mesectoderm or ectomesenchyme).
In lower vertebrates, the mesectoderm arises from
the whole length of the neural axis. In amniotes,
this capacity is restricted to the cephalic neural
tube.
The mesenchymal derivatives of the NC have been
discovered, in 1893, in living embryo of Necturus by
Julia Platt who noticed that cells migrating from the
dorsal aspect of the neural tube contributed to the
skeleton of the mandible (Platt, 1893). In our labo-
ratory, the migration of NCC was followed in inter-
specific chimeras constructed between two species of
birds, the chick (Gallus gallus) and the Japanese quail
(Coturnix coturnix japonica) (Couly et al., 1993; Le Fig. 4.1 Cephalic NCC migration and contribution to the
Livre, 1978). It turned out that the entire facial skele- head skeleton. (a) Migration streams of cephalic NCC to the
naso-frontal bud (NFB) and branchial arches (BA), according to
ton and part of the cranial vault are of NC origin their level of origin (R, rhombomere). (b) Composite origin of
(Fig. 4.1). Moreover, the dermis, connective and fat the avian craniofacial skeleton. The contribution of the cephalic
tissues of the vertebrate head are also NC-derived NC (red), cephalic paraxial mesoderm (blue), and somitic meso-
(Billon et al., 2007; Le Douarin, 1982; Le Douarin and derm (green) is indicated on lateral view of the skull at E14
(upper panel) and on ventral view of E10 chondrocranium
Kalcheim, 1999). (lower panel)
Thus, the NC is a highly pluripotent structure
which, in this respect, can be compared to the
hematopoetic system. The problem was thus raised cells, chondrocytes and osteocytes on the other, could
as to whether all the NC derivatives can arise from have a common precursor in the NC.
one single type of pluripotent stem cells. It was par- In this review, we will envision first the genetic
ticularly interesting to ask whether the neural and pathways, which are responsible for the early speci-
melanocytic derivatives on the one hand, and the fication of the NC itself within the dorsal embryonic
mesectodermal cell types including connective tissue ectoderm. Then we will report the exploration of the
4 Cell Diversification During Neural Crest Ontogeny 49

differentiative capabilities of single NCC by in vitro whose actions are strikingly coordinated in time and
clonal cultures, which revealed the existence, among space. The interplay between these factors is relatively
cells derived from the NC, of intermediate precursors conserved between different vertebrate species, and
able to self-renew. has been especially demonstrated in Xenopus laevis
The persistence in adulthood of such stem-like cells model. The neural plate border is specified by BMP
has been demonstrated in several systems in birds and and Wnt signals originating in the non-neural ecto-
mammals and represents a potential source of stem derm and by FGF signals and BMP antagonists (such
cells for regenerative medicine. as chordin, noggin and follistatin) which are derived
A fourth part will deal with a notion demonstrated in from the paraxial mesoderm (Garcia-Castro et al.,
vivo and in vitro, that the differentiation of NC deriva- 2002; LaBonne and Bronner-Fraser, 1998; Marchant
tives strongly depends upon environmental cues. The et al., 1998; Mayor et al., 1995, 1997; Monsoro-Burq
nature of these cues is being investigated in our and et al., 2003). These signalling molecules act in concert
other laboratories and signalling molecules are being to determine the identity of neural plate border cells
identified, which play a key role in the differentiation characterized by the expression of Msx1, Msx2, Pax3,
of NC derivatives. Pax7 and Zic1 transcription factors (for a review, see
Finally we will discuss the problem of the Sauka-Spengler et al., 2007). In turn Pax3 and Zic1
reversibility of the differentiated state. This question act synergistically in a Wnt-dependent manner, up-
was documented, in our laboratory, for melanocytes regulating some NC-specific marker genes (e.g., Snail2
and glial cells. and FoxD3) in the NF (Hong and Saint-Jeannet, 2007;
Sato et al., 2005). These NC-specific genes (and oth-
4.2 Formation of the Neural Crest, ers like c-Myc, Id, AP-2, Twist and Sox9) control later
a Structure Between CNS cellular events like cell proliferation, delamination and
epithelial-mesenchymal transition. This results in the
and Epidermis in Vertebrate
segregation of the NC from both the future epidermal
Embryos ectoderm and the dorsal neural epithelium (for reviews,
see Kuriyama and Mayor, 2008; Sauka-Spengler and
At the early gastrula stage of vertebrate development, Bronner-Fraser, 2008).
the ectoderm can be subdivided in three different
regions: the neural plate, its lateral borders (or NF) 4.3 Identification of Neural Crest
and the presumptive epidermis. The neural plate
Progenitors and Stem Cells
border cells (which develop at the junction between
the neural and epidermal ectoderm) give rise to NC by In Vitro Single Cell Cultures
and placodal cells (Le Douarin, 1982; Le Douarin and
Kalcheim, 1999; Sauka-Spengler and Bronner-Fraser, The wide diversity of NC-derived cell types raises the
2008). Fate map studies have shown that the NC issue of how cell diversification is produced during NC
originates from the neural plate border, except from ontogeny. Are NCC multipotent stem cells or do the
the most anterior NF, which generates olfactory and various phenotypes arise from precursors already com-
lens placodes (Couly and Le Douarin, 1985) and a mitted in the NF? These issues have been for long a
variety of epithelial and glandular structures (Couly subject of interest for our laboratory and have required
et al., 1993). The absence of NCC from this specific to developing methods in order to investigate the dif-
region was recently shown to be due to an inhibitory ferentiation potentials of single NCC. By analogy to
signal emanating from the prechordal mesoderm the in vitro assay for colony-forming units, which was
and mediated by the signalling molecule Dickkopf devised in the 1950s onward to decipher the origin of
(Carmona-Fontaine et al., 2007). hemopoetic cell diversity, we have designed an in vitro
The establishment in the dorsal aspect of the embryo clonal analysis of NCC isolated from the quail embryo.
of definite ectodermal regions including the prospec- According to the method pioneered by Cohen and
tive NC, occurs already at the late gastrula stage Konisgberg (1975), the NCC can be obtained as adher-
(Basch et al., 2006) and depends largely upon a ent cells when they have migrated out from explanted
set of signalling molecules and transcription factors neural primordium onto culture substrate. Following
50 E. Dupin et al.

trypsin treatment, a pure population of NCC can thus have shown that subsets of clonogenic cells generated
be isolated for subculture experiments. myofibroblasts or chondrocytes together with neu-
Clonogenic assays of avian NCC directly isolated ronal, glial and melanocytic cells (Baroffio et al., 1988,
from the embryo as they migrate (for the mesen- 1991; Dupin et al., 1990; Dupin and Le Douarin,
cephalic NC) or after they have spread from neural 1995; Trentin et al., 2004). These findings provided
tube explants, have provided important information first evidence that segregation of neural-melanocytic
concerning their developmental potential. The migra- and mesenchymal cell lineages is not completed at the
tory NCC constitute a heterogeneous population with time NCC start to migrate, and indeed, few clono-
respect to their proliferation and differentiation poten- genic cells were found to be already committed to
tials. A variety of progenitors can be evidenced in the the chondrocytic phenotype. The majority of the pro-
avian trunk NCC (Cohen and Konigsberg, 1975; Dupin genitors recorded showed diverse combinations of two
and Le Douarin, 1995; Lahav et al., 1998; Sieber-Blum and three phenotypes in their progeny. Among these
and Cohen, 1980; Sieber-Blum, 1989, 1991; Trentin intermediate progenitors, glial-melanocytic (GM) and
et al., 2004). They are either restricted to the pigment glial-myofibroblastic (GF) bipotent progenitors exhib-
cell fate, or bipotent, i.e., able to give rise either to both ited stem cell properties as they could self-renew along
pigment and glial cells, to glial cells and neurones or successive rounds of in vitro subcloning (Trentin et al.,
to glial cells and myofibroblasts. Others yield three and 2004). Another striking result was that all the inter-
more phenotypes, thus recapitulating the repertoire of mediate (including bipotent) precursors were able to
trunk NC derivatives (Trentin et al., 2004). yield glial cells, suggesting that the gliogenic differen-
The in vitro culture method of avian NCC was later tiation potential could constitute a general marker of
adapted to mammalian cells and similarly led to iden- uncommitted NCC.
tify pluripotent progenitors in mouse and rat trunk The existence of a clonogenic cell able to yield
NC (Ito and Sieber-Blum, 1991; Paratore et al., 2001; all the phenotypes normally derived from the NC
Rao and Anderson, 1997; Shah et al., 1994, 1996; in vivo, i.e., a cell yielding glia (G), neurones (N),
Stemple and Anderson, 1992). Precursors exiting from melanocytes (M), myofibroblasts (F), chondrocytes
the rodent trunk NC are able to yield autonomic neu- (C) and osteoblasts (O): GNMFCO, could not be
rones, glial cells and myofibroblasts. They were shown proven until recently. We showed first that chondro-
to self-renew in vitro, thus deserving to be considered genic cells differentiated from cultured cephalic NCC
as NC stem cells (Stemple and Anderson, 1992). if the cells had been harvested after they had migrated
In vitro clonal assays have also shown the pres- out of the neural tube for only 15 h (Calloni et al.,
ence of common progenitors for neurones, pigment 2007). Using these conditions (see Fig. 4.2), bona
cells as well as fibroblasts and chondrocytes, in the fide chondrocytes (expressing Sox9 transcription fac-
cardiac NC of the quail (Ito and Sieber-blum, 1991) tor, collagen-2a and chondroitin sulfate markers) dif-
and mouse (Youn et al., 2003). This cardiac subset of ferentiated and formed aggregates in about 15% of
the NCC is formed at the post-otic level of the neural single NCC cultures. Then the analysis of clonal cul-
axis and yield migratory cells that colonize the cardiac tures showed that nearly all chondrogenic progenitors
outflow tract and have an important role in forma- were multipotent cells that, in addition to chondro-
tion of heart septation. Even after they have colonized cytes, gave rise to glia, melanocytes, myofibroblasts
the posterior branchial arches, post migratory cardiac and neurones (Calloni et al., 2007) (Fig. 4.3).
NC-derived cells retain common progenitors for neu- The expression of both neural-melanocytic and
rones and chondrocytes and/or myofibroblasts (Ito and mesenchymal differentiation potencies by a large sub-
Sieber-Blum, 1993). set of early cephalic NCC has been further documented
The even greater diversity of cell types originating recently by investigating the osteogenic properties of
from the cephalic as compared to the trunk NC led us to cultured NCC (Calloni et al., 2009). The NCC iso-
explore whether the cell types belonging to mesenchy- lated after 15 h of migration from explanted mes-
mal lineages (e.g., smooth muscle cells, chondrocytes encephalon yielded two types of osteoblasts in day
and osteocytes) were derived from multipotent progen- 10-cultures: one, endochondral-like type differenti-
itors in the cephalic NC. In clonal cultures carried out ated in the perichondrium of already formed cartilage
with quail mesencephalic-rhombencephalic NCC, we nodules, whereas the other developed independently
4 Cell Diversification During Neural Crest Ontogeny 51

Fig. 4.2 Method for isolation and culture of quail NCC. The out from the explant (c) and are then harvested to be replated
neural primordium (including premigratory NC) is isolated at in secondary cultures on previously established feeder-layers
the mesencephalic level in 68 somite-stage (a) or at the trunk of growth-arrested 3T3 fibroblasts (d). After 610 days, mass
level in 2025 somite-stage (b) quail embryos following enzy- cultures and colonies derived from single NCC are analysed
matic treatment to remove surrounding tissues. The neural tube by immunocytochemistry and in situ hybridization to identify
(NT) is cultured for 1524 h; during this period NCC migrate differentiated cell types (e)

of the presence of chondrocytes and hence can be 4.4 Pluripotent Neural Crest Stem Cells
assigned to a membranous-type of bone-forming cells. in Tissues and Organs;
The presence of osteoblasts, as identified by expres-
Developmental Remnant
sion of the Runx2 gene, was investigated in colonies
derived from cephalic NCC (Fig. 4.3). It turned out that
and Potential Source of Stem Cells
more than 90% of the clones contained osteoblasts, for Regenerative Medicine
together with one or several other NC-derived differen-
tiated cell types. As shown in Fig. 4.4, the clonogenic In addition to the differentiated cells derived from the
NCC endowed with the capacity to yield osteocytes NC that are present in various organs and tissues, sev-
belong to diverse types of bi- and multipotent pro- eral types of NC stem cells and progenitors have been
genitors. Upstream of these progenitors, we identi- shown to persist in the adult body. The best function-
fied a highly multipotent NCC that has the ability to ally characterized is a melanocyte stem cell identified
generate all the recorded phenotypes, i.e., glia, neu- in the permanent lower portion of the mammalian
rones, melanocytes, myofibroblasts, chondrocytes and hair follicle designated as the bulge, which replen-
osteocytes (GNMFCO; see Fig. 4.4) (Calloni et al., ishes melanocytes during the hair cycle (Nishimura
2009). et al., 2002). Other NC-derived undifferentiated cells
These highly multipotent GNMFCO progenitors are present at postnatal stages in birds and rodents
present at an early migratory stage in the cephalic in several NC derivatives, like the PNS ganglia and
NC are presumably at the origin of all the more nerves (Le Livre and Le Douarin, 1982; Duff et al.,
restricted NC precursor cells. Although they have not 1991; Dupin, 1984; Hagedorn et al., 1999; Joseph
been shown so far to self-renew, the GNMFCO pro- et al., 2004; Li et al., 2007; Morrison et al., 1999;
genitors could thus be designated as the NC stem cells Nagoshi et al., 2008; Schweizer et al., 1983; Sextier-
able to generate all types (both neural and mesenchy- Sainte-claire Deville et al., 1992) and the enteric ner-
mal/skeletogenic) of NC derivatives in the developing vous system (Kruger et al., 2002; Sextier-Sainte-claire
head. Deville et al., 1994).
52 E. Dupin et al.

Fig. 4.3 NC-derived


phenotypes recorded in the
progeny of quail cephalic
NCC. Colonies derived from
single cephalic NCC are
analysed after 10 days for the
presence of six different cell
types. Immunocytochemistry
to HNK1 (a), neurofilament
proteins and III-tubulin (b),
and smooth muscle actin (d)
is used to identify glial cells,
neurones and
myofibroblasts/smooth
muscle cells, respectively.
Pigmented melanocytes (c)
and chondrocyte aggregates
(e) can be detected in
bright-field and phase-contrast
microscopy. Osteoblasts are
identified by expression of
Runx2 transcription factor (f)
(in situ hybridization)

The persistence of NC stem cells in several other The carotid body is therefore a niche for adult neu-
tissues of adult mammals, such as the skin, carotid rogenic stem cells in the PNS (Pardal et al., 2007).
body, bone marrow, heart and cornea, has been demon- However, unlike the neural stem cells of the subven-
strated in transgenic mice using NC-specific promoter- tricular zone in the CNS, NC progenitors in the carotid
Cre and floxed reporter systems, in which NC-derived body are not permanently active but are turned on dur-
cells are permanently labelled (Fernandes et al., 2004; ing adaptive response to changes in the oxygen level of
Nagoshi et al., 2008; Pardal et al., 2007; Sieber-Blum arterial blood.
and Grim, 2004; Sieber-Blum et al., 2004; Tomita The dental pulp also contains NC-derived precur-
et al., 2005; Wong et al., 2006; Yoshida et al., 2006). sors that differentiate into odontoblasts and are respon-
When isolated and cultured in vitro, these NC-derived sible for dentinal repair in adults. Highly proliferative
progenitors can generate various NC derivatives like clonogenic dental pulp stem cells have been isolated
neurones, glial cells and myofibroblasts. Their function in humans from adult molar and exfoliated deciduous
in vivo however remains to be established. teeth (Gronthos et al., 2000, 2002; Miura et al., 2003).
In vivo neurogenesis by rodent NC stem cells in These cells can be expanded ex vivo and reconstitute
the carotid body offers a unique example demonstrat- dentin in transplantation models, thus offering promise
ing physiological function of adult NC progenitors for dental tissue engineering and tooth regeneration in
in vivo (Pardal et al., 2007). During adaptation to the next future.
hypoxia, these cells of the carotid body undergo pro- The adult skin is another source where NC-derived
liferation, convert into intermediate progenitors and stem cells can be isolated with minimally invasive pro-
eventually lead to de novo differentiation of dopamin- cedures. The multipotent NCC populations recently
ergic chemosensory neurones that will activate the identified in the murine skin are located in several epi-
brainstem respiratory centre to increase ventilation. dermal and dermal structures of the whisker follicle,
4 Cell Diversification During Neural Crest Ontogeny 53

They therefore represent a potentially attractive source


of adult somatic stem cells for autologous transplanta-
tion and future cell replacement therapy in a variety of
human tissues.

4.5 In Vivo and In Vitro Demonstration


of the Influence of Environmental
Cues on the Differentiation of Neural
Crest Derivatives

4.5.1 In Vivo Studies

Fig. 4.4 Schematic representation of the various progeni- Transplantation experiments performed in the 1970s
tors identified by in vitro clonal analysis of quail cephalic
and 1980s in the avian embryo have established a fate
NCC. The progenitors are classified according to the number
of cell phenotypes recorded in the clones following analysis map of the various NC derivatives along the neural
of the presence of glial cells (G), neurones (N), melanocytes axis and revealed that the environment into which NCC
(M), myofibroblasts (F), chondrocytes (C) and osteoblasts (O) home at the term of their migration strongly influ-
(see also Fig. 4.3). Subsets of precursors that yield neural and
ences their fate (Le Douarin, 1982; Le Douarin and
melanocytic cells only (G, N and/or M) (in yellow) or mesenchy-
mal cell types (F, C and/or O) only (in blue) are indicated. The Kalcheim, 1999).
great majority of clonogenic NCC generated both neural and One striking example is the ability of the NC to
mesenchymal cells (progenitors in grey), including a variety of give rise either to enteric cholinergic neurones or to
osteogenic NCC (progenitors surrounded in light brown). These
catecholaminergic cells of the adrenal medullary gland
experiments led to identify a highly multipotent cell (GNMFCO
progenitor), which is upstream in the hierarchy of all NC-derived according to the migratory pathway taken by the cells
progenitors known so far when they leave the neural tube. Although regionalized
along the anterior-posterior axis, the NCC fates are not
specified before they start migrating. The differentia-
which are derived from the cephalic NC. In the back
tion choice of NCC depends on the level of the trunk
skin, trunk NC-derived multipotent cells are present
from which they migrate. This level dictates the migra-
in the hair bulge and share early markers of glial and
tion pathway and the tissue of the embryo to which
melanocytic cell lineages (Wong et al., 2006). These
NCC home.
NC stem cells isolated in rodent and human skin dis-
The influence of environmental cues in neurotrans-
play neural and mesenchymal (including skeletogenic)
mitter choice by sympathetic neurones was also clearly
differentiation properties and can be expanded in vitro
documented in newborn rat sympathetic neurones,
(Fernandes et al., 2004; Nagoshi et al., 2008; Sieber-
which can switch their neurotransmitter to acetyl-
Blum and Grim, 2004; Sieber-Blum et al., 2004;
choline if they are co-cultured with glial cells or
Toma et al., 2001, 2005; Wong et al., 2006). The
cardiac muscle (Furshpan et al., 1976; Patterson and
expanded skin-derived NC progenitors were efficient
Chun, 1977).
to repair nerve tissue when tested in murine models of
spinal cord and nerve injury (Biernaskie et al., 2007;
McKenzie et al., 2006; Sieber-Blum et al., 2006). The
skeletogenic potential of human skin-derived NC pro- 4.5.2 In Vitro Studies
genitors has also been exploited recently to repair bone
fracture after transplantation in immuno-compromised The paramount influence of external factors on NCC
mice (Lavoie et al., 2008). differentiation and their action on particular types of
The NC-derived stem cells of the adult skin are NC progenitors was underlined by a number of in vitro
easily accessible; they share some characteristics with culture studies and thanks to the analysis of single
pluripotent stem cells without being tumorigenic as are NCC cultures performed in avian and mammalian
embryonic stem cells when transplanted into the adult. species (Le Douarin and Dupin, 2003).
54 E. Dupin et al.

In the rat model, Anderson and collaborators have facial morphogenesis has been previously recognized
identified NC stem cells yielding glia, autonomic neu- (Ahlgren and Bronner-Fraser, 1999; Chiang et al.,
rones and myofibroblasts (Stemple and Anderson, 1996). A series of in vitro culture experiments led
1992) and they have deciphered the molecular path- to the demonstration that Shh has several effects on
ways involved in controlling their fate. BMP2 sig- mesencephalic NCC. First, the exposure of these cells
nals promote neuronal autonomic development at the to Shh during the first two days of culture strongly
expense of glial cell fate (Shah et al., 1994). The promoted the chondrocytic cell fate, as assessed by
acquisition of a glial phenotype is controlled by increased number of Sox9-expressing chondrocyte pre-
Neuregulin-1 (Shah et al., 1996) and Notch activation cursors and later, of differentiated cartilage cells that
(Morrison et al., 2000) whereas transforming growth formed numerous aggregates in the NCC cultures
factor (TGF)- has been suggested to instruct mul- (Calloni et al., 2007). This effect on the differenti-
tipotent NCC to adopt a myofibroblast fate. Signals ation of chondrocytes was further supported by the
that drive multipotent NCC to form sensory neurones results of clonal NCC cultures, showing that Shh
involve activation of Wnt/-catenin signalling, which highly enhances the total number of the chondrocyte-
is not only required but also sufficient to trigger sen- containing colonies.
sory neurogenesis. Together with BMP, Wnt factors Second, these experiments also provided evidence
play an important role in the maintenance of multipo- that, among clonogenic cephalic NCC, Shh specifi-
tent NC stem cells, as shown recently in the mouse cally increased the number of multipotent progenitors
(Kleber et al., 2005; Lee et al., 2004). These find- that are endowed with both neural and mesenchy-
ings have been discussed in detail in a recent review mal (including chondrocytic) differentiation potentials
(Sommer, 2006). (Calloni et al., 2007). Notably, this increase of the
In the avian model, we could demonstrate the strong proportion of mesenchymal-neural progenitors coin-
implication of the vasoactive peptide endothelin-3 cided with a decrease in the rate of the only-neural
(ET3) in the development of the melanocytic and glial ones while the total number of clones remained similar
lineages both in culture and in vivo (Lahav et al., 1996, in Shh-treated and untreated cultures, arguing that Shh
1998; Lecoin et al., 1998; Nataf et al., 1996, 1998). enhances NCC multipotentiality. Recently we found
The ET3 peptide is produced by the skin and the gut that Shh also acts at a later stage to enhance the devel-
wall during the migration of NCC, which express the opment of cultured cephalic NCC into endochondral-
receptors of ET3 (ETR), and ET3/ETR signalling path- like osteoblasts (Calloni et al., 2009). When NCC are
way is required cell-autonomously in NCC for proper exposed to Shh between 7 and 10 days of culture,
development of pigmentation and enteric nerve cells the differentiation of osteoblasts in the perichondrium
(Gershon, 1999; McCallion and Chakravarti, 2001). At of cartilage nodules is highly promoted whereas Shh
early migration stages, ET3 has a strong mitogenic does not act on the dermal-like type of osteoblasts,
effect on avian NCC (Lahav et al., 1996). However which form independently of chondrocytes. By study-
it does not act similarly on all types of NC progen- ing the effect of Shh treatment on osteogenic progen-
itors; the particular target of ET3 turned out to be a itors in NCC clonal cultures, we found that it raised
bipotent glial-melanocytic stem cell, whose survival, significantly the frequency of the highly multipotent
proliferation and self-renewal were upregulated when GNMFCO progenitors (Calloni et al., 2009), thus pro-
individual NCC were treated in vitro with ET3 (Lahav viding further evidence for a crucial role of Shh on
et al., 1998). Other types of progenitors, such as a glial- multipotent osteo-chondrogenic NCC.
myofibroblast precursor, were not responsive to this
factor.
In a recent work we sought for the putative fac- 4.6 Plasticity and Dedifferentiation
tors involved in growth and differentiation of the
mesenchymal precursors of the cephalic NC, which
Ability of Neural Crest-Derived
are responsible for the development of the facial Differentiated Cells
skeleton (Calloni et al., 2007). We investigated the
possible role of Sonic hedgehog (Shh) on the differ- The plasticity of NCC fate shown in vivo and in
entiation of quail cephalic NCC in vitro. The require- vitro raises the issue as to whether the differentia-
ment of this morphogen for early NCC survival and tion state of NCC is stable during development and
4 Cell Diversification During Neural Crest Ontogeny 55

how differentiated cell phenotypes are maintained 4.7 Concluding Remarks


in NC derivatives. The traditional view that cell
differentiation is unidirectional and irreversible has Although debate still exists as to whether most indi-
been challenged by several lines of evidence such vidual NC cells or only a small subset of them are
as the transdifferentiation and dedifferentiation pro- multipotent after emigrating from the dorsal neural
cesses observed in ocular tissues in vivo and in primordium, several lines of evidence have convinc-
vitro (reviewed by Blau et al., 2001; Egushi and ingly demonstrated the multipotency and self-renewal
Kodama, 1993). Another striking example is the capacity of various subsets of NCC in avian and
recently demonstrated reprogramming of adult fibrob- mammalian species. In the trunk of vertebrates, glial
lasts into pluripotent ES-like cells following forced cells and neurones of the PNS as well as pigment
expression of only four transcription factors (Okita cells originate from multipotent NC stem cells, which
et al., 2007; Takahashi and Yamanaka, 2006; Takahashi then undergo fate restrictions to give rise to appro-
et al., 2007). priate committed precursors in their various sites of
In the NC lineages, we have shown that differen- differentiation. Although the NC and the CNS are
tiated glial and melanocytic cells exhibit phenotype both of ectodermal origin during embryogenesis, the
plasticity in vitro. Schwann cells of the sciatic nerve NCC exhibit a wider range of derivatives than the
and epidermal pigment cells isolated from the quail neural stem cells of the brain since they not only
until late embryonic stages are able to convert into form the PNS component cells but also contribute
each other when treated in culture with ET3, which mesenchymal and skeletogenic cells to the vertebrate
displays mitogenic activity on NCC (Lahav et al., head. One of the significant advances towards deci-
1996, 1998). Schwann cells in clonal cultures yielded phering the mechanisms of lineage diversification in
a multi-phenotypic progeny of melanocytes and myofi- the NC has been recently to show that these different
broblasts in addition to parental-like glial cells (Dupin lineages, the neural and mesenchymal ones, actually
et al., 2003; Real et al., 2005). Moreover, when grafted are derived from common and multipotent progeni-
into the first branchial arch of chick host embryos, tors. This unique characteristic of the cephalic NCC
Schwann cell progeny contributed to the smooth mus- in higher vertebrates should make this cell popula-
cle wall of cranial blood vessels (Real et al., 2005). tion of potential interest for investigations in both the
In vitro experiments aimed at challenging the sta- neural stem cell and mesenchymal stem cell fields.
bility of the pigment cell phenotype have revealed Much remains however to be known with respect to
that, while proliferating, most of isolated pigment cells the molecular regulation of stem cell properties and
can dedifferentiate and reacquire an immature NC-like differentiation of the various progenitors present in
state (Dupin et al., 2000; Real et al., 2006). In clonal the target tissues of post-migratory NCC. These issues
cultures, dedifferentiated melanocytes recapitulate the are of clinical importance to understand the alterations
expression of early NC marker genes and revert to mul- in NCC number and function that occur in human
tipotent NC-like progenitors able to differentiate into neurocristopathies. The recent discovery of resident
melanocytes, glial cells and myofibroblasts, and which NCC with stem cell properties in adult tissues such
self-renew along successive subcloning (Real et al., as the skin may offer a source for isolating autolo-
2006). gous NC-derived stem cells with great promise for cell
These findings show that the differentiated state replacement therapy.
of NC-derived cells is not fixed. If submitted to
Acknowledgements The authors acknowledge the support of
new environmental influences or when displaced out-
the Centre National de la Recherche Scientifique, Foundation
side their niche, differentiated glial and pigment cells Bettencourt Schueller and Association pour la Recherche con-
undergo reversion of their differentiation programme tre le Cancer. G.W.C. was recipient of a post-doctoral fellowship
and recover stem cell properties similar to their mul- from ARC.
tipotent NC ancestors. This phenotypic plasticity also
suggests that, in several adult tissues and organs, ded- References
ifferentiated NC-derived cells as well as persistent
undifferentiated NC stem cells, could be of poten-
Ahlgren SC, Bronner-Fraser M (1999) Inhibition of sonic hedge-
tial efficiency in cell replacement after injury or in hog signaling in vivo results in craniofacial neural crest cell
diseases. death. Curr Biol 9:13041314.
56 E. Dupin et al.

Baroffio A, Dupin E, Le Douarin NM (1988) Clone-forming Dupin E, Glavieux C, Vaigot P, Le Douarin NM (2000)
ability and differentiation potential of migratory neural crest Endothelin 3 induces the reversion of melanocytes to glia
cells. Proc Natl Acad Sci USA 85:53255329. through a neural crest-derived glial-melanocytic progenitor.
Baroffio A, Dupin E, Le Douarin NM (1991) Common precur- Proc Natl Acad Sci USA 97:78827887.
sors for neural and mesectodermal derivatives in the cephalic Dupin E, Le Douarin NM (1995) Retinoic acid promotes the
neural crest. Development 112:301305. differentiation of adrenergic cells and melanocytes in quail
Basch ML, Bronner-Fraser M, Garcia-Castro MI (2006) neural crest cultures. Dev Biol 168:529548.
Specification of the neural crest occurs during gastrulation Dupin E, Real C, Glavieux-Pardanaud C, Vaigot P, Le Douarin
and requires Pax7. Nature 441:218222. NM (2003) Reversal of developmental restrictions in neu-
Biernaskie J, Sparling JS, Liu J, Shannon CP, Plemel JR, Xie Y, ral crest lineages: transition from Schwann cells to glial-
Miller FD, Tetzlaff W (2007) Skin-derived precursors gen- melanocytic precursors in vitro. Proc Natl Acad Sci USA
erate myelinating Schwann cells that promote remyelination 100:52295233.
and functional recovery after contusion spinal cord injury. J Eguchi G, Kodama R (1993) Transdifferentiation. Curr Opin
Neurosci 27:95459559. Cell Biol 5:10231028.
Billon N, Iannarelli P, Monteiro MC, Glavieux-Pardanaud C, Fernandes KJ, McKenzie IA, Mill P, Smith KM, Akhavan M,
Richardson WD, Kessaris N, Dani C, Dupin E (2007) The Barnabe-Heider F, Biernaskie J, Junek A, Kobayashi NR,
generation of adipocytes by the neural crest. Development Toma JG, Kaplan DR, Labosky PA, Rafuse V, Hui CC, Miller
134:22832292. FD (2004) A dermal niche for multipotent adult skin-derived
Blau HM, Brazelton TR, Weimann JM (2001) The evolv- precursor cells. Nat Cell Biol 6:10821093.
ing concept of a stem cell: entity or function?. Cell 105: Furshpan EJ, MacLeish PR, OLague PH, Potter DD (1976)
829841. Chemical transmission between rat sympathetic neurons and
Calloni GW, Glavieux-Pardanaud C, Le Douarin NM, Dupin cardiac myocytes developing in microcultures: evidence for
E (2007) Sonic Hedgehog promotes the development of cholinergic, adrenergic, and dual-function neurons. Proc Natl
multipotent neural crest progenitors endowed with both mes- Acad Sci USA 73:42254229.
enchymal and neural potentials. Proc Natl Acad Sci USA Garcia-Castro MI, Marcelle C, Bronner-Fraser M (2002)
104:1987919884. Ectodermal Wnt function as a neural crest inducer. Science
Calloni GW, Le Douarin NM, Dupin E (2009) High frequency of 297:848851.
cephalic neural crest cells shows coexistence of neurogenic, Gershon MD (1999) Endothelin and the development of the
melanogenic and osteogenic differentiation capacities. Proc enteric nervous system. Clin Exp Pharmacol Physiol 26:
Natl Acad Sci USA 106:89478952. 985988.
Carmona-Fontaine C, Acuna G, Ellwanger K, Niehrs C, Mayor Gronthos S, Brahim J, Li W, Fisher LW, Cherman N, Boyde
R (2007) Neural crests are actively precluded from the ante- A, DenBesten P, Robey PG, Shi S (2002) Stem cell prop-
rior neural fold by a novel inhibitory mechanism dependent erties of human dental pulp stem cells. J Dent Res 81:
on Dickkopf1 secreted by the prechordal mesoderm. Dev 531535.
Biol 309:208221. Gronthos S, Mankani M, Brahim J, Robey PG, Shi S (2000)
Chiang C, Litingtung Y, Lee E, Young KE, Corden JL, Westphal Postnatal human dental pulp stem cells (DPSCs) in vitro and
H, Beachy PA (1996) Cyclopia and defective axial pattern- in vivo. Proc Natl Acad Sci USA 97:1362513630.
ing in mice lacking Sonic hedgehog gene function. Nature Hagedorn L, Suter U, Sommer L (1999) P0 and PMP22 mark
383:407413. a multipotent neural crest-derived cell type that displays
Cohen AM, Konigsberg IR (1975) A clonal approach to the community effects in response to TGF-beta family factors.
problem of neural crest determination. Dev Biol 46:262280. Development 126:37813794.
Couly GF, Coltey PM, Le Douarin NM (1993) The triple ori- Hong CS, Saint-Jeannet JP (2007) The activity of Pax3 and Zic1
gin of skull in higher vertebrates: a study in quail-chick regulates three distinct cell fates at the neural plate border.
chimeras. Development 117:409429. Mol Biol Cell 18:21922202.
Couly GF, Le Douarin NM (1985) Mapping of the early neu- Ito K, Sieber-Blum M (1991) In vitro clonal analysis of quail
ral primordium in quail-chick chimeras. I. Developmental cardiac neural crest development. Dev Biol 148:95106.
relationships between placodes, facial ectoderm, and pros- Ito K, Sieber-Blum M (1993) Pluripotent and developmen-
encephalon. Dev Biol 110:422439. tally restricted neural-crest-derived cells in posterior visceral
Duff RS, Langtimm CJ, Richardson MK, Sieber-Blum M (1991) arches. Dev Biol 156:191200.
In vitro clonal analysis of progenitor cell patterns in dorsal Joseph NM, Mukouyama YS, Mosher JT, Jaegle M, Crone SA,
root and sympathetic ganglia of the quail embryo. Dev Biol Dormand EL, Lee KF, Meijer D, Anderson DJ, Morrison
147:451459. SJ (2004) Neural crest stem cells undergo multilineage
Dupin E (1984) Cell division in the ciliary ganglion of quail differentiation in developing peripheral nerves to gener-
embryos in situ and after back-transplantation into the neu- ate endoneurial fibroblasts in addition to Schwann cells.
ral crest migration pathways of chick embryos. Dev Biol Development 131:55995612.
105:288299. Kleber M, Lee HY, Wurdak H, Buchstaller J, Riccomagno MM,
Dupin E, Baroffio A, Dulac C, Cameron-Curry P, Le Douarin Ittner LM, Suter U, Epstein DJ, Sommer L (2005) Neural
NM (1990) Schwann-cell differentiation in clonal cultures crest stem cell maintenance by combinatorial Wnt and BMP
of the neural crest, as evidenced by the anti-Schwann cell signaling. J Cell Biol 169:309320.
myelin protein monoclonal antibody. Proc Natl Acad Sci Kruger GM, Mosher JT, Bixby S, Joseph N, Iwashita T,
USA 87:11191123. Morrison SJ (2002) Neural crest stem cells persist in the adult
4 Cell Diversification During Neural Crest Ontogeny 57

gut but undergo changes in self-renewal, neuronal subtype Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi
potential, and factor responsiveness. Neuron 35:657669. S (2003) SHED: stem cells from human exfoliated deciduous
Kuriyama S, Mayor R (2008) Molecular analysis of neural teeth. Proc Natl Acad Sci USA 100:58075812.
crest migration. Philos Trans R Soc Lond B Biol Sci 363: Monsoro-Burq AH, Fletcher RB, Harland RM (2003) Neural
13491362. crest induction by paraxial mesoderm in Xenopus embryos
LaBonne C, Bronner-Fraser M (1998) Neural crest induction requires FGF signals. Development 130:31113124.
in Xenopus: evidence for a two-signal model. Development Morrison SJ, Perez SE, Qiao Z, Verdi JM, Hicks C, Weinmaster
125:24032414. G, Anderson DJ (2000) Transient Notch activation initiates
Lahav R, Dupin E, Lecoin L, Glavieux C, Champeval D, Ziller an irreversible switch from neurogenesis to gliogenesis by
C, Le Douarin NM (1998) Endothelin 3 selectively promotes neural crest stem cells. Cell 101:499510.
survival and proliferation of neural crest-derived glial and Morrison SJ, White PM, Zock C, Anderson DJ (1999)
melanocytic precursors in vitro. Proc Natl Acad Sci USA Prospective identification, isolation by flow cytometry, and
95:1421414219. in vivo self-renewal of multipotent mammalian neural crest
Lahav R, Ziller C, Dupin E, Le Douarin NM (1996) Endothelin stem cells. Cell 96:737749.
3 promotes neural crest cell proliferation and mediates a vast Nagoshi N, Shibata S, Kubota Y, Nakamura M, Nagai Y, Satoh
increase in melanocyte number in culture. Proc Natl Acad E, Morikawa S, Okada Y, Mabuchi Y, Katoh H, Okada S,
Sci USA 93:38923897. Fukuda K, Suda T, Matsuzaki Y, Toyama Y, Okano H (2008)
Lavoie JF, Biernaskie JA, Chen Y, Bagli D, Alman B, Kaplan Ontogeny and multipotency of neural crest-derived stem cells
DR, Miller FD (2009) Skin-derived precursors differentiate in mouse bone marrow, dorsal root ganglia, and whisker pad.
into skeletogenic cell types and contribute to bone repair. Cell Stem Cell 2:392403.
Stem Cells Dev 18:893906. Nataf V, Grapin-Botton A, Champeval D, Amemiya A,
Le Douarin N (1982) The Neural Crest. Cambridge University Yanagisawa M, Le Douarin NM (1998) The expression pat-
Press, Cambridge. terns of endothelin-A receptor and endothelin 1 in the avian
Le Douarin NM, Dupin E (2003) Multipotentiality of the neural embryo. Mech Dev 75:145149.
crest. Curr Opin Genet Dev 13:529536. Nataf V, Lecoin L, Eichmann A, Le Douarin NM (1996)
Le Douarin N, Kalcheim C (1999) The Neural Crest, 2nd edn. Endothelin-B receptor is expressed by neural crest cells
Cambridge University Press, Cambridge; New York. in the avian embryo. Proc Natl Acad Sci USA 93:
Le Lievre CS, Le Douarin NM (1982) The early development 96459650.
of cranial sensory ganglia and the potentialities of their Nishimura EK, Jordan SA, Oshima H, Yoshida H, Osawa
component cells studied in quail-chick chimeras. Dev Biol M, Moriyama M, Jackson IJ, Barrandon Y, Miyachi
94:291310. Y, Nishikawa S (2002) Dominant role of the niche
Le Livre CS (1978) Participation of neural crest-derived cells in melanocyte stem-cell fate determination. Nature 416:
in the genesis of the skull in birds. J Embryol Exp Morphol 854860.
47:1737. Okita K, Ichisaka T, Yamanaka S (2007) Generation of germline-
Lecoin L, Sakurai T, Ngo MT, Abe Y, Yanagisawa M, Le competent induced pluripotent stem cells. Nature 448:313
Douarin NM (1998) Cloning and characterization of a novel 317.
endothelin receptor subtype in the avian class. Proc Natl Paratore C, Goerich DE, Suter U, Wegner M, Sommer L (2001)
Acad Sci USA 95:30243029. Survival and glial fate acquisition of neural crest cells are
Lee HY, Kleber M, Hari L, Brault V, Suter U, Taketo MM, regulated by an interplay between the transcription factor
Kemler R, Sommer L (2004) Instructive role of Wnt/beta- Sox10 and extrinsic combinatorial signaling. Development
catenin in sensory fate specification in neural crest stem cells. 128:39493961.
Science 303:10201023. Pardal R, Ortega-Saenz P, Duran R, Lopez-Barneo J (2007) Glia-
Li HY, Say EH, Zhou XF (2007) Isolation and characterization like stem cells sustain physiologic neurogenesis in the adult
of neural crest progenitors from adult dorsal root ganglia. mammalian carotid body. Cell 131:364377.
Stem Cells 25:20532065. Patterson PH, Chun LL (1977) The induction of acetylcholine
Marchant L, Linker C, Ruiz P, Guerrero N, Mayor R (1998) synthesis in primary cultures of dissociated rat sympa-
The inductive properties of mesoderm suggest that the neu- thetic neurons. II. Developmental aspects. Dev Biol 60:
ral crest cells are specified by a BMP gradient. Dev Biol 473481.
198:319329. Platt JB (1893) Ectodermic origin of the cartilage of the head.
Mayor R, Guerrero N, Martinez C (1997) Role of FGF Anat Anz 8:506509.
and noggin in neural crest induction. Dev Biol 189: Rao MS, Anderson DJ (1997) Immortalization and controlled in
112. vitro differentiation of murine multipotent neural crest stem
Mayor R, Morgan R, Sargent MG (1995) Induction of the cells. J Neurobiol 32:722746.
prospective neural crest of Xenopus. Development 121: Real C, Glavieux-Pardanaud C, Le Douarin NM, Dupin E (2006)
767777. Clonally cultured differentiated pigment cells can dedifferen-
McCallion AS, Chakravarti A (2001) EDNRB/EDN3 and tiate and generate multipotent progenitors with self-renewing
Hirschsprung disease type II. Pigment Cell Res 14:161169. potential. Dev Biol 300:656669.
McKenzie IA, Biernaskie J, Toma JG, Midha R, Miller Real C, Glavieux-Pardanaud C, Vaigot P, Le-Douarin N, Dupin
FD (2006) Skin-derived precursors generate myelinating E (2005) The instability of the neural crest phenotypes:
Schwann cells for the injured and dysmyelinated nervous Schwann cells can differentiate into myofibroblasts. Int J Dev
system. J Neurosci 26:66516660. Biol 49:151159.
58 E. Dupin et al.

Sato T, Sasai N, Sasai Y (2005) Neural crest determination by crest stem cell (EPI-NCSC) grafts in the lesioned spinal cord.
co-activation of Pax3 and Zic1 genes in Xenopus ectoderm. Mol Cell Neurosci 32:6781.
Development 132:23552363. Sommer L (2006) Growth factors regulating neural crest cell fate
Sauka-Spengler T, Bronner-Fraser M (2008) A gene regulatory decisions. Adv Exp Med Biol 589:197205.
network orchestrates neural crest formation. Nat Rev Mol Stemple DL, Anderson DJ (1992) Isolation of a stem cell for
Cell Biol 9:557568. neurons and glia from the mammalian neural crest. Cell
Sauka-Spengler T, Meulemans D, Jones M, Bronner-Fraser M 71:973985.
(2007) Ancient evolutionary origin of the neural crest gene Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T,
regulatory network. Dev Cell 13:405420. Tomoda K, Yamanaka S (2007) Induction of pluripotent stem
Schweizer G, Ayer-Le Lievre C, Le Douarin NM (1983) cells from adult human fibroblasts by defined factors. Cell
Restrictions of developmental capacities in the dorsal root 131:861872.
ganglia during the course of development. Cell Differ Takahashi K, Yamanaka S (2006) Induction of pluripotent stem
13:191200. cells from mouse embryonic and adult fibroblast cultures by
Sextier-Sainte-Claire Deville F, Ziller C, Le Douarin N (1992) defined factors. Cell 126:663676.
Developmental potentialities of cells derived from the trun- Toma JG, Akhavan M, Fernandes KJ, Barnabe-Heider F, Sadikot
cal neural crest in clonal cultures. Brain Res Dev Brain Res A, Kaplan DR, Miller FD (2001) Isolation of multipotent
66:110. adult stem cells from the dermis of mammalian skin. Nat Cell
Sextier-Sainte-Claire Deville F, Ziller C, Le Douarin NM (1994) Biol 3:778784.
Developmental potentials of enteric neural crest-derived cells Toma JG, McKenzie IA, Bagli D, Miller FD (2005) Isolation and
in clonal and mass cultures. Dev Biol 163:141151. characterization of multipotent skin-derived precursors from
Shah NM, Groves AK, Anderson DJ (1996) Alternative neu- human skin. Stem Cells 23:727737.
ral crest cell fates are instructively promoted by TGFbeta Tomita Y, Matsumura K, Wakamatsu Y, Matsuzaki Y, Shibuya I,
superfamily members. Cell 85:331343. Kawaguchi H, Ieda M, Kanakubo S, Shimazaki T, Ogawa S,
Shah NM, Marchionni MA, Isaacs I, Stroobant P, Anderson DJ Osumi N, Okano H, Fukuda K (2005) Cardiac neural crest
(1994) Glial growth factor restricts mammalian neural crest cells contribute to the dormant multipotent stem cell in the
stem cells to a glial fate. Cell 77:349360. mammalian heart. J Cell Biol 170:11351146.
Sieber-Blum M (1989) Commitment of neural crest cells to the Trentin A, Glavieux-Pardanaud C, Le Douarin NM, Dupin E
sensory neuron lineage. Science 243:16081611. (2004) Self-renewal capacity is a widespread property of var-
Sieber-Blum M (1991) Role of the neurotrophic factors BDNF ious types of neural crest precursor cells. Proc Natl Acad Sci
and NGF in the commitment of pluripotent neural crest cells. USA 101:44954500.
Neuron 6:949955. Wong CE, Paratore C, Dours-Zimmermann MT, Rochat A, Pietri
Sieber-Blum M, Cohen AM (1980) Clonal analysis of quail T, Suter U, Zimmermann DR, Dufour S, Thiery JP, Meijer
neural crest cells: they are pluripotent and differentiate D, Beermann F, Barrandon Y, Sommer L (2006) Neural
in vitro in the absence of noncrest cells. Dev Biol 80: crest-derived cells with stem cell features can be traced
96106. back to multiple lineages in the adult skin. J Cell Biol
Sieber-Blum M, Grim M (2004) The adult hair follicle: cradle 175:10051015.
for pluripotent neural crest stem cells. Birth Defects Res Part Yoshida S, Shimmura S, Nagoshi N, Fukuda K, Matsuzaki Y,
C Embryo Today 72:162172. Okano H, Tsubota K (2006) Isolation of multipotent neural
Sieber-Blum M, Grim M, Hu YF, Szeder V (2004) Pluripotent crest-derived stem cells from the adult mouse cornea. Stem
neural crest stem cells in the adult hair follicle. Dev Dyn Cells 24:27142722.
231:258269. Youn YH, Feng J, Tessarollo L, Ito K, Sieber-Blum M (2003)
Sieber-Blum M, Schnell L, Grim M, Hu YF, Schneider R, Neural crest stem cell and cardiac endothelium defects in the
Schwab ME (2006) Characterization of epidermal neural TrkC null mouse. Mol Cell Neurosci 24:160170.
Chapter 5

Intermediate Filament Expression in Mouse Embryonic


Stem Cells and Early Embryos

Zhigang Xue, Vivaldo Moura-Neto, Araksya Izmiryan, Sheila Cristina de Souza


Martins, Jean Christophe Larcher, Denise Paulin, and Zhenlin Li

Contents early embryogenesis and differentiation. Finally, we


point out some questions to glial tumors and cytoskele-
5.1 Intermediate Filaments . . . . . . . . . . . 59 tal markers of cells potentially pluripotent present in
5.2 Intermediate Filament Protein Synthesis in these tumors, as nestin and synemin proteins.
Mouse Oocytes and Preimplantation Murine
Embryos . . . . . . . . . . . . . . . . . . 61 Keywords Intermediate filaments Nestin Synemin
5.3 Epithelial Differentiation and Intermediate-Sized
Filaments in Early Postimplantation Embryos . 61 Stem cells Embryonic development Glial tumors
5.4 Intermediate Filaments in Primary Mesenchymal
Cells in Mouse Embryo . . . . . . . . . . . 62 Abbreviations
5.5 Expression of Nestin and Synemin During Early
Embryogenesis and Differentiation . . . . . . 62
5.5.1 Nestin and Synemin Genes . . . . . . . 62 ES cells embryonic stem cells
5.5.2 Nestin Expression . . . . . . . . . . . 63
5.5.3 Synemin Expression . . . . . . . . . . 64 EB embryonic bodies
5.6 Expression of Nestin and Synemin in Tumoral GFAP glial fibrillary acidic protein
Cells of the CNS . . . . . . . . . . . . . . . 67 ICM inner cell mass
5.6.1 Glial Tumors . . . . . . . . . . . . . 67 IF intermediate filament
5.6.2 Nestin in Glioma . . . . . . . . . . . 68 KO knock-out
5.6.3 Synemin Expression in Glioma . . . . . 68
5.6.4 And Now . . . . . . . . . . . . . . . 68 NF neurofilament
References . . . . . . . . . . . . . . . . . . . . 69 PE parietal endoderm
PNS peripheral nervous system
VE visceral endoderm
Abstract In this work we discuss the expression of
intermediate filament protein and synthesis in mouse
oocytes and preimplantation murine embryos. Also
we touch to epithelial differentiation and intermediate- 5.1 Intermediate Filaments
sized filaments in early postimplantation embryos.
After we analyse the intermediate filaments expression
Cellular differentiation in vitro and in vivo is closely
in primary mesenchymal cells in mouse embryo, we
connected with morphological changes based on inter-
look to the expression of nestin and synemin during
mediate filament (IF) protein remodeling. Intermediate
filaments are expressed in cell-type-specific patterns
following and demarcating pathways of embryonic
development and cell differentiation (Herrmann and
Aebi, 2000).
V. Moura-Neto ()
In recent years, intermediate filaments (IFs) have
Instituto de Cincias Biomdicas-Universidade Federal de Rio
de Janeiro, Rio de Janeiro, Brazil attracted much interest, largely because their consti-
e-mail: vivaldo@anato.ufrj.br tutive polypeptide units are specifically expressed in

H. Ulrich (ed.), Perspectives of Stem Cells, 59


DOI 10.1007/978-90-481-3375-8_5, Springer Science+Business Media B.V. 2010
60 Z. Xue et al.

various cell types and thus represent excellent differ- Specific markers of neuronal and glial cell types are
entiation markers (Zehner, 1991; Oshima, 2007). Data powerful tools for studying the mechanisms generat-
obtained through biochemical studies and molecular ing cellular diversification in the nervous system. So
cloning have allowed the classification of IFs into six far, different types of IFs have been identified in the
types according to their primary sequence and pro- nervous system (Table 5.1).
tein structure. The expression of most IF types is The cloning and sequencing of IF mRNAs start-
characteristic of a given cell type: cytokeratins (IF ing in 1981 (Lazarides, 1981) revealed the conserved
types I and II) are produced in epithelia; neurofil- central coiled-coil 1A, 1B and 2A, 2B rod domains.
aments, -internexin (type IV) and peripherin (type Subsequent cDNA sequences of other IFs and gene
III) in neurons; synemin, nestin (type VI) and syn- structure facilitated evolutionary studies. In 1986,
coilin (type IV) in neuroblast and myoblast. On the McKeon discovered that the nuclear lamins were
other hand, the type III IFs are highly related proteins members of the IF superfamily (type V) but con-
which are expressed in different cell types (Peripherin tained extended helical segments not found in other
in neurons, glial fibrillary acidic protein [GFAP] in IF (McKeon et al., 1986). The extended helical seg-
glia, desmin in muscle, vimentin with a widespread ments of the nuclear lamins were found in IFs for
distribution). invertebrates suggesting an evolutionary path from
Tissues without IFs fall apart; they are mechanically nuclear lamins to cytoplasmic IFs. By examining the
unstable, unable to resist physical stress, and this leads existence and structure of IF in a variety of differ-
to cell degeneration. By maintaining the shape and ent animals (Fuchs and Weber, 1994), identification
plasticity of the cell, the intermediate filament network of 11 IF genes, 5 of which are uniquely essential, in
acts as an integrator within the cell space. The state Caenorhabditis elegans and 65 coding genes in the
of mechanical force imposed on a tissue or a cell can human genome contrasts with the absence of cytoplas-
alter the shape of certain elements of the cytoskeleton mic IF in Drosophila (Goldman, 2001).
and thus participate to the control of cell functions.

Table 5.1 Intermediate filament expression in mammal embryo nervous system


Earliest stage
IF Cell types detection References IF assembly MW (kDa)
NF-L Neurons E9 Cochard and Paulin (1984) and Homopolymer 70
Hoffman and Lasek (1975)
NF-M Neurons E9.5 Cochard and Paulin (1984) and Heteropolymer 140
Hoffman and Lasek (1975)
NF-H Neurons E10.5 Cochard and Paulin (1984) and Heteropolymer 210
Hoffman and Lasek (1975)
Peripherin Neurons E12 Escurat et al. (1990, 1988) and Homopolymer 61, 58, 55
Portier et al. (1983)
-Internexin Neurons E12 Kaplan et al. (1990) and Pachter Homopolymer 66
and Liem (1985)
Vimentin Glia cells, astrocytes E5 Izmiryan et al. (2006, 2009) Homopolymer 54
Nestin Glia cells, astrocytes, E5 Dahlstrand et al. (1995), Heteropolymer 240
neuroepithelial cells Frederiksen and McKay
(1988), Izmiryan et al. (2009)
and Tohyama et al. (1992)
H synemin Glia cells, astrocytes, E9 Hirako et al. (2003) and Heteropolymer 180
neurons Izmiryan et al., (2006, 2009)
M synemin Glia cells, astrocytes, E5 Hirako et al. (2003) and Heteropolymer 150
neurons Izmiryan et al., (2006, 2009)
L synemin Neurons E11 Izmiryan et al. (2006, 2009) Heteropolymer 41
GFAP Glia cells, astrocytes E10 Galou et al. (1994) Homopolymer 50
5 IF Expression in Mouse Embryonic Stem Cells and Early Embryos 61

5.2 Intermediate Filament Protein a nonfilamentous pool of cytokeratin in oocytes and


Synthesis in Mouse Oocytes cleavage-stage embryos.
The synthesis of two extra-embryonic endodermal
and Preimplantation Murine
cytoskeletal proteins (Endo A, Mr = 55,000; Endo B,
Embryos Mr = 50,000) was detected by immunoprecipitation at
the 4- to 8-cell stage of preimplantation mouse devel-
The presence and distribution of IF proteins in mouse opment (Oshima et al., 1983). The first detectable syn-
oocytes and preimplantation embryos were studied by thesis of both proteins occurs at about the same time as
several groups. First, to the setting up of asymmetries the earliest allocation of cells to the trophectodermal
within cells, a process called polarization, which first lineage (Brulet et al., 1980). Indirect immunofluores-
takes place during the 8-cell stage at compaction. cence microscopy has been used to detect cytoskeletal
Cytoskeletal elements and organelles, such as clathrin proteins, which allow a distinction between the two-
vesicles and endosomes, accumulate first in an apical cell types present in mouse blastocyst: i.e. the cells of
focus, while the cell nucleus tends to migrate basally the inner cell mass (ICM) and the outer trophoblastic
and gap junctions form. Secondly, to the existence of cells. Antibodies against prekeratin stain the outer tro-
asymmetrical (or differentiative) cell divisions giving phoblastic cells but not the ICM in agreement with the
rise to the two cell types present in the 16-cell stage findings on adult tissues that cytokeratins are a marker
embryo, polarized outer cells and non-polarized inner for various epithelial cells. Interestingly, vimentin fil-
cells. This is due to the fact that some elements of aments typical of mesenchymal cells as well as of
polarity are maintained throughout division and that cells growing in culture seem to be absent in both cell
the cleavage plane may be roughly orthogonal to the types of the blastocyst. Thus, cytokeratins of the tro-
axis of polarity (Maro et al., 1990). phoblastic cells seem to be the first intermediate-sized
Among the three types of cytoskeletal elements, filaments expressed in embryogenesis. Antibodies to
two are clearly present in mouse oocytes: microtubules tubulin and actin show that microtubules and micro-
and microfilaments, the presence of IFs being more filaments are ubiquitous structures, although microfil-
controversial. In both cases, their organization differs aments have a noticeably different organization in the
from that observed in somatic cells. The organiza- two cell types (Paulin et al., 1980).
tion and role of the cytoskeletal networks (mainly
microtubules and microfilaments) during oogenesis,
fertilization and preimplantation development of the
mouse are described given the importance of cell 5.3 Epithelial Differentiation
cell interactions and of the subcellular organization in and Intermediate-Sized Filaments
events leading to the formation of the first two lineages
of the mouse embryo.
in Early Postimplantation Embryos
In immunoblotting analysis of electrophoretically
separated polypeptides, a distinct doublet of polypep- It is concluded that early postimplantation embryonic
tides with Mr of 54K and 57K, reactive with cytok- development, up to mesoderm formation, is character-
eratin antibodies, was detected in oocytes and in ized by the exclusive presence, in both embryonic ecto-
cleavage-stage embryos (Lehtonen et al., 1983b). A derm and proximal endoderm, of differentiated epithe-
similar doublet of polypeptides, reactive with cytok- lial cells containing desmosome-cytokeratin filament
eratin antibodies, was also detected in late morula- complexes and that other types of intermediate-sized
and blastocyst-stage embryos. Immunoblotting with filaments are not yet expressed.
vimentin antibodies gave negative results in both Two layers of extra-embryonic endoderm, the pari-
cleavage-stage and blastocyst-stage embryos. The etal endoderm (PE) and the visceral endoderm (VE),
electron microscopy observations show that these early arise in mouse embryo shortly after implantation. Both
stages also contain detergent-resistant 10- to 11-nm cell populations apparently originate from the primi-
filaments. The relative scarcity of these filaments, as tive endoderm of the blastocyst. The PE and VE of
compared to the high intensity in the immunoblotting mouse conceptuses differ in their expression of IFs:
and immunofluorescence stainings, speaks in favor of while both cell types contain cytokeratin, expression
62 Z. Xue et al.

of vimentin was only revealed in the cells of the PE fibrillar fluorescence in cells of the proximal endo-
(Lehtonen et al., 1983a). This population of individual, derm and weak, predominantly subapical staining in
motile cells seems to be derived from a conventional embryonic ectoderm. Correspondingly, antibodies to
epithelium by migration and differentiation. All results desmoplakins, the major proteins of the desmosomal
support the idea that vimentin expression is specifically plaque, show punctate fluorescence in both embryonic
related to reduced cell-to-cell contact, and to the inde- epithelia. These epithelial cells are not significantly
pendent existence of a cell following detachment from stained with antibodies to other IF proteins such as
an epithelial sheet. The co-expression of cytokeratins vimentin and desmin. However, antibodies to vimentin
and vimentin was found in the parietal endoderm of the show positive fluorescence, often in fibrillar tangles, in
mouse embryo E8.5-E13.5 (Lane et al., 1983). Among primary mesenchymal cells which in turn are negative
the six classes of IFs found in vertebrate tissues, the with cytokeratin and desmin antibodies. The obser-
cytokeratins are considered unique to epithelial tis- vations indicate that during embryogenesis synthesis
sues, while vimentin is expressed by endothelial and of vimentin occurs, for the first time, in the primitive
mesenchymal cells. streak stage and is restricted to the primary mesenchy-
The characteristic epithelia formed by the embry- mal cells. Concomitantly, these cells cease to produce
onic ectoderm and proximal (visceral) endoderm cytokeratins and desmoplakin (Franke et al., 1982a).
present well-developed junction complexes and vari-
ous differentiated membrane structures. Several api-
cal differentiations of the proximal endodermal cells,
such as brush border-like microvilli, the endocytotic 5.5 Expression of Nestin and Synemin
labyrinthum, and the supranuclear vacuoles resemble During Early Embryogenesis
the organization of epithelial cells of the ileum of and Differentiation
neonatal mammals (Jackson et al., 1981). Both embry-
onic epithelia show typical desmosomes and attached
intermediate-sized filaments of the cytokeratin type 5.5.1 Nestin and Synemin Genes
(Franke et al., 1982b). Other types of intermediate-
sized filaments, such as synemin, nestin and vimentin Upon its identification, nestin was designated as the
mRNA, have been detected in E5 embryos by RT-PCR, prototype of a new IF protein group (type VI) because
and in the embryonic ectoderm and mesoderm at E7 it did not fall clearly into any of the previously
using in situ hybridization (Kawaguchi et al., 2001; described types (Lendahl et al., 1990). Some debate
Izmiryan et al., 2009). arose on this classification since nestin gene structure
is closely related to the neurofilament (NF) branch
in having two of its three intron positions in com-
mon with NF genes (Dahlstrand et al., 1992b). The
nestin and synemin genes show structural similarities
5.4 Intermediate Filaments in Primary
to type III (e.g. vimentin, desmin) and especially type
Mesenchymal Cells in Mouse Embryo IV (e.g. neurofilaments, alpha-internexin) IF proteins
(Herrmann and Aebi, 2000), which led to its classifi-
The cytoskeletal composition of the primitive streak cation as a type IV IF protein. Due to a low degree of
stage of mouse embryos, i.e. at late day 8 (day 8.5) protein sequence homology of the rod domain in com-
of gestation corresponds to the onset of vimentin. At parison to the five IF protein classes, accordingly, it
this stage primary mesenchymal cells are observed had been proposed to re-classify nestin as a type VI
in the posterior part of the embryo, which seem to IF protein (Herrmann and Aebi, 2000). Shortly after
migrate toward the anterior region. For most of the nestin was sequenced, the gene structure of synemin
embryo, these mesenchymal cells are separated from (Bellin et al., 1999; Titeux et al., 2001; Xue et al.,
the embryonic ectoderm by a continuous basal lam- 2004) was also described. According to their sequence
ina. Mesenchymal cells also can form junctions of similarities, nestin and synemin were then grouped
the fascia adhaerens-type but appear to be devoid of with transitin and tanabin, as type VI IF proteins
desmosomes. Antibodies to cytokeratins reveal strong (Guerette et al., 2007).
5 IF Expression in Mouse Embryonic Stem Cells and Early Embryos 63

Nestin was first identified with a monoclonal anti- alternative splicing, resulting in 3 synemin isoforms of
body (Hockfield and McKay, 1985), and then identified 180 (H or alpha), 150 (M or beta) and 41 (L) kDa. The
in neuroepithelial stem cells of rat (Lendahl et al., structure of mammal synemin gene consists of four
1990). The rat (chromosome 2q34) and human (chro- exons and three introns. The isoform M has 5 exons
mosome 1q23.1) nestin genes contain five exons span- and 4 introns, because the exon 4b becomes the inton
ning four introns (Dahlstrand et al., 1992b; Lendahl 4 by splicing. In the isoform L, the exon 4 and 4b are
et al., 1990), whereas the mouse nestin gene (chro- spliced and resulting a reading frame change in its C-
mosome 3F1) contains eight exons. Transcriptional terminal (Xue et al., 2004). All three isoforms have the
regulation of the nestin gene is unique. The 240- same head and rod domains and different tail domain
kDa protein nestin contains a short N-terminus and an sequences (Titeux et al., 2001; Xue et al., 2004), but
unusually long C-terminus. Analyses of the rat nestin each is expressed in a developmentally specific man-
promoter in transgenic mice indicate that the region ner (Xue et al., 2004). The avian synemin has only one
upstream of the first exon does not contain any iden- protein (230 kDa), equivalent to mammal H synemin
tifiable regulatory elements (Zimmerman et al., 1994). isoform (Bellin et al., 1999). As the nestin, the synemin
In fact, nestin expression in muscle precursor and proteins belong to type VI of IF family.
neuroepithelial stem cells of the central nervous sys- As is typical for IF proteins, nestin and synemin
tem (CNS) is independently regulated by temporally are characterized by a central alpha-helical region of
and spatially restricted enhancer elements in the first about 310 amino acids flanked by non-a-helical head
and second introns, respectively (Zimmerman et al., and tail domains (Dahlstrand et al., 1992b; Herrmann
1994). The second intron contains two highly con- and Aebi, 2004; Strelkov et al., 2003). The nestin
served elements, a mid-brain and a CNS enhancer and synemin proteins contain a short N-terminus (8
element that are activated and independently regulated 11 amino acids) and an unusually long C-terminus
in rat and human (Lothian et al., 1999; Yaworsky and (Dahlstrand et al., 1992b; Titeux et al., 2001; Xue et al.,
Kappen, 1999) and which are not strong enhancers 2004), which interacts with other cytoskeleton compo-
in the peripheral nervous system (Zimmerman et al., nents, such as microfilaments, microtubules and cell
1994). The CNS enhancer is active in neural stem cells adhesion molecules (Bellin et al., 2001, 1999; Bhosle
of the developing CNS (Lothian and Lendahl, 1997). et al., 2006; Lendahl et al., 1990; Sun et al., 2008a, b).
Only limited data are available on the transcriptional Nestin and synemin are unable to self-assemble (Bellin
regulation of nestin in other tissues or in vitro. et al., 1999; Herrmann and Aebi, 2000; Khanamiryan
Synemin is another type VI IF protein. Synemin et al., 2008; Titeux et al., 2001), most likely because
was originally identified in avian muscle, along with of its very short N-terminus (a domain necessary
desmin and vimentin (Bellin et al., 1999; Granger and for IF assembly) and the property of their 2A and
Lazarides, 1980). It has also been found in avian and 2B regions of the rod domain (Khanamiryan et al.,
cane toad erythrocytes (Centonze et al., 1986; Granger 2008); therefore, nestin and synemin require an appro-
and Lazarides, 1982), chicken and human lens and the priate copolymerization partner, such as vimentin or
retina cells (Granger and Lazarides, 1984; Tawk et al., desmin (Bellin et al., 1999; Guerette et al., 2007;
2003), in a subpopulation of rodent astrocytes that con- Khanamiryan et al., 2008; Marvin et al., 1998; Steinert
tains GFAP, vimentin and nestin (Izmiryan et al., 2006; et al., 1999; Titeux et al., 2001), to assemble into
Sultana et al., 2000), and in neurons (Izmiryan et al., heteropolymers.
2006).
Three synemin isoforms have characterized in
the human (GenBank accession numbers: AJ310521, 5.5.2 Nestin Expression
AJ310522, and AJ697971) (Titeux et al., 2001)
and mouse (GenBank accession numbers: AJ579700, The IF protein nestin has been detected in a multitude
AJ579701 and AJ579702) (Xue et al., 2004). This gene of cellular phenotypes in embryonic and adult tissues
is localized in human chromosome 15 (Chr.15q26.3) in vivo and in vitro. Cells expressing nestin show char-
and mouse chromosome 7 (Chr.7B5) (Titeux et al., acteristic features of progenitor cells, such as multi-
2001; Xue et al., 2004). The synemin gene is one par- potency, high proliferation, limited self-renewal and
ticular IF gene which produces 3 major mRNAs by regeneration capacity. After differentiation induction
64 Z. Xue et al.

of embryonic stem (ES) cells by tissue-specific growth and Lendahl, 1993). Importantly, nestin is abundant
and extracellular matrix factors, nestin expression is during early neurogenesis (Frederiksen and McKay,
significantly upregulated. In a transient ES cell differ- 1988) and in the peripheral and central nervous sys-
entiation stage, nestin expression is found in different tems, suggesting that nestin-positive cells are critical
subpopulations of potent progenitor cells differentiat- for embryonic neural development (Table 5.2).
ing into mesenchymal/mesodermal, neural, pancreatic In adult organisms, nestin-expressing cells are
endocrine and hepatic cell lineages. restricted to defined locations, where they may func-
Nestin was abundant in embryonic bodies (EB) and tion as a (quiescent) cellular reserve capable of
early EB outgrowths. A significant number of nestin- proliferation, differentiation and migration after re-
positive cells was detected in day 2 and day 6 EB with a activation during tissue regeneration.
significant concentration of nestin-positive cells at the In addition to nestin expression during embryogen-
ectodermal rim of EB (Wiese et al., 2004). esis and in adult tissues, numerous cell types express
After spontaneous differentiation or specific differ- nestin after in vitro cultivation. This could suggest that
entiation induction into neural, pancreatic and hepatic nestin expression may be a consequence of in vitro
cell lineages, a partial and transient co-expression of cultivation conditions that are not necessarily represen-
nestin with lineage-specific marker proteins, including tative of in vivo expression dynamics. Nestin expres-
glial fibrillary acidic protein (GFAP, neuroglial cells), sion is therefore widespread both in vivo and after in
C-peptide (pancreatic cells), albumin (hepatic cells) vitro cultivation of multiple cell lineages. Although,
and desmin (mesenchymal/mesodermal cells). The co- many questions concerning the functional role and
expression of nestin with lineage-specific marker pro- transcriptional regulation of nestin expression in var-
teins was only found at time points that define the onset ious cells and tissues remain open, the data obtained
of cell lineage specification before terminal differenti- until today can lead us to propose nestin as a marker of
ation. With continued differentiation into specialized multi-lineage progenitor cells.
cell types, nestin expression was downregulated in all
differentiated cells.
Nestin-positive neuroglial progenitor cells were 5.5.3 Synemin Expression
found as early as 3 days after the onset of differen-
tiation of ES cells, indicating that ES cells at first The synemins are expressed principally in muscle
differentiate into nestin-expressing cells and later into and nervous system. The expression of synemins are
neuronal and glial cells (Strubing et al., 1995). The dynamic and varied depend the stages of development.
differentiation of ES cells into functional insulin- By means of RT-PCR, the genes encoding M syne-
producing pancreatic cells occurs also via a transient min isoform were already active at E5 when the mouse
stage of progenitor cells expressing C-peptide and embryo has been just implanted (Izmiryan et al., 2009).
nestin (Wiese et al., 2004). Also, spontaneous dif- H synemin mRNA was found later, in E9 embryos, at
ferentiation of ES cells without selection for nestin- a time when vasculogenesis, somitogenesis, the migra-
expressing cells results in hepatic cells that develop tion of neural crest cells and other major activities
from nestin-positive progenitor cells co-expressing are under way in the embryo. L synemin was absent
albumin (Wiese et al., 2004). Altogether, the data from until E11 (Izmiryan et al., 2009). In toto hybridiza-
ES cell in vitro differentiation models suggest that at tion experiment showed that M synemin mRNA was
least neural, pancreatic and hepatic cells are all derived principally accumulated within the embryonic ecto-
via nestin-positive intermediates. derm of E7 embryos (Izmiryan et al., 2009). Strong
During mammalian embryogenesis, nestin is widely labelling was found in the cardiac zone of E8 embryos,
expressed in a variety of embryonic and fetal tis- while E8.5 embryos showed synemin reactivity in the
sues. It is first detected in E5 by RT-PCR (Izmiryan cephalic region, the prosencephalon, mesencephalon
et al., 2009), in the one-cell layer of neural ecto- and rhombencephalon. M synemin mRNA synthesis
derm at day E7 (Kawaguchi et al., 2001; Izmiryan et was also detected at E8.5 in the presomitic mesoderm
al., 2009), in neuroepithelial cells of E7.75 embryos though not in the segmented somites. At E9.5, M syne-
(Dahlstrand et al., 1995), in presomitic mesoderm and min mRNA could be found in the cephalic region of
in the myotome layer of the somites in mice (Sejersen embryos, although it was mostly concentrated in the
5 IF Expression in Mouse Embryonic Stem Cells and Early Embryos 65

Table 5.2 Nestin and synemin expression in mammal embryonic and fetal tissues
Cell types Nestin H synemin M synemin L synemin
Radial glia cells + + +
(Frederiksen and (Hirako et al., 2003; (Hirako et al., 2003; (Izmiryan et al.,
McKay, 1988; Izmiryan et al., Izmiryan et al., 2006)
Tohyama et al., 2006) 2006)
1992)
Astrocytes + + +
(Izmiryan et al., (Hirako et al., 2003; (Hirako et al., 2003; (Izmiryan et al.,
2006) Izmiryan et al., Izmiryan et al., 2006)
2006) 2006)
Schwann cells + + +
(Hockfield and (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
McKay, 1985) 2006) 2006) 2006)
Neuron (CNS) +
(Izmiryan et al., (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
2006) 2006) 2006) 2006)
Neuron (PNS) + + +
(Izmiryan et al., (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
2006) 2006) 2006) 2006)
Neuroepithelial cells + + +
(Dahlstrand et al., (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
1995) 2009) 2009) 2009)
Neural crest cells + + +
(Dahlstrand et al., (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
1995; Hockfield 2009) 2009) 2009)
and McKay,
1985)
Oligodendrocyte + ni ni ni
precursors (Gallo and
Armstrong, 1995)
Sensorial neurons + + +
(Izmiryan et al., (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
2006) 2006) 2006) 2006)
Sympathetic neurons + +
(Izmiryan et al., (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
2006) 2006) 2006) 2006)
Retina + + +
(Walcott and Provis, (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
2003) 2006; Tawk et al., 2006; Tawk et al., 2006)
2003) 2003)
Lens + + +
(Mokry and (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
Nemecek, 1998b) 2006; Tawk et al., 2006; Tawk et al., 2006)
2003) 2003)
Motoneurons ni
(Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
2006) 2006) 2006)
Presomitic mesoderm + +
(Hockfield and (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
McKay, 1985; 2009) 2009) 2009)
Zimmerman
et al., 1994)
Myotome + + +
(Hockfield and (Izmiryan et al., (Izmiryan et al., (Xue et al., 2004)
McKay, 1985; 2009) 2009)
Zimmerman
et al., 1994)
66 Z. Xue et al.

Table 5.2 (continued)


Cell types Nestin H synemin M synemin L synemin
Dermatome + + + ni
(Kachinsky et al., (Izmiryan et al., (Izmiryan et al.,
1994) 2009) 2009)
Striated muscle ni + +
(Xue et al., 2004) (Xue et al., 2004) (Xue et al., 2004)
Smooth muscle ni + +
(Xue et al., 2004) (Xue et al., 2004) (Xue et al., 2004)
Endothelial cells of + + +
developing blood (Mokry and (Izmiryan et al., (Izmiryan et al., (Izmiryan et al.,
vessels Nemecek, 1998a) 2009) 2009) 2009)
Hepatic oval cells + ni ni ni
(Sun and An, 2004)
Hepatic stellate cells ni + + ni
(Schmitt-Graeff (Schmitt-Graeff
et al., 2006; et al., 2006;
Uyama et al., Uyama et al.,
2006) 2006)
Pancreatic epithelial + ni ni ni
progenitor cells (Delacour et al.,
2004; Esni et al.,
2004)
The references are shown in parentheses. ni: no information available.

prosencephalon. At the same stage, the labelling of E17 and in adults, where GFAP was also found. The L
H and M synemin extended to the first two branchial synemin was distributed in different neurons in adults,
arches and the ventricular zone of the heart. In E11.5 including the mouse cortex neurons, hippocampus and
embryos, the limb buds and dorsal root ganglia were the granular neurons of the cerebellum. In contrast,
heavily stained in addition to the heart and cephalic Purkinje cells were immunostained for the H/M iso-
region. At the 25-somite stage, synemins were concen- forms. The L synemin protein was also detected in
trated in the dermomyotomes (Izmiryan et al., 2009). the neurons of the motor column of the spinal cord
However, in contrast to the nestin gene, first expressed from E17, newborns, and adult mice. In the periph-
in neuroepithelial cells and later activated in somites eral nervous system (PNS), L synemin appeared from
originating from the mesoderm at E9 (Lendahl, E13 where it was confined to the neurons of spinal
1997), the M synemin isoform is present from ganglia. In the meantime, the H/M synemin isoforms
both the neuroectoderm and the mesoderm at E7.5 were found in both the neurons and Schwann cells
(Table 5.2). of the sensorial ganglia from E11 (Izmiryan et al.,
In the nervous system, the synthesis of the three 2006). This complex dynamic pattern of synemin
synemin isoforms was probably selected by cell types, isoform synthesis may reflect the reorganization of
and their temporal and spatial distributions. The the cytoskeleton during nervous system development
expression of synemins are dynamic and varied depend (Table 5.2).
the stages of development. H/M synemins occurred The complex distribution patterns of the syne-
together with nestin and vimentin in glial progeni- min isoforms in neural cells raises several questions
tors during the early differentiation of the developing about the regulation of the synemin gene during the
mouse CNS. They are later found in GFAP-labeled determination of glial and neuronal cell lineages in
cells. In contrast, the L isoform appeared only in the CNS and PNS. First, an unexpected finding is the
neurons, together with neurofilaments and beta III- selective synthesis of the two high molecular weight
tubulin in the brain after birth. H/M isoforms were (H/M) synemin isoforms in CNS astrocytes, while the
detected in the medulla oblongata, glia limitans, the smallest synemin isoform (L) is present only in neu-
retina and lens from E13, in ependymal cells from rons. This selectivity suggests that the commitment of
5 IF Expression in Mouse Embryonic Stem Cells and Early Embryos 67

CNS precursor cells to form glia or neuron involves endothelial cells where it was associated with vimentin
the direct regulation of the single synemin gene. As (Izmiryan et al., 2009).
the H/M synemins are present mainly in glial cells and However, the transcriptional regulation of synemin
the L synemin isoform in neurons, it may be partic- in vivo or in vitro is remained to determine.
ularly important to identify the mechanisms whereby
the synemin gene is regulated as part of general inves-
tigation of how stem cells differentiate. Second, the 5.6 Expression of Nestin and Synemin
PNS sensory neurons can contain all three isoforms in Tumoral Cells of the CNS
(H, M, and L), indicating that the production of the
H/M isoforms by glial cells and the L isoform by neu-
rons is not strictly linked to the glia or neuron fates of 5.6.1 Glial Tumors
precursor cells. Instead, it indicates a flexible program
that can be adjusted to each type of nervous system The theory that tumors derive from stem cells is
and may result from the differentiation of radial glial now well established. Stem cells occur in the sub-
cells, which can give rise to both astrocytes and neu- granular layer of the hippocampus and subependymal
rons (Bibel et al., 2004). This modulation of synemin zone, from where they could migrate and colonize
gene regulation within neuron subtypes is also illus- other brain regions. Established neuroectodermal brain
trated by the presence of the H/M synemin isoforms tumors could originate from these migrating stem cells,
in Purkinje cells that lack the L isoform, although it and the tumor mass could be continuously replenished
is present in neighboring granular neurons. We there- with these cells from those clinically silent regions
fore believe that the production of synemin proteins is (Berger et al., 2004). Nestin and synemin are present
closely associated with the early specialization of the in the cells of these silent regions.
cytoplasm of cells in the nervous system. We have immunostained synemin in the ependymal
Out of the nervous system, synemin isoforms are cells from embryonic and adult brain, and a differ-
also expressed in striated muscle (Table 5.2). In muscle ent variant, L synemin, is also present in the mouse
tissues, the synemins are specifically associated with adult neurons of the hippocampus (Izmiryam et al.,
the desmin and vimentin (Xue et al., 2004). Normally, 2006). This may suggest that this marker exists in the
the synemin is associated with desmin in the line Z of ependymal zone earlier during development, present
the sacromere of skeletal muscle and in the interca- in cells that could populate the brain regions, and co-
late disc of cardiac muscle. In the desmin myopathy exists in the glial and neuronal cells during the course
patient, the synemin is colocalized with desmin in of life. Presence of self-renewing neural stem cells in
the desmin aggregates. Ours studies with desmin and the CNS of both children and adults suggests that they
vimentin knock-out (KO) mouse have demonstrated play some role in tumorigenesis (Lewis, 1968) and in
that the synemin is specifically associated with the resistance to current therapeutic strategies (Pilkington,
desmin in the skeletal and cardiac muscle. When the 2005).
desmin is absent, the synemin is also absent in the Glial tumors or gliomas originating from astrocytes
skeletal and cardiac muscle. In the smooth muscle, the are recognized as astrocytomas. Gliomas are the most
synemin is associated with both desmin and vimentin. frequent primary tumors of the CNS and are an impor-
In desmin KO mouse, the synemin is detected in the tant cause of mental impairment and death. They are
smooth muscle that is associated with the vimentin. among the most difficult neoplasms to treat effectively;
In vimentin KO mouse, the expression of synemin is being resistant to different therapeutic procedures. It
decreased in the smooth muscle, but still detected. This is due the combination of their rate of proliferation,
is the synemin associated with the desmin. When both cellular heterogeneity, and capacity to invade dif-
the desmin and the vimentin are absent, the synemin fusely contiguous nervous tissues (Schiffer, 1997).
is absent in all the muscle tissues (Xue et al., 2004). The classification of glioma subfamilies is based on
The synemin was also detected in human liver hep- specific histopathological characteristics (cellularity,
atic stellate cells, where it was co-localized with focal nuclear atypia, mitotic activity, microvascular prolif-
adhesion proteins in long slender processes (Schmitt- eration, and necrosis) in agreement with the World
Graeff et al., 2006; Uyama et al., 2006), and mouse Health Organization (WHO); see Kleihues et al. (2002)
68 Z. Xue et al.

and Kleihues et al. (1995). However, the characteris- with malignant primary tumors of the CNS (Strojnik
tics used to classify these tumors generate complex et al., 2007).
interpretations and differences in diagnoses (Daumas- Nestin, as indicated above, has been detected in pri-
Duport et al., 2000). mary CNS tumors, but not in carcinoma metastases
One of the challenges for cancer control is its (Dahlstrand et al., 1992a; Ikota et al., 2006; Tohyama
early detection. Molecular markers, which could be et al., 1992). More recently, synemin was also detected
signposts to detect tumor cells and indicate their ori- in gliomas.
gin, phenotype, and function, based for instance on
recently developed technologies such as cDNA and
oligonucleotide microarrays on chips, serial analysis
of gene expression (SAGE), and proteomic methods 5.6.3 Synemin Expression in Glioma
to detect cellular proteins implicated in a pathologi-
cal transition, are currently in progress (Snirivas et al.,
As shown earlier, H/M synemin expression is related
2001).
to glial cell fate during mouse CNS development
(Izmiryan et al., 2006). However, H synemin is not
found in the astrocytes of the adult normal human
brain, H synemin is de novo expressed in response to
5.6.2 Nestin in Glioma pathological situations, such as brain injury (reactive
astrocytes) and gliomas (Jing et al., 2005, 2007). In
glioma cells, synemin interacts with -actinin at the
Structural proteins such as cytoskeletal components
leading edge (Jing et al., 2005). It can decorate glial
have been used for many years to characterize the phe-
tumor cells in vitro, and in the tumor in vivo.
notype and tissue origin of the tumor glial cells. GFAP
Recently, it was shown that the presence of synemin
and neurofilament proteins are useful to identify the
at the leading edge also correlated with a high migra-
glial and neuronal origin of the cells present in brain
tory potential of the cells (Pan et al., 2008). In fact, a
tumors (Schiffer, 1997; Zehner, 1991). Recently, intro-
role for synemin in glioma migration was suggested
duction of the nestin protein (Strubing et al., 1995;
by its down-regulation, which sharply decreased the
Wiese et al., 2004) to decorate tumor cells indicated the
migration of gliomas. Therefore, H/M synemin seems
possibility that primitive cells are present in the tumor.
to be an important glioma marker for its invasive-
This nestin expression, in isolation or co-existing with
ness capacity, but this hypothesis needs experimental
GFAP and vimentin, in glial tumors opens this question
demonstration.
(Bao et al., 2006; Faria et al., 2006; Singh et al., 2004,
2003). In fact, nestin has been detected in all the differ-
ent grades of gliomas. Interestingly, the highest level of
nestin-positive cells (mean 25.3%) is found in glioblas-
toma, the most malignant glial tumor (Ma et al., 2005; 5.6.4 And Now
Strojnik et al., 2007). On the other hand, in neuroblas-
toma, another CNS tumor entity, it is very difficult to These are golden days for stem cells. Their contri-
confirm a correlation between nestin expression and its butions to the development and construction of the
malignancy (Korja et al., 2005; Thomas et al., 2004). human body, to regeneration, and the design of new
Co-expression of nestin and vimentin in different therapies have attracted much attention to these cells.
astrocytoma cell lines has been related to a migratory New markers are welcome to identify and follow these
cell phenotype with increased motility and invasive- cells guiding the construction of tissues and organs. A
ness, which revels a metastatic potential of different well-exploited example is the neural crest cells, multi-
astrocytoma cell lines (Rutka et al., 1999). Moreover, potent cells that migrate during early development to
the high level of nestin in tumor cells indicates a construct different tissues and organs (Le Douarin and
prognosis for significantly shorter survival of glioma Kalcheim, 1999; Trentin et al., 2004).
patients. An intense immunostaining of nestin in tumor The cytoskeletal proteins constitute a useful arse-
cells may be used to predict the risk of death in patients nal to recognize cells from different tissues, in order to
5 IF Expression in Mouse Embryonic Stem Cells and Early Embryos 69

analyze cell migration, cell morphology, and morpho- Berger F, Gay E, Pelletier L, Tropel P, Wion D (2004)
logical changes (Zehner, 1991). Development of gliomas: potential role of asymmetrical cell
division of neural stem cells. Lancet Oncol 5:511514.
These proteins are expressed from different genes
Bhosle RC, Michele DE, Campbell KP, Li Z, Robson RM (2006)
and different splices, generating isovariants with dif- Interactions of intermediate filament protein synemin with
ferent isoelectric points or molecular weights. These dystrophin and utrophin. Biochem Biophys Res Commun
variations can be specifically expressed in the cells. 346:768777.
Bibel M, Richter J, Schrenk K, Tucker KL, Staiger V, Korte M,
This is the case for a variant of Tubulin, beta III
Goetz M, Barde YA (2004) Differentiation of mouse embry-
tubulin, which is expressed in the neuronal precursor onic stem cells into a defined neuronal lineage. Nat Neurosci
cells (Menezes and Luskin, 1994). Another example 7:10031009.
is GFAP, which is expressed by five different gene Blechingberg J, Holm IE, Nielsen KB, Jensen TH, Jrgensen
AL, Nielsen AL (2007) Identification and characterization of
transcripts, generated by alternate transcriptional start GFAPkappa, a novel glial fibrillary acidic protein isoform.
sites or by different splicing, alpha (the predominant Glia 55:497507.
isoform of GFAP), beta, gamma, delta, and kappa Brulet P, Babinet C, Kemler R, Jacob F (1980) Monoclonal
(Blechingberg et al., 2007; Condorelli et al., 1999; antibodies against trophectoderm-specific markers during
mouse blastocyst formation. Proc Natl Acad Sci USA 77:
Feinstein et al., 1992; Galea et al., 1995; Zelenika 41134117.
et al., 1995). One of these, GFAP gamma, seems to be Centonze VE, Ruben GC, Sloboda RD (1986) Structure and
a potential marker for a restricted astrocytic population composition of the cytoskeleton of nucleated erythrocytes:
located specifically in the adult human subventricular, III. Organization of the cytoskeleton of Bufo marinus
erythrocytes as revealed by freeze-dried platinum-carbon
subgranular, and subpial zones (Roelofs et al., 2005). replicas and immunofluorescence microscopy. Cell Motil
Furthermore, the antibodies against GFAP delta can Cytoskeleton 6:376388.
immunostain subpial gliosis (Andreiuolo et al., 2009). Cochard P, Paulin D (1984) Initial expression of neurofilaments
Synemin isotypes, H/M and L, have also differ- and vimentin in the central and peripheral nervous system of
the mouse embryo in vivo. J Neurosci 4:20802094.
ent cellular distribution in the CNS. This distribution Condorelli DF, Nicoletti VG, Barresi V, Conticello SG, Caruso
seems correlated with development, with a precocious A, Tendi EA, Giuffrida Stella AM (1999) Structural features
expression in embryonic life. We cannot eliminate of the rat GFAP gene and identification of a novel alternative
the possibility that synemin could with nestin be a transcript. J Neurosci Res 56:219228.
Dahlstrand J, Collins VP, Lendahl U (1992a) Expression of
precocious marker of multipotent cells. the class VI intermediate filament nestin in human central
Now, these isoforms of cytoskeletal proteins could nervous system tumors. Cancer Res 52:53345341.
be a point of investigation toward a possible use of Dahlstrand J, Lardelli M, Lendahl U (1995) Nestin mRNA
these markers in different periods of development or expression correlates with the central nervous system pro-
genitor cell state in many, but not all, regions of develop-
at the moment of commitment of the cells. ing central nervous system. Brain Res Dev Brain Res 84:
109129.
Dahlstrand J, Zimmerman LB, McKay RD, Lendahl U (1992b)
Characterization of the human nestin gene reveals a close
References evolutionary relationship to neurofilaments. J Cell Sci
103:589597.
Andreiuolo F, Junier M-P, Hol EM, Miquet C, Chimelli Daumas-Duport C, Beuvon F, Varlet P, Fallet-Bianco C (2000)
L, Leonard N, Chneiweiss H, Daumas-Duport C, Varlet Gliomas: WHO and Sainte-Anne Hospital classifications [in
P (2009) GFAP immunostaining improves visualiza- French]. Ann Pathol 20:413428.
tion of normal and pathological astrocytic heterogeneity. Delacour A, Nepote V, Trumpp A, Herrera PL (2004) Nestin
Neuropathology 29:3139. expression in pancreatic exocrine cell lineages. Mech Dev
Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, 121:314.
Dewhirst MW, Bigner DD, Rich JN (2006) Glioma stem Le Douarin NM, Kalcheim C (1999) The Neural Crest.
cells promote radioresistance by preferential activation of the Cambridge University Press, Cambridge, Second Edition.
DNA damage response. Nature 444:756760. Escurat M, Djabali K, Gumpel M, Gros F, Portier MM
Bellin RM, Huiatt TW, Critchley DR, Robson RM (2001) (1990) Differential expression of two neuronal intermediate-
Synemin may function to directly link muscle cell interme- filament proteins, peripherin and the low-molecular-mass
diate filaments to both myofibrillar Z-lines and costameres. neurofilament protein (NF-L), during the development of the
J Biol Chem 276:3233032337. rat. J Neurosci 10:764784.
Bellin RM, Sernett SW, Becker B, Ip W, Huiatt TW, Robson Escurat M, Gumpel M, Lachapelle F, Gros F, Portier MM
RM (1999) Molecular characteristics and interactions of (1988) Comparative expression of 2 intermediate filament
the intermediate filament protein synemin. Interactions with proteins, peripherin and the 68 kDa neurofilament protein,
alpha-actinin may anchor synemin-containing heterofila- during embryonal development of the rat. C R Acad Sci III
ments. J Biol Chem 274:2949329499. 306:447456.
70 Z. Xue et al.

Esni F, Stoffers DA, Takeuchi T, Leach SD (2004) Origin of (2003) Characterization of mammalian synemin, an interme-
exocrine pancreatic cells from nestin-positive precursors in diate filament protein present in all four classes of muscle
developing mouse pancreas. Mech Dev 121:1525. cells and some neuroglial cells: co-localization and interac-
Faria J, Romo L, Martins S, Alves T, Mendes FA, de Faria tion with type III intermediate filament proteins and keratins.
GP, Hollanda R, Takiya C, Chimelli L, Morandi V, de Souza Cell Tissue Res 313:195207.
JM, Abreu JG, Moura Neto V (2006) Interactive properties Hockfield S, McKay RD (1985) Identification of major cell
of human glioblastoma cells with brain neurons in cul- classes in the developing mammalian nervous system. J
ture and neuronal modulation of glial laminin organization. Neurosci 5:33103328.
Differentiation 74:562572. Hoffman PN, Lasek RJ (1975) The slow component of axonal
Feinstein DL, Weinmaster GA, Milner RJ (1992) Isolation of transport. Identification of major structural polypeptides of
cDNA clones encoding rat glial fibrillary acidic protein: the axon and their generality among mammalian neurons. J
expression in astrocytes and in Schwann cells. J Neurosci Res Cell Biol 66:351366.
32:114. Ikota H, Kinjo S, Yokoo H, Nakazato Y (2006) Systematic
Franke WW, Grund C, Kuhn C, Jackson BW, Illmensee K immunohistochemical profiling of 378 brain tumors with
(1982a) Formation of cytoskeletal elements during mouse 37 antibodies using tissue microarray technology. Acta
embryogenesis. III. Primary mesenchymal cells and the first Neuropathol 111:475482.
appearance of vimentin filaments. Differentiation 23:4359. Izmiryan A, Cheraud Y, Khanamiryan L, Leterrier JF, Federici
Franke WW, Schmid E, Schiller DL, Winter S, Jarasch ED, T, Peltekian E, Moura-Neto V, Paulin D, Li Z, Xue ZG
Moll R, Denk H, Jackson BW, Illmensee K (1982b) (2006) Different expression of synemin isoforms in glia
Differentiation-related patterns of expression of proteins of and neurons during nervous system development. Glia 54:
intermediate-size filaments in tissues and cultured cells. Cold 204213.
Spring Harb Symp Quant Biol 46:431453. Izmiryan A, Franco CA, Paulin D, Li Z, Xue Z (2009) Synemin
Frederiksen K, McKay RD (1988) Proliferation and differentia- isoforms during mouse development: multiplicity of partners
tion of rat neuroepithelial precursor cells in vivo. J Neurosci in vascular and neuronal systems. Exp Cell Res 315:769
8:11441151. 783.
Fuchs E, Weber K (1994) Intermediate filaments: struc- Jackson BW, Grund C, Winter S, Franke WW, Illmensee
ture, dynamics, function, and disease. Annu Rev Biochem K (1981) Formation of cytoskeletal elements during
63:345382. mouse embryogenesis. II. Epithelial differentiation and
Galea E, Dupouey P, Feinstein DL (1995) Glial fibrillary acidic intermediate-sized filaments in early postimplantation
protein mRNA isotypes: expression in vitro and in vivo. J embryos. Differentiation 20:203216.
Neurosci Res 41:452461. Jing R, Pizzolato G, Robson RM, Gabbiani G, Skalli O (2005)
Gallo V, Armstrong RC (1995) Developmental and growth Intermediate filament protein synemin is present in human
factor-induced regulation of nestin in oligodendrocyte lin- reactive and malignant astrocytes and associates with ruffled
eage cells. J Neurosci 15:394406. membranes in astrocytoma cells. Glia 50:107120.
Galou M, Pournin S, Ensergueix D, Ridet JL, Tchelingerian JL, Jing R, Wilhelmsson U, Goodwill W, Li L, Pan Y, Pekny M,
Lossouarn L, Privat A, Babinet C, Dupouey P (1994) Normal Skalli O (2007) Synemin is expressed in reactive astrocytes
and pathological expression of GFAP promoter elements in in neurotrauma and interacts differentially with vimentin
transgenic mice. Glia 12:281293. and GFAP intermediate filament networks. J Cell Sci 120:
Goldman RD (2001) Worms reveal essential functions for inter- 12671277.
mediate filaments. Proc Natl Acad Sci USA 98:76597661. Kachinsky AM, Dominov JA, Miller JB (1994) Myogenesis and
Granger BL, Lazarides E (1980) Synemin: a new high molec- the intermediate filament protein, nestin. Dev Biol 165:216
ular weight protein associated with desmin and vimentin 228.
filaments in muscle. Cell 22:727738. Kaplan MP, Chin SS, Fliegner KH, Liem RK (1990) Alpha-
Granger BL, Lazarides E (1982) Structural associations of syne- internexin, a novel neuronal intermediate filament pro-
min and vimentin filaments in avian erythrocytes revealed by tein, precedes the low molecular weight neurofilament pro-
immunoelectron microscopy. Cell 30:263275. tein (NF-L) in the developing rat brain. J Neurosci 10:
Granger BL, Lazarides E (1984) Expression of the intermediate- 27352748.
filament-associated protein synemin in chicken lens cells. Kawaguchi A, Miyata T, Sawamoto K, Takashita N, Murayama
Mol Cell Biol 4:19431950. A, Akamatsu W, Ogawa M, Okabe M, Tano Y, Goldman SA,
Guerette D, Khan PA, Savard PE, Vincent M (2007) Molecular Okano H (2001) Nestin-EGFP transgenic mice: visualization
evolution of type VI intermediate filament proteins. BMC of the self-renewal and multipotency of CNS stem cells. Mol
Evol Biol 7:164164. Cell Neurosci 17:259273.
Herrmann H, Aebi U (2000) Intermediate filaments and their Khanamiryan L, Li Z, Paulin D, Xue Z (2008) Self-assembly
associates: multi-talented structural elements specifying incompetence of synemin is related to the property of its head
cytoarchitecture and cytodynamics. Curr Opin Cell Biol and rod domains. Biochemistry 47:95319539.
12:7990. Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger
Herrmann H, Aebi U (2004) Intermediate filaments: molecular G, Burger PC, Cavenee WK (2002) The WHO classification
structure, assembly mechanism, and integration into func- of tumors of the nervous system. J Neuropathol Exp Neurol
tionally distinct intracellular Scaffolds. Annu Rev Biochem 61:215225.
73:749789. Kleihues P, Soylemezoglu F, Schauble B, Scheithauer BW,
Hirako Y, Yamakawa H, Tsujimura Y, Nishizawa Y, Okumura M, Burger PC (1995) Histopathology, classification, and grading
Usukura J, Matsumoto H, Jackson KW, Owaribe K, Ohara O of gliomas. Glia 15:211221.
5 IF Expression in Mouse Embryonic Stem Cells and Early Embryos 71

Korja M, Finne J, Salmi TT, Kalimo H, Karikoski R, Tanner Pachter JS, Liem RK (1985) Alpha-internexin, a 66-kD inter-
M, Isola J, Haapasalo H (2005) Chromogenic in situ mediate filament-binding protein from mammalian central
hybridization-detected hotspot MYCN amplification asso- nervous tissues. J Cell Biol 101:13161322.
ciates with Ki-67 expression and inversely with nestin Pan Y, Jing R, Pitre A, Williams BJ, Skalli O (2008) Intermediate
expression in neuroblastomas. Mod Pathol 18:15991605. filament protein synemin contributes to the migratory prop-
Lane EB, Hogan BL, Kurkinen M, Garrels JI (1983) Co- erties of astrocytoma cells by influencing the dynamics of the
expression of vimentin and cytokeratins in parietal endoderm actin cytoskeleton. FASEB J 22:31963206.
cells of early mouse embryo. Nature 303:701704. Paulin D, Babinet C, Weber K, Osborn M (1980) Antibodies as
Lazarides E (1981) Intermediate filaments chemical hetero- probes of cellular differentiation and cytoskeletal organiza-
geneity in differentiation. Cell 23:649650. tion in the mouse blastocyst. Exp Cell Res 130:297304.
Lehtonen E, Lehto VP, Paasivuo R, Virtanen I (1983a) Parietal Pilkington GJ (2005) Cancer stem cells in the mammalian
and visceral endoderm differ in their expression of interme- central nervous system. Cell Prolif 38:423433.
diate filaments. EMBO J 2:10231028. Portier MM, de Nechaud B, Gros F (1983) Peripherin, a new
Lehtonen E, Lehto VP, Vartio T, Badley RA, Virtanen I (1983b) member of the intermediate filament protein family. Dev
Expression of cytokeratin polypeptides in mouse oocytes and Neurosci 6:335344.
preimplantation embryos. Dev Biol 100:158165. Roelofs RF, Fischer DF, Houtman SH, Sluijs JA, Van Haren
Lendahl U (1997) Transgenic analysis of central nervous sys- W, Van Leeuwen FW, Hol EM (2005) Adult human sub-
tem development and regeneration. Acta Anaesthesiol Scand ventricular, subgranular, and subpial zones contain astrocytes
110:116118. with a specialized intermediate filament cystoskeleton. Glia
Lendahl U, Zimmerman LB, McKay RD (1990) CNS stem cells 52:289300.
express a new class of intermediate filament protein. Cell Rutka JT, Ivanchuk S, Mondal S, Taylor M, Sakai K, Dirks P, Jun
60:585595. P, Jung S, Becker LE, Ackerley C (1999) Co-expression of
Lewis PD (1968) Mitotic activity in the primate subependymal nestin and vimentin intermediate filaments in invasive human
layer and the genesis of gliomas. Nature 217:974975. astrocytoma cells. Int J Dev Neurosci 17:503515.
Lothian C, Lendahl U (1997) An evolutionarily conserved region Schiffer D (1997) Brain Tumors, Biology, Pathology, and
in the second intron of the human nestin gene directs gene Clinical References. Springer Verlag, Berlin, Second Revised
expression to CNS progenitor cells and to early neural crest Edition.
cells. Eur J Neurosci 9:452462. Schmitt-Graeff A, Jing R, Nitschke R, Desmouliere A, Skalli O
Lothian C, Prakash N, Lendahl U, Wahlstrom GM (1999) (2006) Synemin expression is widespread in liver fibrosis and
Identification of both general and region-specific embryonic is induced in proliferating and malignant biliary epithelial
CNS enhancer elements in the nestin promoter. Exp Cell Res cells. Hum Pathol 37:12001210.
248:509519. Sejersen T, Lendahl U (1993) Transient expression of the inter-
Ma DK, Ming GL, Song H (2005) Glial influences on neural mediate filament nestin during skeletal muscle development.
stem cell development: cellular niches for adult neurogene- J Cell Sci 106:12911300.
sis. Curr Opin Neurobiol 15:514520. Singh SK, Clarke ID, Hide T, Dirks PB (2004) Cancer stem cells
Maro B, Kubiak J, Gueth C, De Pennart H, Houliston E, in nervous system tumors. Oncogene 23:72677273.
Weber M, Antony C, Aghion J (1990) Cytoskeleton orga- Singh SK, Clarke ID, Terasaki M, Bonn VE, Hawkins C, Squire
nization during oogenesis, fertilization and preimplantation J, Dirks PB (2003) Identification of a cancer stem cell in
development of the mouse. Int J Dev Biol 34:127137. human brain tumors. Cancer Res 63:58215828.
Marvin MJ, Dahlstrand J, Lendahl U, McKay RD (1998) A Srinivas PR, Kramer BS, Srivastava S (2001) Trends in
rod end deletion in the intermediate filament protein nestin biomarker research for cancer detection. Lancet Oncol
alters its subcellular localization in neuroepithelial cells of 2:698704.
transgenic mice. J Cell Sci 111:19511961. Steinert PM, Chou YH, Prahlad V, Parry DA, Marekov LN,
McKeon FD, Kirschner MW, Caput D (1986) Homologies in Wu KC, Jang SI, Goldman RD (1999) A high molecular
both primary and secondary structure between nuclear enve- weight intermediate filament-associated protein in BHK-
lope and intermediate filament proteins. Nature 319:463 21 cells is nestin, a type VI intermediate filament protein.
468. Limited co-assembly in vitro to form heteropolymers with
Menezes JR, Luskin MB (1994) Expression of neuron-specific type III vimentin and type IV alpha-internexin. J Biol Chem
tubulin defines a novel population in the proliferative layers 274:98819890.
of the developing telencephalon. J Neurosci 14:53995416. Strelkov SV, Herrmann H, Aebi U (2003) Molecular architecture
Mokry J, Nemecek S (1998a) Angiogenesis of extra- and of intermediate filaments. Bioessays 25:243251.
intraembryonic blood vessels is associated with expression Strojnik T, Rosland GV, Sakariassen PO, Kavalar R, Lah
of nestin in endothelial cells. Folia Biol (Praha) 44:155161. T (2007) Neural stem cell markers, nestin and musashi
Mokry J, Nemecek S (1998b) Immunohistochemical detec- proteins, in the progression of human glioma: correlation
tion of intermediate filament nestin. Acta Medica (Hradec of nestin with prognosis of patient survival. Surg Neurol
Kralove) 41:7380. 68:133143.
Oshima RG (2007) Intermediate filaments: a historical perspec- Strubing C, Ahnert-Hilger G, Shan J, Wiedenmann B,
tive. Exp Cell Res 313:19811994. Hescheler J, Wobus AM (1995) Differentiation of pluripotent
Oshima RG, Howe WE, Klier FG, Adamson ED, Shevinsky LH embryonic stem cells into the neuronal lineage in vitro gives
(1983) Intermediate filament protein synthesis in preimplan- rise to mature inhibitory and excitatory neurons. Mech Dev
tation murine embryos. Dev Biol 99:447455. 53:275287.
72 Z. Xue et al.

Sultana S, Sernett SW, Bellin RM, Robson RM, Skalli O Uyama N, Zhao L, Van Rossen E, Hirako Y, Reynaert H,
(2000) Intermediate filament protein synemin is transiently Adams DH, Xue Z, Li Z, Robson R, Pekny M, Geerts
expressed in a subset of astrocytes during development. Glia A (2006) Hepatic stellate cells express synemin, a pro-
30:143153. tein bridging intermediate filaments to focal adhesions. Gut
Sun XY, An J (2004) Expression of nestin, an intermediate fila- 55:12761289.
ment protein, in human fetal hepatic stem cells. Di Yi Jun Yi Walcott JC, Provis JM (2003) Muller cells express the neuronal
Da Xue Xue Bao 24:207209. progenitor cell marker nestin in both differentiated and undif-
Sun N, Critchley DR, Paulin D, Li Z, Robson RM (2008a) ferentiated human foetal retina. Clin Experiment Ophthalmol
Human alpha-synemin interacts directly with vinculin and 31:246249.
metavinculin. Biochem J 409:657667. Wiese C, Rolletschek A, Kania G, Blyszczuk P, Tarasov
Sun N, Critchley DR, Paulin D, Li Z, Robson RM (2008b) KV, Tarasova Y, Wersto RP, Boheler KR, Wobus
Identification of a repeated domain within mammalian alpha- AM (2004) Nestin expression a property of
synemin that interacts directly with talin. Exp Cell Res multi-lineage progenitor cells? Cell Mol Life Sci
314:18391849. 61:25102522.
Tawk M, Titeux M, Fallet C, Li Z, Daumas-Duport C, Xue ZG, Cheraud Y, Brocheriou V, Izmiryan A, Titeux M,
Cavalcante LA, Paulin D, Moura-Neto V (2003) Synemin Paulin D, Li Z (2004) The mouse synemin gene encodes
expression in developing normal and pathological human three intermediate filament proteins generated by alternative
retina and lens. Exp Neurol 183:499507. exon usage and different open reading frames. Exp Cell Res
Thomas SK, Messam CA, Spengler BA, Biedler JL, Ross RA 298:431444.
(2004) Nestin is a potential mediator of malignancy in human Yaworsky PJ, Kappen C (1999) Heterogeneity of neural progen-
neuroblastoma cells. J Biol Chem 279:2799427999. itor cells revealed by enhancers in the nestin gene. Dev Biol
Titeux M, Brocheriou V, Xue Z, Gao J, Pellissier JF, Guicheney 205:309321.
P, Paulin D, Li Z (2001) Human synemin gene generates Zehner ZE (1991) Regulation of intermediate filament gene
splice variants encoding two distinct intermediate filament expression. Curr Opin Cell Biol 3:6774.
proteins. Eur J Biochem 268:64356449. Zelenika D, Grima B, Brenner M, Pessac B (1995) A novel glial
Tohyama T, Lee VM, Rorke LB, Marvin M, McKay RD, fibrillary acidic protein mRNA lacking exon 1. Brain Res
Trojanowski JQ (1992) Nestin expression in embryonic Mol Brain Res 30:251258.
human neuroepithelium and in human neuroepithelial tumor Zimmerman L, Parr B, Lendahl U, Cunningham M, McKay
cells. Lab Invest 66:303313. R, Gavin B, Mann J, Vassileva G, McMahon A (1994)
Trentin A, Glavieux-Pardanaud C, Le Douarin NM, Dupin E Independent regulatory elements in the nestin gene direct
(2004) Self-renewal capacity is a widespread property of var- transgene expression to neural stem cells or muscle precur-
ious types of neural crest precursor cells. Proc Natl Acad Sci sors. Neuron 12:1124.
USA 101:44954500.
Chapter 6

Aneuploidy in Embryonic Stem Cells

Rafaela C. Sartore, Priscila B. Campos, Michael J. McConnell, and Stevens K. Rehen

Contents great potential for use in both basic science and thera-
peutic strategies, including transplantation for regen-
6.1 Introduction . . . . . . . . . . . . . . . . 74 erative medicine. A challenge for cell therapy using
6.2 A Brief History of Aneuploidy . . . . . . . . 74 hES is the maintenance of stable cell lines, particularly
6.3 Cell Cycle Checkpoints Maintain Genome following extended passaging. Chromosomal instabil-
Integrity . . . . . . . . . . . . . . . . . . 74
ity has recently been reported in hES, resulting from
6.4 Increased Levels of Aneuploidy Indicates
Reduced Checkpoint Fidelity in clonal expansion of aneuploid cells. It includes hyper-
Stem/Progenitor Cells . . . . . . . . . . . . 76 ploidies, particularly trisomies of chromosomes 12, 17
6.5 DNA Damage Signaling and Aneuploidy . . . 77 or 20 and, to a lesser extent, chromosome loss. The
6.6 Does Aneuploidy in Stem and/or Progenitor
generality of this phenomenon is uncertain and it is
Cells Have Consequences for Development and
Disease? . . . . . . . . . . . . . . . . . . 79 currently unclear whether aneuploidy in stem cell lines
6.7 Aneuploidy and Cancer Stem Cells . . . . . 80 is in fact deleterious. What is the physiological signif-
6.8 Telomeres and Telomerase Under Genomic icance or therapeutic risk associated with hES aneu-
Stability Control . . . . . . . . . . . . . . 80
ploidy? Here, features of cell cycle and the relevance
6.9 Aneuploidy and Cell-Based Therapy . . . . . 81
6.9.1 Mechanical Versus Enzymatic Methods 81 of aneuploidy are discussed on regard of its implica-
6.9.2 Risks and Benefits of Aneuploidy to tions for the physiology and therapeutic purposes of
Cell-Based Therapies . . . . . . . . 82 embryonic stem cells.
References . . . . . . . . . . . . . . . . . . . . 83
Keywords Aneuploidy Cancer stem cells Cell cycle
checkpoint Embryonic stem cells Genome integrity
Abstract Aneuploidy is defined as the loss and/or
gain of chromosomes to produce a numerical deviation Abbreviations
from multiples of the haploid chromosomal comple-
ment. This phenomenon can be classified as a net AD Alzheimers disease
increase in chromosome number, referred to as hyper- APC/C anaphase promoting complex-cyclosome
ploidy, or a net decrease, referred to as hypoploidy. ATM ataxia telangiectasia-mutated protein
Biologically, chromosome number can range from kinase
0 (red blood cells) to polyploidy (hepatocytes, neo- Bub Budding Uninhibited by Benzimidazole
plasms). Human embryonic stem cells (hES) have CSC cancer stem cell(s)
Cdc Cell division control protein
Chk Checkpoint kinase
CDK Cyclin-dependent kinase activity
CKI Cyclin Kinase Inhibitors
dNTP deoxyribonucleotide triphosphate(s)
S.K. Rehen ()
Instituto de Cincias Biomdicas, Universidade Federal do Rio EB Embryoid Bodies
de Janeiro, Rio de Janeiro, Brazil EC cell(s) Embryonal Carcinoma cell(s)
e-mail: srehen@anato.ufrj.br ES cell(s) Embryonic Stem cell(s)

H. Ulrich (ed.), Perspectives of Stem Cells, 73


DOI 10.1007/978-90-481-3375-8_6, Springer Science+Business Media B.V. 2010
74 R.C. Sartore et al.

FISH Fluorescence In Situ Hybridization leads to four, rather than two, blastomeres after the first
G phasesGap phases mitosis. Given that this unexpected fertilization pro-
pRb Retinoblastoma protein cess was initiated with two centrosomes (two sperm
IVF In Vitro Fertilization cells) and three chromosomal complements, Boveri
mEF mouse embryonic fibroblast(s) deduced that a quadripolar mitotic spindle might
CENP-E Microtubule-dependent motor have been generated which in turn segregated the 3n
centromere-associated protein E genome into four unequal, or at least non-euploid,
Mps1 Mitogen-activated protein kinase 1 fractions. Boveri suggested that aneuploid blastomeres
M phase Mitosis phase might have limited developmental potential relative to
Mad Mitotic arrest deficient euploid blastomeres.
MVA Mosaic Variegated Aneuploidy
By testing this hypothesis, he found that blas-
NPC neural progenitor cell(s)
tomeres of dispermic eggs differed from each other
OCT-4 Octamer-binding transcription factor 4
in their development. Dispermic-derived blastomeres
p19/ARF p19 alternative reading frame protein
arrested during development at the blastula stage,
SAC Spindle Assembly Checkpoint
SSEA-1 Stage-Specific Embryonic Antigen 1 whereas those derived from normal eggs completed
S phase Synthesis phase development. Boveri based the explanation of this
p53 (53 kilodalton) tumor suppressor protein phenomenon on the distribution of the chromosomes.
Furthermore, he suggested that the function of bipolar
mitotic figure is to multiply successively the nucleus in
its totality and transfer of the qualities present in one
6.1 Introduction nucleus into many nuclei (Boveri, 1902). From this,
Boveri postulated that not only the number but also
The organization of mammalian genomes is such that the quality of the chromosomes was crucial for nor-
an individual cell has two copies of each chromo- mal development, calling attention to the importance of
some. At fertilization, each copy comes from a parental maintaining genomic integrity during cell proliferation
gamete. When this event occurs, typically the result- (Manchester, 1995; Satzinger, 2008).
ing blastomere is euploid and its genome is diploid Soon after, Boveri identified a correlation between
(2n): 2n represents 40 chromosomes in mice and 46 aneuploidy and tumor formation (Nyberg et al., 2002).
chromosomes in human. Chromosomal aberrations, including aneuploidy, are
Although euploidy predominates, certain deviations hallmark of many types of cancer. However, it is still
can be observed. When an entire chromosome comple- debated whether aneuploidy could be a determinant in
ment is over duplicated, these cells become polyploid cellular transformation (Rasnick and Duesberg, 1999;
(i.e. 3n is triploid, 4n is tetraploid). However, occur- Duesberg et al., 2005) or whether it could even con-
ring more frequently during mammalian development tribute to tumor avoidance (Weaver et al., 2007; Torres
are single chromosome losses and gains, referred to as et al., 2008). Indeed, aneuploidy on primary mouse
aneuploidies, found in embryonic stem (ES) cell and cells impairs proliferation and altered metabolic prop-
neural progenitor cell (NPC) populations. Herein, we erties. Immortalization, the acquisition of the ability
review the mechanisms by which altered cell cycle to proliferate indefinitely, was also abrogated by the
controls can give rise to aneuploidy, discuss certain presence of an additional copy of certain chromosomes
consequences of aneuploidy in human development (Williams et al., 2008).
and disease, and introduce some of the challenges that
aneuploidy brings to cell-based therapies.
6.3 Cell Cycle Checkpoints Maintain
Genome Integrity
6.2 A Brief History of Aneuploidy
In order to generate two daughter cells with identi-
Aneuploidy was first observed during early embryoge- cal genomes and equal size relative to the original
nesis in sea urchins. As reported by Theodor Boveri mother cell, a proliferating cell must coordinate
in 1902, the fertilization of one egg by two sperm cells growth with genome duplication and genome
6 Aneuploidy in Embryonic Stem Cells 75

segregation. Over the course of a cell cycle, genome Cell cycle transitions are driven by cyclin-
duplication is performed during the S (synthesis) phase dependent kinase activity (CDK); checkpoints inhibit
and genome segregation occurs during the M (mitosis) Cdk activity to prevent cell cycle progression. Cdk
phase. These two phases are separated by two gaps of complexes are comprised of an enzymatic subunit, the
variable lengths of time, named G1, located between Cdk core, and an activating subunit called cyclin. The
the M and S phases, and G2, located between the S molecular machinery that promotes and inhibits cellu-
and M phases. These gap phases allow cell growth and lar proliferation includes fluctuations between cyclin
are monitored by surveillance mechanisms known as synthesis and degradation, deactivating phosphoryla-
checkpoints. tion and activating dephosphorylation of cyclin-Cdk
Checkpoints control major transitions in the cell complexes and the presence of cyclin-Cdk inhibitory
cycle, verifying that all processes related to each proteins, called cyclin kinase inhibitors (CKI), such
phase have been appropriately completed before the as p16, p18, p21 and p27 (van den Heuvel, 2005)
next phase is initiated. Furthermore, they function (Fig. 6.1).
to ensure that genome integrity is maintained, as Different cyclin-Cdk complexes are formed and
well as to delay crucial events like DNA synthe- activated in each phase of the cell cycle. In response
sis or chromosomal segregation into daughter cells to mitogenic and growth factors in early G1 phase,
in response to abnormal conditions. For example, cyclin D is expressed and its gene product forms a
upon detection of DNA damage, cells arrest cell complex with Cdk4 and Cdk6 to promote G1 phase
cycle progression in G1 or G2 phases (Lew and progression. The G1/S transition is regulated by the
Burke, 2003; Nyberg et al., 2002). In order to pre- retinoblastoma tumor suppressor protein (pRb). E2F-
vent aneuploidy, the spindle assembly checkpoint DP-dependent transcription promotes entry into S
(SAC) delays anaphase until all sister chromatids phase, and hypophosphorylated Rb represses the E2F-
have been attached to mitotic spindle microtubules, DP family of transcription factors. Active cyclin D-
thus providing time for repairing errors and avoid- Cdk4/6 phosphorylates pRb to allow the transcription
ing chromosomal aberrations (Lew and Burke, 2003). of cyclin E. Cyclin E-Cdk2, in turn, hyperphosphory-
In the presence of unattached chromosomes to the lates pRb, leading to its complete dissociation from
mitotic spindle, anaphase is inhibited by kinetochore- E2F-DP and the derepression of S phase promoting
associated mitotic checkpoint machinery including genes, notably cyclin A. The cyclin A-Cdk2 complex
Bub1, Bub3, BubR1, Mad1, Mad2, Mad3, Mps1 and sustains S phase progression and cyclin A-Cdk1 will
CENP-E. More specifically, this accumulation inhibits be required later on during the G2 phase. In order to
the anaphase promoting complex-cyclosome (APC/C), promote mitosis, cyclin B-Cdk1 is activated and trig-
whose activity is required for securin degradation gers a variety of processes that peak with cytokinesis
and anaphase progression (Musacchio and Salmon, (Schwartz and Shah, 2005; Rane and Reddy, 2000)
2007). (Fig. 6.1).

Fig. 6.1 Simple


representation of a typical
cell cycle and checkpoints
76 R.C. Sartore et al.

In mammalian somatic cells, the fidelity of chro- mostly hypoploidy, in normal mouse NPC results from
mosome transmission is secured by checkpoints, and chromosome missegregation and multipolar spindles
when there is an ablation in checkpoint functions, ane- are readily observed in these cells (Rehen et al., 2001;
uploidy can arise (Weaver et al., 2007).Although it Yang et al., 2003). Although the exact mechanisms by
is assumed that all normal mammalian cells maintain which NPC cell cycles permit aneuploidy is not yet
genomic integrity through these control mechanisms, clear, it is likely related to the suspension of the decate-
compelling evidence shows that this is not a common nation checkpoint as observed in these cells and also
feature in embryonic stem (ES) cells (Neganova and ES cells (Damelin et al., 2005).
Lako, 2008). In early development, the strict requirements for cell
growth are suspended to allow rapid cell proliferation.
Rapid cell division leads to an unusual cell cycle dis-
tribution: more than 50% of the ES cell populations
6.4 Increased Levels of Aneuploidy observed are found in S phase, whereas smaller frac-
Indicates Reduced Checkpoint tions (~25%) are found in G1 or in G2/M phases (Stead
Fidelity in Stem/Progenitor Cells et al., 2002; Fluckiger et al., 2006). It is important to
note that early NPC cell cycles are also very rapid
(~8 h), followed by a progressive lengthening of G1
Despite robust checkpoint control described in sev-
phase to ~24 h by the end of cortical neurogenesis
eral cell types, aneuploidy is still observed in certain
(Miyama, Cerebral Cortex, 1997).
stem/progenitor cell populations. Specifically, ~35%
Rapid ES cell cycle is achieved through the irregu-
of mouse neural progenitor cells (NPC) are aneuploid
lar use of cell cycle control molecules (e.g. cyclins and
(Rehen et al., 2001). These aneuploid NPC give rise to
CDK) (Fluckiger et al., 2006). ES cells derived from
aneuploid neurons in mouse and in human (Kingsbury
mice and primates exhibit elevated levels of cyclin E
et al., 2005; Rehen et al., 2005) (Fig. 6.2). Aneuploidy,
and cyclin A throughout the whole cell cycle, whereas
the functions of these cyclins are typically restricted
to G1 or S phases (Sancar et al., 2004). Mouse ES
cells also show high Cdk2 activity in a cell cycle-
independent manner, which can be explained in part
by the elevated expressions of cyclin E and cyclin A.
Likewise, the Cdk inhibitors p21 and p27 are almost
undetectable, hence favoring the absence of Cdk2 reg-
ulation throughout the cell cycle (Linke et al., 1996;
Stead et al., 2002) (Fig. 6.3).
Constitutive expression of cyclin E results in kary-
otypic instability in mammalian cells (Spruck et al.,
1999). Similarly, high levels of cyclin E are correlated
to breast, endometrial and skin cancers and also with
aneuploidization (Dutta et al., 1995; Keyomarsi et al.,
1995; Hubalek et al., 2004; Bito et al., 1997).
In addition to cyclins, the Rb protein, whose phos-
phorylation leads to S phase progression, is mainly
found hyperphosphorylated in undifferentiated primate
ES cells (Fluckiger et al., 2006). Moreover, down-
Fig. 6.2 Aneuploid neurons are present in the brains of
adult mice. Cells from the cerebral cortex of a male mouse (box stream E2F-DP target genes, such as B-myb, RRMP2
in bottom image) can be hybridized with chromosome paints and cdc2, are continuously transcribed throughout the
(top image) to show variations in the chromosome number (red, ES cell cycle (Stead et al., 2002). Together, consti-
X chromosome; green, Y chromosome; blue, DAPI stain). Note
tutive cyclin A/E-Cdk2 activity and a silenced pRb
that the cell on the left has one Y chromosome but two X
chromosomes while the other two cells have a single X and Y control provide rapid ES cell division (Stead et al.,
chromosome. Provided courtesy of Marcy Kingsbury 2002; Fluckiger et al., 2006) (Fig. 6.3).
6 Aneuploidy in Embryonic Stem Cells 77

Fig. 6.3 Embryonic cell


cycle representation
composed basically by M
and S phases. Note
constitutive activation of
cyclin-Cdk complexes typical
of G1/S and S phases and the
predominance of the
hyperphosphorylated form of
pRb. ES cells are refractory to
CKI inhibition (p16, p21,
p27)

The Rb pathway is often inactivated in cancer. particularly prone to differentiate between the M and S
Interestingly, Rb disruption generates chromosomal phases, that is, during G1 phase, and become less sus-
instability by destabilizing Mad2 expression, a com- ceptible to differentiate upon S phase (Mummery et al.,
ponent of SAC. Ablation of Rb function produces 1987a).
deregulated E2F-DP activity and consequently the Taken together, the emerging data point out that
overexpression of Mad2, an E2F-DP target gene, thus ES cells guarantee rapid cell division by bypassing
leading to uncontrolled mitosis and finally resulting in certain mechanisms of cell cycle control and they
aneuploidy (Hernando et al., 2004). are primed to comprise of a lengthened S phase and
It was initially thought that cyclin D-associated shortened gap phases. In other words, they are inces-
activities were absent in ES cells (Savatier et al., santly ready to replicate. Like ES cells, other pluripo-
1996). However, subsequent studies have shown that tent cell types also exhibit an extensive proportion
cyclin D1 and cyclin D3 are expressed, but at low of cells in S phase. This characteristic is common
levels in murine ES cells. On the other hand, high to some embryonic carcinoma (EC) cells (Mummery
activity of cyclin D3-Cdk6 is present, but this com- et al., 1987b; Kranenburg et al., 1995), embryonic
plex is not regulated by p16 (Faast et al., 2004) germ cells (Resnick et al., 1992) and embryonic epi-
(Fig. 6.3). blast cells (Mac Auley et al., 1993), and is postu-
When ES cells are induced to differentiate into lated to be a property of stemness (Fluckiger et al.,
embryoid bodies (EB), G1 phase is lengthened con- 2006).
comitantly with the loss of pluripotency markers and
increased levels of differentiation markers (White
et al., 2005). At the same time, the high activity of
Cdk2, cyclin A and cyclin E, whose activities are
6.5 DNA Damage Signaling and
cell cycle-independent in ES cells, are reduced and
become cell cycle-regulated upon differentiation. It
Aneuploidy
is underpinned by elevated levels of p21 and p27
CKIs and the establishment of cell cycle regulated A striking difference has been demonstrated between
pRb-E2F activity (White et al., 2005). ES cells also somatic and embryonic stem cells: Checkpoints con-
become sensitive to p16 activity (Savatier et al., trolling the different phases transitions are neglected
1996). in ES cells.
Pluripotent cells may shorten their G1 phase and Cells have a normal load of DNA damage that is
reject their delaying components to preserve their managed by cell cycle checkpoints. In the presence
pluripotency properties. In line with this idea, it has of DNA damage, entry into S phase is prevented by
been observed that P19 embryonal carcinoma cells are the G1/S checkpoint that arrests cell cycle progression
78 R.C. Sartore et al.

at the G1 phase. The checkpoint response is initiated which is responsible for inducing G1 arrest (Hong and
when the DNA-damage response kinase ATM senses Stambrook, 2004).
DNA damage and phosphorylates target molecules Another event that indicates the uncoupling of
including Chk1/2 and p53. Activated Chk1/2 phospho- checkpoint-apoptosis in ES cells can be observed
rylates Cdc25A phosphatase, targeting it for ubiquitin- by testing the SAC and the checkpoint induced by
dependent degradation. In turn, the phosphorylated DNA double-strand breaks. The treatment of mouse
form of Cdk2 (inactive) accumulates, caused by the and human ES cells with drugs that promote micro-
lack of Cdc25A activity and thus, is unable to trig- tubule disruption and SAC activation do not hinder
ger DNA replication. The phosphorylation of p53 them from achieving mitosis. While prolonged expo-
obstructs its nuclear export and degradation, leading sure of normal somatic cells to microtubule disrupting
to its accumulation in the nucleus and the activation of agents induces apoptosis or senescence, embryonic
target genes, notably p21. The Cdk inhibitor, p21, ulti- stem cells bypass mitotic delay and complete cytokine-
mately blocks cell proliferation by inhibiting the cyclin sis, resulting in polyploidy/aneuploidy cells which do
E-Cdk2 complex, maintaining G1 arrest (Sancar et al., not elicit apoptosis. Similarly, mouse ES cells exposed
2004). to double-strands breaks become both aneuploid and
Another signal that promotes G1 arrest and is inde- resistant to apoptosis (Mantel et al., 2007). Therefore,
pendent of DNA damage occurs by the depletion of ES cells are allowed to enter into an aneuploid cell
cellular deoxyribonucleotide triphosphates (dNTP). In cycle.
this condition, somatic cells inhibit DNA synthesis via In 2005, Damelin suggested that deficiency in
p53 and p21 activities and prevent defects during cell checkpoints could be a feature of progenitor cells. In
division (Linke et al., 1996). his work, the decatenation checkpoint efficiency was
In this scenario, the shortage of nucleotides does tested by inhibiting topoisomerase II on mouse ES
not elicited cell cycle arrest in ES cells and they cells, mouse NPC and human haematopoietic progeni-
continue to enter into S phase, even in such a subop- tor cells. The decatenation checkpoint delays the entry
timal growth condition (Hong and Stambrook, 2004; into mitosis during the G2 phase if any chromosome
Aladjem et al., 1998). Also, when submitted to DNA has not yet been appropriately disentangled by topoi-
lesions, ES cells enter into S phase in a propor- somerase II (Damelin and Bestor, 2007). Stem and
tion similar to cells that have not been exposed to progenitor cells are observed to enter mitosis even
any DNA challenges and no accumulation of cells in the presence of entangled chromosomes, reflecting
in G1 or G2 phases is observed (Aladjem et al., a silenced decatenation checkpoint control, possibly
1998). leading to aneuploid daughter cells (Damelin et al.,
The inability of ES cells to undergo cell cycle 2005).
arrest stimulated by either dNTP depletion or DNA Hence, ES cells have inefficient checkpoints that
damage infers that ES cells do not present a robust p53- allow them to progress through cell cycle even under
mediated DNA-damage-response pathway (Aladjem et abnormal conditions and enable them to escape apop-
al., 1998). ES cells do express the p53 protein however, tosis. However, as they differentiate into EB, the
because it is inefficiently translocated into the nucleus checkpoints are restored and aneuploid cells initiate
after DNA damage, it remains predominantly cytoplas- robust apoptosis, consistent with other somatic cells.
mic. This is consistent with the undetectable levels of SAC and DNA-damage checkpoint activation then
p21 in these situations. The occurrence of apoptosis become coupled to apoptosis (Mantel et al., 2007)
through other p53 independent pathways, confirm that and differentiation of ES cells is accompanied by an
ES cells have an inefficient p53 response (Aladjem increased efficiency of the decatenation checkpoint
et al., 1998). (Damelin et al., 2005). Thus, emerging data suggests
A parallel signaling pathway involving ATM, Chk2, that the switch from aneuploidy tolerance/survival to
Cdc25A and Cdk2 is also shown to be corrupted in aneuploidy intolerance/elimination is discerned upon
ES cells. This is due to the restricted localization differentiation (Mantel et al., 2007) and that check-
of Chk2 to the centrosomes, rendering it incapable point inefficiency could be a feature of the undiffer-
of phosphorylating its substrates, primarily Cdc25A, entiated state.
6 Aneuploidy in Embryonic Stem Cells 79

6.6 Does Aneuploidy in Stem and/or aneuploid (Ambartsumyan and Clark, 2008). Mosaic
Progenitor Cells Have Consequences Variegated Aneuploidy (MVA) is a detrimental exam-
ple of mosaicism whereby chromosome missegrega-
for Development and Disease?
tion occurs throughout development. MVA (Kajii et al.,
2001; Jacquemont et al., 2002) and several cases of
Aneuploidy in ES cells and NPC might reflect the related syndromes (Hunter, 2003) have been reported
necessity of obtaining high quantities of cells dur- in at least 15 individuals and are recently linked to a
ing early stages of development; another non-mutually genetic deficit in the SAC protein BubR1 (Kajii et al.,
exclusive possibility is that aneuploidy plays an impor- 2001; Ikeuchi et al., 2004).
tant role in normal development. Towards the latter, it Similar phenotypes have also been observed for
has been proposed that genetically dissimilar cells are hereditary deficits that compromise DNA damage sig-
essential during normal development to influence mor- naling (Rolig and McKinnon, 2000; Baker et al., 2007)
phogen diffusion and gradient formation (Mantel et al., and may point out to a common cellular mechanism
2007; Gurdon and Bourillot, 2001). Only after the that controls the generation and survival of aneuploid
acquisition of a sufficient number of cells the embryo cells during both tumorigenesis and neurogenesis.
can eliminate aberrant cells without interfering with Although aneuploidy at fertilization can account for
the overall patterning (Mantel et al., 2007). ~35% of spontaneous abortions, this is not always nec-
This idea is supported by studies carried out in essarily lethal (Hassold et al., 1980; Griffin, 1996).
developing embryos generated by in vitro fertilization Human development is attainable with the gain or loss
(IVF) and assessed by fluorescence in situ hybridiza- of certain chromosomes (e.g. Chromosomes 21, X, Y).
tion (FISH) technique. It revealed a high frequency Given that there is a certain level of aneuploidy in the
of chromosomal mosaicism in normal human preim- healthy human brain, it is worth noting that individuals
plantation embryos. However, when comparing early with trisomy 21 (Downs syndrome) and sex chromo-
developmental stages with embryos in the blastocyst some aneuploidies (Klinefelters Syndrome, Turners
stage, it was found that embryos undergo a natural Syndrome) frequently present neurological symptoms.
selection of euploid cells, resulting in a decreased num- In addition to mental retardation, older Downs syn-
ber of aneuploid cells throughout the blastocyst stage drome patients invariably develop Alzheimers disease
(Frumkin et al., 2008; Gonzalez-Merino et al., 2003). (AD) (Potter, 1991). The amyloid precursor protein
Cell biologic mechanisms managed to rescue ane- gene, linked to the early onset of AD, is located on
uploidy at conception have been observed (Kalousek chromosome 21. Thus, an increased gene dosage due
and Vekemans, 1996). Trisomic zygotic rescue can to trisomy 21 might predispose Downs syndrome
occur when chromosome missegregation generates patients to AD (Li et al., 1997; Geller and Potter,
aneuploid ES cells. Here, an euploid cell can arise from 1999).
a trisomic cell through the loss of the additional chro- When chromosome missegregation occurs later in
mosome (i.e. trisomic rescue). However, depending development, both cell types can contribute to the
on the lost chromosome, the embryo can remain with embryo and mosaic individuals are born. In Downs
chromosomes of both parental origin (i.e. biparental syndrome individuals, when development proceeds
disomy) or of only one parental origin (i.e. uniparental from an aneuploid zygote, a beneficial consequence
disomy). In these instances, aneuploid ES cells are of mosaicism might be an increased prevalence of the
confined, in an unspecified way, to the placenta while euploid cell line, which could lessen the severity of the
the euploid cell line gives rise to the embryo proper disease.
(Kalousek and Dill, 1983; Kalousek, 2000). Thus, In the case of NPCs, loss of heterozygosity due
early development has certain mechanisms for distin- to aneuploidy can alter gene expression differently
guishing between aneuploid and euploid cells. in individual neurons (Kaushal et al., 2003). Such
Chromosomal abnormalities affecting either one mosaicism could modulate the structure and/or func-
or a few blastomeres during cleavage manifest as a tion of neural networks.
mosaic pattern in the developing embryo, in which a Despite its characteristic failure in checkpoint
fraction of cells is euploid and the remaining ones are functions, a p53-dependent suppression of nanog
80 R.C. Sartore et al.

expression has been suggested as a mechanism to In most of cases, cancer develops from CSC and
avoid genomic instability in ES cells (Lin et al., 2005). contains alterations such as chromosome transloca-
Nanog is a protein exclusively expressed in ES cells tions and aneuploidy. Boveris observation of an unde-
and is essential to maintain self-renewal and pluripo- fined cell mass at the center of aneuploid blasto-
tency (Mitsui et al., 2003). It has been demonstrated cysts, known today as a tumor, provided the foun-
that after DNA damage, such as DNA double-strands dation, which has allowed researchers to later form
break, p53 binds the nanog promoter and represses its a correlation between aneuploidy and cancer. He
expression to induce differentiation (Lin et al., 2005). also noted that these aneuploid cells had a growth
Consequently, efficient p53-dependent cell cycle arrest advantage comparable to wild type cells, and that
and apoptosis can be restored, given that this is a this advantage could contribute to tumor progres-
common phenomenon reached upon differentiation sion (Boveri, 1902). However, one question remains
(Mantel et al., 2007; Damelin et al., 2005). Thus, the unclear: are tumor formations always accompanied by
commitment of ES cells to differentiation can be a use- aneuploidy?
ful strategy adopted by ES cells to circumvent inherited In a recent study, Miuras group showed that the
genomic errors by subsequent generations. continuous passage in vitro of bone marrow mes-
Another possible strategy adopted by ES cells to enchymal stem cells spontaneously transformed them
escape genotoxic stress is presented in both mouse into malignant cells. This transformation was usually
and human ES cells which, in contrast to differenti- accompanied by chromosomal abnormalities, includ-
ated cells, are comprised of a higher repair capacity ing chromosome gains (Miura et al., 2006).
(Saretzki et al., 2004; Maynard et al., 2008). When On the other hand, many other groups provide evi-
compared to differentiated cells, enzymes that correct dence that aneuploidy suppresses the formation of spe-
DNA damage are found at elevated levels after damage cific kinds of tumors. Individuals with trisomy 21 have
has been induced in ES cells, thus demonstrating that lower risks of developing solid tumors, but are more
these cells respond more dramatically to DNA stress prone to developing leukemias (Hasle et al., 2000;
(Maynard et al., 2008). These data exemplify another Satge et al., 2003). Weaver also showed that a reduc-
attempt of ES cells to attenuate genomic alterations tion in CENP-E leads to spontaneous tumorigenesis
resulting from an inefficient checkpoint surveillance during aging and enhances aneuploidy and transfor-
system. mation in culture. Contrary to this, aneuploidization
caused by the reduction of CENP-E in cells depleted
of the tumor suppressing protein p19/ARF had dimin-
ished tumor formation. Hence aneuploidy has been
shown to both promote and/or to block tumor forma-
6.7 Aneuploidy and Cancer Stem Cells tion (Weaver et al., 2007).

As described in the beginning of this chapter, aneu-


ploidy is usually associated with tumorigenesis. The
idea that cancer arises from a small population of cells
6.8 Telomeres and Telomerase Under
that possess stem cell properties was first proposed 150
years ago, but it is only recently that this hypothesis is Genomic Stability Control
able to be tested. In general, there are three possible
events that can lead to cancer: (1) Cancer arises from Telomeres are specialized structures at the end of
the proliferation of normal cells that have acquired eukaryotic chromosomes and protect the ends against
DNA damage; (2) Cancer originates from mutant tis- chromosomal fusion, recombination and DNA degra-
sue stem cells otherwise known as cancer stem cells dation. In most vertebrates, they are composed of
(CSC) that have the same properties of stem cells and non-coding TTAGGG repetitive sequences and shorten
can escape from the aging process (Rajaraman et al., during each cell division due to the absence of telom-
2006); and (3) through asymmetric division, can gen- erase and the end replication problem. Telomerase is
erate one CSC and one differentiated tissue-specific the enzyme responsible for the maintenance of telom-
cancer cell (Wicha et al., 2006). eres, but are not present in most somatic cells. This fact
6 Aneuploidy in Embryonic Stem Cells 81

suggests that its activity is related to undifferentiated fusions, complete absence of telomeric repeats, as well
and high proliferating cells such as embryonic stem as aneuploidy could be observed (Blasco et al., 1997).
cells and cancer cells (Aubert and Lansdorp, 2008). Telomeres are involved in almost all types of chro-
The concept that cancer cells are immortal comes mosomal alterations. It is most likely for this rea-
from the primary observations made by Gey`s group. son that Pathak asked the question Centromere or
By culturing normal epithelial cells and cells taken telomere: who is the boss?, and many groups now
from an invasive carcinoma, they observed that cancer are trying to answer it. For many years, centromeres
cells could be cultured with continuous dividing poten- have been considered as essential actors of the cell-
tial, and then established the oldest carcinoma cell line, division machinery. Today, most groups are consider-
the HeLa cells (Gey et al., 1952). About ten years later, ing the hypothesis that telomere disruption followed
with improved media for culture and enhanced sterile by endomitosis and centrosome amplification, is in
conditions, Hayflick published his study on embry- fact the first event in aneuploidization (Pathak, 1995;
onic lung fibroblasts, concluding that somatic cells Pathak et al., 2002).
had a limited number of divisions later known as the
Hayflick limit (Hayflick and Moorhead, 1961). Human
somatic cells usually stop dividing before they reach
this limit, entering into an irreversible growth arrest. 6.9 Aneuploidy and Cell-Based Therapy
Upon reaching this limit, telomeres may initiate a DNA
damage response through the activation of the p53- and
As recombinant DNA revolutionized drug develop-
Rb-dependent checkpoints, leading to senescence and
ment in the 1990s, cell-based therapeutic technologies
the inhibition of tumorigenesis. However, when the
are now poised to drive biotechnology through the next
p53 and Rb checkpoint arrest pathways are corrupted,
decade. Perhaps the biggest challenge in advancing
the cells continue to proliferate, resulting in genetic
this field would involve the ability to culture embry-
instability and the survival of cells with chromosomal
onic stem cells without changing its basic characteris-
rearrangements and/or aneuploidy. This stage, termed
tics. Despite the fact that human and mouse ES cells
crisis, and its associated genomic instability, leads to
have been shown to be diploid with normal karyotype
genetic chaos and cell death. Telomere stabilization
(Thomson et al., 1998; Evans and Kaufman, 1981),
from exogenous telomerase or spontaneous expression
many studies are now reporting that when human
could rescue certain cells from crisis by bypassing
and/or mouse ES cells are cultured in vitro for pro-
senescence, resulting in their immortalization (Akimov
longed periods of time, they become susceptible to
et al., 2005; Bodnar et al., 1998).
genomic alterations such as aneuploidy (Maitra et al.,
Is there a link between telomeres and aneuploidy?
2005; Baker et al., 2007).
In fact, for many years, researchers have been focusing
on the way in which centromeres have been responsi-
ble for the correct running of cell division, and how one
simple failure could result in abnormal chromosome
6.9.1 Mechanical Versus Enzymatic
segregation, resulting in aneuploidy. On the other hand,
other groups described the importance of telomeres for Methods
the generation of aneuploid cells. The answer lies sim-
ply on the fact that telomeres maintain the integrity of There is an interest to optimize ES cell culturing pro-
chromosome ends. cedures because of their limitless therapeutic potential
Plentz showed, in a recent work, a link between in degenerative diseases. The use of an enzyme was
telomere shortening and aneuploidy (Plentz et al., the first method described for expanding cells in vitro
2005). While in the process of studying mice with (Grover, 1961). This technique results in single-cell
deficient telomerase production, Blasco confirmed that dissociation, but this fact may contribute to the gen-
telomeres were shorter and shorter in each consec- eration of chromosomal alterations. Trypsin is one of
utive generation. Although these telomerase-deficient the most common enzyme used to pass cells in cul-
mice were viable for about six generations, many alter- ture (Fig. 6.4). Even though trypsin is used extensively,
ations such as chromosomal abnormalities, end-to-end many groups are now finding traces suggesting that this
82 R.C. Sartore et al.

Fig. 6.4 Schematic model


of embryonic stem cell
culture. (a) Embryonic stem
cells plated on inactivated
murine embryonic fibroblasts
(mEF) and dissociated by
enzymatic method resulting in
chromosome alterations
during culture. (b)
Maintenance of pluripotency
and chromosome integrity by
mechanical dissociation

passage practice should be monitored. Chans group the differentiation potential of these abnormal cells, no
showed that when human ES cells are cultured for long alteration was observed (Liu et al., 1997; Park et al.,
periods of time with the use of trypsin, the cells begin 1998).
to acquire chromosomal alterations, most of which Catalinas group showed in a recent study that both
result in trisomy 12/17 (Chan et al., 2008; Gertow manual and enzymatic methods (Fig. 6.4) for dissociat-
et al., 2007). Although, Thomsons group showed that ing human ES maintained a stable karyotype for up to
human ES cells can be cultured using trypsin for 180 passages in feeders. Surprisingly, when these same
long periods of time without affecting their karyotype cells were submitted to a non-feeder culture condition
(Thomson et al., 2008). and cultured for up to 30 passages, only one out of
In his recent work, Rebuzzinis group monitored a the three lineages studied remained unaltered (Catalina
mouse ES cell line, called UPV04, during 40 passages et al., 2008).
while using trypsin. They observed that at passage 6, Altogether, these data suggest a straightforward
100% of cells in metaphase had the correct number technique such as cell passage, needs to be highly mon-
of chromosomes (2n=40) and the correct chromosome itored to ensure chromosomal stability and to assure
complement. At passage 34, 65% of the cells had the the future application of ES cells in therapy and drug
correct chromosome number but only 50% of them had screening.
the correct chromosome complement. Even with such
chromosomal alterations, the cells presented markers
indicative of an undifferentiated state such as OCT-
4, SSEA-1 and FOM-1 antigens as well as alkaline
6.9.2 Risks and Benefits of Aneuploidy to
phosphatase activity (Rebuzzini et al., 2008).
Sugawaras group analyzed the karyotype of mouse Cell-Based Therapies
ES cells lines obtained in Japan, and showed that
abnormal karyotype occurred in 35 out of the 88 mouse As aneuploidy has been shown to be a common event
ES cells lines analyzed. They also observed that tri- in preimplantation embryos, as well as an ordinary
somy 11 was related to the acceleration of growth, and condition in ES cell culture, yet at the same time, it
hence cell growth advantage (Sugawara et al., 2006). has been negatively linked to cancer. Thus, a lingering
This fact correlates to a previous study that relates question remains to be clarified: could aneuploid cells
higher growth rates with trissomy 8, and in relation to be compatible for cell-based therapy?
6 Aneuploidy in Embryonic Stem Cells 83

Monitoring ES cell karyotypes has been deemed in benign and premalignant keratinocytic lesions. J Cutan
essential in making their application feasible for ther- Pathol 24:305308.
Blasco MA, Lee HW, Hande MP, Samper E, Lansdorp PM,
apy. Cells with an abnormal number of chromosomes,
DePinho RA, Greider CW (1997) Telomere shortening and
usually gain, could present a threat by causing uncon- tumor formation by mouse cells lacking telomerase RNA.
trolled proliferation and tumor formation (Zuber et al., Cell 91:2534.
2002; Liu et al., 1997). Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin
GB, Harley CB, Shay JW, Lichtsteiner S, Wright WE (1998)
However, contrary to the presumed concept that
Extension of life-span by introduction of telomerase into
aneuploidy is a sign of cellular transformation, high normal human cells. Science 279:349352.
levels of aneuploidy have been suggested to increase Boveri T (1902) ber mehrpolige Mitosen als Mittel zur
cell death and suppress tumor development (Weaver Analyse des Zellkerns. Verhandlungen der physikalisch-
medizinischen Gesellschaft zu Wrzburg 35:3790.
et al., 2003, 2008). Moreover, defined aneuploidy may Catalina P, Montes R, Ligero G, Sanchez L, Cueva TD, Bueno C,
be used as the determinant in driving the differentia- Leone PE, Menendez P (2008) Human ESCs predisposition
tion process, e.g. trisomy 12 prompts human ES cells to karyotypic instability: Is a matter of cell culture adapta-
towards the renal phenotype (Gertow et al., 2007). In tion or differential vulnerability among hESC lines due to
inherent properties? Mol Cancer 7:7676.
this context, the persistence of aneuploid cells within Chan EM, Yates F, Boyer LF, Schlaeger TM, Daley GQ (2008)
the neural circuitry of normal brains suggests that some Enhanced plating efficiency of trypsin-adapted human
types of aneuploid cells might further be harmless. embryonic stem cells is reversible and independent of tri-
Further research is necessary to determine when and somy 12/17. Cloning Stem Cells 10:107118.
Damelin M, Bestor TH (2007) The decatenation checkpoint. Br
how aneuploidy can be considered a real barrier to J Cancer 96:201205.
ES cell-based therapies and whether particular cases of Damelin M, Sun YE, Sodja VB, Bestor TH (2005) Decatenation
defined aneuploidies could even be used as a strategy checkpoint deficiency in stem and progenitor cells. Cancer
for obtaining a cell type of interest. Cell 8:479484.
Duesberg P, Li R, Fabarius A, Hehlmann R (2005) The chromo-
somal basis of cancer. Cell Oncol 27:293318.
Acknowledgements We thank Stacie Ngo Abdalla for Dutta A, Chandra R, Leiter LM, Lester S (1995) Cyclins
manuscript editing. This work was supported by grants from as markers of tumor proliferation: immunocytochemical
Faperj Fundao Carlos Chagas Filho de Amparo Pesquisa studies in breast cancer. Proc Natl Acad Sci USA 92:
do Estado do Rio de Janeiro (S.R.), CNPq Conselho Nacional 53865390.
para o Desenvolvimento Cientfico e Tecnolgico (R.S., P.B., Evans MJ, Kaufman MH (1981) Establishment in culture
S.R.), Pew Latin American Program in Biomedical Sciences of pluripotential cells from mouse embryos. Nature 292:
(S.R.) and Brazilian Ministry of Health/DECIT (S.R.). 154156.
Faast R, White J, Cartwright P, Crocker L, Sarcevic B, Dalton S
(2004) Cdk6-cyclin D3 activity in murine ES cells is resistant
to inhibition by p16(INK4a). Oncogene 23:491502.
Fluckiger AC, Marcy G, Marchand M, Negre D, Cosset FL,
References Mitalipov S, Wolf D, Savatier P, Dehay C (2006) Cell
cycle features of primate embryonic stem cells. Stem Cells
24:547556.
Akimov SS, Ramezani A, Hawley TS, Hawley RG (2005) Frumkin T, Malcov M, Yaron Y, Ben-Yosef D (2008) Elucidating
Bypass of senescence, immortalization, and transforma- the origin of chromosomal aberrations in IVF embryos
tion of human hematopoietic progenitor cells. Stem Cells by preimplantation genetic analysis. Mol Cell Endocrinol
23:14231433. 282:112119.
Aladjem MI, Spike BT, Rodewald LW, Hope TJ, Klemm M, Geller LN, Potter H (1999) Chromosome missegregation and tri-
Jaenisch R, Wahl GM (1998) ES cells do not activate p53- somy 21 mosaicism in Alzheimers disease. Neurobiol Dis
dependent stress responses and undergo p53-independent 6:167179.
apoptosis in response to DNA damage. Curr Biol 8:145155. Gertow K, Cedervall J, Unger C, Szoke K, Blennow E, Imreh
Ambartsumyan G, Clark AT (2008) Aneuploidy and early MP, Ahrlund-Richter L (2007) Trisomy 12 in HESC leads
human embryo development. Hum Mol Genet 17:R10R15. to no selective in vivo growth advantage in teratomas, but
Aubert G, Lansdorp PM (2008) Telomeres and aging. Physiol induces an increased abundance of renal development. J Cell
Rev 88:557579. Biochem 100:15181525.
Baker DE, Harrison NJ, Maltby E, Smith K, Moore HD, Shaw Gey GO, Coffman W, Kubicek MT (1952) Tissue culture studies
PJ, Heath PR, Holden H, Andrews PW (2007) Adaptation to of the growth properties of cervical carcinoma and normal
culture of human embryonic stem cells and oncogenesis in epithelium. Cancer Res 12:264265.
vivo. Nat Biotechnol 25:207215. Gonzalez-Merino E, Emiliani S, Vassart G, Van den Bergh M,
Bito T, Ueda M, Ito A, Ichihashi M (1997) Less expression Vannin AS, Abramowicz M, Delneste D, Englert Y (2003)
of cyclin E in cutaneous squamous cell carcinomas than Incidence of chromosomal mosaicism in human embryos at
84 R.C. Sartore et al.

different developmental stages analyzed by fluorescence in Kingsbury MA, Friedman B, McConnell MJ, Rehen SK, Yang
situ hybridization. Genet Test 7:8595. AH, Kaushal D, Chun J (2005) Aneuploid neurons are func-
Griffin DK (1996) The incidence, origin, and etiology of aneu- tionally active and integrated into brain circuitry. Proc Natl
ploidy. Int Rev Cytol 167:263296. Acad Sci USA 102:61436147.
Grover JW (1961) The enzymatic dissociation and reproducible Kranenburg O, de Groot RP, Van der Eb AJ, Zantema A (1995)
reaggregation in vitro of 11-day embryonic chick lung. Dev Differentiation of P19 EC cells leads to differential modula-
Biol 3:555568. tion of cyclin-dependent kinase activities and to changes in
Gurdon JB, Bourillot PY (2001) Morphogen gradient interpreta- the cell cycle profile. Oncogene 10:8795.
tion. Nature 413:797803. Lew DJ, Burke DJ (2003) The spindle assembly and spindle
Hasle H, Clemmensen IH, Mikkelsen M (2000) Risks of position checkpoints. Annu Rev Genet 37:251282.
leukaemia and solid tumours in individuals with Downs Li J, Xu M, Zhou H, Ma J, Potter H (1997) Alzheimer prese-
syndrome. Lancet 355:165169. nilins in the nuclear membrane, interphase kinetochores, and
Hassold T, Chen N, Funkhouser J, Jooss T, Manuel B, Matsuura centrosomes suggest a role in chromosome segregation. Cell
J, Matsuyama A, Wilson C, Yamane JA, Jacobs PA (1980) A 90:917927.
cytogenetic study of 1000 spontaneous abortions. Ann Hum Lin T, Chao C, Saito S, Mazur SJ, Murphy ME, Appella E, Xu
Genet 44:151178. Y (2005) p53 induces differentiation of mouse embryonic
Hayflick L, Moorhead PS (1961) The serial cultivation of human stem cells by suppressing Nanog expression. Nat Cell Biol 7:
diploid cell strains. Exp Cell Res 25:585621. 165171.
Hernando E, Nahl Z, Juan G, Diaz-Rodriguez E, Alaminos Linke SP, Clarkin KC, Di Leonardo A, Tsou A, Wahl GM (1996)
M, Hemann M, Michel L, Mittal V, Gerald W, Benezra R, A reversible, p53-dependent G0/G1 cell cycle arrest induced
Lowe SW, Cordon-Cardo C (2004) Rb inactivation promotes by ribonucleotide depletion in the absence of detectable
genomic instability by uncoupling cell cycle progression DNA damage. Genes Dev 10:934947.
from mitotic control. Nature 430:797802. Liu X, Wu H, Loring J, Hormuzdi S, Disteche CM, Bornstein
Hong Y, Stambrook PJ (2004) Restoration of an absent G1 P, Jaenisch R (1997) Trisomy eight in ES cells is a common
arrest and protection from apoptosis in embryonic stem potential problem in gene targeting and interferes with germ
cells after ionizing radiation. Proc Natl Acad Sci USA 101: line transmission. Dev Dyn 209:8591.
1444314448. Mac Auley A, Werb Z, Mirkes PE (1993) Characterization
Hubalek MM, Widschwendter A, Erdel M, Gschwendtner A, of the unusually rapid cell cycles during rat gastrulation.
Fiegl HM, Mller HM, Goebel G, Mueller-Holzner E, Marth Development 117:873883.
C, Spruck CH, Reed SI, Widschwendter M (2004) Cyclin Maitra A, Arking DE, Shivapurkar N, Ikeda M, Stastny V,
E dysregulation and chromosomal instability in endometrial Kassauei K, Sui G, Cutler DJ, Liu Y, Brimble SN, Noaksson
cancer. Oncogene 23:41874192. K, Hyllner J, Schulz TC, Zeng X, Freed WJ, Crook J,
Hunter A (2003) High risk of malignancy in mosaic variegated Abraham S, Colman A, Sartipy P, Matsui S, Carpenter M,
aneuploidy syndrome. Am J Med Genet 117A:199199. Gazdar AF, Rao M, Chakravarti A (2005) Genomic alter-
Ikeuchi T, Yang ZQ, Wakamatsu K, Kajii T (2004) Induction ations in cultured human embryonic stem cells. Nat Genet
of premature chromatid separation (PCS) in individuals 37:10991103.
with PCS trait and in normal controls. Am J Med Genet Manchester KL (1995) Theodor Boveri and the origin of malig-
127A:128132. nant tumours. Trends Cell Biol 5:384387.
Jacquemont S, Boceno M, Rival JM, Mechinaud F, David A Mantel C, Guo Y, Lee MR, Kim MK, Han MK, Shibayama
(2002) High risk of malignancy in mosaic variegated ane- H, Fukuda S, Yoder MC, Pelus LM, Kim KS, Broxmeyer
uploidy syndrome. Am J Med Genet 109:1721. HE (2007) Checkpoint-apoptosis uncoupling in human and
Kajii T, Ikeuchi T, Yang ZQ, Nakamura Y, Tsuji Y, Yokomori mouse embryonic stem cells: a source of karyotpic instabil-
K, Kawamura M, Fukuda S, Horita S, Asamoto A (2001) ity. Blood 109:45184527.
Cancer-prone syndrome of mosaic variegated aneuploidy and Maynard S, Swistowska AM, Lee JW, Liu Y, Liu ST, Da
total premature chromatid separation: report of five infants. Cruz AB, Rao M, de Souza-Pinto NC, Zeng X, Bohr
Am J Med Genet 104:5764. VA (2008) Human embryonic stem cells have enhanced
Kalousek DK (2000) Pathogenesis of chromosomal mosaicism repair of multiple forms of DNA damage. Stem Cells 26:
and its effect on early human development. Am J Med Genet 22662274.
91:3945. Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M,
Kalousek DK, Dill FJ (1983) Chromosomal mosaicism con- Takahashi K, Maruyama M, Maeda M, Yamanaka S (2003)
fined to the placenta in human conceptions. Science 221: The homeoprotein Nanog is required for maintenance of
665667. pluripotency in mouse epiblast and ES cells. Cell 113:
Kalousek DK, Vekemans M (1996) Confined placental 631642.
mosaicism. J Med Genet 33:529533. Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B,
Kaushal D, Contos JJ, Treuner K, Yang AH, Kingsbury MA, Patel V, Seo BM, Sonoyama W, Zheng JJ, Baker CC,
Rehen SK, McConnell MJ, Okabe M, Barlow C, Chun J Chen W, Ried T, Shi S (2006) Accumulated chromoso-
(2003) Alteration of gene expression by chromosome loss in mal instability in murine bone marrow mesenchymal stem
the postnatal mouse brain. J Neurosci 23:55995606. cells leads to malignant transformation. Stem Cells 24:
Keyomarsi K, Conte D Jr, Toyofuku W, Fox MP (1995) 10951103.
Deregulation of cyclin E in breast cancer. Oncogene 11: Mummery CL, van den Brink CE, de Laat SW (1987a)
941950. Commitment to differentiation induced by retinoic acid in
6 Aneuploidy in Embryonic Stem Cells 85

P19 embryonal carcinoma cells is cell cycle dependent. Dev Sancar A, Lindsey-Boltz LA, Unsal-Kacmaz K, Linn S (2004)
Biol 121:1019. Molecular mechanisms of mammalian DNA repair and
Mummery CL, van Rooijen MA, van den Brink SE, de Laat SW the DNA damage checkpoints. Annu Rev Biochem 73:
(1987b) Cell cycle analysis during retinoic acid induced dif- 3985.
ferentiation of a human embryonal carcinoma-derived cell Saretzki G, Armstrong L, Leake A, Lako M, von Zglinicki T
line. Cell Differ 20:153160. (2004) Stress defense in murine embryonic stem cells is
Musacchio A, Salmon ED (2007) The spindle-assembly check- superior to that of various differentiated murine cells. Stem
point in space and time. Nat Rev Mol Cell Biol 8: Cells 22:962971.
379393. Satge D, Sasco AJ, Lacour B (2003) Are solid tumours differ-
Neganova I, Lako M (2008) G1 to S phase cell cycle tran- ent in children with Downs syndrome? Int J Cancer 106:
sition in somatic and embryonic stem cells. J Anat 213: 297298.
3044. Satzinger H (2008) Theodor and Marcella Boveri: chromosomes
Nyberg KA, Michelson RJ, Putnam CW, Weinert TA (2002) and cytoplasm in heredity and development. Nat Rev Genet
Toward maintaining the genome: DNA damage and replica- 9:231238.
tion checkpoints. Annu Rev Genet 36:617656. Savatier P, Lapillonne H, van Grunsven LA, Rudkin BB,
Park JI, Yoshida I, Tada T, Takagi N, Takahashi Y, Kanagawa H Samarut J (1996) Withdrawal of differentiation inhibitory
(1998) Trisomy 8 does not affect differentiative potential in activity/leukemia inhibitory factor up-regulates D-type
a murine parthenogenetic embryonic stem cell line. Jpn J Vet cyclins and cyclin-dependent kinase inhibitors in mouse
Res 46:2935. embryonic stem cells. Oncogene 12:309322.
Pathak S (1995) Centromere or telomere: who is the boss? Schwartz GK, Shah MA (2005) Targeting the cell cycle: a new
Anticancer Res 15:25492550. approach to cancer therapy. J Clin Oncol 23:94089421.
Pathak S, Multani AS, Furlong CL, Sohn SH (2002) Telomere Spruck CH, Won KA, Reed SI (1999) Deregulated
dynamics, aneuploidy, stem cells, and cancer (review). Int J cyclin E induces chromosome instability. Nature 401:
Oncol 20:637641. 297300.
Plentz RR, Schlegelberger B, Flemming P, Gebel M, Kreipe Stead E, White J, Faast R, Conn S, Goldstone S, Rathjen J,
H, Manns MP, Rudolph KL, Wilkens L (2005) Telomere Dhingra U, Rathjen P, Walker D, Dalton S (2002) Pluripotent
shortening correlates with increasing aneuploidy of chro- cell division cycles are driven by ectopic Cdk2, cyclin A/E
mosome 8 in human hepatocellular carcinoma. Hepatology and E2F activities. Oncogene 21:83208333.
42:522526. Sugawara A, Goto K, Sotomaru Y, Sofuni T, Ito T (2006)
Potter H (1991) Review and hypothesis: Alzheimer dis- Current status of chromosomal abnormalities in mouse
ease and Down syndromechromosome 21 nondisjunc- embryonic stem cell lines used in Japan. Comp Med 56:
tion may underlie both disorders. Am J Hum Genet 48: 3134.
11921200. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA,
Rajaraman R, Guernsey DL, Rajaraman MM, Rajaraman SR Swiergiel JJ, Marshall VS, Jones JM (1998) Embryonic
(2006) Stem cells, senescence, neosis and self-renewal in stem cell lines derived from human blastocysts. Science
cancer. Cancer Cell Int 6:2525. 282:11451147.
Rane SG, Reddy EP (2000) Cell cycle control of pancreatic beta Thomson A, Wojtacha D, Hewitt Z, Priddle H, Sottile V,
cell proliferation. Front Biosci 5:D1D19. Di Domenico A, Fletcher J, Waterfall M, Corrales NL,
Rasnick D, Duesberg PH (1999) How aneuploidy affects Ansell R, McWhir J (2008) Human embryonic stem
metabolic control and causes cancer. Biochem J 340: cells passaged using enzymatic methods retain a normal
621630. karyotype and express CD30. Cloning Stem Cells 10:
Rebuzzini P, Neri T, Mazzini G, Zuccotti M, Redi CA, 89106.
Garagna S (2008) Karyotype analysis of the euploid Torres EM, Williams BR, Amon A (2008) Aneuploidy: cells
cell population of a mouse embryonic stem cell line losing their balance. Genetics 179:737746.
revealed a high incidence of chromosome abnormalities Van den Heuvel S (2005) Cell-cycle regulation. Worm Book Sep
that varied during culture. Cytogenet Genome Res 121: 21:116.
1824. Weaver BA, Bonday ZQ, Putkey FR, Kops GJ, Silk AD,
Rehen SK, McConnell MJ, Kaushal D, Kingsbury MA, Yang Cleveland DW (2003) Centromere-associated protein-E is
AH, Chun J (2001) Chromosomal variation in neurons of the essential for the mammalian mitotic checkpoint to prevent
developing and adult mammalian nervous system. Proc Natl aneuploidy due to single chromosome loss. J Cell Biol
Acad Sci USA 98:1336113366. 162:551563.
Rehen SK, Yung YC, McCreight MP, Kaushal D, Yang AH, Weaver BA, Silk AD, Cleveland DW (2008) Low rates of
Almeida BS, Kingsbury MA, Cabral KM, McConnell MJ, aneuploidy promote tumorigenesis while high rates of ane-
Anliker B, Fontanoz M, Chun J (2005) Constitutional uploidy cause cell death and tumor suppression. Cell Oncol
aneuploidy in the normal human brain. J Neurosci 25: 30:453.
21762180. Weaver BA, Silk AD, Montagna C, Verdier-Pinard P, Cleveland
Resnick JL, Bixler LS, Cheng L, Donovan PJ (1992) Long- DW (2007) Aneuploidy acts both oncogenically and as a
term proliferation of mouse primordial germ cells in culture. tumor suppressor. Cancer Cell 11:2536.
Nature 359:550551. White J, Stead E, Faast R, Conn S, Cartwright P, Dalton S
Rolig RL, McKinnon PJ (2000) Linking DNA damage and (2005) Developmental activation of the Rb-E2F pathway
neurodegeneration. Trends Neurosci 23:417424. and establishment of cell cycle-regulated cyclin-dependent
86 R.C. Sartore et al.

kinase activity during embryonic stem cell differentiation. Yang AH, Kaushal D, Rehen SK, Kriedt K, Kingsbury MA,
Mol Biol Cell 16:20182027. McConnell MJ, Chun J (2003) Chromosome segregation
Wicha MS, Liu S, Dontu G (2006) Cancer stem cells: an old idea defects contribute to aneuploidy in normal neural progenitor
a paradigm shift. Cancer Res 66:18831890; discussion cells. J Neurosci 23:1045410462.
18951886. Zuber MA, Koschny R, Koschny T, Froster UG (2002)
Williams BR, Prabhu VR, Hunter KE, Glazier CM, Whittaker Gain of chromosome 7 detected by comparative genomic
CA, Housman DE, Amon A (2008) Aneuploidy affects pro- hybridization accumulates with age in patients with
liferation and spontaneous immortalization in mammalian glioblastoma multiforme. Cancer Genet Cytogenet 136:
cells. Science 322:703709. 9294.
Chapter 7

Retrotransposition and Neuronal Diversity

Maria C. N. Marchetto, Fred H. Gage, and Alysson R. Muotri

Contents plasticity. However, the extent of the impact of L1 on


the neuronal genome is unknown. In this chapter we
7.1 Introduction . . . . . . . . . . . . . . . . . 87 will discuss the potential influence of L1 retrotrans-
7.2 Silencing and Activation of L1 Retrotransposons 89 position during brain development and the evolution-
7.3 L1 Targets in Neuronal ary pressures that may have selected this unexpected
Progenitor Cells . . . . . . . . . . . . . . . 91
machinery of diversity in neuronal precursor cells.
7.4 Environmental Regulation of L1 Activity
in the Brain . . . . . . . . . . . . . . . . . 93
7.5 L1 Activity and Disease . . . . . . . . . . . 93 Keywords Brain evolution Genetic mosaicism L1
7.6 Evolutionary Consequences of L1 Impact retrotransposon Neural stem cells Selfish gene
in Neuronal Genomes . . . . . . . . . . . . 94
References . . . . . . . . . . . . . . . . . . . . 95 Abbreviations

LINE-1 or L1 Long Interspersed Nucleotide


Abstract The generation of specialized cell types,
Elements-1
such as neurons, derived from stem cells has been
NSC neural stem cell(s)
proposed as a groundbreaking technology for regener-
EGFP enhanced green fluorescent protein
ative medicine. Unfortunately, subtype-specific differ- EN endonuclease
entiation of functional neurons is extremely difficult. NRSE neuron-restrictive silencer element
Understanding the mechanisms of neuronal diversi- RT reverse transcriptase
fication is not only relevant for the proper differ- TPRT Target Primed Reverse Transcription
entiation of complex neuronal types but might also UTR untranslated terminal repeats
shed light on normal brain development and cognitive
diversification. The recent finding that LINE-1 (Long
Interspersed Nucleotide Elements-1, or L1) retroele-
ments are active in somatic neuronal progenitor cells 7.1 Introduction
has provided a potential additional mechanism for gen-
erating neuronal diversification. L1 retrotransposition
It is known for more than a century, through the work
in the nervous system challenges the idea of static neu-
of Camilo Golgi and Santiago Ramon y Cajal, that
ronal genomes, adding a new element for neuronal
neurons are specialized cells with a huge diversity of
shapes and connections. It is estimated that the human
brain contains more than 10,000 different morpho-
A.R. Muotri () logical types of neurons. However, neuronal diversity
University of California at San Diego, School of Medicine, cannot be defined only by morphology or anatomic
Department of Pediatrics/Rady Childrens Hospital San Diego
position. Similar cells, located at the same brain region,
and Department of Cellular and Molecular Medicine, La Jolla,
CA 92093, MC 0695, USA may have distinct electrophysiological properties and
e-mail: muotri@ucsd.edu unique connection within other neurons. Moreover,

H. Ulrich (ed.), Perspectives of Stem Cells, 87


DOI 10.1007/978-90-481-3375-8_7, Springer Science+Business Media B.V. 2010
88 M.C.N. Marchetto et al.

neurons are extremely plastic cells, allowing extraor-


dinary response upon micro and macro environmental
stimulation. Any attempt to understand how the brain
works must take into account neuronal diversity. Such
diversity is likely the reason why each one of us is
unique; even genetically identical twins have different
preferences or opinions. But the fundamental mech-
anisms by which neural stem cells produce such a
variety of neuronal types are slowly being revealed.
In contrast to the single mechanism for the pro-
duction of antibodies (VDJ recombination) in the
immune system, several molecular mechanisms con-
tribute to the generation of neuronal diversity (Muotri
and Gage, 2006). Those mechanisms not only act
on the DNA, but also act on the RNA and pro-
tein level, allowing epigenetic modifications to take
place. Among these mechanisms, alternative splic-
ing, promoter usage, alternate polyadenylation, RNA
editing and post-translation modifications are all part
of the genetic tool box present in neuronal precur-
sor cells. However, even with such repertoire, this
is still not enough to justify the observed constella-
tion of neuronal types. New mechanisms are likely
to be uncovered. We anticipate that novel strategies
for the neuronal diversity contribution are hidden in
non-coding regions of the genome (Cao et al., 2006;
Muotri and Gage, 2006). We have recently shown that
an engineered human L1 element could retrotranspose
in neuronal precursor cells, changing neuronal-related
gene expression, which, in turn, can influence neu-
ronal cell fate in vitro (Muotri et al., 2005). Long
dismissed as selfish or junk DNA, retroelements
are thought to be intracellular parasites from our dis- Fig. 7.1 A model for generation of neuronal diversity by
L1 retrotransposition. In neural stem cells, Sox2 expression
tant evolutionary past. Together with their mutated is correlated with a repression of L1 retrotransposons and
relatives, retroelement sequences constitute 45% of neuronal genes. During early phases of neuronal differentiation,
the mammalian genome, with L1 alone representing there is a reduction in the expression of Sox2 and other neuronal
20%. Full-length L1s are ~6 Kb long, and encode two stem cell genes. As a result, L1 transcription can be activated,
allowing subsequent retrotransposition into neuronal genes such
ORFs (ORF1 and ORF2), which code for an RNA- as for the Psd-93 gene (Muotri et al., 2005). The resulting retro-
binding protein with nucleic acid chaperone activity transposition events can alter gene expression, which, in turn,
and a 150-kDa protein with both endonuclease (EN) can influence the phenotype of the resulting cell. The functional
and reverse transcriptase (RT) activities (Fig. 7.1). L1s variability in gene expression induced by L1 retrotransposition
could also contribute, in principle, to the high cell death rate
mobilize via an RNA intermediate to integrate them- observed in adult neurogenesis, where only a few newly born
selves into genomic DNA at the target site (Fig. 7.1). neurons successfully integrate into the pre-existing neuronal
Most of the L1s in the mammalian genome are unable network
to retrotransposed due to mutations. Consequently,
only 80150 retrotransposition competent L1s capa-
ble of autonomous retrotransposition are located in the The fact that L1 can retrotranspose in a defined
human genome (Brouha et al., 2003; Sassaman et al., window of neuronal differentiation, changing the
1997). genetic information in single neurons in an arbitrary
7 Retrotransposition and Neuronal Diversity 89

fashion, could allow the brain to develop in distinctly somatic tissues. This assumption is based on sev-
different ways. These characteristics of variety and eral arguments. First, there is no detectable or low
flexibility may contribute to the uniqueness of an indi- level of retrotransposon expression in most somatic tis-
vidual brain. L1 retrotransposition causes a neuronal sues. However, only a few tissues have been subjected
genetic mosaicism, i.e., the presence of more than to meticulous analysis, including subtype cell differ-
one genetically distinct neuronal type. Such mosaicism ences. Second, the somatic silencing of L1s fits well in
might be undetectable unless closely inspected. In the selfish DNA hypothesis, where the mobile ele-
fact, genetic mosaicism is frequently overlooked or ments exist merely to propagate themselves, so there is
interpreted as normal variation caused by stochas- no reason to transpose in somatic cells. Finally, there
tic developmental factors or unequal influence of is a clear detection bias of somatic retrotransposition
the environment. However, depending on the mosaic since only visible mutants, usually leading to human
nature, frequency, environmental cues, and tissues diseases, such as cancer, are detectable (Kazazian,
of origin, even subtle alterations in gene expression 2001; Kazazian et al., 1988). The lack of experimen-
can contribute to detectable phenotypic alterations in tal data and the paucity of natural evidence for somatic
the organism. Normal processes, such as aging, the L1 retrotransposition have led to the view that L1
generation of immune diversity, and the pheno- activity is restricted to early embryonic and germ line
typic variability between monozygotic twins (such cells, suggesting that intrinsic factors may be present
as schizophrenia) can be due to somatic genetic or absent in certain cell types responsible for trans-
mosaicism (Dipple and McCabe, 2000; Machin, 1996; position (Mathias and Scott, 1993; Prak et al., 2003).
Vijg, 2000). The stochastic nature of retrotransposon Nonetheless, retrotransposon silencing could be phys-
activity, and the large number of genes that this process iologically attenuated. DNA methylation is likely the
may affect, could produce an ample spectrum of neu- most effective and global strategy against retrotrans-
ronal diversity, which may affect behavior, cognition poson mobility (Yoder et al., 1997). Accordingly, DNA
and disease risk. methyltransferase-1 (Dmnt1)-deficient mouse embryos
have much higher levels of IAP (Intracisternal A-
particle) retrotransposon transcripts than their wild-
type littermates (Walsh et al., 1998). Repression of
7.2 Silencing and Activation of L1
retrotransposition is removed under definite conditions
Retrotransposons during a specific developmental window. One example
is the specific induction of IAP elements in the stem
L1 retrotransposons can threaten the structure and reg- cells of the male germ line at undifferentiated stages
ulate the expression of the genome in different ways, when they are de-methylated. This leads to the hypoth-
such as creating new splicing forms, promoter activa- esis that, a similar mechanism may be found in somatic
tion, skipping exons or gene inactivation among others tissues.
(Gilbert et al., 2005; Kazazian, 2004). Such a variety of One useful approach to track somatic retrotrans-
strategies make L1 retrotransposons the most creative position is the analysis of the L1-enhanced green
force shaping the genomes during evolution. fluorescent protein (EGFP) transgenic animal (Muotri
Deleterious retrotransposition events in the germ et al., 2005; Prak et al., 2003). These mice were
line or in early development have resulted in a variety engineered to carry an active L1 retrotransposon
of genetic disorders, and a somatic L1 retrotransposi- with an EGFP indicator cassette that only expresses
tion in man has resulted in a sporadic case of colon EGFP after retrotransposition and de novo insertions
cancer (Kazazian, 1998; Miki et al., 1992; Ostertag (Fig. 7.2). Because the assay uses the strong and ubiq-
and Kazazian, 2001). In plants and other organisms in uitous CMV promoter, it is expected to express EGFP
which transposition is not restricted to the germ line, in a large spectrum of somatic cells, if retrotranspo-
somatic activity of transposable elements provides the sition indeed occurs. Obviously, the system will not
opportunity for a phenotypic variability that can some- detect truncated or silenced insertions of the reporter
times be stunning with regard to individual genome cassette. Of the several somatic tissues analyzed by
flexibility (Lisch, 2002). In contrast, retrotransposons immunohistochemistry, brain tissue was the only tis-
are frequently assumed to be silenced in mammalian sue where EGFP expression was detected, specifically
90 M.C.N. Marchetto et al.

in neurons (Muotri et al., 2005). The in vitro cellu- events could alter the expression of neuronal genes.
lar assay indicates that L1 retrotransposition actually Such mechanism could, presumably, generate a large
happened in precursor cells rather than postmitotic spectrum of genetically distinct neurons, adding to
neurons. Therefore, neuronal precursor cells may have the great neuronal variation that is currently observed
a greater frequency of L1 retrotransposition than other in the adult CNS. L1 activation is likely regulated
cell types and/or this finding may be due to the long by host factors in equilibrium: too much L1 retro-
life of neurons, in contrast to the continuous renewal transposition can cause cell damage and induce the
of other cell types. Either way, the presence of EGFP- cells to die (Haoudi et al., 2004); too little can limit
positive cells indicates that somatic L1 silencing is neuronal diversity. The identification of neuronal host
incomplete in the brain. This observation suggests that factors responsible for L1 repression and/or activa-
L1 retrotransposons might be activated in neuronal tion will be extremely important to understand how
precursor cells and the resultant retrotransposition retrotransposition is regulated.

Fig. 7.2 Detection of L1


retrotransposition in the
brains of transgenic mice.
The structure of the
L1RP -EGFP transgene is
indicated at the top of the
figure. The
retrotransposition-competent
human L1 (L1RP ) contains a
5 untranslated region (UTR)
that harbors an internal
promoter, two open reading
frames (ORF1 and ORF2; not
drawn to scale), and a 3 UTR
that ends in a poly (A) tail.
The EGFP retrotransposition
indicator cassette consists of a
backward copy of the EGFP
gene whose expression is
controlled by the human
cytomegalovirus major
immediate early promoter
(pCMV) and the herpes
simplex virus thymidine
kinase polyadenylation
sequence (pA). This
arrangement ensures that
EGFP expression will only
become activated upon L1
retrotransposition. The black
arrows indicate PCR primers
flanking the intron present in
the EGFP gene
7 Retrotransposition and Neuronal Diversity 91

Fig. 7.3 A model for L1 retrotransposition. After transcrip- the new insertion is frequently mutated on its 5 UTR. Other
tion, the L1 RNA forms a complex with ORF1 and ORF2 in retroelements, such as Alus, can hijack the L1 machinery and
the cytoplasm (RNP complex). The complex travels back to the retrotranspose into the genome. Once inserted, the new insertion
nucleus where the endonuclease domain of ORF2 (EN) nicks can impact the genome in different ways, such as by the creation
the target site on the genomic DNA. The de novo insertion prob- of new insertions and deletions
ably occurs by Target Primed Reverse Transcription (TPRT) and

L1 expression is dependent on the activation of its may trigger neuronal differentiation (Fig. 7.3). Such
own 5 untranslated terminal repeat (UTR) sequence, a mechanism preserves genetic stability in NSC but
which acts as a promoter. The human L1 5 UTR is 1 kb allow instability to happen in neuronal committed
long, harboring one YY1-binding site that is required cells.
for proper transcriptional initiation (Athanikar et al.,
2004; Swergold, 1990) two Sox (sex determining
region of Y-chromosome, SRY, related HMG-box)
binding sites (Tchenio et al., 2000) and a runt-domain 7.3 L1 Targets in Neuronal
transcription factor 3 (RUNX3) binding site (Yang
Progenitor Cells
et al., 2003). Interestingly, none of these factors is
germ-cell specific, suggesting the presence of other,
unknown factors. Sox proteins are expressed in a To cause a significant impact on neuronal genomes,
variety of tissues, including NSC and testis (Wegner, new L1 insertions must target important regulatory
1999). The lack of Sox2 allowed activation of neu- regions or genes that are being expressed at the
ronal genes and differentiation, suggests that Sox2 moment of neuroblast differentiation. Likely, only the
may function as a repressor of differentiation in neu- combination of multiple L1 events, and not an even-
ral stem cells (Graham et al., 2003). We demonstrated tual catastrophic insertion in single neurons will be
that a decrease in Sox2 expression during the early ultimately responsible for any change in the neu-
stages of neuronal differentiation is correlated with an ronal network. But L1 retrotransposition is a dan-
increase in both L1 transcriptional activity and retro- gerous situation for the cell, since L1 insertions
transposition (Muotri et al., 2005). We propose that can hit essential genes that may induce cell death
L1 retrotransposons are silenced in NSC due to Sox2- or even target oncogenes, leading to a neoplastic
mediated transcriptional repression. Down-regulation transformation.
of Sox2 accompanies chromatin modifications, such as Despite the low number of examples, the sequence
DNA de-methylation and histone acetylation, which data from target insertional sites in rat neuroblasts
92 M.C.N. Marchetto et al.

were often close to or inside neuron-associated genes integrated into the genomes of individual neurons dur-
(Muotri et al., 2005). Even with a small sample, ing the entire life of the organism. These insertions
two L1 insertions were located in the same gene, then act in a stochastic fashion, working as control-
indicating that the integration process might not ling elements, fine-tuning to increase the probability
be completely random. Some of these target genes that genes will be differentially transcribed. The model
included an olfactory receptor, ion channel-associated is consistent with neuroblast differentiation, in which
genes and a cadherin receptor (Muotri et al., 2005). similar cells are subjected to the same environmental
An L1 insertion in the promoter region of the Psd- stimuli but do not respond uniformly. Thus, new inser-
93 gene, encoding a post-synaptic density protein tions in neurons represent genomic scars that may
involved in different aspect of synapse formation, have the potential to influence the fate of the resultant
significantly increased gene expression level and, con- cells and, consequently, the function of the neuronal
sequently, accelerated neuronal maturation in culture. network.
Despite the fact that randomness seems to be the The study of the human L1 5 UTR promoter dur-
best way for L1 to survive during evolution when ing neuronal differentiation revealed that L1 activation
they are active in germ cells, somatic insertions might occurs in the initial stages of cell differentiation. That
be controlled by local microvariations in DNA chro- is exactly the same time that several neuronal genes,
matin structure that depend on different host factors such as NeuroD1, are upregulated and several cell
in specific subsets of cell types. Thus, we propose cycle genes are downregulated (Hsieh et al., 2004a;
that L1 insertions in the nervous system are some- Zhao and Gage, 2002). Additionally, the strong anti-
how guided to specific gene targets. In a similar way, mitotic small modulatory neuron-restrictive silencer
the yeast Ty1 transposon is highly nonrandom in element (NRSE) dsRNA, responsible for the neuronal
vivo, being preferentially inserted upstream of tRNA fate of NSC, is expressed in initial steps of differ-
genes (Bachman et al., 2004; Devine and Boeke, entiation, activating several NRSE-containing neuron-
1996). specific genes and stopping the cell cycle (Kuwabara
In the L1-EGFP transgenic mice, we followed et al., 2004). These data suggest that there is an
the retrotransposition of a single human L1 element orchestrated regulation during neuronal differentia-
and retrotransposition was detected by EGFP expres- tion, avoiding an eventual cell transformation. Such
sion. However, the indicator cassette did not reflect an idea conforms with the low incidence of neu-
a direct measurement of the 3,000 estimated endoge- roblastomas (Zhu and Parada, 2002) but does not
nous active L1 retrotransposons (DeBerardinis et al., exclude the possibility that an abnormal L1 retrotrans-
1998; Goodier et al., 2001). Moreover, as pointed out position leads to a neoplastic transformation in CNS
before, the L1 retrotransposition assay did not report cells.
EGFP-truncated or silenced insertions. Additionally, Taken together, a specific regulation of L1 retro-
it certainly did not account for the indirect, in trans, transposon activity that takes into account its non-
L1-mediated insertions of Alus, retrotransposition- random neuronal insertion and a specific window of
defective L1s, and other non-autonomous RNAs. time during cell differentiation may turn a potentially
Virtually any RNA molecule can be subject to retro- harmful phenomenon into a useful one. The problem
transposition if hijacked by L1 machinery. In this now, as with most novel scientific debates, is one of
regard, every single developing neuron can potentially quantification and significance. Future technologies for
carry L1-mediated events, and if part of the resultant single-cell endogenous L1 activity assays will bring
insertions occurs in genes expressed during neuronal new insights into the problem. Moreover, the genera-
development, altering gene expression, then it is pos- tion of three-dimensional brain mapping depicting the
sible that brain development could be significantly occurrence of L1 retrotransposition will allow the visu-
affected by L1 retrotransposition. It has been proposed alization of preferential target neuronal subtypes. The
that stochastic gene expression might be a fundamental comparison of normal brains with brains where L1
part of development and differentiation and, where it is activity is mis-regulated, will provide the structural
advantageous, these stochastic patterns are retained in organization for the design of algorithms that predict
the adult organism (Fiering et al., 2000). We speculate eventual retrotransposition-affected neuronal networks
that these new L1 retrotransposition events are stably or systems.
7 Retrotransposition and Neuronal Diversity 93

7.4 Environmental Regulation of L1 most of the EGFP-positive cells co-localized with neu-
Activity in the Brain ronal markers, but not with glial markers, in both
treatments, consistent with recent reports demonstrat-
ing that epigenetic modifications accompany neuronal
L1 somatic retrotransposition may be regulated by differentiation of NSC (Hsieh and Gage, 2004; Hsieh
environmental signals. Because adult neurogenesis can et al., 2004b).
be influenced by a wide variety of environmental and Barbara McClintock first proposed the genomic
behavioral cues, and since adult neuroblasts can sup- shocks hypothesis, saying that environmental factors
port L1 transposition, it is possible that L1 retrotrans- could induce transposition of controlling elements
position is also affected during neurogenesis. Recently, (McClintock, 1984). In fact, a recent finding demon-
we described the effects of voluntary running (a potent strated that the copy number of the plant BARE-1
neurogenic activity) in the L1-EGFP transgenic ani- retrotransposon correlates with a sharp microclimate
mals. Surprisingly, the number of EGFP-positive cells divergence, suggesting stress-induced mobilization of
increased about twofold in the runners brains (Muotri retroelements in response to environmental stimuli
et al., 2009). It is still premature to speculate how run- (Kalendar et al., 2000). The experiment with the
ning can induce L1 retrotransposition or the survival of L1-EGFP animal illustrates how neuronal genomes
L1-inserted cells. It could be an indirect observation, might modify themselves when confronted with a new
since running increases the number of cell divisions or unfamiliar environment. Some of these changes
in neurogenic areas, thus increasing the chances of L1 are irreversible (L1 insertions) but others may be
insertions in newborn cells or that another factor, such reversible (EGFP re-expression), resetting specific
as hormones, could be acting to stimulate L1 transcrip- sequences of the genome to its early state. The fact that
tion, as previously suggested (Morales et al., 2002; genetic modifications caused by L1 elements specif-
Trelogan and Martin, 1995). ically happen in neurons is certainly associated with
Interestingly, EGFP-positive cells were increased the enormous somatic plasticity observed in the ner-
not only in the adult neurogenic areas but also in vous system. The implications of these observations
non-neurogenic areas, such as the cortex or amyg- are potentially important for the definition of indi-
dala. Because preliminary data suggest that L1 retro- vidual organisms as well as the meaning of neural
transposition does not occur in postmitotic neurons plasticity. Such plasticity predicts that the brain and
(Muotri et al., 2005), this finding indicates that run- the neuronal network will never be the same after a
ning not only increased the number of new inser- new experience, even in genetically identical twins.
tions in the brain but also activated EGFP expression Moreover, individuals lacking factors involved with
in mature neurons from silenced L1 insertions. The chromatin remodeling might have abnormal genome
mechanism for this activation is unknown; however, reprogramming that can give rise to a wide range of
the L1-EGFP insertion is likely working as a real altered phenotypes.
time gene-trap system, indicating that several regions
of the neuronal genome can be re-expressed dur-
ing exercise. Such re-expression is probably taking
place by chromatin remodeling factors that expose 7.5 L1 Activity and Disease
the silenced DNA region to transcription factors. A
similar phenomenon was observed in neural clones The idea that retroviral elements are relevant for cer-
that harbor L1-EGFP insertions (Muotri et al., 2005). tain brain disorders has its origins in the search for
After single-cell cloning in the presence of FGF-2, a viral pathogen for schizophrenia in the early 1970s
the EGFP expression are silenced, generating EGFP- (Torrey and Peterson, 1976). The research brought
variegated clones in which EGFP expression will be evidences that endogenous retrovirus transcripts were
completely attenuated after a prolonged period in cul- expressed in the brains and cerebrospinal fluids of
ture. Curiously, EGFP expression could be restored affected individuals (Frank et al., 2005; Karlsson
only during neuronal differentiation, suggesting that et al., 2001; Kim et al., 1999; Yee and Yolken,
the L1-inserted loci are activated in the neuronal but 1997). Interestingly, RNA expression in libraries gen-
not in the glial lineage. Immunostaining revealed that erated from the frontal cortex regions of individuals
94 M.C.N. Marchetto et al.

with schizophrenia revealed an increased level of L1 that gave rise to the mitochondria, generating the
sequences, in particular from ORF2 sequences (Yolken modern-day spliceosomal introns (Zimmerly et al.,
et al., 2000). Furthermore, retrotransposition activity, 1995). Further acquisition of an endonuclease enzyme
measured by the presence of processed pseudogenes and a promoter sequence certainly represented impor-
without introns, is higher in genomic DNA from tant steps in the evolution of L1 retrotransposons, pro-
individuals with schizophrenia. Accordingly, individ- viding autonomy for L1s to insert into many locations
uals with schizophrenia and other psychiatric disor- throughout the genome.
ders have increased levels of RT activity as com- The apparent lack of obvious function of retroele-
pared to unaffected controls (Yolken et al., 2000). ments in the genome suggests that transposable ele-
Taken together, this evidence identifies retroelements ments are selfish DNA, acting as parasites in the
as important somatic components of neuropsychiatric genome in order to propagate themselves. This idea
diseases that are consistent with genetic, environmen- has long puzzled scientists and inspired the con-
tal, and neurodevelopmental aspects of the disease cept of junk DNA to illustrate the idea that such
process observed in schizophrenia, autism and oth- sequences were mere evolutionary remnants (Doolittle
ers syndromes where brain waves may fall out of and Sapienza, 1980; Orgel and Crick, 1980). However,
synchrony during the process of perception. the recognition that retrotransposons can actively
Successful identification of susceptible target genes reshape the genome is slowly challenging this termi-
seems unlikely to lead to gene therapy for psychi- nology. Moreover, the mammalian genome has suf-
atric illnesses. However, it may be important to deter- fered waves of transposon bombardment, but the con-
mine the potential biochemical pathways involved in stant, single lineage of L1 history reveals that active
producing particular symptoms, which could be down- L1 were never absent from mammals genomes dur-
stream of a group of genes with individually weak ing evolution, suggesting an inextricable link between
effects. The characterization of the role of mobile ele- L1 and their host (Furano et al., 2004). The relation-
ments in the etiopathogenesis of psychotic diseases ship between transposons and their hosts is probably
might lead to new methods for diagnosis, treatment, not entirely antagonistic, as several host genes have a
and prevention. high degree of homology to one or more transposable
elements. Evidence in the literature points to a somatic
function for L1 transcripts, involving cell prolifera-
tion (Kuo et al., 1998), differentiation (Mangiacasale
7.6 Evolutionary Consequences of L1
et al., 2003) and early embryo development (Pittoggi
Impact in Neuronal Genomes et al., 2003). Moreover, it is difficult to reconcile why
the genome would need so many copies of retrotrans-
One of the most remarkable findings from the sequenc- posons and whether this expansion has any correlation
ing of the human genome is that retrotransposable ele- with retrotransposition itself. The restricted activity
ments make up a significant portion of the human DNA of retrotransposons in germ or early embryonic cells
(Deininger et al., 2003). Based on reverse transcriptase apparently fits well with the selfish DNA concept,
(RT) phylogeny, L1 elements are most closely related since new insertions will be passed to the next gen-
to the group II introns of mitochondria and eubacte- erations, but somatic insertions pose a conundrum.
ria (Cavalier-Smith, 1991; Xiong and Eickbush, 1990). According to the symbiotic theory, it is advantageous
These studies revealed that the RT enzyme is extremely to any transposable element to promote host mating,
old and that retroelements can be viewed as relics or securing the propagation of the master elements to
molecular fossils of the first primitive replication sys- the next generations. From this perspective, it is not
tems in the progenote. The origin of retroelements surprising that advantageous insertional events in the
possibly traces back to the conversion of RNA-based brain, resulting in a better (cultural and social) fitness
systems, the RNA World (Orgel, 2004), to modern of the individual organism, also can contribute to the
DNA-based systems. Current models suggest that host mating.
these mobile introns of eubacteria were transmitted to The evolution of the CNS provided a notable selec-
eukaryotes during the initial fusion of the eubacterial tive advantage, as information about the environment
and archaebacterial genomes or during the symbiosis could be processed rapidly and would allow organisms
7 Retrotransposition and Neuronal Diversity 95

to more readily meet the challenges of ever-changing References


environmental conditions. Moreover, epigenetic mod-
ification allowed the non-genetic transfer of informa- Athanikar JN, Badge RM, Moran JV (2004) A YY1-binding
tion or transmission of culture at an unprecedented site is required for accurate human LINE-1 transcription
magnitude. Such specialization is highly dependent initiation. Nucleic Acids Res 32:38463855.
on the cognitive levels acquired by the species that Bachman N, Eby Y, Boeke JD (2004) Local definition of Ty1 tar-
get preference by long terminal repeats and clustered tRNA
are directly linked to the complexity of the neuronal genes. Genome Res 14:12321247.
network. Therefore, the advantages gained by retain- Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH,
ing the mechanisms for somatic retrotransposition may Moran JV, Kazazian HH Jr. (2003) Hot L1s account for the
outweigh the cost of a less plastic nervous system. In bulk of retrotransposition in the human population. Proc Natl
Acad Sci USA 100:52805285.
fact, such strategy expands the number of function- Cao X, Yeo G, Muotri AR, Kuwabara T, Gage FH (2006)
ally distinct neurons that could be produced from a Noncoding RNAs in the mammalian central nervous system.
given stem cell gene pool (Muotri and Gage, 2006). Annu Rev Neurosci 29:77103.
This characteristic of variety and flexibility may con- Cavalier-Smith T (1991) Intron phylogeny: a new hypothesis.
Trends Genet 7:145148.
tribute to the uniqueness of an individual brain, even DeBerardinis RJ, Goodier JL, Ostertag EM, Kazazian HH Jr.
between genetically identical twins. Mobile elements (1998) Rapid amplification of a retrotransposon subfamily is
in the brain may be part of the conserved core pro- evolving the mouse genome. Nat Genet 20:288290.
cess responsible for evoking facilitated complex non- Deininger PL, Moran JV, Batzer MA, Kazazian HH Jr. (2003)
Mobile elements and mammalian genome evolution. Curr
random phenotypical variation on which selection may Opin Genet Dev 13:651658.
act. It is remarkable to imagine that the brain is a Devine SE, Boeke JD (1996) Integration of the yeast retro-
consequence of ancient retrotransposition in eukary- transposon Ty1 is targeted to regions upstream of genes
otic cells. Such possibility had not, at any time, been transcribed by RNA polymerase III. Genes Dev 10:
620633.
considered before, but it was suggested to us by the Dipple KM, McCabe ER (2000) Phenotypes of patients with
experimental results. simple Mendelian disorders are complex traits: thresh-
The identification of L1 elements as potential cre- olds, modifiers, and systems dynamics. Am J Hum Genet
ative somatic shapers of transcriptional complexity in 66:17291735.
Doolittle WF, Sapienza C (1980) Selfish genes, the phenotype
neuronal genomes may be an important phenomenon paradigm and genome evolution. Nature 284:601603.
for developmental neurosciences. The hypothesis that Fiering S, Whitelaw E, Martin DI (2000) To be or not to be
L1 activity is responsible for fine-tuning neuronal active: the stochastic nature of enhancer action. Bioessays
wiring waves requires the merger of different fields 22:381387.
Frank O, Giehl M, Zheng C, Hehlmann R, Leib-Mosch C,
and may consequently open new ways of consider- Seifarth W (2005) Human endogenous retrovirus expression
ing individual differences and the neuronal correlates profiles in samples from brains of patients with schizophrenia
of human cognition. Rigorous experimental proof of and bipolar disorders. J Virol 79:1089010901.
this model will require attenuation of retrotransposi- Furano AV, Duvernell DD, Boissinot S (2004) L1 (LINE-1)
retrotransposon diversity differs dramatically between mam-
tion activity from the mammalian genome and com- mals and fish. Trends Genet 20:914.
paring their behavior to that of wild type animals. Gilbert N, Lutz S, Morrish TA, Moran JV (2005) Multiple fates
Nonetheless, the experimental approach presents a of L1 retrotransposition intermediates in cultured human
major methodological challenge for molecular biolo- cells. Mol Cell Biol 25:77807795.
Goodier JL, Ostertag EM, Du K, Kazazian HH Jr. (2001) A
gists, since a canonical single-gene knockout strategy novel active L1 retrotransposon subfamily in the mouse.
is no longer suitable. On the other hand, the study Genome Res 11:16771685.
of abnormal activation of L1 retrotransposition in the Graham V, Khudyakov J, Ellis P, Pevny L (2003) Sox2 func-
brain may elucidate complex neurological syndromes, tions to maintain neural progenitor identity. Neuron 39:
749765.
permitting an understanding of diseases at a different Haoudi A, Semmes OJ, Mason JM, Cannon RE (2004)
level. Retrotransposition-competent human LINE-1 induces apop-
tosis in cancer cells with intact p53. J Biomed Biotechnol
Acknowledgments M.C.N.M. and F.H.G. are supported by the 2004:185194.
Lookout Fund and the National Institutes of Health: National Hsieh J, Gage FH (2004) Epigenetic control of neural stem cell
Institute on Aging and National Institute of Neurological fate. Curr Opin Genet Dev 14:461469.
Disease and Stroke. The authors would like to thank M.L. Gage Hsieh J, Nakashima K, Kuwabara T, Mejia E, Gage FH
for editorial comments. (2004a) Histone deacetylase inhibition-mediated neuronal
96 M.C.N. Marchetto et al.

differentiation of multipotent adult neural progenitor cells. Muotri AR, Zhao C, Marchetto MC, Gage FH (2009)
Proc Natl Acad Sci USA 101:1665916664. Environmental influence on L1 retrotransposons in the adult
Hsieh J, Nakashima K, Kuwabara T, Mejia E, Gage FH (2004b) hippocampus. Hippocampus 19:10021007.
Histone deacetylase inhibition-mediated neuronal differenti- Orgel LE (2004) Prebiotic chemistry and the origin of the RNA
ation of multipotent adult neural progenitor cells. Proc Natl world. Crit Rev Biochem Mol Biol 39:99123.
Acad Sci USA 101:1665916664. Orgel LE, Crick FH (1980) Selfish DNA: the ultimate parasite.
Kalendar R, Tanskanen J, Immonen S, Nevo E, Schulman AH Nature 284:604607.
(2000) Genome evolution of wild barley (Hordeum spon- Ostertag EM, Kazazian HH Jr. (2001) Biology of mammalian L1
taneum) by BARE-1 retrotransposon dynamics in response retrotransposons. Annu Rev Genet 35:501538.
to sharp microclimatic divergence. Proc Natl Acad Sci USA Pittoggi C, Sciamanna I, Mattei E, Beraldi R, Lobascio AM,
97:66036607. Mai A, Quaglia MG, Lorenzini R, Spadafora C (2003) Role
Karlsson H, Bachmann S, Schroder J, McArthur J, Torrey EF, of endogenous reverse transcriptase in murine early embryo
Yolken RH (2001) Retroviral RNA identified in the cere- development. Mol Reprod Dev 66:225236.
brospinal fluids and brains of individuals with schizophrenia. Prak ET, Dodson AW, Farkash EA, Kazazian HH Jr. (2003)
Proc Natl Acad Sci USA 98:46344639. Tracking an embryonic L1 retrotransposition event. Proc
Kazazian HH Jr. (1998) Mobile elements and disease. Curr Opin Natl Acad Sci USA 100:18321837.
Genet Dev 8:343350. Sassaman DM, Dombroski BA, Moran JV, Kimberland ML,
Kazazian HH (2001) Retrotransposon insertions in germ cells Naas TP, DeBerardinis RJ, Gabriel A, Swergold GD,
and somatic cells. Dev Biol (Basel) 106:307313. Kazazian HH Jr. (1997) Many human L1 elements are
Kazazian HH Jr. (2004) Mobile elements: drivers of genome capable of retrotransposition. Nat Genet 16:3743.
evolution. Science 303:16261632. Swergold GD (1990) Identification, characterization, and cell
Kazazian HH Jr., Wong C, Youssoufian H, Scott AF, Phillips specificity of a human LINE-1 promoter. Mol Cell Biol
DG, Antonarakis SE (1988) Haemophilia A resulting from 10:67186729.
de novo insertion of L1 sequences represents a novel mecha- Tchenio T, Casella JF, Heidmann T (2000) Members of the SRY
nism for mutation in man. Nature 332:164166. family regulate the human LINE retrotransposons. Nucleic
Kim HS, Takenaka O, Crow TJ (1999) Isolation and phylogeny Acids Res 28:411415.
of endogenous retrovirus sequences belonging to the HERV- Torrey EF, Peterson MR (1976) The viral hypothesis of
W family in primates. J Gen Virol 80:26132619. schizophrenia. Schizophr Bull 2:136146.
Kuo KW, Sheu HM, Huang YS, Leung WC (1998) Expression of Trelogan SA, Martin SL (1995) Tightly regulated, developmen-
transposon LINE-1 is relatively human-specific and function tally specific expression of the first open reading frame from
of the transcripts may be proliferation-essential. Biochem LINE-1 during mouse embryogenesis. Proc Natl Acad Sci
Biophys Res Commun 253:566570. USA 92:15201524.
Kuwabara T, Hsieh J, Nakashima K, Taira K, Gage FH (2004) Vijg J (2000) Somatic mutations and aging: a re-evaluation.
A small modulatory dsRNA specifies the fate of adult neural Mutat Res 447:117135.
stem cells. Cell 116:779793. Walsh CP, Chaillet JR, Bestor TH (1998) Transcription of IAP
Lisch D (2002) Mutator transposons. Trends Plant Sci 7: endogenous retroviruses is constrained by cytosine methyla-
498504. tion. Nat Genet 20:116117.
Machin GA (1996) Some causes of genotypic and phenotypic Wegner M (1999) From head to toes: the multiple facets of Sox
discordance in monozygotic twin pairs. Am J Med Genet proteins. Nucleic Acids Res 27:14091420.
61:216228. Xiong Y, Eickbush TH (1990) Origin and evolution of retroele-
Mangiacasale R, Pittoggi C, Sciamanna I, Careddu A, Mattei ments based upon their reverse transcriptase sequences.
E, Lorenzini R, Travaglini L, Landriscina M, Barone C, Embo J 9:33533362.
Nervi C, Lavia P, Spadafora C (2003) Exposure of normal Yang N, Zhang L, Zhang Y, Kazazian HH Jr. (2003) An
and transformed cells to nevirapine, a reverse transcriptase important role for RUNX3 in human L1 transcription and
inhibitor, reduces cell growth and promotes differentiation. retrotransposition. Nucleic Acids Res 31:49294940.
Oncogene 22:27502761. Yee F, Yolken RH (1997) Identification of differentially
Mathias SL, Scott AF (1993) Promoter binding proteins of expressed RNA transcripts in neuropsychiatric disorders.
an active human L1 retrotransposon. Biochem Biophys Res Biol Psychiatry 41:759761.
Commun 191:625632. Yoder JA, Walsh CP, Bestor TH (1997) Cytosine methylation
McClintock B (1984) The significance of responses of the and the ecology of intragenomic parasites. Trends Genet
genome to challenge. Science 226:792801. 13:335340.
Miki Y, Nishisho I, Horii A, Miyoshi Y, Utsunomiya J, Kinzler Yolken RH, Karlsson H, Yee F, Johnston-Wilson NL, Torrey
KW, Vogelstein B, Nakamura Y (1992) Disruption of the EF (2000) Endogenous retroviruses and schizophrenia. Brain
APC gene by a retrotransposal insertion of L1 sequence in Res Brain Res Rev 31:193199.
a colon cancer. Cancer Res 52:643645. Zhao X, Gage FH (2002) Expressing the central nervous system.
Morales JF, Snow ET, Murnane JP (2002) Environmental fac- Neurochem Res 27:953954.
tors affecting transcription of the human L1 retrotrans- Zhu Y, Parada LF (2002) The molecular and genetic basis of
poson. I. Steroid hormone-like agents. Mutagenesis 17: neurological tumours. Nat Rev Cancer 2:616626.
193200. Zimmerly S, Guo H, Perlman PS, Lambowitz AM (1995) Group
Muotri AR, Gage FH (2006) Generation of neuronal variability II intron mobility occurs by target DNA-primed reverse
and complexity. Nature 441:10871093. transcription. Cell 82:545554.
Chapter 8

Directing Differentiation of Embryonic Stem Cells


into Distinct Neuronal Subtypes

Noelle Ammon, Nathaniel Hartman, and Laura Grabel

Contents learned about the conditions that promote emergence


of these lineages in the embryo.
8.1 Introduction . . . . . . . . . . . . . . . . . 98
8.2 Identifying the Desired ESC-Derived Cell Type Keywords Embryonic stem cell Neural stem cell
for Transplantation . . . . . . . . . . . . . 98 Neurodegenerative disease Neuron
8.3 Generating Neural Progenitors: Back to the
Embryo . . . . . . . . . . . . . . . . . . . 100 Abbreviations
8.4 Midbrain Dopaminergic Neurons . . . . . . . 102
8.5 GABAergic Interneurons . . . . . . . . . . . 104
8.6 Spinal Cord Motor Neurons . . . . . . . . . 106 5-HT 5-hydroxytryptamine (serotonin)
8.7 Serotonergic Neurons . . . . . . . . . . . . 108
8.8 Basal Forebrain Cholinergic Neurons . . . . . 109
AA ascorbic acid
8.9 Conclusions . . . . . . . . . . . . . . . . . 110 AADC amino acid decarboxylase
References . . . . . . . . . . . . . . . . . . . . 110 ACh acetylcholine
AD Alzheimers disease
ALS Amyotrophic Lateral Sclerosis
Abstract There is great interest in testing the effi- BAC bacterial artificial chromosome
cacy of treating neurodegenerative diseases using BDNF brain derived neurotrophic factor
embryonic stem cell derivatives. A first step towards BMP bone morphogenic protein
this goal is demonstrating that embryonic stem cells cAMP cyclic adenosine monophosphate
can produce, in culture, the specific cell types lost CB calbindin
in the various diseases. We describe here currently CCK cholecystokinin
used approaches for generating neural stem cells, CGE caudal ganglionic eminences
CNS central nervous system
as well as specific neuronal subtypes, from mouse
CR calretinin
and human embryonic stem cells. Based upon their
DA dopaminergic
demonstrated role in neurodegenerative disease and
DV dorsal-ventral
the reports documenting their derivation from embry-
EGF epidermal growth factor
onic stem cells, we focus on dopaminergic neurons, EPL primitive ectoderm-like
GABAergic interneurons, spinal cord motor neurons, ESC embryonic stem cell(s)
serotonergic neurons, and basal forebrain cholinergic FACS fluorescence activated cell sorting
neurons. Protocols are all based upon what has been FGF fibroblast growth factor
GABA -aminobutyric acid
GAD glutamic acid decarboxylase
GDNF glial cell derived neurotrophic factor
Glu glutamate
L. Grabel ()
Hall-Atwater Laboratories, Biology Department, Wesleyan hESC human embryonic stem cell(s)
University, Middletown, CT, USA Hh hedgehog
e-mail: lgrabel@wesleyan.edu iPS induced pluripotent stem cell

H. Ulrich (ed.), Perspectives of Stem Cells, 97


DOI 10.1007/978-90-481-3375-8_8, Springer Science+Business Media B.V. 2010
98 N. Ammon et al.

MAP2 microtubule associated protein 2 also necessary to identify the most appropriate cell
mDA midbrain dopaminergic type for transplant and to be able to generate large
mES cells mouse embryonic stem cells numbers of relatively pure populations of these cells
MGE medial ganglionic eminences readily in vitro. Our goal in this chapter is to summa-
MN motor neuron(s) rize the existing literature documenting the derivation
MPTP 1-methyl-4-phenyl-1,2,3,6- of some neuronal subtypes: midbrain dopaminergic
tetrahycopyridine neurons, forebrain -aminobutyric acid (GABA)ergic
NGF nerve growth factor interneurons, and motor neurons. We also briefly
NPY neuropeptide Y describe the derivation of serotonergic and cholinergic
NSC neural stem cell(s) neurons. These subtypes were chosen because of their
NT-4 neurotrophin 4
potential therapeutic uses; dopaminergic midbrain
PD Parkinsons disease
neurons for PD, inhibitory GABAergic interneurons
PDD Parkinsonian disease dementia
for epilepsy, serotonergic neurons for psychiatric
pMN motor neuron progenitor(s)
disorders, cholinergic neurons for Alzheimers disease
PV parvalbumin
RA retinoic acid (AD), and motor neurons for ALS or spinal cord injury
SCI spinal cord injuries (SCI). We will focus on the derivation of neurons
Shh sonic hedgehog from ESC, though glial derivatives can also be readily
SMA Spinal Muscular Atrophy generated.
SNi substantia nigra This chapter is not meant to provide specific proto-
SSRI serotonin selective reuptake inhibitors cols for the derivation of neurons, but rather to outline
SST somatostatin the conceptual framework behind the approaches used.
TGF- transforming growth factor Citations of the original research literature should pro-
TH tyrosine hydroxylase vide the reader with the necessary references to design
TLE temporal lobe epilepsy a relevant experimental approach for production of the
VIP vasoactive intestinal peptide desired neuronal cell type (see Table 8.1).
VMAT2 vesicular monoamine transporter 2

8.2 Identifying the Desired ESC-Derived


8.1 Introduction Cell Type for Transplantation

With the report of the isolation of the first human Ideally a stem cell-based therapy would replace the
embryonic stem cells (hESC) (Thomson et al., 1998), dead or damaged tissue with viable cells that could
now over 10 years ago, their potential use as ther- integrate and function after transfer to the brain. Given
apeutic agents became apparent, and an exciting ESC as the starting material, it is not clear at what
new era in stem cell biology began. There has been stage in their trajectory from pluripotent stem cells or
particular interest in using ESC-derived neural pro- neural stem cells (NSC) to differentiated neurons they
genitors and neurons for the treatment of a variety should be transplanted. The answer to this question
of neurodegenerative diseases and central nervous will depend, to some extent, on the specific disease
system injuries that have proven somewhat intractable under treatment, but some considerations generally
to other treatment approaches. Candidate disorders for apply. It may be desirable to have a self-renewing
ESC-based cell therapy include Parkinsons Disease source of the desired cell type, particularly if the disor-
(PD), amyotrophic lateral sclerosis (ALS), and spinal der is characterized by an ongoing neurodegenerative
cord lesions. There are many scientific roadblocks that environment, which is likely to promote the death
must be overcome before we can approach the clinic, of transplanted derivatives as well as the endogenous
including genetic compatibility of transplant material neuronal population. Transplantation of undifferenti-
with the patient, the tendency of ESC to form tumors ated ESC, however, frequently results in the formation
in vivo, and the contamination of many existing hESC of teratocarcinomas, tumors that contain undifferenti-
lines with animal products (Gruen and Grabel, 2006). ated ESC as well as differentiated derivatives of all
Work along all of these fronts is proceeding, but it is three primary germ layers (Brustle et al., 1997; Deacon
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 99

Table 8.1 Overview of selected protocols for derivation of specific neuronal subtypes from mouse and human embryonic stem
cells
Desired Cell Growth factor and other Molecular markers Molecular References
neuron type source molecular treatments (intermediates) markers (mature)
Dopaminergic mES AA, cAMP, and FGF2 Lmx1a and Msx1, TH Lee et al.
then AA, FGF2, then Nurr1 and (2000)
FGF8, and Shh, then Pitx3
only AA and cAMP

hES Wnt1 (from MS5 Lmx1a and Msx1, TH Perrier et al.


stromal cells) then then Nurr1 and (2004)
AA, BDNF, cAMP, Pitx3
FGF8, GDNF, Shh,
and TGF3 then AA,
BDNF, FGF8, and
Shh then AA, BDNF,
cAMP, GDNF, and
TGF3

Serotonergic mES FGF4 and Shh then GATA2, Pet1, and 5-HT and SERT Barberi et al.
FGF2, FGF4, and Shh Lmx1b (2003)
then only AA and
BDNF

hES BDNF, FGF1, Forskolin, GATA2, Pet1, and TPH and 5-HT Kumar et al.
GDNF, and 5HT Lmx1b (2008)

Cholinergic mES FGF2 and RA then Nkx2.1 and Lhx8 ChAT Okada et al.
BMP4, Shh, or Wnt3a (2008)

GABAergic mES FGF2 then FGF2, FGF8, GAD67 GAD67 and Barberi et al.
(cortical) and Shh then BDNF GABA (2003)
and NT-4

mES FGF2 then only N2B27 GAD67 GAD67, GABA, Okabe et al.
and FCS and calbindin (1996)

GABAergic mES BMP2, RA, Shh, and MAP-2 Lim2, GAD67, Murashov
(spinal) Wnt3a and GABA et al.
(2004)

Motor mES RA then Shh (or Pax6, Nkx6.1, and HB9 Wichterle
synthetic Hh agonist) Olig2 et al.
(2002)

hES RA and Shh then BDNF, Pax6, Sox1, Olig2 HB9 and Islet1/2 Li et al.
GDNF, and IGF-1 then ChAT (2005)

hES AA, BDNF, RA, and Nkx6.1 and Olig2 HB9, ChAT, and Lee et al.
Shh then AA, BDNF Lhx3 (2007)
and GDNF

iPS RA and Shh Pax6 and Olig2 HB9, Islet1/2, Dimos et al.
(human) and ChAT (2008)

et al., 1998; Wernig et al., 2004). This is obviously not transplanted is proliferative, however, it remains capa-
a desired outcome, and most approaches have therefore ble of forming tumors consisting of predominantly
focused on using NSC, neural progenitors, or differ- NSC and progenitors (Carpentino et al., 2008). If ter-
entiated neurons for grafts. As long as the cell type minally differentiated neurons are grafted, tumor risk
100 N. Ammon et al.

decreases, but these cells are no longer capable of self- that promote the emergence of these cell types in
renewal and may not even survive in certain areas of the embryo and during adult neurogenesis (Cai and
the brain. The relative threat of neural tumor forma- Grabel, 2007) (Fig. 8.1). During mammalian embryo-
tion by proliferative progenitors would depend upon genesis the epiblast, or primitive ectoderm, is the
the host environment and whether it could support source of all three primary germ layers; ectoderm,
appropriate terminal differentiation. Here the niche mesoderm and endoderm. The signals that direct the
into which the ESC-derived grafts are transplanted ectoderm to make neurectoderm versus epidermis or
may be key, and this will vary for different disor- other lineages have been studied extensively in many
ders. Evidence suggests that both NSC and specific species (Stern, 2005). Initial analyses suggested that
progenitors are able to respond to certain host environ- the absence of instructions, a so-called default state,
ments and integrate and function following transplant, led to specification of neurectoderm. In particular,
and some examples are discussed in subsequent sec- organizer regions associated with neural induction con-
tions. If the host environment is unable to support tain molecules that inhibit both bone morphogenic
the differentiation and integration of transplanted NSC protein (BMP) (Sasai et al., 1995, 1994; Smith and
or progenitors, terminally differentiated neurons may Harland, 1992) and Wnt signaling (Wilson et al.,
provide the most suitable graft material, although a 2001). Additional studies have implicated a positive
limited lifespan in vivo may necessitate multiple grafts role for Notch (Ross et al., 2003) as well as fibrob-
administered periodically. Thus, in selecting the cell last growth factor (FGF) (De Robertis and Kuroda,
type for transplant, one must weigh relative tumor risk 2004) and Wnts (Baker et al., 1999) in the initial
with the ability of the cells to survive and function in phase of neural specification, and Hedgehog(Hh) in
the host environment. As we identify factors that pro- the proliferation and survival of embryonic and adult
mote successful transplant incorporation, a subject of neural stem cells. Studies suggest that a similar array
intense investigation, these may be used to facilitate of factors, including Hh and Wnts, supports the sur-
functional integration under unfavorable conditions. vival and proliferation of NSC in the two regions
of adult neurogenesis, the subventricular zone of the
lateral ventricles and the subgranular zone of the hip-
8.3 Generating Neural Progenitors: Back pocampus dentate gyrus (Ahn and Joyner, 2005; Lie
to the Embryo et al., 2005; Machold et al., 2003; Palma et al., 2005).
Protocols for generating NSC from ESC can be
Directing the differentiation of pluripotential ESC to divided into two classes, those that rely upon an embry-
specific neuronal subtypes begins with their transition oid body intermediate, and those that do not (Cai and
to multipotential NSC that can differentiate into oligo- Grabel, 2007). When ESC are removed from their
dendrocytes, astrocytes, or neurons (Temple, 2001). feeder layer, and placed in suspension culture, they
Successful methodologies for producing NSC and neu- form aggregates, which within a few days consist of
rons from either mouse or human ESC have been an outer layer of hypoblast-like cells (extraembry-
based on a thorough understanding of the conditions onic visceral endoderm) surrounding an epiblast-like

Fig. 8.1 From ESC to


neuron. Production of
neurons or glia from
embryonic stem cells involves
production of NSC
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 101

core (Martin et al., 1977; Maye et al., 2004; Rathjen suspension assay for production of NSC (Smukler et
et al., 2002). At early stages these embryoid bodies al., 2006). Production of NSC, however, is a rare event
resemble the anterior pre-streak stage embryo, with under these conditions, with only 0.2% of ESC form-
the epiblast-like core able to generate derivatives of all ing neurospheres. More efficient direct differentiation
three primary germ layers (Keller, 2005). The interac- of ESC to NSC can also be obtained under serum-free
tion of the distinct lineages within the embryoid body conditions in monolayer culture at moderate cell den-
provides the positive and negative instructions needed sities (Ying et al., 2003). By 5 days, up to 75% of
to promote neurectoderm differentiation. This lineage the cells express NSC markers. The transition to NSC
commitment can be enhanced in vitro by a number of is accompanied by the death of significant numbers
treatments (Maye et al., 2004; Rathjen et al., 2002). of cells in the cultures, suggesting that these condi-
The signaling molecule Indian hedgehog, secreted by tions are effective, at least in part, due to selective
the visceral endoderm, promotes neurectoderm differ- survival of the NSC. Under these conditions, emerging
entiation in vitro (Maye et al., 2004), as does the addi- NSC form rosettes, in which cells elongate and align
tion of retinoic acid (RA) (Bain et al., 1995; Gottlieb, radially, in a manner reminiscent of neural tube for-
2002; Guan et al., 2001). RA addition also increases mation, as well as of neurectoderm differentiation in
the yield of neural derivatives, including motor neu- ESC embryoid bodies (Fig. 8.1b). These ESC-derived
rons (Bibel et al., 2004; Li et al., 2005; Wichterle NSC share morphological and biochemical character-
et al., 2002). Conditioned medium from a hepatocel- istics of radial glia, the NSC of the embryonic neural
lular carcinoma cell line can promote the homoge- tube. Co-culture of ESC at very low, clonal densities on
neous differentiation of primitive ectoderm-like (EPL) stromal cell lines (Table 8.1) also promotes NSC pro-
cells from mouse ESC in suspension culture (Rathjen duction (Barberi et al., 2003; Kawasaki et al., 2000).
et al., 2002, 1999). The conditioned medium contains The identity of the factor, or factors, provided by the
unidentified signals that induce neurectoderm differen- stromal cells has not been determined (Kawasaki et al.,
tiation at the expense of mesodermal and endodermal 2000).
derivatives. Given the goal to produce a variety of neuronal
An additional protocol combines use of an embry- cell types for transplantation therapies, it is impor-
oid body intermediate with a neural lineage-specific tant to determine whether the ESC-derived NSC have
selection step (Guan et al., 2001; Okabe et al., 1996). restricted potential or can generate all desired progen-
The selection step deprives the cells of growth fac- itors and differentiated cell types. During embryoge-
tors and signaling molecules, mimicking a default state nesis, the potential of emerging NSC, as measured
under which non-neural lineages die and NSC survive. by their ability to generate different classes of neu-
These conditions remove inhibitory factors for neu- rons both in vitro (Fig. 8.2) and in vivo, is dependent
ral differentiation, such as BMPs, as well as signals upon embryonic age as well as their site of ori-
required for the differentiation and survival of other gin along both the rostral/caudal and dorsal/ventral
lineages. Early stage embryoid bodies are plated on axes. Identification of neuronal subtype is based upon
adhesive substrates in a minimal serum-free medium. the expression profile of region specific transcrip-
During several days of culture, non-neurectoderm tion factors and neurotransmitters. Care must be taken
derivatives die, leaving a neurectoderm-derived neural to not rely on a single marker, including neuro-
progenitor population (Okabe et al., 1996). Subsequent transmitters, since, for example, GABA characterizes
plating on laminin substrates in the presence of FGF2 interneurons of both the telencephalon and spinal
and epidermal growth factor (EGF) promotes NSC cord.
proliferation, and at this stage cultures may be 8095% What is known about the potential of NSC gen-
NSC (Okabe et al., 1996). erated by the various protocols described? The NSC
NSC can be generated directly from ESC without an produced using the RA protocol may have restricted
embryoid body intermediate by plating the cells at very pluripotency, in one case to make dorsal, Pax6-positive
low clonal densities (120 cells/well) under feeder-free progenitors that differentiate in vitro into pyramidal
conditions in serum-free defined medium (Tropepe et neurons (Bibel et al., 2004). When these progenitors
al., 2001). This approach uses a neurosphere-based were transplanted to the embryonic chick neural tube,
102 N. Ammon et al.

Fig. 8.2 Mouse


ESC-derived NSC and
neurons. Nestin+ neural stem
cells derived using the
embryoid body-based defined
medium protocol, (a);
Sox1-GFP+ rosettes of
ESC-derived NSC formed
using the monolayer defined
medium protocol, (b);
III-tubulin+, (c); MAP2+
ESC-derived neurons, (d);
glutamatergic (Glu) neuron,
E; cluster of GABAergic
III-tubulin+ neurons

they showed limited capacity to differentiate, com- early post-mitotic neurons express tyrosine hydrox-
pared to ESC-derived neural progenitors generated in ylase (TH), which catalyzes the production of DA
the absence of RA (Plachta et al., 2004). RA has (Marin et al., 2005). Degeneration of SNi DA neurons
also been demonstrated to caudalize the neural tube, has been linked to PD, and defective dopaminergic
as well as ESC-derived neurons (Okada et al., 2004). transmission in the nucleus accumbens can contribute
Figure 8.1c, d shows III tubulin-positive and MAP2- to the development of depression, schizophrenia and
positive neurons as well as a glutamatergic and drug addiction (Nestler, 2000; Sulzer, 2007). As DA
GABAergic neurons derived from mouse ESC. neurons in the SNi deteriorate, levels of dopamine
Although not investigated extensively, the other release in the striatum plummet. This leads to motor
neurogenesis protocols appear to produce progenitors deficits and cognitive impairments at later stages.
with anterior, dorsal properties. Treatment with growth Patients exhibit rigidity, tremor and dyskinesia (Samii
factors or expression of transcription factors known et al., 2004). Current treatment with L-dopa can help
to provide regional identity during embryogenesis can reduce impairments, but eventually the drug loses effi-
be used to direct differentiation toward specific pro- cacy and the disease progresses. Understanding the
genitor and neuron fates. For example, treatment of factors that influence DA neuron differentiation may
ESC-derived neural progenitors with Wnt3A promotes lead to better cell therapies for the treatment of PD.
a dorsal telencephalic fate, whereas sonic hedgehog Several growth factors contribute to the produc-
(Shh) promotes a ventral fate (Watanabe et al., 2005). tion of DA neurons. During the development of
Approaches for the derivation of specific subtypes of the forebrain, Shh and FGF8, which is dependent
neurons, as well as the role of these cells in neuro- upon Shh for its induction (Aoto et al., 2002),
logical disorders are described below. Progress in stem are both expressed at the midbrain-hindbrain bor-
cell-based therapeutics to treat these conditions is also der, specifying mesencephalic progenitors (Abeliovich
summarized. and Hammond, 2007). Expression of both FGF8 and
Shh continues into adulthood in the SNi, with a
decrease of FGF8 during the onset of PD (Tanaka
et al., 2001). Also, Transforming Growth Factor-
8.4 Midbrain Dopaminergic Neurons (TGF-) family members are expressed in the floor
plate during early midbrain development and can pro-
Motor control, coordinated by the striatum, is influ- mote the survival of cultured mesencephalic DA neu-
enced by dopamine release from neurons of the sub- rons. TGF- and Shh appear to act independently of
stantia nigra (SNi). Dopaminergic (DA) neurons of the each other for the induction of midbrain dopaminer-
SNi are derived from the developing midbrain where gic (mDA) neurons (Roussa and Krieglstein, 2004).
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 103

Taken together, Shh, FGF8 and TGF- temporal and establish a niche for the development of DA pre-
regional expression in the early midbrain overlap, cursors. Neural progenitors exposed to all three fac-
and these factors appear to establish a niche envi- tors as well as glial cell derived neurotrophic factor
ronment for the differentiation of DA neurons in the (GDNF) and brain derived neurotrophic factor (BDNF)
midbrain. can differentiate into cultures that are 80% DA neu-
At this point, there is a second wave of expression rons (Perrier et al., 2004). It is clear from these
of regional markers that influences the differentia- studies that ESC-derived neural progenitors rely on
tion of neurons in the ventral midbrain into mature similar environmental cues as neural progenitors in
DA neurons. Precursor cells are still dividing as the vivo.
homeobox transcription factors Lmx1a and Msx1 are Utilizing transgenic approaches, recent studies have
expressed. Lmx1a is a crucial transcription factor for been able to purify, via fluorescence activated cell
the emergence of DA neurons in the midbrain. Loss sorting (FACS), populations of neural progenitors that
of function studies for Lmx1using RNAi have shown a express Pitx3. Mouse ESC-derived neural progeni-
reduction of DA neurons in the midbrain, while gain tors that express eGFP under the Pitx3 promoter were
of function studies using ESC yield an increase of enriched from 2 to 3% of the total population to
DA neurons in vitro (Andersson et al., 2006). As DA over 90% after FACS (Hedlund et al., 2008). The
progenitors become post-mitotic another set of tran- resulting population also expressed Nurr1, Engrailed-
scription factors are expressed including Nurr1 and 1, Lmx1a, TH, l-aromatic amino acid decarboxy-
Pitx3. Nurr1 is expressed in DA neurons in adulthood lase (AADC), and vesicular monoamine transporter
and is responsible for vesicle packaging and axonal 2 (VMAT2), all of which are expressed in mature
transport of dopamine. When neuronal progenitors are midbrain DA neurons. Future strategies for the deriva-
co-cultured with Nurr1-overexpressing astrocytes, a tion of midbrain DA neurons should focus on mark-
fourfold increase in DA markers is observed (Chung ers that are expressed in committed DA progenitors,
et al., 2002). Pitx3 expression increases following such as Limx1a. Once a transgenic line is created
Nurr1, is highly expressed by DA neurons in the adult with fluorescent protein expression under the con-
SNi, and contains a high-affinity binding site for the trol of a promoter like Limx1a, cells can be sorted
TH promoter, which is expressed by mature mDA and expanded in order to produce larger numbers of
neurons (Cazorla et al., 2000). pure post-mitotic neurons that express markers for
The pathways for in vivo differentiation of mid- midbrain DA neurons. The resulting cell population
brain DA neurons can be applied in vitro to direct ESC would provide a highly enriched graft of DA neu-
toward a DA neuron fate. Early methods for producing rons that could potentially restore function and alle-
neural progenitors from mouse ESC lines required the viate symptoms of PD when transplanted into the
addition of FGF2 and EGF, which bias the population striatum.
of neural progenitors towards a forebrain fate (Okabe Animal models for PD were developed in the 1980s
et al., 1996). Later methods utilized cAMP and ascor- when it was discovered that the drug 1-methyl-4-
bic acid (AA) to bias in favor of a ventral phenotype phenyl-1,2,3,6-tetrahydropyridine (MPTP) selectively
(Lee et al., 2000). As Shh and FGF8 are expressed by destroys mDA neurons in rodents and primates result-
the midbrain-hindbrain organizer, several studies have ing in hypokinesia and rigidity (Doudet et al., 1985).
shown that addition of these factors during neural dif- Initially, cell replacement therapy for PD was tested
ferentiation in vitro increases the yield of DA neurons on patients who had ingested MPTP and devel-
substantially (Geeta et al., 2008; Sonntag et al., 2007). oped Parkinsonian symptoms. Bilaterally grafting fetal
However, only 40% of the neurons were TH positive in mesencephalic progenitors to the caudate putamen
these cultures, possibly due to the inclusion of FGF2 enabled the patients to regain motor function and
in the protocols. Eliminating FGF2 and co-culturing reduce their L-dopa regimen (Widner et al., 1992).
neural progenitors with Wnt1-overexpressing stromal However, transplantation of the same cell population
cells has been shown to dramatically increase TH- in Parkinsonian patients has yielded mixed results due
positive populations (Castelo-Branco et al., 2003). to poor grafts or subsequent inflammation (Piccini
During early ventral midbrain development, Shh, et al., 2005). Because of the ethical complications
FGF8 and the TGF- family members cooperate to concerning the origin of the graft population and
104 N. Ammon et al.

the difficulty of obtaining a substantial amount of (TLE). In the hippocampus, a tri-synaptic circuit exists
DA neurons from fetal ventral mesencephalic tis- in which excitatory signals coming from the entorhi-
sue for transplant, deriving a pure population of DA nal cortex activate granule cells in the dentate gyrus.
neurons from ESC offers more promise for a cell These cells then synapse onto pyramidal cells in the
therapy. CA3 region, which in turn activate pyramidal cells in
the CA1 region. Excitatory signals from CA1 are sent
back to the entorhinal cortex and the relay continues
(Danglot et al., 2006). The GABAergic interneurons
8.5 GABAergic Interneurons present in the hippocampus are damaged as a result of
overactivation in this circuit (Magloczky et al., 2000).
-aminobutyric acid (GABA)ergic neurons are the As the loss of interneurons results in lessened inhibi-
major inhibitory neurons in the mammalian nervous tion of other excitatory neurons of the hippocampus,
system. By activating GABA receptors on target glu- firing continues and the interneurons are further dam-
tamatergic neurons, they provide inhibitory regulation aged (Morimoto et al., 2004). The result is seizures,
in neuronal circuits. GABAergic neurons are typi- which can vary in severity and ultimately lead to
cally categorized based on their morphology, location, hippocampal sclerosis. GABAergic interneurons gen-
synaptic targets (whether they synapse on the soma, erated in vitro could be used to replace the damaged
axons, or dendrites of target cells), and the calcium neurons in the hippocampus, thereby alleviating the
binding proteins and neuropeptides they express. In seizures. There is hope that GABAergic interneurons
this chapter, we focus on the expression of calcium generated from human ESC could provide the neu-
binding proteins and neuropeptides as indicators of ronal replacement cells to treat human patients with
interneuronal subtype generated in in vitro culture. For TLE.
a more detailed description of categorization based on Changes in the number of specific types of
morphology and synaptic targets refer to Freund and GABAergic interneurons as well as errors in their
Buzsaki (Freund and Buzsaki, 1996). function have also been implicated in psychological
All GABAergic neurons express the enzyme glu- diseases such as schizophrenia and bipolar disorder
tamic acid decarboxylase (GAD), which is necessary (Benes and Berretta, 2001). In cases of schizophre-
for synthesis of the inhibitory neurotransmitter GABA. nia, analysis of post-mortem human brain tissue
Different subtypes of interneurons express the calcium has indicated a decrease in PV- and SST-expressing
binding proteins parvalbumin (PV), calretinin (CR), interneurons in various layers of the prefrontal cor-
or calbindin (CB) (Freund and Buzsaki, 1996). While tex (Hashimoto et al., 2003; Morris et al., 2008).
PV interneurons tend to be large and have elaborate Another study of post-mortem tissue from both
axons, CR positive interneurons are smaller, bipolar, schizophrenic and bipolar disorder patients showed
and are usually vertically oriented (Tamamaki variations in number of interneurons representative
et al., 2003; Wonders and Anderson, 2006). of all three calcium binding protein subtypes when
Interneurons are also classified based upon expression compared to control subjects (Sakai et al., 2007).
of the neuropeptides somatostatin (SST), vasoactive These data suggest that transplantation of healthy, in
intestinal peptide (VIP), cholecystokinin (CCK) or vitro-derived GABAergic interneurons could provide
neuropeptide Y (NPY) (Markram et al., 2004). possible therapy for patients with either psychological
As the main role of GABAergic interneurons is to disorder.
regulate neuronal activity by providing inhibition of Huntingtons disease is a genetic disorder caused by
circuit firing, damage to or loss of these neurons can a mutation in the huntingtin gene which results in an
result in uncontrolled activity of excitatory neurons. abnormally high number of CAG repeats in the coding
This hyperactivity can lead to excitotoxicity and neu- sequence of the gene and aggregation of the mutant
ronal death, uncontrolled body movement, and other protein product in affected cells (Walker, 2007). In
neurological and psychological disorders. Huntingtons patients, the GABAergic medium spiny
Many studies indicate that loss of GABAergic projection neurons of the striatum (comprising about
interneurons is central to seizures originating in the 95% of total striatal neurons) are progressively lost.
hippocampus in human cases of temporal lobe epilepsy Many events have been proposed that ultimately cause
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 105

the death of these neurons, including excitotoxicity et al., 2007). Interestingly, CR-positive interneurons
and disruption of cellular metabolism (Gil and Rego, appear to be specified independent of Nkx2.1 activity
2008). This disease is manifested by cognitive and as they are still generated in Nkx2.1 mutant mice
emotional deficits as well as progressive loss of coor- (Nery et al., 2002). Strong evidence exists that PV-
dination and uncontrolled body movements (chorea), and SST-positive interneurons derive from the MGE,
eventually leading to death (Walker, 2007). whereas CR-positive interneurons derive from the
In addition to the documented degeneration of Nkx2.1-negative CGE (Xu et al., 2004). CR-positive
motor neurons in the neurodegenerative disorders ALS interneurons appear to be more dependent on activity
and spinal muscular atrophy (SMA), there has recently of the distal-less homeobox transcription factor Dlx1/2
been interest in the effect of these diseases on interneu- for their specification, as they are nearly eliminated
rons present in the adult spinal cord (Stephens et al., in Dlx1/2 null mice (Xu et al., 2004) while SST- and
2006). The GABAergic neurons of the ventral horn NPY-positive interneurons are only reduced in num-
of the spinal cord appear to be damaged to a similar ber. Dlx1/2 is required in all interneurons arising from
extent as the motor neurons in this region, indicating both the MGE and CGE for proper migration into cor-
that in order to provide cell therapy for these diseases, tical regions (Anderson et al., 1999; Panganiban and
the inhibitory neurons that regulate motor neuron Rubenstein, 2002).
function will also need to be generated and introduced GABAergic interneurons present in the mammalian
into patients for successful treatment. spinal cord are specified during neural tube pattern-
During mammalian embryonic development, cor- ing in response to opposing gradients of BMPs from
tical GABAergic interneurons are born in ventral the overlying ectoderm and roof plate and Shh from
regions of the telencephalon called the medial and cau- the notochord (Briscoe and Ericson, 1999; Helms
dal ganglionic eminences (MGE and CGE) (Sussel and Johnson, 2003; Jessell, 2000). The dorsal half of
et al., 1999; Wonders and Anderson, 2006). The the spinal cord contains eight populations of neurons
interneurons, which make up about 20% of the total referred to as dI1 through dI6 (from dorsal to ventral)
cortical neuron population, then migrate tangentially and dILA and dILB , which are subsequently born in
towards their final locations in the cortex. Many the region of dI4 through dI6. The ventral spinal cord
steps are required for the specification of cortical is comprised of five domains of neurons; three pools
GABAergic interneurons. Activation and maintenance of ventral interneurons called V0 through V3 and a
of the homeodomain-containing transcription factor pool of motor neurons (MN). Of the thirteen total neu-
Nkx2.1 in interneuron precursors comprising the MGE ronal populations present in the spinal cord, only the
is required for their initial specification, as Nkx2.1 dI4, dI6, dILA , V0, and V1 neurons are GABAergic
mutant mice lack a proper MGE and have a dra- inhibitory neurons (Glasgow et al., 2005; Wenner et al.,
matic reduction in GABA positive interneurons (Sussel 2000). The early transcription factor profile of the pro-
et al., 1999). Shh, secreted from the prechordal genitors for the six dorsal populations includes Math1
mesendoderm, is involved in the activation of Nkx2.1 (expressed by dI1 progenitors), Ngn1/2 (expressed by
(Ericson et al., 1995) during early patterning and main- dI2 and dI6 progenitors), Dbx2 (expressed by dI6
tenance of the subpallium (Gulacsi and Anderson, progenitors), and Mash1 and Pax7 (expressed by dI3
2006) during interneuron neurogenesis (E12 to E16 in through dI5 progenitors) (Helms and Johnson, 2003;
mouse). Proper dorsal/ventral patterning of the MGE Muller et al., 2002; Wilson and Maden, 2005). The
also involves strict regulation of gradients of BMPs progenitors of V0 and V1 GABAergic neurons in the
and FGF8, as loss of BMP inhibition leads to lack of ventral neural tube express Dbx2, Pax6, Irx3, and
Nkx2.1 expression (Anderson et al., 2002). A recent Dbx1 (the latter only in V0 progenitors)(Briscoe and
study on the role of Nkx2.1 in cortical interneuron Ericson, 2001). The early and late born GABAergic
specification demonstrates that it directly activates neurons themselves go on to express varied transcrip-
the LIM-homeodomain transcription factor Lhx6 (Du tion factors such as Lbx1, Isl1/2, Lim1/2, Lmx1b,
et al., 2008), which is expressed in migrating PV and Brn3a, Evx1/2, and En1 (Muller et al., 2002; Wilson
SST positive interneurons and is believed to be essen- and Maden, 2005). Pax2 and transcription factors,
tial for proper GABAergic interneuron migration from such as Ptf1, Lhx1, and Lhx5 that regulate its expres-
the MGE and incorporation into the cortex (Liodis sion in the spinal cord, are necessary for GABAergic
106 N. Ammon et al.

specification of spinal cord progenitors (Cheng et al., and mouse adult NSC have also been used in animal
2004, 2005; Glasgow et al., 2005; Pillai et al., 2007). models of Huntingtons disease and have demonstrated
Based on knowledge of how GABAergic interneu- varying degrees of graft incorporation and differentia-
rons develop during embryogenesis, protocols for in tion of the cells into more mature neurons (Clelland
vitro differentiation from pluripotent ESC have been et al., 2008). Adult NSC need to be obtained from
developed to enrich for these neurons. One such human patients by biopsy and are not as plentiful
protocol makes use of the embryoid body interme- or malleable in culture as ESC. This puts further
diate method of deriving NSC from mouse ESC emphasis on our need to better understand the in vivo
(Okabe et al., 1996). Following expansion of NSC development of striatal GABAergic neurons so that
in FGF2, growth factors were withdrawn and cells ESC-derived neurons can efficiently be generated for
were differentiated in neurobasal medium supple- transplantation approaches.
mented with B27 and fetal calf serum (Okabe et al., Transplantation paradigms for treatment of epilepsy
1996; Westmoreland et al., 2001). This differentiation have focused mainly on transplanting multipotent NSC
method resulted in cultures containing cells that were obtained from embryonic mouse or human brain tis-
positive for the GAD67 protein, which is necessary sue into the hippocampus or other brain regions to
for production and packaging of GABA. Sequential alleviate seizure activity. Replacement of GABA inhi-
treatment of ESC-derived NSC with Shh and FGF8 fol- bition in epileptic hippocampi seems to be critical for
lowed by withdrawal of these mitogens and treatment preventing seizures by inhibiting the overactivity of
with neurotrophin 4 (NT-4) and BDNF has improved the hippocampal circuitry characteristic of epilepsy.
upon this method, resulting in greater than 60% of Previous studies have demonstrated that transplanted
total neurons generated that were positive for GABA embryonic or adult NSC are able to survive to vari-
(Barberi et al., 2003). ous extents, differentiate into a selection of neuronal
Efforts have also been made to derive GABAergic subtypes including astrocytes and some GABAergic
interneurons specific to the dorsal spinal cord neurons, and provide initial relief of seizure activity
(Murashov et al., 2004). By mimicking the patterning (Chu et al., 2004; Hattiangady et al., 2008; Shetty and
of spinal interneuron progenitors using BMP signal- Hattiangady, 2007). As ESC can proliferate in culture
ing and treatment with Wnts, which are believed to act to generate large numbers of cells, and many pro-
downstream of BMP in dorsal spinal cord specification tocols have been devised to differentiate these ESC
(Muroyama et al., 2002), ESC can be differentiated into defined neuronal lineages, transplantation studies
into Lim2-positive neurons that also express GAD67 using ESC-derived NSC from mouse and human have
at a rate of about 55% of total neurons produced been increasing. Interestingly, it appears that the spe-
(Murashov et al., 2004). This protocol involves sequen- cific location into which NSC are transplanted plays
tial treatment with RA to induce a caudalized fate, a role in determining what type of neurons they will
recombinant Shh, BMP2, and Wnt3a. Importantly, mature into, as the host environment may send sig-
treatment of the NSC with these factors individu- nals to the transplanted cells to instruct their fate
ally was observed to be inefficient at inducing dorsal (Carpentino et al., 2008). This indicates that NSC may
GABAergic interneuron fate. The order and timing of need to be patterned into specific progenitors (such as
treatment is critical to reproducing the appropriate in GABAergic progenitors) or into more mature neurons
vivo environment. in order to generate the cells of interest following trans-
Cell transplantation studies for Huntingtons dis- plantation. To date, there are no reports of ESC-derived
ease began in the 1980s using fetal striatal cells and mature GABAergic neurons transplanted into epilepsy
were shown to generate healthy grafts and improve animal models.
motor function in both rodent and primate models
of Huntingtons disease and later in human patients
(Freeman et al., 2000, 1995; Hantraye et al., 1992; 8.6 Spinal Cord Motor Neurons
Isacson et al., 1986; Kendall et al., 1998). The use of
fetal derived striatal neurons for transplantation faces Motor neurons are generally cholinergic neurons and
many ethical issues and is not practical for widespread project from the four sections of the spinal cord
use in the treatment of Huntingtons disease. Human (cervical, thoracic, lumbar, and sacral) and innervate
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 107

musculature of the body needed for locomotion of the neural tube, with the highest concentration in
(somatic motor neurons) or various smooth muscle the most ventral region (Roelink et al., 1995). The
organs and glands (visceral motor neurons). These varying concentrations of Shh present along the DV
neurons either project from the brain stem to other axis are responsible for activation of specific home-
motor neurons of the spinal cord, or directly synapse obox transcription factors in each of the five domains
onto their target muscles to produce neuromuscu- of the ventral neural tube termed V0, V1, V2, MN,
lar junctions where neurotransmitters such as acetyl- and V3 (from dorsal to ventral). The combination of
choline are released. Loss of muscle innervation transcription factors indicates the type of neuron that
by motor neurons can lead to muscle atrophy, and will develop from progenitors in each region (Briscoe
improper regulation of motor neuron activity can cause et al., 2000; Wilson and Maden, 2005). The homeobox
uncontrolled muscle contraction. transcription factors Pax6 and Nkx2.2 are expressed
Cell replacement therapy using ESC-derived motor in response to Shh, with Pax6 expressed in a high
neurons has potential for treatment of patients suffer- to low gradient from dorsal to ventral and Nkx2.2
ing from motor neuron diseases such as ALS and SMA expressed in the ventral-most region where Pax6 is
as well as SCI (Hedlund et al., 2007). ALS, a late- absent (Ericson et al., 1997; Price and Briscoe, 2004).
onset disease typically affecting middle-aged people, Motor neurons are specified from progenitors (pMN)
is characterized by loss of motor neurons in the cere- in the ventral neural tube that express the transcription
bral cortex, brain stem, and spinal cord (Boillee et al., factors Olig2, Nkx6.1, and Pax6 (Briscoe et al., 2000;
2006; Mitchelle and Borasio, 2007). In SMA, which Wilson and Maden, 2005). As these motor neurons
mostly affects infants, the spinal cord motor neurons mature, they go on to express the molecular markers
selectively degenerate (Lunn and Wang, 2008). As MNR2 and HB9 (Wilson and Maden, 2005).
these neurons degenerate, the muscles that they inner- In vitro protocols aimed at directing ESC toward
vate lose function, resulting in paralysis and muscle a motor neuron fate take advantage of the depen-
deterioration. While many drug therapies have been dence of motor neuron progenitors on distinct con-
proposed, most offer little or no benefit to ALS and centrations of Shh. A protocol established by the
SMA patients. Cell therapy, however, could combine Jessell group (Wichterle et al., 2002) demonstrated
transplantation of ESC-derived neurons with growth that in order to generate motor neurons from mouse
factor treatments, potentially holding more promise ESC, three steps were necessary. First, ESC are dif-
(Boillee et al., 2006; Hedlund et al., 2007; Lunn and ferentiated into neuroectoderm via embryoid body
Wang, 2008). formation. These neurectodermal cells are then cau-
SCIs occur mostly in young adults, however they dalized by RA treatment to give them spinal cord
can affect people of all ages. In SCI, compression identity. Lastly, a ventral motor neuron identity is
to or lesion of the spinal cord can occur at various induced by treatment with either Shh peptide or a
levels of the spinal cord, often resulting in paralysis synthetic Hh agonist (Wichterle et al., 2002). Similar
below the point of injury. Following initial damage, protocols have been developed working with human
inflammation, ischemia, and glial scarring occur, ulti- ESC (Lee et al., 2007; Li et al., 2005) with some
mately leading to loss of myelination and neuronal modifications, including treatment with BDNF and
death (Schwab and Bartholdi, 1996). The ability to GDNF (Lee et al., 2007). Recently, motor neurons
implant healthy neurons near the site of SCI and direct have been derived, using the above guidelines, from
growth of their axons to restore neurological and motor induced pluripotent stem (iPS) cells that were gener-
activity is the ultimate goal of stem cell therapy for ated from fibroblast cells of an elderly patient with
SCI. ALS (Dimos et al., 2008). This work will allow fur-
Motor neurons are specified in the ventral half of ther study of the progression of motor neuron diseases
the posterior developing neural tube in response to like ALS and improve our ability to produce the proper
patterning cues emanating from the floor plate and cell types and conditions needed for effective cell
the underlying notochord. The main factor involved therapy.
in patterning the ventral neural tube is Shh, which is Several transplantation studies using animal mod-
produced by both the notochord and floor plate and is els of ALS, SMA, and SCI have had some success in
distributed in a gradient along the dorsal ventral axis functional incorporation of transplanted ESC-derived
108 N. Ammon et al.

motor neurons and other stem cells including mes- are dependent upon Gata2, which, in chicks, is suffi-
enchymal stem cells and adult NSC (Deshpande cient for the induction of both Pet1 and Lmx1b (Craven
et al., 2006; Harper et al., 2004; Hedlund et al., et al., 2004). Not all neurons from this developmental
2007; Mazzini et al., 2008; Vercelli et al., 2008). niche differentiate into 5-HT neurons. The repression
In a recent study using a rat motor neuron disease of this fate is dependent on the action of the transcrip-
model in which ventral motor neurons are destroyed tion factor Phox2b, as loss of function of Phox2b leads
by the Neuroadapted Sindbis virus, mouse ESC- to a vast increase in the number of 5-HT neurons in the
derived motor neurons transplanted into the spinal cord brain (Pattyn et al., 2003).
were able to survive and form functional neuromus- Thus far, there have been few attempts to dif-
cular junctions with skeletal muscle (Deshpande et ferentiate 5-HT neurons from ES cells lines. Most
al., 2006). Some treated rats even exhibited hindlimb protocols that influence the production of DA neurons
improvement. In another study using human ESC- result in the formation of a small number (23%) of
derived motor neurons transplanted into rodent spinal 5-HT neurons. This is most likely due to the addi-
cord, cells were able to form healthy grafts, send tion of Shh and FGF8, crucial in both DA and 5-HT
processes out from the graft site, and continued to differentiation. Induction of 5-HT neurons from mono-
mature in vivo (Lee et al., 2007). Further stud- layer ESC cell lines occurs in the presence of Shh,
ies are needed to demonstrate whether these human FGF2 and BDNF (Ying et al., 2003). However, these
ESC-derived neurons can make functional connections conditions produce only small numbers of 5-HT neu-
with the musculature and relieve symptoms of motor rons. From mouse ESC, optimal enrichment for 5-HT
neuron diseases. neurons resulted from co-culture on a stromal cell layer
and treatment with Shh and FGF4 several days before
exposure to FGF8 (Barberi et al., 2003). Recently,
5-HT neurons were derived from human ESC with
8.7 Serotonergic Neurons a somewhat greater efficiency (Kumar et al., 2008).
Sixty percent of the neurons in these cultures were
The dorsal Raphe nuclei are comprised of about 20,000 5-HT positive.
serotonergic (5-HT) neurons that project to most areas Clues of how to derive purer populations of 5-HT
of the brain. Because of their extensive innervation, neurons from ESC may be gained from observing how
5-HT neurons can influence many modes of behav- these cells arise during development. DA and 5-HT
ior and homeostasis. Disruptions of 5-HT signaling neurons share similar initial induction steps. From here
characterize many neuropsychiatric disorders such as the two populations diverge. DA neuron differentia-
depression, schizophrenia and bipolar disorder. Recent tion relies upon the expression of Pitx3 and Lmx1a,
reports have shown dystrophic 5-HT axons in patients whereas 5-HT neurons require the expression of Pet1
with PD, frontal lobe dementia and Lewy-body demen- and Lmx1b, both of which are controlled by Gata2.
tia (Azmitia and Nixon, 2008). As conventional med- Future strategies targeting 5-HT populations could
ication does little to repair or replace atrophied 5-HT focus on both driving the expression of 5-HT pro-
neurons, cell therapy may become an invaluable tool. genitor transcription factors, through proper timing of
Neural progenitors caudal to the midbrain-hindbrain FGF4 and/or other growth factors or by introducing
border give rise to the pool of 5-HT neurons in the genetic modifications that increase the expression of
adult brain. Similar to the induction of DA progenitors, Pet1 and Lmx1b, and inhibiting the production of DA
Shh and FGF8 play roles in 5-HT neuron specification, progenitors.
as does FGF4 (Ye et al., 1998). Together, these fac- 5-HT neurons have not been widely used in
tors define the initial inductive center of 5-HT neuronal transplantation studies. It has been shown, however,
differentiation. Pet1 is an important transcription fac- that 5-HT increases the proliferation and survival of
tor that marks early 5-HT progenitors into adulthood. transplanted neuroepithelial cells into the primordial
Mutations of Pet1 in mice lead to a severe reduction of neocortex of rats (Petrova and Otellin, 2007). Thus
5-HT neurons (Hendricks et al., 2003). In addition, the transplantation of 5-HT neurons could function to
ablation of Lmx1b leads to the absence of 5-HT neu- improve engraftment of other cell populations, such
rons in the brain (Ding et al., 2003). These two factors as DA neurons in patients with PD or GABAergic
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 109

interneurons in patients with TLE. It is also possible neurons. Cholinergic progenitors arise in the MGE, a
that 5-HT neurons may be used to treat neuropsychi- region that expresses the transcription factor Nkx2.1
atric disorders without the risk of side effects from and is exposed to Shh released from the prechordal
conventional treatments. As increases in synaptic 5- plate. In addition to cholinergic progenitors, the MGE
HT are linked to the function of serotonin selective gives rise to the majority of GABAergic interneu-
reuptake inhibitors (SSRIs), transplanting 5-HT neu- rons, as previously described. Nkx2.1 represses dor-
rons directly into brain regions like the hippocampus sal transcription factors like Pax6 while enhancing
and the anterior cingulated cortex may decrease the the expression of the LIM-homeodomain transcription
severity of the disorder when drug regimens fail. factors Lhx6 and Lhx8 (also known as Lhx7 and L3)
(Sussel et al., 1999). While Lhx6 is critical for the for-
mation of GABAergic interneurons and is expressed
by cholinergic progenitors, only Lhx8 is critical to
8.8 Basal Forebrain Cholinergic Neurons the development of forebrain cholinergic neurons.
Lhx8 deficient mice completely lose both the cholin-
Cholinergic neurons in the CNS contribute to ergic interneurons and cholinergic projection neurons
the regulation of cortical activation, behavioral of the striatum (Fragkouli et al., 2005; Zhao et al.,
arousal and memory formation. Located in the basal 2003).
forebrain, clusters of cholinergic neurons project axons To date, protocols for the efficient derivation of
into the limbic system, including the amygdala and cholinergic neurons from ESC are lacking. It has been
the hippocampus, and all cortical areas. Cholinergic observed that several factors can increase the pop-
neurons play an important role in learning and mem- ulation of cholinergic neurons from varying starting
ory formation whereby acetylcholine (ACh) release cell populations. Fetal basal forebrain tissue from rats
can contribute to long-term potentiation and affect the exposed in utero to BNDF followed by later addition of
plasticity of synapses of neurons in the hippocam- nerve growth factor (NGF) increased the emergence of
pus (Sarter and Bruno, 2000). Activity of cholinergic cholinergic neurons (Knusel et al., 1991). Endogenous
neurons is increased during arousal and attention. adult NSC were induced to a cholinergic neuron fate
Diseases associated with memory deficits and with both NGF and RA in rats with a lesioned basal
dementia often involve the degeneration of choliner- forebrain (Calza et al., 2003). In mouse ESC, RA
gic neurons. In Parkinsonian Disease Dementia (PDD), also increases the yield of cholinergic neurons while
cholinergic deficits appear early in the disease and expanding the population of GAD67 positive interneu-
are widespread throughout the brain. The loss of rons (Okada et al., 2008). As both cholinergic and
cholinergic innervation in the cortex and hippocam- GABAergic neurons develop in the same regional
pus is often associated with the formation of Lewy niche in vitro, factors that encourage the differentia-
Bodies, aggregates of protein in the cell bodies of tion of one cell type should also bias in favor of the
neurons. Plaque formation, a build up of amyloid- differentiation of the other in vitro. In order to select
peptide, is minimal in PDD patient brains when for cholinergic neurons over GABAergic interneurons,
compared to the brains of patients with AD (Farlow it will be necessary to identify the conditions that
and Cummings, 2008). Plaque formation during the increase the expression of Lhx8. In addition, pure
progression of AD increases in the medial tempo- population of cholinergic neurons could be induced
ral lobe and cortical structures. In addition, cognitive using a bacterial artifical chromosome (BAC) encod-
deficits in patients with AD appear when cholin- ing Lhx8. As cholinergic progenitors are derived, cell
ergic transduction declines. The neuropathology of sorting techniques could be employed to enhance the
AD shows an increased loss of cholinergic neu- cholinergic population for transplantation.
rons in the basal forebrain and hippocampus lead- ESC-derived cholinergic neurons could provide a
ing to deficits in cognition, memory, language and valuable tool for treating patients with AD, as the
behavior. loss of cholinergic innervation leads to the most pro-
Most cholinergic neurons in the forebrain, found in nounced symptoms of the disease. Using mouse adult
the striatum, can be classified into two distinct groups: NSC, cortical grafts differentiated into cholinergic
cholinergic interneurons and cholinergic projection neurons and improved working memory in a mouse
110 N. Ammon et al.

model of AD (Wang et al., 2006). Additionally, human subtype specification of fertilization and nuclear transfer
NSC transplantation also leads to a reduction in work- embryonic stem cells and application in parkinsonian mice.
Nat Biotechnol 21: 12001206.
ing memory error as the transplanted cells migrate
Benes FM, Berretta S (2001) GABAergic interneurons: implica-
through the cortex and hippocampus (Marutle et al., tions for understanding schizophrenia and bipolar disorder.
2007). While transplanting NSC improves the cogni- Neuropsychopharmacology 25: 127.
tive abilities in mouse models, the grafted cells also Bibel M, Richter J, Schrenk K, Tucker KL, Staiger V, Korte M,
Goetz M, Barde YA (2004) Differentiation of mouse embry-
differentiate into glia and neurons that are not cholin-
onic stem cells into a defined neuronal lineage. Nat Neurosci
ergic. By focusing on producing pure populations of 7: 10031009.
cholinergic neurons for engraftment, the beneficial Boillee S, Velde CV, Cleveland DW (2006) ALS: a disease of
effects of ESC-derived transplants could be increased. motor neurons and ther nonneuronal neighbors. Neuron 52:
3959.
Briscoe J, Ericson J (1999) The specification of neuronal identity
by graded sonic hedgehog signalling. Semin Cell Dev Biol
10: 353362.
8.9 Conclusions Briscoe J, Ericson J (2001) Specification of neuronal fates in the
ventral neural tube. Curr Opin Neurobiol 11: 4349.
Briscoe J, Pierani A, Jessell TM, Ericson J (2000) A home-
The next decade may well see the application of human odomain protein code specifies progenitor cell identity and
ESC-based therapies. It is likely that neurological dis- neuronal fate in the ventral neural tube. Cell 101: 435445.
orders will be among the first conditions treated, and Brustle O, Spiro AC, Karram K, Choudhary K, Okabe S, McKay
RD (1997) In vitro-generated neural precursors participate in
with care taken to avoid the potential pitfalls, we will mammalian brain development. Proc Natl Acad Sci U S A
hopefully enter a new era of regenerative medicine. 94: 1480914814.
Cai C, Grabel L (2007) Directing the differentiation of embry-
onic stem cells to neural stem cells. Dev Dyn 236:
References 32553266.
Calza L, Giuliani A, Fernandez M, Pirondi S, DIntino G,
Aloe L, Giardino L (2003) Neural stem cells and choliner-
Abeliovich A, Hammond R (2007) Midbrain dopamine neuron gic neurons: regulation by immunolesion and treatment with
differentiation: factors and fates. Dev Biol 304: 447454. mitogens, retinoic acid, and nerve growth factor. Proc Natl
Ahn S, Joyner AL (2005) In vivo analysis of quiescent adult Acad Sci U S A 100: 73257330.
neural stem cells responding to Sonic hedgehog. Nature 437: Carpentino JE, Hartman NW, Grabel LB, Naegele JR (2008)
894897. Region-specific differentiation of embryonic stem cell-
Anderson RM, Lawrence AR, Stottmann RW, Bachiller D, derived neural progenitor transplants into the adult mouse
Klingensmith J (2002) Chordin and noggin promote orga- hippocampus following seizures. J Neurosci Res 86:
nizing centers of forebrain development in the mouse. 512524.
Development 129: 49754987. Castelo-Branco G, Wagner J, Rodriguez FJ, Kele J, Sousa K,
Anderson S, Mione M, Yun K, Rubenstein JL (1999) Differential Rawal N, Pasolli HA, Fuchs E, Kitajewski J, Arenas E
origins of neocortical projection and local circuit neurons: (2003) Differential regulation of midbrain dopaminergic neu-
role of Dlx genes in neocortical interneurogenesis. Cereb ron development by Wnt-1, Wnt-3a, and Wnt-5a. Proc Natl
Cortex 9: 646654. Acad Sci U S A 100: 1274712752.
Andersson E, Tryggvason U, Deng Q, Friling S, Alekseenko Z, Cazorla P, Smidt MP, OMalley KL, Burbach JP (2000) A
Robert B, Perlmann T, Ericson J (2006) Identification of response element for the homeodomain transcription fac-
intrinsic determinants of midbrain dopamine neurons. Cell tor Ptx3 in the tyrosine hydroxylase gene promoter. J
124: 393405. Neurochem 74: 18291837.
Aoto K, Nishimura T, Eto K, Motoyama J (2002) Mouse GLI3 Cheng L, Arata A, Mizuguchi R, Qian Y, Karunaratne A, Gray
regulates Fgf8 expression and apoptosis in the developing PA, Arata S, Shirasawa S, Bouchard M, Luo P, et al. (2004)
neural tube, face, and limb bud. Dev Biol 251: 320332. Tlx3 and Tlx1 are post-mitotic selector genes determining
Azmitia EC, Nixon R (2008) Dystrophic serotonergic axons in glutamatergic over GABAergic cell fates. Nat Neurosci 7:
neurodegenerative diseases. Brain Res 1217: 185194. 510517.
Bain G, Kitchens D, Yao M, Huettner JE, Gottlieb DI (1995) Cheng L, Samad OA, Xu Y, Mizuguchi R, Luo P, Shirasawa S,
Embryonic stem cells express neuronal properties in vitro. Goulding M, Ma Q (2005) Lbx1 and Tlx3 are opposing
Dev Biol 168: 342357. switches in determining GABAergic versus glutamatergic
Baker JC, Beddington RS, Harland RM (1999) Wnt signaling transmitter phenotypes. Nat Neurosci 8: 15101515.
in Xenopus embryos inhibits bmp4 expression and activates Chu K, Kim M, Jung KH, Jeon D, Lee ST, Kim J, Jeong SW,
neural development. Genes Dev 13: 31493159. Kim SU, Lee SK, Shin HS et al. (2004) Human neural stem
Barberi T, Klivenyi P, Calingasan NY, Lee H, Kawamata H, cell transplantation reduces spontaneous recurrent seizures
Loonam K, Perrier AL, Bruses J, Rubio ME, Topf N, Tabar V, following pilocarpine-induced status epilepticus in adult rats.
Harrison NL, Beal MF, Moore MAS, Studer L (2003) Neural Brain Res 1023: 213221.
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 111

Chung S, Sonntag KC, Andersson T, Bjorklund LM, Park JJ, Freeman TB, Sanberg PR, Isacson O (1995) Development of the
Kim DW, Kang UJ, Isacson O, Kim KS (2002) Genetic engi- human striatum: implications for fetal striatal transplantation
neering of mouse embryonic stem cells by Nurr1 enhances in the treatment of Hungtingtons disease. Cell Transplant 4:
differentiation and maturation into dopaminergic neurons. 539545.
Eur J Neurosci 16: 18291838. Freund T, Buzsaki G (1996) Interneurons of the Hippocampus.
Clelland CD, Barker RA, Watts C (2008) Cell therapy in Hippocampus 6: 347470.
Huntington disease. Neurosurg Focus 24: E9. Geeta R, Ramnath RL, Rao HS, Chandra V (2008) One year
Craven SE, Lim KC, Ye W, Engel JD, de Sauvage F, Rosenthal A survival and significant reversal of motor deficits in parkin-
(2004) Gata2 specifies serotonergic neurons downstream of sonian rats transplanted with hESC derived dopaminergic
sonic hedgehog. Development 131: 11651173. neurons. Biochem Biophys Res Commun 373: 258264.
Danglot L, Triller A, Marty S (2006) The development of Gil JM, Rego AC (2008) Mechanisms of neurodegeneration in
hippocampal interneurons in rodents. Hippocampus 16: Huntingtons disease. Eur J Neurosci 27: 28032820.
10321060. Glasgow SM, Henke RM, Macdonald RJ, Wright CV, Johnson
De Robertis EM, Kuroda H (2004) Dorsal-ventral patterning and JE (2005) Ptf1a determines GABAergic over glutamatergic
neural induction in Xenopus embryos. Annu Rev Cell Dev neuronal cell fate in the spinal cord dorsal horn. Development
Biol 20: 285308. 132: 54615469.
Deacon T, Dinsmore J, Costantini LC, Ratliff J, Isacson O (1998) Gottlieb DI (2002) Large-scale sources of neural stem cells.
Blastula-stage stem cells can differentiate into dopaminergic Annu Rev Neurosci 25: 381407.
and serotonergic neurons after transplantation. Exp Neurol Gruen L, Grabel L (2006) Concise review: scientific and ethi-
149: 2841. cal roadblocks to human embryonic stem cell therapy. Stem
Deshpande DM, Kim Y-S, Martinez T, Carmen J, Dike S, Cells 24: 21622169.
Shats I, Rubin LL, Drummond J, Krishnan C, Hoke A, et al. Guan K, Chang H, Rolletschek A, Wobus AM (2001) Embryonic
(2006) Recovery from paralysis in adult rats using embryonic stem cell-derived neurogenesis. Retinoic acid induction and
stem cells. Ann Neurol 60: 3244. lineage selection of neuronal cells. Cell Tissue Res 305:
Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto 171176.
H, Chung W, Croft GF, Saphier G, Leibel R, Goland R, Gulacsi A, Anderson SA (2006) Shh maintains Nkx2.1 in the
et al. (2008) Induced pluripotent stem cells generated from MGE by a Gli3-independent mechanism. Cereb Cortex 16:
patients with ALS can be differentiated into motor neurons. i89i95.
Science 321: 12181221. Hantraye P, Riche D, Maziere M, Isacson O (1992) Intrastriatal
Ding YQ, Marklund U, Yuan W, Yin J, Wegman L, Ericson J, transplantation of cross-species fetal striatal cells reduces
Deneris E, Johnson RL, Chen ZF (2003) Lmx1b is essential abnormal movements in a primate model of Huntington
for the development of serotonergic neurons. Nat Neurosci 6: disease. Proc Natl Acad Sci U S A 89: 41874191.
933938. Harper JM, Krishnan C, Darman JS, Deshpande DM, Peck S,
Doudet D, Gross C, Lebrun-Grandie P, Bioulac B (1985) MPTP Shats I, Backovic S, Rothstein JD, Kerr DA (2004) Axonal
primate model of Parkinsons disease: a mechanographic and growth of embryonic stem cell-derived motoneurons in vitro
electromyographic study. Brain Res 335: 194199. and in motoneuron-injured adult rats. Proc Natl Acad Sci
Du T, Xu Q, Ocbina PJ, Anderson SA (2008) Nkx2.1 specifies U S A 101: 71237128.
cortical interneuron fate by activating Lhx6. Development Hashimoto T, Volk DW, Eggan SM, Mirnics K, Pierri JN, Sun Z,
135: 15591567. Sampson AR, Lewis DA (2003) Gene expression deficits
Ericson J, Muhr J, Placzek M, Lints T, Jessell T, Edlund T (1995) in a subclass of GABA neurons in the prefrontal cortex of
Sonic hedgehog induces the differentiation of ventral fore- subjects with schizophrenia. J Neurosci 23: 63156326.
brain neurons: a common signal for ventral patterning within Hattiangady B, Rao MS, Shetty AK (2008) Grafting of striatal
the neural tube. Cell 81: 747756. precursor cells into hippocampus shortly after status epilep-
Ericson J, Rashbass P, Schedl A, Brenner-Morton S, ticus restrains chronic temporal lobe epilepsy. Exp Neurol
Kawakami A, Heyningen Vv, Jessell T, Briscoe J (1997) 212: 468481.
Pax6 controls progenitor cell identity and neuronal fate in Hedlund E, Hefferan MP, Marsala M, Isacson O (2007) Cell
response to graded Shh signaling. Cell 90: 169180. therapy and stem cells in animal models of motor neuron
Farlow MR, Cummings J (2008) A modern hypothesis: the dis- disorders. Eur J Neurosci 26: 17211737.
tinct pathologies of dementia associated with Parkinsons Hedlund E, Pruszak J, Lardaro T, Ludwig W, Vinuela A,
disease versus Alzheimers disease. Dement Geriatr Cogn Kim KS, Isacson O (2008) Embryonic stem cell-derived
Disord 25: 301308. Pitx3-enhanced green fluorescent protein midbrain dopamine
Fragkouli A, Hearn C, Errington M, Cooke S, Grigoriou M, neurons survive enrichment by fluorescence-activated cell
Bliss T, Stylianopoulou F, Pachnis V (2005) Loss of fore- sorting and function in an animal model of Parkinsons
brain cholinergic neurons and impairment in spatial learning disease. Stem Cells 26: 15261536.
and memory in LHX7-deficient mice. Eur J Neurosci 21: Helms AW, Johnson JE (2003) Specification of dorsal spinal
29232938. cord interneurons. Curr Opin Neurobiol 13: 4249.
Freeman TB, Cicchetti F, Hauser RA, Deacon TW, Li XJ, Hersch Hendricks TJ, Fyodorov DV, Wegman LJ, Lelutiu NB, Pehek
SM, Nauert GM, Sanberg PR, Kordower JH, Saporta S, et al. EA, Yamamoto B, Silver J, Weeber EJ, Sweatt JD, Deneris
(2000) Transplanted fetal striatum in Huntingtons disease: ES (2003) Pet-1 ETS gene plays a critical role in 5-HT neu-
phenotypic development and lack of pathology. Proc Natl ron development and is required for normal anxiety-like and
Acad Sci U S A 97: 1387713882. aggressive behavior. Neuron 37: 233247.
112 N. Ammon et al.

Isacson O, Dunnett SB, Bjorklund A (1986) Graft-induced Markram H, Toledo-Rodriguez M, Wang Y, Gupta A,
behavioral recovery in an animal model of Huntington dis- Silberberg G, Wu C (2004) Interneurons of the neocortical
ease. Proc Natl Acad Sci U S A 83: 27282732. inhibitory system. Nat Rev Neurosci 5: 793807.
Jessell TM (2000) Neuronal specification in the spinal cord: Martin GR, Wiley LM, Damjanov I (1977) The development of
inductive signals and transcriptional codes. Nat Rev Genet cystic embryoid bodies in vitro from clonal teratocarcinoma
1: 2029. stem cells. Dev Biol 61: 230244.
Kawasaki H, Mizuseki K, Nishikawa S, Kaneko S, Kuwana Y, Marutle A, Ohmitsu M, Nilbratt M, Greig NH, Nordberg A,
Nakanishi S, Nishikawa SI, Sasai Y (2000) Induction of Sugaya K (2007) Modulation of human neural stem cell
midbrain dopaminergic neurons from ES cells by stromal differentiation in Alzheimer (APP23) transgenic mice by
cell-derived inducing activity. Neuron 28: 3140. phenserine. Proc Natl Acad Sci U S A 104: 1250612511.
Keller G (2005) Embryonic stem cell differentiation: emer- Maye P, Becker S, Siemen H, Thorne J, Byrd N, Carpentino J,
gence of a new era in biology and medicine. Genes Dev 19: Grabel L (2004) Hedgehog signaling is required for the dif-
11291155. ferentiation of ES cells into neurectoderm. Dev Biol 265:
Kendall AL, Rayment FD, Torres EM, Baker HF, Ridley RM, 276290.
Dunnett SB (1998) Functional integration of striatal allo- Mazzini L, Mareschi K, Ferrero I, Vassallo E, Oliveri G,
grafts in a primate model of Huntingtons disease. Nat Med Nasuelli N, Oggioni GD, Testa L, Fagioli F (2008) Stem
4: 727729. cell treatment in Amyotrophic Lateral Sclerosis. J Neurol Sci
Knusel B, Winslow JW, Rosenthal A, Burton LE, Seid DP, 265: 7883.
Nikolics K, Hefti F (1991) Promotion of central choliner- Mitchelle J, Borasio G (2007) Amyotrophic lateral sclerosis.
gic and dopaminergic neuron differentiation by brain-derived Lancet 369: 20312041.
neurotrophic factor but not neurotrophin 3. Proc Natl Acad Morimoto K, Fahnestock M, Racine RJ (2004) Kindling and sta-
Sci U S A 88: 961965. tus epilepticus models of epilepsy: rewiring the brain. Prog
Kumar M, Kaushalya SK, Gressens P, Maiti S, Mani S (2008) Neurobiol 73: 160.
Optimized derivation and functional characterization of 5- Morris HM, Hashimoto T, Lewis DA (2008) Alterations in
HT neurons from human embryonic stem cells. Stem Cells somatostatin mRNA expression in the dorsolateral prefrontal
Dev. cortex of subjects with schizophrenia or schizoaffective dis-
Lee H, Shamy GA, Elkabetz Y, Schofield CM, Harrison order. Cereb Cortex 18: 15751587.
NL, Panagiotakos G, Socci ND, Tabar V, Studer L Muller T, Brohmann H, Pierani A, Heppenstall PA, Lewin
(2007) Directed differentiation and transplantation of human GR, Jessell TM, Birchmeier C (2002) The homeodomain
embryonic stem cell-derived motoneurons. Stem Cells 25: factor lbx1 distinguishes two major programs of neu-
19311939. ronal differentiation in the dorsal spinal cord. Neuron 34:
Lee SH, Lumelsky N, Studer L, Auerbach JM, McKay RD 551562.
(2000) Efficient generation of midbrain and hindbrain neu- Murashov AK, Pak ES, Hendricks WA, Owensby JP, Sierpinski
rons from mouse embryonic stem cells. Nat Biotechnol 18: PL, Tatko LM, Fletcher PL (2004) Directed differentiation
675679. of embyronic stem cells into dorsal interneurons. FASEB J
Li XJ, Du ZW, Zarnowska ED, Pankratz M, Hansen LO, Pearce 2: 252254.
RA, Zhang SC (2005) Specification of motoneurons from Muroyama Y, Fujihara M, Ikeya M, Kondoh H, Takada S (2002)
human embryonic stem cells. Nat Biotechnol 23: 215221. Wnt signaling plays an essential role in neuronal specifica-
Lie DC, Colamarino SA, Song HJ, Desire L, Mira H, tion of the dorsal spinal cord. Genes Dev 16: 548553.
Consiglio A, Lein ES, Jessberger S, Lansford H, Dearie Nery S, Fishell G, Corbin JG (2002) The caudal ganglionic emi-
AR, et al. (2005) Wnt signalling regulates adult hippocampal nence is a source of distinct cortical and subcortical cell
neurogenesis. Nature 437: 13701375. populations. Nat Neurosci 5: 12791287.
Liodis P, Denaxa M, Grigoriou M, Akufo-Addo C, Yanagawa Y, Nestler EJ (2000) Genes and addiction. Nat Genet 26: 277281.
Pachnis V (2007) Lhx6 activity is required for the neuronal Okabe S, Forsberg-Nilsson K, Spiro AC, Segal M, McKay RD
migration and specification of cortical interneuron subtypes. (1996) Development of neuronal precursor cells and func-
J Neurosci 27: 30783089. tional postmitotic neurons from embryonic stem cells in
Lunn MR, Wang CH (2008) Spinal muscular atrophy. Lancet vitro. Mech Dev 59: 89102.
371: 21202133. Okada Y, Matsumoto A, Shimazaki T, Enoki R, Koizumi A,
Machold R, Hayashi S, Rutlin M, Muzumdar MD, Nery S, Ishii S, Itoyama Y, Sobue G, Okano H (2008) Spatio-
Corbin JG, Gritli-Linde A, Dellovade T, Porter JA, Rubin temporal recapitulation of central nervous system develop-
LL, et al. (2003) Sonic hedgehog is required for progenitor ment by murine ES cell-derived neural stem/progenitor cells.
cell maintenance in telencephalic stem cell niches. Neuron Stem Cells 26: 30863098.
39: 937950. Okada Y, Shimazaki T, Sobue G, Okano H (2004) Retinoic-acid-
Magloczky Z, Wittner L, Borhegyi Z, Halasz P, Vajda J, concentration-dependent acquisition of neural cell identity
Czirjak S, Freund T (2000) Changes in the distribution and during in vitro differentiation of mouse embryonic stem cells.
connectivity of interneurons in the epileptic human dentate Dev Biol 275: 124142.
gyrus. Neuroscience 96: 725. Palma V, Lim DA, Dahmane N, Sanchez P, Brionne TC,
Marin F, Herrero MT, Vyas S, Puelles L (2005) Ontogeny of Herzberg CD, Gitton Y, Carleton A, Alvarez-Buylla A, Ruiz
tyrosine hydroxylase mRNA expression in mid- and fore- i Altaba A (2005) Sonic hedgehog controls stem cell behav-
brain: neuromeric pattern and novel positive regions. Dev ior in the postnatal and adult brain. Development 132:
Dyn 234: 709717. 335344.
8 Directing Differentiation of ESC into Distinct Neuronal Subtypes 113

Panganiban G, Rubenstein JL (2002) Developmental functions Sasai Y, Lu B, Steinbeisser H, De Robertis EM (1995)


of the Distal-less/Dlx homeobox genes. Development 129: Regulation of neural induction by the Chd and Bmp-4 antag-
43714386. onistic patterning signals in Xenopus. Nature 377: 757.
Pattyn A, Vallstedt A, Dias JM, Samad OA, Krumlauf R, Rijli Sasai Y, Lu B, Steinbeisser H, Geissert D, Gont LK, De Robertis
FM, Brunet JF, Ericson J (2003) Coordinated temporal and EM (1994) Xenopus chordin: a novel dorsalizing factor
spatial control of motor neuron and serotonergic neuron gen- activated by organizer-specific homeobox genes. Cell 79:
eration from a common pool of CNS progenitors. Genes Dev 779790.
17: 729737. Schwab ME, Bartholdi D (1996) Degeneration and regenera-
Perrier AL, Tabar V, Barberi T, Rubio ME, Bruses J, Topf N, tion of axons in the lesioned spinal cord. Physiol Rev 76:
Harrison NL, Studer L (2004) Derivation of midbrain 319370.
dopamine neurons from human embryonic stem cells. Proc Shetty AK, Hattiangady B (2007) Concise review: prospects of
Natl Acad Sci U S A 101: 1254312548. stem cell therapy for temporal lobe epilepsy. Stem Cells 25:
Petrova ES, Otellin VA (2007) Serotonin is involved in the reg- 23962407.
ulation of histogenetic processes in rat embryonic neocortex. Smith WC, Harland RM (1992) Expression cloning of noggin, a
Bull Exp Biol Med 143: 372375. new dorsalizing factor localized to the Spemann organizer in
Piccini P, Pavese N, Hagell P, Reimer J, Bjorklund A, Oertel Xenopus embryos. Cell 70: 829840.
WH, Quinn NP, Brooks DJ, Lindvall O (2005) Factors Smukler SR, Runciman SB, Xu S, van der Kooy D (2006)
affecting the clinical outcome after neural transplantation in Embryonic stem cells assume a primitive neural stem cell
Parkinsons disease. Brain 128: 29772986. fate in the absence of extrinsic influences. J Cell Biol 172:
Pillai A, Mansouri A, Behringer R, Westphal H, Goulding M 7990.
(2007) Lhx1 and Lhx5 maintain the inhibitory- Sonntag KC, Pruszak J, Yoshizaki T, van Arensbergen J,
neurotransmitter status of interneurons in the dorsal Sanchez-Pernaute R, Isacson O (2007) Enhanced yield of
spinal cord. Development 134: 357366. neuroepithelial precursors and midbrain-like dopaminergic
Plachta N, Bibel M, Tucker KL, Barde YA (2004) neurons from human embryonic stem cells using the bone
Developmental potential of defined neural progenitors morphogenic protein antagonist noggin. Stem Cells 25:
derived from mouse embryonic stem cells. Development 411418.
131: 54495456. Stephens B, Guiloff RJ, Navarrete R, Newman P, Nikhar N,
Price SR, Briscoe J (2004) The generation and diversification of Lewis P (2006) Widespread loss of neuronal populations in
spinal motor neurons: signals and responses. Mech Dev 121: the spinal ventral horn in sporadic motor neuron disease. A
11031115. morphometric study. J Neurol Sci 244: 4158.
Rathjen J, Haines BP, Hudson KM, Nesci A, Dunn S, Rathjen Stern CD (2005) Neural induction: old problem, new findings,
PD (2002) Directed differentiation of pluripotent cells to yet more questions. Development 132: 20072021.
neural lineages: homogeneous formation and differenti- Sulzer D (2007) Multiple hit hypotheses for dopamine neuron
ation of a neurectoderm population. Development 129: loss in Parkinsons disease. Trends Neurosci 30: 244250.
26492661. Sussel L, Marin O, Kimura S, Rubenstein JL (1999) Loss of
Rathjen J, Lake JA, Bettess MD, Washington JM, Chapman G, Nkx2.1 homeobox gene function results in a ventral to dorsal
Rathjen PD (1999) Formation of a primitive ectoderm molecular respecification within the basal telencephalon: evi-
like cell population, EPL cells, from ES cells in response dence for a transformation of the pallidum into the striatum.
to biologically derived factors. J Cell Sci 112 (Pt 5): Development 126: 33593370.
601612. Tamamaki N, Yanagawa Y, Tomioka R, Miyazaki J-I, Obata K,
Roelink H, Porter J, Chiang C, Tanabe Y, Chang D, Beachy P, Kaneko T (2003) Green fluorescent protein expression and
Jessell T (1995) Floor plate and motor neuron induction colocalization with calretinin, parvalbumin, and somatostatin
by different concentrations of the amino-terminal cleav- in the GAD67-GFP knock-on mouse. J Comp Neurol 467:
age product of sonic hedgehog autoproteolysis. Cell 81: 6079.
445455. Tanaka A, Kamiakito T, Hakamata Y, Fujii A, Kuriki K,
Ross SE, Greenberg ME, Stiles CD (2003) Basic helix-loop- Fukayama M (2001) Extensive neuronal localization and
helix factors in cortical development. Neuron 39: 1325. neurotrophic function of fibroblast growth factor 8 in the
Roussa E, Krieglstein K (2004) Induction and specification of nervous system. Brain Res 912: 105115.
midbrain dopaminergic cells: focus on SHH, FGF8, and Temple S (2001) The development of neural stem cells. Nature
TGF-beta. Cell Tissue Res 318: 2333. 414: 112117.
Sakai T, Oshima A, Nozaki Y, Ida I, Haga C, Akiyama H, Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA,
Nakazato Y, Mikuni M (2007) Changes in density of Swiergiel JJ, Marshall VS, Jones JM (1998) Embryonic
calcium-binding-protein-immunoreactive GABAergic neu- stem cell lines derived from human blastocysts. Science 282:
rons in prefrontal cortex in schizophrenia and bipolar disor- 11451147.
der. Neuropathology 28: 143150. Tropepe V, Hitoshi S, Sirard C, Mak TW, Rossant J, van der
Samii A, Nutt JG, Ransom BR (2004) Parkinsons disease. Kooy D (2001) Direct neural fate specification from embry-
Lancet 363: 17831793. onic stem cells: a primitive mammalian neural stem cell stage
Sarter M, Bruno JP (2000) Cortical cholinergic inputs mediat- acquired through a default mechanism. Neuron 30: 6578.
ing arousal, attentional processing and dreaming: differential Vercelli A, Mereuta OM, Garbossa D, Muraca G, Mareschi K,
afferent regulation of the basal forebrain by telencephalic and Rustichelli D, Ferrero I, Mazzini L, Madon E, Fagioli F
brainstem afferents. Neuroscience 95: 933952. (2008) Human mesenchymal stem cell transplantation
114 N. Ammon et al.

extends survival, improves motor performance and decreases Bilateral fetal mesencephalic grafting in two patients
neuroinflammation in mouse model of amyotrophic lateral with parkinsonism induced by 1-methyl-4-phenyl-1,2,3,6-
sclerosis. Neurobiol Dis 31: 395405. tetrahydropyridine (MPTP). N Engl J Med 327: 15561563.
Walker FO (2007) Huntingtons disease. Lancet 369: 218228. Wilson L, Maden M (2005) The mechanisms of dorsoven-
Wang Q, Matsumoto Y, Shindo T, Miyake K, Shindo A, tral patterning in the vertebrate neural tube. Dev Biol 282:
Kawanishi M, Kawai N, Tamiya T, Nagao S (2006) Neural 113.
stem cells transplantation in cortex in a mouse model of Wilson SI, Rydstrom A, Trimborn T, Willert K, Nusse R, Jessell
Alzheimers disease. J Med Invest 53: 6169. TM, Edlund T (2001) The status of Wnt signalling regulates
Watanabe K, Kamiya D, Nishiyama A, Katayama T, Nozaki S, neural and epidermal fates in the chick embryo. Nature 411:
Kawasaki H, Watanabe Y, Mizuseki K, Sasai Y (2005) 325330.
Directed differentiation of telencephalic precursors from Wonders CP, Anderson SA (2006) The origin and spec-
embryonic stem cells. Nat Neurosci 8: 288296. ification of cortical interneurons. Nat Rev Neurosci 7:
Wenner P, ODonovan MJ, Matise MP (2000) Topographical 687695.
and physiological characterization of interneurons that Xu Q, Cobos I, Cruz EDL, Rubenstein JL, Anderson SA (2004)
express engrailed-1 in the embryonic chick spinal cord. J Origins of cortical interneuron subtypes. J Neurosci 24:
Neurophysiol 84: 26512657. 26122622.
Wernig M, Benninger F, Schmandt T, Rade M, Tucker KL, Ye W, Shimamura K, Rubenstein JL, Hynes MA, Rosenthal A
Bussow H, Beck H, Brustle O (2004) Functional integration (1998) FGF and Shh signals control dopaminergic and sero-
of embryonic stem cell-derived neurons in vivo. J Neurosci tonergic cell fate in the anterior neural plate. Cell 93:
24: 52585268. 755766.
Westmoreland JJ, Hancock CR, Condie BG (2001) Neuronal Ying QL, Stavridis M, Griffiths D, Li M, Smith A (2003)
development of embryonic stem cells: a model of Conversion of embryonic stem cells into neuroectodermal
GABAergic neuron differentiation. Biochem Biophys Res precursors in adherent monoculture. Nat Biotechnol 21:
Commun 284: 674680. 183186.
Wichterle H, Lieberam I, Porter JA, Jessell TM (2002) Directed Zhao Y, Marin O, Hermesz E, Powell A, Flames N, Palkovits M,
differentiation of embryonic stem cells into motor neurons. Rubenstein JL, Westphal H (2003) The LIM-homeobox gene
Cell 110: 385397. Lhx8 is required for the development of many cholinergic
Widner H, Tetrud J, Rehncrona S, Snow B, Brundin P, neurons in the mouse forebrain. Proc Natl Acad Sci U S A
Gustavii B, Bjorklund A, Lindvall O, Langston JW (1992) 100: 90059010.
Chapter 9

Neurotransmitters as Main Players in the Neural


Differentiation and Fate Determination Game

Katia K. Yuahasi, Katia N. Gomes, Marcelo Campos, Arthur A. Nery, Ariane


Nunes-Alves, Cleber A. Trujillo, and Henning Ulrich

Contents (peaks or waves) leads to initiation of a calcium-


dependent gene expression program essential for the
9.1 Introduction . . . . . . . . . . . . . . . . . 116 progress to the next differentiation stage. We dis-
9.2 An Overview of Neurogenesis . . . . . . . . 116 cuss the evidence pointing to participation of GABA,
9.3 Models of Neuronal Differentiation . . . . . . 118 glutamate, cholinergic and purinergic receptors in neu-
9.3.1 Mesenchymal Stem Cells (MSC) . . . . 118
ronal differentiation of various stem and progenitor
9.3.2 Neural Stem Cells (NSC) . . . . . . . . 119
9.3.3 Embryonic Stem (ES) and Embryonal cell models.
Carcinoma (EC) Cells . . . . . . . . . 119
9.4 Participation of Neurotransmitters in Neural Keywords Calcium signalling Neural
Differentiation . . . . . . . . . . . . . . . . 120 differentiation Neurotransmitter Phenotype
9.4.1 -Aminobutyric Acid (GABA) . . . . . 120
fate Stem cells
9.4.2 Acetylcholine . . . . . . . . . . . . . 121
9.4.3 Glutamate . . . . . . . . . . . . . . 122 Abbreviations
9.4.4 Purines . . . . . . . . . . . . . . . . 124
9.5 Calcium Signaling and Neuronal Differentiation 125
9.6 Conclusions . . . . . . . . . . . . . . . . . 128 ADSC adipose-derived stem cells
References . . . . . . . . . . . . . . . . . . . . 128 AMPA -amino-3-hydroxy-5-methyl-4-
isoxazolepropionic acid
BMSC bone marrow stromal cells
Abstract The wide phenotypic variety of individ- CICR calcium-induced calcium release
ual cells in the CNS and the enormous complexity of EB embryoid bodies
the network formed between these cells results from [Ca2+ ]i free intracellular calcium concentration
specific development programs which direct differenti- EC cells embryonal carcinoma cells
ation of stem and progenitor cells into numerous types EGF epidermal growth factor
of neurons as well as into astrocytes and oligodendro- ER endoplasmic reticulum
cytes. It is believed that in addition to activation of ES cells embryonic stem cells
FGF fibroblast growth factor
intrinsic genetic programs, extrinsic factors are needed
GABA -aminobutyric acid
for the progress of differentiation and neural pheno-
GluR glutamate receptor(s)
type determination. Recent studies have revealed that
ICM inner cell mass
in addition to classical growth factors, neurotransmit-
IP3 inositol-1,4,5-triphosphate
ters have morphogenic functions. Calcium signaling is MPEP methyl-6-(phenylethynyl)-pyridine
triggered by activation of ion channels or metabotropic mGluR metabotropic glutamate receptor subtype(s)
receptors that codified in a frequency of transients MSC mesenchymal stem cell(s)
nAChR nicotinic acetylcholine receptor(s)
H. Ulrich () NMDA N-methyl-D-aspartate
Departamento de Bioqumica, Instituto de Quimica, NSC Neural stem cells
Universidade de So Paulo, So Paulo, Brazil RA retinoic acid
e-mail: henning@iq.usp.br RyR ryanodine receptor(s)

H. Ulrich (ed.), Perspectives of Stem Cells, 115


DOI 10.1007/978-90-481-3375-8_9, Springer Science+Business Media B.V. 2010
116 K.K. Yuahasi et al.

9.1 Introduction effort has been devoted to characterize intrinsic and


extrinsic factors involved in proliferation and differ-
entiation programs of stem cells and neural progeni-
The development of the central nervous system is
tors. Several soluble and membrane-bound molecules,
one of the most important and complex morpho-
including growth factors, hormones and neurotrans-
genetic events occurring in the embryo. This process
mitters, have been implied in the extrinsic regulation
is the result of the standard events which begins with
of these processes (for a review see Cameron et al.,
proliferation and specification of neural cell identity
1998).
(neuronal or glial), followed by cell migration and mat-
While classic roles of neurotransmitters and their
uration, neurite extension and nerve guidance, synapse
receptors in the adult brain related to synaptic com-
formation, plasticity and network implementation and
munication are well understood, the comprehension
finally concludes with the refinement of synaptic con-
of their functions in the development of the nervous
nections through the elimination of axonal branches
system is only beginning and has been more inten-
or cell death (Frisn et al., 1998). In this way, the
sively investigated during the last years (for a review
mature nervous system arises from few embryonic
see Nguyen et al., 2001). It has become evident that
cells, reaches billions of cells in adult life and forms
in order to achieve its enormous complexity, the devel-
trillions of high precision circuits.
oping neuronal system must rely on cellular interac-
Due to the immense complexity of this system, the
tions and subsequent intracellular signal transduction.
general opinion had been that birth of new neurons or
Neurotransmitters are part of diverse classes of dif-
replacement of lost ones did not exist in the adult cen-
fusible factors capable to activate membrane-bound
tral nervous system, according to the neuronal doctrine
receptors, thereby regulating the progress of neuronal
elaborated by Santiago Ramon y Cajal at the end of
differentiation and determining the final phenotypic
the nineteenth century. For many following years only
fate. In view of that, neurotransmitters function as
a handful of biologists, including Joseph Altman and
morphogens during development.
Michael Kaplan, suggested the existence of adult neu-
In this chapter we will review experimental evi-
rogenesis (Altman and Das, 1965; Kaplan and Hinds,
dence pointing to the participation of neurotransmitter
1977). However, during the last decades, the study
receptors and their agonists in the developing neuronal
of neurogenesis in adult brains of birds (Nottebohm
system as well as in neuronal differentiation of stem
and Goldman, 1983), rodents (Kuhn et al., 1996),
cell models. The selected neurotransmitter receptors
primates (Gould et al., 1999) and finally in humans
whose functions will be discussed are -aminobutyric-
(Eriksson et al., 1998) overturned this ancient dogma
acid (GABA), glutamate, acetylcholine and purinergic
of neuroscience.
receptors. Without any doubts, the selected recep-
The discovery of neurogenesis in some regions of
tors and ligands are components of diverse classes
the adult brain, in the subgranular zone of the dentate
of interconnected extrinsic factors responsible for the
gyrus and the earlier part of the subventricular zone
immense complexity of brain development.
(Ming and Song, 2005), and advances in the study of
embryonic and adult stem cells brought new insights
regarding the use of these cells for the treatment of neu-
rological injuries of traumatic or degenerative nature.
However, although the capacity of stem cells of gen- 9.2 An Overview of Neurogenesis
erating new neurons has raised numerous expectations
in therapeutic intervention, the complex mechanisms The diversity of neural cells in embryonic development
underlying proliferation and differentiation of stem arises from proliferative regions in the neural tube.
cells, such as the elucidation of the factors involved New neurons are continuously added to the neural cir-
in the establishment of the individual neuronal phe- cuits in two restricted areas of the adult mammalian
notypes, need yet to be consolidated. Recent studies brain: the subventricular zone and the hippocampal
indicate that the final neuronal fate can be experi- subgranular zone. Outside of these two regions, prolif-
mentally determined by in vitro manipulation of the erating cells give rise to new glia but not to neurons
environment of stem cell cultures. Therefore, much in the adult CNS. The molecular mechanisms and
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 117

the cross-talk between multiple signaling pathways, involves a return to the symmetric division of progeni-
transcription factors and cellcell interactions control- tors, controlled by intrinsic and extrinsic factors.
ling cell fate have lately attracted much attention. Glial cells and in particular radial glia are now con-
Many studies revealed interactions among astrocytes, sidered as a source of neuronal progenitors (Song et al.,
radial glial cells and neural progenitor cells. These 2002). According to Doetsch (2003b), radial glial cells
interactions can promote and control neurogenesis are a widespread non-neuronal cell type in the devel-
and gliogenesis of resident precursor cells in their oping CNS, appearing at the onset of neurogenesis
niches (Doetsch, 2003a). The mechanisms underlying and giving rise, directly or through progenitor forma-
these remarkable changes in progenitor behavior and tion, to most neurons of the cortex. Radial glial cells,
fate during development are not understood, but are defined by their radial morphology and glial properties,
thought to include both changes in the intrinsic prop- appear during early brain development where they sup-
erties of neural progenitors, as well as changes in their port migrating neurons (Levitt and Rakic, 1980). In the
signaling environment (Guillemot, 2007). embryonic telencephalon, radial glial cells reach the
The direction of cell fates into neurons or glia is inner ventricular zone, extend radial process at the pial
also related to symmetric and asymmetric cell divi- surfaces of the neural tube and divide at the ventricular
sion, characterized by equal and unequal cytoplasmic surface (Cameron and Rakic, 1991). Gtz and Huttner
distributions to progeny. Symmetric or proliferative (2005) have published a comprehensive review of this
cell division results in a rapid expansion of stem cells complex mechanism of neurogenesis.
and progenitors. Neurogenesis starts by changing from At this point, with these recent findings that glia
symmetric to asymmetric and neurogenic division, can act as neural stem cells (Goldman, 2003), we
whereby stem cells produce another stem cell and a are experiencing one of the biggest revolutions in
neuron or a neural progenitor. Asymmetric division the field of neural stem cell research. The traditional
is important for the generation of cellular diversity in opinion was that neuroepithelial cells from the early
the mammalian cortex (Fishell and Kriegstein, 2003; neural tube generate two separate groups of glial and
Miyata et al., 2004). The transition to gliogenesis neuronal progenitors. The current accepted view of

Fig. 9.1 Novel hypothesis on the lineage of neural stem progenitors. According to the oak tree model, neuroepithelial
cells. In 2001, Alvarez-Buylla and coworkers proposed a uni- cells originate radial glia which then differentiates into astro-
fied hypothesis based on oak tree model of the neural stem cells cytes. Radial glia can divide asymmetrically to produce neurons
lineage, abandoning the classical pine tree model. According and glia and might produce these cells directly or through transit
to the pine tree model, neuroepithelial cells in the neural tube amplifying cells
generate two separate groups of committed glial and neuronal
118 K.K. Yuahasi et al.

Fig. 9.2 In vitro models


for the study of
neurogenesis. P19 embryonal
carcinoma, embryonic stem,
neural progenitor and
mesenchymal stem cells have
the capacity to differentiate in
vitro into neural phenotypes.
Neurotransmitter receptors are
functional along the process
of differentiation

neurogenesis proposes that neuroepithelial cells at the in vitro stem cell models were established to study
base of the lineage develop into radial glia and then the actions of these factors in a simplified environ-
into astrocytes. Neurons and glia are produced by ment. This chapter will focus on mesenchymal stem
asymmetric division of radial glial cells, which occurs cells (MSC), neural stem cells (NSC), embryonic stem
directly or through intermediated precursors. Radial (ES) and embryonal carcinoma (EC) cells as models
glial cells disappear after birth in mammals, when for neuronal differentiation (Fig. 9.2).
they are thought to transform into astrocytes (Fig. 9.1)
(Alvarez-Buylla et al., 2001).
Even with recent data providing evidence that neu-
ral stem cells have some characteristics of glia, it is
extremely improbable that all glial cells are neural 9.3.1 Mesenchymal Stem Cells (MSC)
stem cells. The quest of highly efficient cell-specific
markers and isolation methods together with investiga- MSC, recognized as adherent stromal cells, are derived
tions into the mechanisms of extrinsic factors, such as from embryonic mesoderm. The existence of stem
neurotransmitters, growth factors and their receptors, cells for non-hematopoietic cells in bone marrow
and induction of intracellular calcium transients con- was proposed over 100 years ago, but the isola-
tributing to neurogenesis promises to reveal how these tion and differentiation of marrow stromal cells into
cells decide their fates. osteoblasts, chondroblasts, adipocytes and myoblasts
was only recently demonstrated (reviewed by Prockop,
1997). Non-hematopoietic precursors from bone
marrow stroma have been referred to as colony-
9.3 Models of Neuronal Differentiation forming-unit fibroblasts, MSC or bone marrow stro-
mal cells (BMSC). Although BMSC can naturally be
Understanding the mechanism controlling neuronal expected to be a source of surrounding tissue of bone,
differentiation and behavior has raised much interest cartilage, and fat, several recent reports demonstrate
over recent years. A large variety of extrinsic and that these cells, under specific experimental conditions,
intrinsic factors are being characterized regarding their can differentiate into muscle, glia and hepatocytes
functions during neuronal development. However, due (Ferrari et al., 1998; Azizi et al., 1998; Peterson et al.,
to the enormous complexity of such studies in vivo, 1999; Sanchez-Ramos et al., 2000).
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 119

Cultures of mesenchymal stem cells have also been neurons. For instance, astrocytes (one of three cell
studied for capacity of differentiation into cell types of types obtained during neurosphere differentiation) pro-
non-mesenchymal origin, including neurons (reviewed mote formation and function of excitatory synapses
by Jackson et al., 2007). This trans-differentiation (Haber et al., 2006).
potential has not only been observed for bone-marrow
stem cells but also for adipose-derived stem cells
9.3.3 Embryonic Stem (ES) and Embryonal
(ADSC). These cells are characterized by their capa-
bility to differentiate into adipocytes, osteoblasts, Carcinoma (EC) Cells
myocytes, chondrocytes, endothelial cells and car-
diomyocytes (Zuk et al., 2002; Gimble et al., 2007). Pluripotent embryonic carcinoma (EC) and embry-
Cultures of ADSC can also be induced to form neu- onic stem (ES) cells possess characteristics of early
rospheres (Kang et al., 2004; Nagase et al., 2007) and embryonic cells (Kahan and Ephrussi, 1970; Evans and
further differentiate into cells with neuronal morphol- Kaufman, 1981; Martin, 1981). ES and EC cells are
ogy or even Schwann cells (Safford et al., 2002; Xu able to undergo unlimited self-renewal and differenti-
et al., 2008). Intracerebral transplantation of human ation into the three primary germ layers (endoderm,
ADSC improved neurological deficits after cerebral mesoderm, and ectoderm) of the embryo. EC cells
ischemia in rats (Kang et al., 2003). In fact, ADSC may being part of teratocarcinomas (malignant germ cell
be a better source for cell therapy as bone-marrow stem tumors) were used for decades as in vitro models for
cells as they can be easily obtained in large amounts by developmental processes. The in vivo counterpart of
liposuction from subcutaneous adipose tissue (Franco EC cells are ES cells, the inner cell mass (ICM) of the
Lambert et al., 2009). blastocyst stage. These cells originate all cells of the
adult body. Mouse EC cells are similar to cells of the
ICM (Gachelin et al., 1977; Solter and Knowles, 1978)
and contribute to differentiation into somatic cell types
9.3.2 Neural Stem Cells (NSC) when injected into the ICM. Thus, EC and ES cells
are a versatile in vitro tool for understanding molecular
Since they were first described by Reynolds and Weiss and cellular controls in early mammalian neurogenesis
(1992), neural stem and progenitor cells have been the (Tropepe et al., 2001; Wichterle et al., 2002; Mizuseki
focus of growing attention due to their potential in the et al., 2003; Ying et al., 2003; Watanabe et al., 2005).
study of neurogenesis and development of future ther- A widely used model for in vitro differentiation is
apy in neurological disorders. In this study, Reynolds the P19 mouse embryonic carcinoma cell line, which
and Weiss isolated striatum cells from adult mouse can be induced to neuroectodermal differentiation by
brain and induced them to proliferate as free-floating culturing free floating embryoid bodies (EB) in serum-
spherical expansion (neurospheres). Neurospheres are free defined medium containing retinoic acid (RA)
clonal expansions of self-renewing neural progenitor (McBurney et al., 1982; Jones-Villeneuve et al., 1982).
cells that form tridimensional cellular aggregates in The obtained differentiated phenotypes are neurons,
the presence of epidermal growth factor (EGF) and astrocytes, oligodendrocytes and fibroblast-like cells
fibroblast growth factor (FGF)-2 (Tropepe et al., 1999; (Jones-Villeneuve et al., 1982). Like EC cells, ES cells
Ostenfeld and Svendsen, 2004; Kelly et al., 2005). can be induced to differentiation into neural pheno-
In their undifferentiated stage, cells originally express types in the presence of RA and by culturing them
nestin (stemness marker) and following attachment as floating or hanging drop cell suspension (Guan
to culture dishes, they change their morphology and et al., 2001). Mechanisms of RA-triggered neural dif-
antigenic properties to those of neurons and astro- ferentiation of ES and EC cells are largely unknown.
cytes. Such cells keep their potential differentiation Nevertheless, gene expression of several transcription
into neurons, astrocytes and oligodendrocytes both factors and signaling molecules such as sonic hedge-
in vitro and in vivo following transplantation into hog, transcription factors Pax6 and Mash1 and the
developing or adult brain. It is believed that the inter- signaling molecule Wnt-1 was induced in ES cells in
action between multiple cell types occurring during the presence of RA. The transcription factors Neuro D,
neurosphere differentiation facilitates maturation of Math1 and NSCL2 were expressed in differentiating
120 K.K. Yuahasi et al.

mouse EC cells following RA application (reviewed 9.4.1 -Aminobutyric Acid (GABA)


by Guan et al., 2001).
-aminobutyric-acid (GABA) is the main fast-
acting inhibitory neurotransmitter in the adult cortex
9.4 Participation of Neurotransmitters (Connors et al., 1988; Krnjevic and Schwartz, 1967),
in Neural Differentiation and it is also one of the most abundant neurotrans-
mitters detected in mammalian brain development
(Cicirata et al., 1991; Miranda-Contreras et al., 1998,
Neurotransmitters and their receptors have well char-
1999, 2000; Nguyen et al., 2001). GABA receptors
acterized functions in neuromodulation, but their par-
are divided into ionotropic GABAA and GABAC sub-
ticipation in directing neural differentiation has only
types and metabotropic GABAB receptors (reviewed
recently been studied. Experimental evidence strongly
by Chebib and Johnston, 1999). The abundance of
suggests that neurotransmitter receptors and their ago-
GABA and its receptors in the early developing rodent
nists could behave as growth regulators during neu-
CNS has raised speculations regarding the role of
ronal differentiation and development (Brezun and
this transmitter in immature neural cell proliferation,
Daszuta, 2000; Butler et al., 1999; Cameron et al.,
migration, differentiation and survival. For instance,
1998; Fiszman et al., 1999; Haydar et al., 2000; Lauder
immunoreactivity for GABA together with glutamate
et al., 1998; LoTurco et al., 1995; Ma et al., 1998,
was detected in the developing cortical marginal zone
2000; Wang et al., 1996; Weiss et al., 1998). As
and subplate cells as examples of transient popula-
we shall discuss, neurotransmitters and their recep-
tions in the developing neuronal system (Del Rio
tors are expressed throughout development where they
et al., 1992). Moreover, GABA was present at the
control cellular processes such as metabolism, pro-
beginning of hypothalamus development (Van den Pol,
liferation, cell survival, migration and differentiation.
1997) pointing to functions of this neurotransmitter in
With on-going neuronal differentiation and matura-
neurogenesis of several regions prior to formation of
tion completed by the refinement of synaptic contacts,
synaptic interactions.
neurotransmitters take over new functions as medi-
Nowadays, it is well established that GABA exerts
ators of synaptic communication (Buznikov et al.,
trophic actions during neural development which may
1996). Since many neurotransmitters and hormones
involve stimulation of all three types of GABA recep-
are known to induce signaling cascades in progeni-
tors (reviewed by Barker et al., 1998; Lauder et al.,
tors and stem cells, a major question remains how
1998). These findings have initiated further studies in
these multiple external signals cooperate throughout
order to elucidate GABA release and GABA-receptor
development of the neuronal system into its immense
interactions during neuronal differentiation of embry-
diversity. Nicolas Spitzer and co-workers have shown
onic and adult stem cells into specific neural phe-
ion channel and receptor activation codes for a spe-
notypes. In young neurons and neuronal precursors,
cific follow-up of [Ca2+ ]i transients as a frequency of
GABA is an excitatory neurotransmitter, becoming
calcium waves that trigger the progress of neural dif-
inhibitory only later (Ben-Ari et al., 1989; Owens
ferentiation (reviewed by Spitzer et al., 2004). This
et al., 1996; Rivera et al., 1999). GABAA receptor sub-
phenotypic transition depends on activation of neuron-
unit expression is differentially regulated during brain
specific gene expression programs at defined check
development, with each subunit exhibiting a unique
points along differentiation.
regional and temporal developmental expression pro-
However, most of the available data of neurotrans-
file. GABAA receptor activity also acts facilitating
mitters and their receptors role in neuronal differenti-
ciliary neurotrophic factor-induced astrocyte differen-
ation as morphogens, by paracrine or autocrine mech-
tiation of neural progenitors isolated from fetal rat
anisms, have been obtained using simplified in vitro
brain (Yoneyama et al., 2007). Although the signifi-
systems. This limitation arises from the complexity of
cance of the differential expression of GABAA recep-
in vivo development, and therefore the participation
tor subunits is not completely understood, it seems that
of neurotransmitter receptors during animal develop-
subunit switching in certain brain regions is essential
ment can only be studied in transgenic and knock-out
for normal development (Culiat et al., 1994; Gnther
animals or by using pharmacological approaches.
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 121

et al., 1995). Furthermore, GABAB receptors have division (Vaca, 1988). The release of acetylcholine
been recently related to modulation of proliferation through growth cones of migrating cholinergic neu-
and differentiation of neural progenitor cells isolated rons suggests trophic roles for this neurotransmitter in
from fetal mouse brain (Fukui et al., 2008). establishing correct synaptic connections. Consistent
In the same way, the differentiation of embry- with this, choline acetyltransferase immunoreactivity
onic or adult tissue-specific stem cells is regulated by was detectable in pre-migratory or early migratory
a microenvironment of interactions between several neurons of rat spinal cord (Phelps et al., 1990). Subunit
neurotransmitters, growth factors and others trophic expression and functional properties of nicotinic recep-
factors defining a specific neuronal phenotype. Cells tors change during neural differentiation and matura-
with neuronal-like morphology obtained by in vitro tion. In this context, Schneider and coworkers (2002)
differentiation of embryonic or mesenchymal stem provided proof for functional nAChR in fetal mouse
cells express functional GABA receptors. Moreover, cerebral cortex as early as on E10. Transient eleva-
GABA-evoked receptor activity increases during pro- tion of intracellular calcium concentration ([Ca2+ ]i )
gressing differentiation of mouse neurospheres, sug- in response to application of nicotinic agonists were
gesting that functional properties of neurons derived markedly prolonged in cells from early embryonic
from fetal mouse neurospheres are compatible with stages when compared to cells from later stages of
those of neuronal precursors in vivo (Pagani et al., development. The effects of nicotine on developing
2006). neurons have been intensively studied during hip-
pocampal formation which is characterized by high
expression of 7-subtype nAChR (Adams et al., 2002).
Due to the high permeability of this ion channel for
9.4.2 Acetylcholine calcium ions, high nicotine concentrations may be
cytotoxic and therefore reduce neurogenesis.
Acetylcholine receptors are classified into the two The participation of nicotinic and muscarinic recep-
classes of ionotropic nicotinic and metabotropic mus- tors in neurogenesis has also been studied in simplified
carinic receptors based on structural and pharmaco- systems suggesting that this neurotransmitter can act
logical properties. Neuronal nicotinic receptors are as morphogen alone or in combination with other
assembled by five homo-or heteromeric receptor sub- extrinsic factors. For instance, mouse ES and P19 EC
units (2-10 and 2-4), while muscarinic subtypes cells synthesize acetylcholine (Paraoanu et al., 2007;
are comprised by five metabotropic receptors coupling Parnas and Lilian, 1995) and the expression of the
to Gq/11 (M1, M3 and M5 receptors) or Gi/o pro- nicotinic and muscarinic receptor subtypes changes
teins (M2, M4). While M1-M4 subtypes are found during RA-induced neuronal differentiation of P19
throughout all tissues, M5 types are only expressed in cells (Resende et al., 2008a, b). Embryonic P19 cells
the neuronal system. Nicotinic acetylcholine receptors already expressed various nicotinic receptor subunits
(nAChR) are widely distributed all over the mam- including the 7 type and responded to nicotine appli-
malian CNS (Quik et al., 2000; Zoli et al., 1995) with cation with transient elevations of free intracellular
high expression levels in adult and developing human calcium concentration ([Ca2+ ]i ). Nicotinic receptor
brain (Court et al., 1995, 1997; Gotti et al., 1997; subunits were differentially expressed along neuronal
Kinney et al., 1993; Paterson and Nordberg, 2000). The differentiation of P19 EC to neurons and increases
activity of these ionotropic receptors contributes to a in nicotinic agonist-induced [Ca2+ ]i responses were
wide range of brain processes from cognitive functions noted when cells acquired neuronal phenotypes. This
such as learning, memory formation or reward (Levin, increased response to nicotine resulted from higher
1992) to cellular events such as neurodegeneration overall nAChR expression and activation of ryanodine-
(Zoli et al., 1999) and neural development (Coronas sensitive intracellular calcium release and voltage-
et al., 2000; Lauder, 1993). gated calcium channels (Resende et al., 2008a).
Nicotinic receptor subunits are among the first Nicotinic receptor stimulation inhibited the prolifer-
membrane proteins to appear during CNS devel- ation of embryonic P19 EC cells in a dose-dependent
opment. Synthesis of acetylcholine initiates dur- manner. P19 neural progenitor cells following 4 days
ing motoneuron development upon cessation of cell of induction to differentiation by RA responded to
122 K.K. Yuahasi et al.

nicotine application with proliferation stimulation and roles in sensory dorsal root ganglion development.
augmented differentiation to neurons as judged by Coordination of neuronal and glial differentiation,
increased expression of high molecular mass neurofil- including Schwann cell formation, is also believed to
ament. Activation of ryanodine-sensitive intracellular involve cholinergic receptor activity. Biagioni and co-
calcium release has been suggested as a possible mech- workers discuss in this context that neuron-produced
anism for nicotine-promoted proliferation stimulation acetylcholine acts on muscarinic receptors expressed
in neural progenitors. This mechanism, also known by Schwann cells, thereby inducing transient changes
as calcium-induced calcium release (CICR), was not in [Ca2+ ]i . Schwann cells participate in regulation of
present in embryonic P19 cells, in agreement with the neuronal differentiation and axon outgrowth (Biagioni
inability of nicotine to stimulate proliferation (Resende et al., 2000). The example of acetylcholine synthe-
et al., 2008b). sis in sensory neurons shows that cholinergic receptor
Similar mechanisms, which also involve liberation function is important for development of neuronal-glial
of intracellular calcium, have been shown for mus- networks.
carinic receptor induced proliferation and differenti-
ation in P19 cells. In contrast to nAChR, muscarinic
receptors were not functional in embryonic P19 cells. 9.4.3 Glutamate
In progenitor cells, muscarine induced proliferation
by activation of Gq/11 -protein coupled M1, M3 and Glutamate is the main mediator of excitatory neu-
M5 receptors and intracellular calcium stores whereas rotransmisson in the adult human brain (Gass and
Gi/o -protein coupled M2 receptor activity mediated Olive, 2008) and can be found in several areas of the
neuronal differentiation (Resende et al., 2008b). mouse CNS during neurogenesis (Miranda-Contreras
Functions of the cholinergic system have also been et al., 1998, 1999, 2000). Glutamatergic receptors are
revealed in other cell types, including neuroblastoma classified by their structural properties into ionotropic
cells used as model of differentiation into choliner- and metabotropic receptors. There are eight types of
gic phenotypes and sensory neurons which express metabotropic receptors: mGluR1-8. Ionotropic recep-
components of the cholinergic system during early tors are subdivided into three families: -amino-3-
development (Biagoni et al., 2000). Neurite outgrowth hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)
induced in neuroblastoma cells by RA or dibutyril- receptors, N-methyl-D-aspartic acid (NMDA) recep-
cAMP depended on the capability of the cells to tors and kainate receptors (Pin and Duvoisin, 1995).
synthesize acetylcholine. Besides the impaired acetyl- Several studies have already confirmed the role
choline synthesis of the mouse neuroblastoma clone of glutamatergic receptors in regulating prolifera-
N18TG2, this cell line was unable to establish synaptic tion during neurogenesis where glutamate may have
contacts. Following hybridization of this neuroblas- proliferation-inducing or inhibiting effects depending
toma cell with a glial component, the generated neu- on the developmental status and localization of glu-
roblastoma X glioma hybrid cell line showed a marked tamate action. Application of glutamate or GABA
increase in acetylcholine synthesis and was also capa- to organotypic slice cultures stimulated proliferation
ble to proceed with neurite outgrowth (Hamprecht, in the ventricular zone by shortening the cell cycle,
1977). As further confirmation for the importance whereas proliferation in the subventricular zone was
of acetylcholine production during differentiation of decreased (Haydar et al., 2000). Based on these results,
neuroblastoma cells, N18TG2 cells could be stimu- the authors concluded that relative contributions of the
lated to fiber outgrowth following transfection with subventricular zone to neocortical growth may be regu-
cDNAs coding for choline acetyltransferase and gene lated by differential effects of glutamate and GABA on
expression of synapsin I (Bignami et al., 1997). As proliferation. Proliferation stimulating effects of glu-
a mechanism for differentiation induction, the group tamate in E12-22 rat striatum neuroblasts were inhib-
of Augusti-Tocco suggested an autocrine loop by ited in the presence of N-methyl-D-aspartate (NMDA)
which muscarinic receptor activity stimulates acetyl- receptor antagonists (Sadikot et al., 1998). Activation
choline synthesis and neuron-specific gene expres- of glutamate receptors in cortical oligodendrocyte
sion (Augusto-Tocco et al., 2006). Acetylcholine and progenitor (O-2A) cells suppressed proliferation and
muscarinic receptor activity were attributed to play blocked lineage progression from the O-2A stage to the
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 123

pro-oligodendroblast stage (Gallo et al., 1996) suggest- in expression levels and responsiveness of such recep-
ing that glutamate also participated in the regulation of tors in developing neurons. Finally, non-NMDA-type
glial cell differentiation. ionotropic and metabotropic glutamate receptors may
Roles for glutamate receptors during neurogene- modulate effects of NMDA receptor activation on
sis include cell survival and migration processes. For neurogenesis (reviewed by Nacher and McEwen,
instance, activation of NMDA- or kainate receptors 2006).
enhanced survival of immature granule neurons of The participation of glutamatergic system has also
P7-P8 rat cerebellum (Balazs et al., 1988, 1990). In been studied during in vitro neuronal differentiation
agreement, blockade of NMDA receptors resulted in using P19 EC and ES cells. The concentration of
increased apoptotic elimination of granule neurons in beta-citryl-L-glutamate, catalyzing the formation of
P7-P11 rat cerebellum (Monti and Contestabile, 2000), L-glutamate, increased (Narahara et al., 2000), and
augmented cell death within the dentate gyrus of many glutamatergic receptors, such as kainate recep-
neonatal rats (Gould et al., 1994) and inhibited migra- tor subunits (GluR5, GluR6 and GluR7) as well as
tion of O-2A cells in P0 rat neurohypophysial explants the AMPA receptor subunits (GluR2 and GluR3), had
(Wang et al., 1996). Enhancement of NMDA receptor their expression changed during the course of differ-
activity promoted neurite outgrowth of rat cerebellar entiation of P19 EC cells into neurons (Minakami
granule cells (Pearce et al., 1987) and increased the rate et al., 1995; Heck et al., 1997; Lee et al., 2003).
of migration of granule cells of the developing mouse Gene expression of metabotropic glutamate receptors
cerebellum (Komuro and Rakic, 1993). Furthermore, (mGluR2, mGluR4 and mGluR8) were detectable in
increase of endogenous extracellular glutamate con- undifferentiated P19 cells and in all stages of differ-
centration by inhibition of its uptake also augmented entiation. However, expression of mGluR3, mGluR7
cell migration rates. These and subsequent studies sug- and mGluR8 became visible during the course of dif-
gested that glutamate, acting through NMDA receptors ferentiation (Heck et al., 1997). Functional NMDA-,
and subsequent calcium signaling, is a chemoattractive kainate- and AMPA receptors were present follow-
factor for neurons in mouse developing cortex, signal- ing three days of differentiation induction by RA.
ing cells to migrate from the ventricular and subven- Glutamate-induced [Ca2+ ]i transients augmented dur-
tricular zones into the cortical plate (Behar et al., 1999, ing on-going differentiation (Lee et al., 2003; Ulrich
reviewed by Nacher and McEwen, 2006). Glutamate- and Majumder, 2006). There is also evidence of gluta-
induced signaling is essential for dendritic and axonal mates regulatory role during ES cell differentiation.
growth (Mattsson et al., 1988a, b) as well as for For instance, gene expression of the NMDA recep-
the refinement and reorganization of synaptic contacts tor subunits NR1, NR2A and NR2B increased during
(Aruffo et al., 1987; Scheetz and Constantine-Paton, neural differentiation of mouse ES cells (Qu et al.,
1994). 2003). Gene expression of metabotropic mGluR5 was
The effects of glutamatergic signaling on neu- the only among metabotropic glutamate receptors
ral gene expression have been studied in hippocam- detectable in mouse ES cells. Blockade of this recep-
pal development. Glutamate promote neurogenesis tor decreased the expression of Oct-4 and Nanog,
in adult hippocampal precursor cells by augmenting two transcription factors involved in maintaining the
[Ca2+ ]i levels which was blocked by NMDA recep- undifferentiated state of ES cells, and of alkaline phos-
tor antagonists (Deisseroth et al., 2004). In agreement, phatase, a marker of ES cells (Cappuccio et al., 2005).
neurospheres from adult mice hippocampus express- Mouse ES cells differentiated with serum-free
ing NR1, NR2A and NR2B subunits of the NMDA N2 and B27 medium expressed mGluR5 through-
receptor were induced to augmented gene expression out the entire differentiation period. Blockade of
of c-fos and c-jun, and neuronal differentiation by sus- the metabotropic glutamatergic subtype mGluR5
tained exposure to NMDA (Kitayama et al., 2004). by methyl-6-(phenylethynyl)-pyridine (MPEP) dur-
However, in dentate gyrus of one week old rats inhi- ing differentiation of ES cells into neuron-like cells
bition of NMDA receptors led to an increase in neu- resulted in an increase of 3-tubulin-positive cells
rogenesis (Poulsen et al., 2005). These contradictory with GABAergic phenotype. At the end of differ-
findings of glutamate-induced NMDA receptor activ- entiation, percentages of cells expressing 3-tubulin
ity on neurogenesis could be in part due to differences were identical when cells had been differentiated in
124 K.K. Yuahasi et al.

the presence or absence of MPEP. These data sug- families: ionotropic, ligand-gated ion channel recep-
gest that metabotropic glutamate receptor blockade tors assembled by P2X1,2,3,4,5,6,7 subunits and the
accelerated differentiation of ES cells into neuron-like metabotropic P2Y family of G protein-coupled recep-
cells (Sarichelou et al., 2008). ES cells differentiat- tors consisting of P2Y1,2,4,6,11,12,13,14 subtypes
ing into embryoid bodies (EB) progressively stopped (reviewed by North, 2002).
expressing mGluR5 but augmented mGluR4 gene ATP is released by a variety of cell types, includ-
and protein expression. Activation of mGluR4 dur- ing neurons, glia, endothelium and blood cells.
ing neural induction of ES cells with RA or medium ATP acts as a primary neurotransmitter or as a
supplemented with insulin, transferrin, selenium and co-transmitter with glutamate, acetylcholine, nora-
fibronectin increased the expression of the early neu- drenaline, 5-hydroxytryptamine, dopamine or GABA.
ral markers Dlx-2 and nestin (Cappuccio et al., 2006), In the synaptic cleft, ATP is enzymatically con-
and differentiation into neurons with a GABAergic verted to ADP, AMP and adenosine (Burnstock, 2004).
phenotype (Sarichelou et al., 2008). This work also Adenosine release is observed in neurons, astrocytes
demonstrated that subtype-selective glutamate recep- and microglia. It is known to exert neuroprotective
tor ligands might be used for in vitro production of function by protecting against excessive glutamate
GABAergic neurons from ES cell cultures. and P2X7 receptor mediated calcium fluxes (Schubert
Depending on the activated subtype (mGluR4 or and Kreutzberg, 1993; Dona et al., 2009). Adenosine
mGluR5), metabotropic glutamate receptors either reverts deleterious membrane depolarization by acti-
stimulated or inhibited the progress of neuronal differ- vating Go protein-coupled A1 receptors followed by
entiation. Activation of mGluR5 favored self-renewal augmented K+ conductance and opening of chlo-
of ES cells and maintenance of the cells in their undif- ride channels (reviewed by Schubert and Kreutzberg,
ferentiated state, whereas differentiation of ES cells 1993). However, it seems that adenosine negatively
into embryoid bodies was associated with mGluR4 interferes with the progress of neuronal development.
activation (Melchiorri et al., 2007). Consequently, A1 subtype gene expression and receptor concentra-
mGluR5 expression with inhibitory action on neuro- tions in the brains of prenatal rodents are consid-
genesis decreased following induction to differentia- erably less than postnatal levels. In addition, during
tion, while mGluR4 expression was enhanced during neonatal period A1 receptors are expressed on axons
on-going differentiation. and growth cones and their activation interferes with
axon and white matter formation. Treatment with an
A1 receptor agonist led to axonal loss and reduced
9.4.4 Purines expression of myelin basic protein in treated animals
(Turner et al., 2002). In agreement, A1 receptor activa-
In the 1970s, the first direct evidence was presented tion also resulted in inhibition of nerve-growth factor
for ATP action as a fast transmitter. This suggested the (NGF)-induced neurite outgrowth in PC12 pheochro-
concept of purinergic nerves and purinergic neuro- mocytoma cells and cultured cortical and hippocampal
transmission (Burnstock, 1972, 1978). Following an neurons (Thevananther et al., 2001). Early develop-
initially slow acceptance, purinergic signaling in neu- mental expression and subsequent alterations in the
rotransmission and neuronal development is a rapidly pattern of A2a adenosine receptors within the rat stria-
expanding field. However, due to the absence of spe- tum also suggested functions for this P1 receptor in
cific agonists and antagonists for many purinergic differentiation and migration events (Weaver, 1993).
receptor subtypes, most cellular functions of these Several studies also indicated functions of puriner-
receptors are not yet understood. gic signaling by P2 (P2X and P2Y) receptors during
Two types of purinoreceptors (P1 and P2) are development. Metabotropic P2Y1, P2Y2, P2Y4 and
distinguished based on their ligand specificity for P2Y6 subtypes were expressed during rat embryoge-
adenosine or ATP and ADP, respectively. Four sub- nesis, with the P2Y4 receptor being the most abundant
types of P1 adenosine receptors have been cloned one during brain development (Cheung et al., 2003).
(A1 and A3 inhibiting and A2a and A2b stimulating The P2Y1 subtype seems to be crucial in early neu-
adenylyl cyclase, respectively) (reviewed by Fredholm rogenesis including neurulation and initiation of eye
et al., 2001). P2 receptors are divided into two development of frog embryos (Bogdanov et al., 1997;
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 125

Mass et al., 2007) and gastrulation of the chicken EGF- and FGF-2-induced neurosphere proliferation
embryo (Lasberg, 1990). In rat embryos of age E14-18, (Mishra et al., 2006).
propagation of calcium waves through radial glial cells Extracellular purines stimulated the synthesis and
in the proliferative cortical ventricular zone was medi- release of polypeptides with trophic actions (Rathbone
ated by P2Y1 receptors. Disruption of these calcium et al., 1999; Neary et al., 2004), and regulated differ-
waves decreased proliferation of the ventricular zone, entiation and neurite outgrowth in various cell lines
demonstrating the importance of this receptor during (Aono et al., 1990; Gysbers and Rathbone, 1992,
embryonic neurogenesis (Weissman et al., 2004). 1996). However, elucidation of the exact roles of many
Ionotropic purinergic P2X also exert several func- purinergic receptors depends on the development of
tions during brain development. Cheung et al. (2005) subtype agonists and antagonists as well as on down-
detected P2X3 and P2X2 receptors first in the embry- regulation of receptor expression by RNA interference
onic rat CNS on E11 and E14, respectively. P2X7 and in vivo studies with knock-out animals.
receptors were also expressed from E14 onwards, and
the existence of functional P2X7 receptors in the mid-
brain synaptic terminals as well as in synaptosomes
9.5 Calcium Signaling and Neuronal
and axodendritic prolongations of cerebellar gran-
ule cells has also been documented (Miras-Portugal Differentiation
et al., 2003). These receptors mediate apoptotic cell
death by inducing cytotoxicity at high ATP concen- Changes in intracellular calcium concentration pro-
trations released during inflammation or upon tissue vide a signaling mechanism responsible for controlling
trauma (Le Feuvre et al., 2002). Since apoptosis is numerous cellular events, including metabolism, cell
a common event in developing brain, activation of death, proliferation and differentiation. The ability of a
P2X7 receptors could participate in cell death during simple ion such as calcium to acquire crucial functions
neurogenesis. in cell biology emerges from the use of an exten-
Purinergic receptors were also studied in cell sive molecular repertoire of signaling components that
lines. Using P19 embryonal carcinoma cells as an can be assembled in combinations to create signals
in vitro model for early neural development, our with widely different spatial and temporal profiles. Its
laboratory has shown that P2Y1, P2Y2 and P2X4 signaling function is activated when its cytosolic con-
subtypes are important in regulating cell prolifera- centration [Ca2+ ]i is transiently elevated from 10100
tion and differentiation by activating intracellular cal- nM to roughly 1000 nM due to spontaneous events or
cium pathways (Resende et al., 2008c). ATP dose- as result of receptor-activated calcium flux. The coor-
dependently stimulated proliferation of P19 embry- dination of calcium signaling occurs through intrinsic
onic and progenitor cells and accelerated differentia- mechanisms of calcium influx into the cell and lib-
tion as judged by augmented expression of neuron- eration or capture of calcium by intracellular stores
specific proteins. These cellular effects were blocked (reviewed by Berridge et al., 2000). The main internal
when cells were treated with purinergic receptor calcium store is the membrane system of the endoplas-
antagonists or with inhibitors of intracellular inositol- mic reticulum (ER). The ER contains various special-
1,4,5-triphosphate (IP3 )-mediated calcium mobiliza- ized channels for calcium release, of which IP3 and
tion. Moreover, purinergic receptor subtype activity ryanodine receptor (RyR) channels have been stud-
contributed to the acquisition of defined neural phe- ied most extensively. The activity of RyR intracellular
notypes. For instance, inhibition of P2Y1 receptor calcium channels is furthermore influenced by [Ca2+ ]i
activity along differentiation of P19 cells resulted levels, a mechanism known as calcium-induced cal-
in loss of NMDA receptor activity. Furthermore, cium release (Berridge, 1993; Clapham, 1995). The
reduction of cholinergic receptor responses in dif- entry of external calcium occurs through calcium chan-
ferentiated P19 cells was due to inhibition of nel proteins in the plasma membranes activated by
P2Y2 receptor activity during differentiation (Resende different mechanisms: (i) voltage-operated channels
et al., 2007). In agreement with growth stimulation activated by membrane depolarization, (ii) receptor-
effects demonstrated in P19 cells (Resende et al., operated channels opening in response to binding of
2008c), P2Y1 and P2Y2 receptor activation enhanced an extracellular ligand, (iii) store-operated channels
126 K.K. Yuahasi et al.

opening in response to depletion of internal calcium Functional consequences of changes of cal-


stores, (iv) and mechano-sensitive ion channels cium spike frequency during development of the
(reviewed by Berridge et al., 2006). GABAergic phenotype were studied by the group
The cellular mechanism employed for removal of an of Dr. Nicolas Spitzer. Developmental expression
excess of calcium from the cytoplasm is based on acti- of GABA in Xenopus spinal neurons was calcium-
vation of various pumps and exchangers. The plasma dependent and involved mRNA transcription (Gu and
membrane Ca2+ -ATPase pumps (Pozzan et al., 1994) Spitzer, 1995; Spitzer et al., 1993). Increased expres-
as well as Na+ /Ca2+ exchangers remove calcium from sion of glutamic acid decarboxylase, responsible for
the cell into the extracellular space whereas the sarco- GABA synthesis, was observed with rising [Ca2+ ]i
endoplasmic reticulum ATPase pumps return calcium levels. Gene expression of glutamic acid decarboxy-
back into internal stores (Blaustein and Lederer, 1999). lase was down-regulated when neurons were grown
Mitochondria also play an important function in regu- in a calcium-free medium, but augmented follow-
lating cytosolic calcium levels. These organelles have ing imposing specific frequencies of calcium tran-
a low affinity, but high-capacity rapid Ca2+ uniporter sients in cultured neurons (Spitzer et al., 2000). In
that can significantly reduce [Ca2+ ]i levels (Rizzuto agreement with this study, depolarization and sub-
et al., 1993). When the cell returns to the resting state, sequent calcium influx through voltage-operated cal-
a mitochondrial Na+ /Ca2+ exchanger pumps the large cium channels also defined neurotransmitter choice
load of calcium back into the cytoplasm, from which in other systems. Depolarization mediated calcium
it either returns to the ER or is removed from the cell influx during early differentiation of cultured sen-
(Duchen, 1999; Bernadi, 1999). In response to stimuli, sory neurons led to an increased number of tyrosine
both entry and release channels mediate calcium fluxes hydroxylase-immunoreactive cells (Brosenitsch et al.,
into the cytoplasm. Since these channels have short 1998). Accordingly, calcium influx also mediated dif-
open-times, they only introduce brief pulses of calcium ferentiation of chick ciliary ganglion and mouse spinal
concentration elevations that then form a small plume neurons (Nishi and Berg, 1981; Ishida and Deguchi,
around the mouth of the channel before diffusing into 1983). Thus, spikes and waves of cytosolic calcium
the cytoplasm. Thus, calcium signals are usually pre- contribute in an important manner to the phenomena
sented as brief local spikes. In some cases, individual of proliferation and neuronal differentiation (Fig. 9.3).
spikes are sufficient to trigger a cellular response such Ligand-gated ion channels and metabotropic receptors
as neurotransmitter release. (glutamate, purinergic, GABA and cholinergic recep-
When longer periods of signaling are necessary, tors among many others) contribute to the generation
spikes are repeated to originate waves with differ- of [Ca2+ ]i transients (waves and spikes) and signal-
ent frequencies (Berridge et al., 2000; Spitzer et al., ing during development and neuronal fate acquisition.
2000). The cell responds to these stimuli by differential Essential functions for calcium have been shown for
gene expression programs. Wave frequencies and peak neuronal motility (Zheng and Poo, 2007), intercellu-
heights are internally encoded in the localization and lar communication (Braet et al., 2004), cell fate and
activity states. It is possible that calcium signals gener- developmental patterning (Whitaker and Smith, 2008;
ated at the cell membrane cross the nuclear envelope to Leclerc et al., 2006). However, despite their well-
trigger gene expression as shown for hippocampal neu- known functions in the mature CNS, the role of these
rons (Eder and Bading, 2007). However, in other cell receptors in the modulation of the calcium waves and
types, i.e. neuroblastoma cells and primary rat sensory spikes during development, directing differentiation
neurons, the nuclear envelope is thought to insulate the and phenotype acquisition, has been the subject of
nucleus from larger activity-induced cytoplasmic cal- recent studies (Spitzer et al., 1994, 2004; Spitzer and
cium transients (Al-Mohanna et al., 1994). As a final Borodinsky, 2008).
result of the translocation of the calcium-induced sig- Receptor-induced calcium waves are not only
naling into the nucleus by incoming calcium transients important for inducing signal transduction in the stim-
or other signaling factors, down-stream genes are acti- ulated cell. These signals are also essential for com-
vated which implement cellular responses promoting munication between neurons and glial cells in the
differentiation. adult nervous system and the developing brain. Such
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 127

Fig. 9.3 Direction


of proliferation and neural
differentiation by
neurotransmitter-induced
pattern of intracellular
calcium spikes and waves.
The progress of neuronal
differentiation is directed
through the different types
of intracellular calcium
transients (spikes and waves)
induced at defined time points
(check points) during
development. This calcium
signaling then activates
transcription factors
responsible for expression
of ion channels specific
for the next, advanced
differentiation stage. These
channels generate patterns
of intracellular calcium spikes
and waves, which again
promote differentiation
through activation
of calcium-dependent
transcription factors and final
maturation into neurons
expressing specific
neurotransmitters
and receptors

intercellular calcium signals are transient elevations 2004). The transmission of calcium signals as part of
in [Ca2+ ]i , which in analogy to action potentials, are intercellular communication involves paracrine mech-
propagated to neighboring cells. The velocity of sig- anisms by calcium-induced release of neurotransmit-
nal propagation is in the order of tens of micrometers ters into the extracellular space, which then bind to
per second, i.e. two to three orders of magnitude receptors on neighboring cells, as well as activation
slower than action potential propagation (Braet et al., of downstream calcium signaling or diffusion of the
128 K.K. Yuahasi et al.

calcium-mobilizing messenger IP3 (Saez et al., 1989) neuron size and functional neural networks (DiCicco-
or calcium itself, through gap junction channels. Bloom et al., 2006).
The importance of intracellular calcium release in
modulation of proliferation and differentiation has also
been shown in various works. Proliferation and dif-
ferentiation induction of P19 cells by metabotropic 9.6 Conclusions
purinergic and muscarinic activation involved libera-
tion of calcium from intracellular pools, since these A large amount of evidence, mostly derived from
effects were abolished when formation of IP3 was diverse in vitro models for neurogenesis indicate that
inhibited (Resende et al., 2008b, c). Moreover, cal- neurotransmitters and other molecules act on cal-
cium influx from the extracellular space into embry- cium entry into the cell and on intracellular calcium
onic P19 cells led to decreased proliferation activity. fluxes throughout development. The early appearance
However, in P19 progenitor cells nicotinic recep- of these signaling factors and their wide distribution
tor activity promoted proliferation and differentiation throughout development of a variety of animal species
since at this differentiation state nicotinic receptor led to suggest that neurotransmitters act as extrinsic
activation involved activation of intracellular RyR factors that trigger differentiation and define final phe-
calcium-channels as calcium-induced calcium release notypes of these cells. Although the mechanism of
(CICR). Cooperation of L-type calcium channels with how these multiple external signals are integrated and
RyR2 was essential for neuronal differentiation of ES cooperate, is far from being understood, it is clear
cells (Yu et al., 2008). Moreover, pituitary adeny- that neuronal diversity is codified by this system, and
late cyclase-activating peptide ligand/type 1 receptor- that any change to the pattern of calcium signaling at
mediated activation of the phospholipase C-/IP3 - defined time points will interfere with the final out-
dependent signaling pathway promoted proliferation come of the differentiation process. Recent studies
and gliogenesis of mouse cortex neural progenitor cells of mechanisms of developmental related neurological
(Nishimoto et al., 2007). diseases have identified mutations in neurotransmitter
Other neurotransmitters are also important for trig- receptors and other ion channels. More studies may
gering calcium signals during neuronal development. elucidate changes in developmental signaling related
For example, calcium spikes initiated by activation to neurological disorder and identify new targets for
of GABA and metabotropic glutamate receptors in therapeutic intervention. Furthermore, the promising
embryonic cells contribute to the determination of the therapeutic applications of embryonic, neural and mes-
final phenotype such as specification of neurotransmit- enchymal stem cells in regeneration therapy require the
ter expression (Root et al., 2008). In fact, non-synaptic elucidation of the composition of niches favoring cell
(paracrine actions) signaling of GABA presents con- survival and differentiation at the localization of cell
comitant effects which provides the timing and instruc- transplantation. The understanding of the mechanisms
tion for cells to properly differentiate and integrate of how stem cells differentiate into defined species of
into an existing synaptic network (reviewed by Bordey, neurons will also contribute to cell regeneration ther-
2007). apy of neurodegenerative illnesses such as Parkinsons
Molecular mechanisms of developmental neural disease, for which prior in vitro differentiation to a
diseases such as autism are suggested to involve homogeneous neural phenotype is crucial.
changes in calcium signaling. Mutations in several
voltage-operated calcium channels and ligand-gated
ion channels (GABA and NMDA receptors) affecting References
neuronal excitability, calcium signaling and conse-
quently neurogenesis have been associated with autism Adams CE, Broide RS, Chen Y, Winzer-Serhan UH, Henderson
and related diseases (reviewed by Krey and Dolmetsch, TA, Leslie FM, Freedman R (2002) Development of the
2007). Such defects in developmental signal transmis- alpha7 nicotinic cholinergic receptor in rat hippocampal
formation. Brain Res Dev Brain Res 139:175187.
sion could contribute to neuroanatomical alterations
Al-Mohanna FA, Caddy KW, Bolsover SR (1994) The nucleus
observed in patients with autism spectrum disorder is insulated from large cytosolic calcium ion changes. Nature
including augmented cell packing density, decreases in 367:745750.
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 129

Altman J, Das GD (1965) Autoradiographic and histological evi- the neural plate of Xenopus embryos. J Biol Chem 272:
dence of postnatal hippocampal neurogenesis in rats. J Comp 1258312590.
Neurol 124:319335. Bordey A (2007) Enigmatic GABAergic networks in adult
Alvarez-Buylla A, Garcia-Verdugo JM, Tramontin AD (2001) neurogenic zones. Brain Res Rev 53:124134.
An unified hypothesis on the lineage of neural stem cells. Braet K, Cabooter L, Paemeleire K, Leybaert L (2004) Calcium
Nat Rev Neurosci 2:287293. signal communication in the central nervous system. Biol
Aono K, Nakanishi N, Yamada S (1990) Increase in intracellu- Cell 96:7991.
lar Ca2+ level and modulation of nerve growth factor action Brezun JM, Daszuta A (2000) Serotonin may stimulate gran-
on pheochromocytoma PC12h cells by extracellular ATP. ule cell proliferation in the adult hippocampus, as observed
Meikai Daigaku Shigaku Zasshi 19:221229. in rats grafted with foetal raphe neurons. Eur J Neurosci
Aruffo C, Ferszt R, Hildebrandt AG, Crvos-Navarro J (1987) 12:391396.
Low doses of L-monosodium glutamate promote neu- Brosenitsch TA, Salgado-Commissariat D, Kunze DL, Katz DM
ronal growth and differentiation in vitro. Dev Neurosci 9: (1998) A role for L-type calcium channels in developmen-
228239. tal regulation of transmitter phenotype in primary sensory
Augusti-Tocco G, Biagioni S, Tata AM (2006) Acetylcholine neurons. J Neurosci 18:10471055.
and regulation of gene expression in developing systems. Burnstock G (1972) Purinergic nerves. Pharmacol Rev 24:
J Mol Neurosci 30:4548. 509581.
Azizi SA, Stokes D, Augelli BJ, DiGirolamo C, Prockop DJ Burnstock G (1978) A basis for distinguishing two types of
(1998) Engraftment and migration of human bone marrow purinergic receptor. In: Cell Membrane Receptors for Drugs
stromal cells implanted in the brains of albino rats similar- and Hormones: A Multidisciplinary Approach (Straub, RW
ities to astrocyte grafts. Proc Natl Acad Sci USA 95:3908 and Bolis, L, eds), pp. 107118. Raven Press, New York.
3913. Burnstock G (2004) Cotransmission. Curr Opin Pharmacol
Balazs R, Hack N, Jorgensen OS (1990) Selective stimulation 4:4752.
of excitatory amino acid receptor subtypes and the survival Butler AK, Uryu K, Rougon G, Chesselet MF (1999) N-Methyl-
of cerebellar granule cells in culture: effect of kainic acid. Daspartate receptor blockade affects polysialylated neural
Neuroscience 37:251258. cell adhesion molecule expression and synaptic density dur-
Balazs R, Jorgensen OS, Hack N (1988) N-Methyl-D-aspartate ing striatal development. Neuroscience 89:11691181.
promotes the survival of cerebellar granule cells in culture. Buznikov GA, Shmukler YB, Lauder JM (1996) From oocyte
Neuroscience 27:437451. to neuron: do neurotransmitters function in the same
Barker JL, Behar T, Li YX, Liu QY, Ma W, Maric D, Maric I, way throughout development? Cell Mol Neurobiol 16:
Schaffner AE, Serafini R, Smith SV, Somogyi R, Vautrin JY, 537559.
Wen XL, Xian H (1998) GABAergic cells and signals in CNS Cameron HA, Hazel TG, McKay RD (1998) Regulation
development. Perspect Dev Neurobiol 5:305322. of neurogenesis by growth factors and neurotransmitters.
Behar TN, Scott CA, Greene CL, Wen X, Smith SV, Maric J Neurobiol 36:287306.
D, Liu QY, Colton CA, Barker JL (1999) Glutamate acting Cameron RS, Rakic P (1991) Glial cell lineage in the cerebral
at NMDA receptors stimulates embryonic cortical neuronal cortex: a review and synthesis. Glia 4:124137.
migration. J Neurosci 19:44494461. Cappuccio I, Spinsanti P, Porcellini A, Desiderati F, De Vita T,
Ben-Ari Y, Cherubini E, Corradetti R, Gaiarsa JL (1989) Giant Storto M, Capobianco L, Battaglia G, Nicoletti F, Melchiorri
synaptic potentials in immature rat CA3 hippocampal neu- D (2005) Endogenous activation of mGlu5 metabotropic glu-
rones. J Physiol 416:303325. tamate receptors supports self-renewal of cultured mouse
Bernadi P (1999) Mitochondrial transport of cations: chan- embryonic stem cells. Neuropharmacology 49(Suppl 1):
nels, exchangers, and permeability transition. Physiol Rev 196205.
79:11271155. Cappuccio I, Verani R, Spinsanti P, Niccolini C, Gradini R,
Berridge MJ (1993) Inositol trisphosphate and calcium sig- Costantino S, Nicoletti F, Melchiorri D (2006) Context-
nalling. Nature 361:315325. dependent regulation of embryonic stem cell differ-
Berridge MJ (2006) Calcium microdomains: organization and entiation by mGlu4 metabotropic glutamate receptors.
function. Cell Calcium 40:405412. Neuropharmacology 51:606611.
Berridge MJ, Lipp P, Bootman MD (2000) The versatility and Chebib M, Johnston GA (1999) The  ABC of GABA receptors:
universality of calcium signaling. Nat Rev Mol Cell Biol a brief review. Clin Exp Pharmacol Physiol 26:937940.
1:1121. Cheung KK, Chan WY, Burnstock G (2005) Expression of
Biagioni S, Tata AM, De Jaco A, Augusti-Tocco G (2000) P2X purinoceptors during rat brain development and their
Acetylcholine synthesis and neuron differentiation. Int J Dev inhibitory role on motor axon outgrowth in neural tube
Biol 44:689697. explant cultures. Neuroscience 133:937945.
Bignami F, Bevilacqua P, Biagioni S, De Jaco A, Casamenti F, Cheung KK, Ryten M, Burnstock G (2003) Abundant and
Felsani A, Augusti-Tocco G (1997) Cellular acetylcholine dynamic expression of G protein-coupled P2Y receptors in
content and neuronal differentiation. J Neurochem 69: mammalian development. Dev Dyn 228:254266.
13741381. Cicirata F, Meli C, Castorina C, Serapide MF, Sorrenti V, Di
Blaustein MP, Lederer WJ (1999) Sodium/calcium exchange: Its Giacomo C, Gambera G, Vanella A (1991) Neurotransmitter
physiological implications. Physiol Rev 79:763854. amino acid levels in rat thalamus and cerebral cortex after
Bogdanov YD, Dale L, King BF, Whittock N, Burnstock G cerebellectomy. Int J Dev Neurosci 9:365369.
(1997) Early expression of a novel nucleotide receptor in Clapham DE (1995) Calcium signaling. Cell 80:259268.
130 K.K. Yuahasi et al.

Connors BW, Malenka RC, Silva LR (1988) Two inhibitory Fiszman ML, Borodinsky LN, Neale JH (1999) GABA induces
postsynaptic potentials, and GABAA and GABAB receptor- proliferation of immature cerebellar granule cells grown in
mediated responses in neocortex of rat and cat. J Physiol vitro. Brain Res Dev Brain Res 115:18.
406:443468. Franco Lambert AP, Fraga Zandonai A, Bonatto D, Cantarelli
Coronas V, Durand M, Chabot JG, Jourdan F, Quirion R (2000) Machado D, Pgas Henriques J (2009) Differentiation of
Acetylcholine induces neuritic outgrowth in rat primary human adipose-derived adult stem cells into neuronal tissue:
olfactory bulb cultures. Neuroscience 98:213219. does it work? Differentiation 77:221228.
Court JA, Lloyd S, Johnson M, Griffiths M, Birdsall NJ, Piggott Fredholm BB, IJzerman AP, Jacobson KA, Klotz KN, Linden
MA, Oakley AE, Ince PG, Perry EK, Perry RH (1997) J (2001) International Union of Pharmacology. XXV.
Nicotinic and muscarinic cholinergic receptor binding in Nomenclature and classification of adenosine receptors.
the human hippocampal formation during development and Pharmacol Rev 53:527552.
aging. Brain Res Dev Brain Res 101:93105. Frisn J, Johansson CB, Lothian C, Lendahl U (1998) Central
Court JA, Perry EK, Spurden D, Griffiths M, Kerwin JM, Morris nervous system stem cells in the embryo and adult. Cell Mol
CM, Johnson M, Oakley AE, Birdsall NJ, Clementi F et al. Life Sci 54:935945.
(1995) The role of the cholinergic system in the develop- Fukui M, Nakamichi N, Yoneyama M, Ozawa S, Fujimori S,
ment of the human cerebellum. Brain Res Dev Brain Res 90: Takahata Y, Nakamura N, Taniura H, Yoneda Y (2008)
159167. Modulation of cellular proliferation and differentiation
Culiat CT, Stubbs LJ, Montgomery CS, Russell LB, Rinchik EM through GABA(B) receptors expressed by undifferentiated
(1994) Phenotypic consequences of deletion of the gamma 3, neural progenitor cells isolated from fetal mouse brain. J Cell
alpha 5, or beta 3 subunit of the type A gamma-aminobutyric Physiol 216:507519.
acid receptor in mice. Proc Natl Acad Sci USA 91: Gachelin G, Kemler R, Kelly F, Jacob F (1997) PCC4, a new cell
28152818. surface antigen common to multipotential embryonal car-
Deisseroth K, Singla S, Toda H, Monje M, Palmer TD, Malenka cinoma cells, spermatozoa, and mouse early embryos. Dev
RC (2004) Excitation-neurogenesis coupling in adult neural Biol 57:199209.
stem/progenitor cells. Neuron 42:535552. Gallo V, Zhou JM, McBain CJ, Wright P, Knutson PL,
Del Rio JA, Soriano E, Ferrer I (1992) Development of GABA- Armstrong RC (1996) Oligodendrocyte progenitor cell pro-
immunoreactivity in the neocortex of the mouse. J Comp liferation and lineage progression are regulated by gluta-
Neurol 326:501526. mate receptor-mediated K+ channel block. J Neurosci 16:
DiCicco-Bloom E, Lord C, Zwaigenbaum L, Courchesne E, 26592670.
Dager SR, Schmitz C, Schultz RT, Crawley J, Young LJ Gass JT, Olive MF (2008) Glutamatergic substrates of
(2006) The developmental neurobiology of autism spectrum drug addiction and alcoholism. Biochem Pharmacol 75:
disorder. J Neurosci 26:68976906. 218265.
Doetsch F (2003a) A niche for adult neural stem cells. Curr Opin Gimble JM, Katz AJ, Bunnell BA (2007) Adipose-derived stem
Genet Dev 13:543550. cells for regenerative medicine. Circ Res 100:12491260.
Doetsch F (2003b) The glial identity of neural stem cells. Nat Goldman S (2003) Glia as neural progenitor cells. Trends
Neurosci 6:11271134. Neurosci 26(11):590596.
Don F, Ulrich H, Persike DS, Conceio IM, Blini JP, Gotti C, Fornasari D, Clementi F (1997) Human neuronal
Cavalheiro EA, Fernandes MJ (2009) Alteration of puriner- nicotinic receptors. Prog Neurobiol 53:199237.
gic P2X4 and P2X7 receptor expression in rats with Gould E, Cameron HA, McEwen BS (1994) Blockade of NMDA
temporal-lobe epilepsy induced by pilocarpine. Epilepsy Res receptors increases cell death and birth in the developing rat
83:157167. dentate gyrus. J Comp Neurol 340:551565.
Duchen MR (1999) Contributions of mitochondria to animal Gould E, Reeves AJ, Fallah M, Tanapat P, Gross CG, Fuchs
physiology: from homeostatic sensor to calcium signalling E (1999) Hippocampal neurogenesis in adult Old World
and cell death. J Physiol 516:117. primates. Proc Natl Acad Sci USA 96:52635267.
Eder A, Bading H (2007) Calcium signals can freely cross Gu X, Spitzer NC (1995) Distinct aspects of neuronal differenti-
the nuclear envelope in hippocampal neurons: somatic cal- ation encoded by frequency of spontaneous Ca2+ transients.
cium increases generate nuclear calcium transients. BMC Nature 375:784787.
Neurosci 8:5757. Guan K, Chang H, Rolletschek A, Wobus AM (2001) Embryonic
Eriksson PS, Perfilieva E, Bjrk-Eriksson T, Alborn AM, stem cell-derived neurogenesis. Retinoic acid induction and
Nordborg C, Peterson DA, Gage FH (1998) Neurogenesis in lineage selection of neuronal cells. Cell Tissue Res 305:
the adult human hippocampus. Nat Med 4:13131317. 171176.
Evans MJ, Kaufman MH (1981) Establishment in culture Guillemot F (2007) Cell fate specification in the mammalian
of pluripotential cells from mouse embryos. Nature 292: telencephalon. Prog Neurobiol 83:3752.
154156. Gysbers JW, Rathbone MP (1992) Guanosine enhances
Ferrari G, Cusella-De Angelis G, Coletta M, Paolucci E, NGFstimulated neurite outgrowth in PC12 cells.
Stornaiuolo A, Cossu G, Mavilio F (1998) Muscle regenera- NeuroReport 3:9971000.
tion by bone marrow-derived myogenic precursors. Science Gysbers JW, Rathbone MP (1996) GTP and guanosine synergis-
279:15281530. tically enhance NGF-induced neurite outgrowth from PC12
Fishell G, Kriegstein AR (2003) Neurons from radial glia: cells. Int J Dev Neurosci 14:19 34.
the consequences of asymmetric inheritance. Curr Opin Gtz M, Huttner WB (2005) The cell biology of neurogenesis.
Neurobiol 13:3441. Nat Rev Mol Cell Biol 6:777788.
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 131

Gnther U, Benson J, Benke D, Fritschy JM, Reyes G, Knoflach Krnjevic K, Schwartz S (1967) The action of gamma-
F, Crestani F, Aguzzi A, Arigoni M, Lang Y, Bluethmann aminobutyric acid on cortical neurons. Exp Brain Res 3:
H, Mohler H, Luscher B (1995) Benzodiazepine-insensitive 320336.
mice generated by targeted disruption of the gamma 2 sub- Kuhn HG, Dickinson-Anson H, Gage FH (1996) Neurogenesis
unit gene of gamma-aminobutyric acid type A receptors. in the dentate gyrus of the adult rat: age-related decrease of
Proc Natl Acad Sci USA 92:77497753. neuronal progenitor proliferation. J Neurosci 16:20272033.
Haber M, Zhou L, Murai KK (2006) Cooperative astrocyte Laasberg T (1990) Ca2(+) -mobilizing receptors of gastrulating
and dendritic spine dynamics at hippocampal excitatory chick embryo. Comp Biochem Physiol C 97:912.
synapses. J Neurosci 26:88818891. Lauder JM (1993) Neurotransmitters as growth regulatory sig-
Hamprecht B (1977) Structural, electrophysiological, biochem- nals: role of receptors and second messengers. Trends
ical, and pharmacological properties of neuroblastoma- Neurosci 16:233240.
glioma cell hybrids in cell culture. Int Rev Cytol 49:99170. Lauder JM, Liu J, Devaud L, Morrow AL (1998) GABA as a
Haydar TF, Wang F, Schwartz ML, Rakic P (2000) Differential trophic factor for developing monoamine neurons. Perspect
modulation of proliferation in the neocortical ventricular and Dev Neurobiol 5:247259.
subventricular zones. J Neurosci 20:57645774. Leclerc C, Nant I, Webb SE, Miller AL, Moreau M (2006)
Heck S, Enz R, Richter-Landsberg C, Blohm DH (1997) Calcium transients and calcium signalling during early neu-
Expression of eight metabotropic glutamate receptor sub- rogenesis in the amphibian embryo Xenopus laevis. Biochim
types during neuronal differentiation of P19 embryocarci- Biophys Acta 1763:11841191.
noma cells: a study by RT-PCR and in situ hybridization. Le Feuvre RA, Brough D, Iwakura Y, Takeda K, Rothwell
Brain Res Dev Brain Res 101:8591. NJ (2002) Priming of macrophages with lipopolysaccha-
Ishida I, Deguchi T (1983) Effect of depolarizing agents on ride potentiates P2X7-mediated cell death via a caspase-1-
choline acetyltransferase and acetylcholinesterase activities dependent mechanism, independently of cytokine produc-
in primary cell cultures of spinal cord. J Neurosci 3: tion. J Biol Chem 277:32103218.
18181823. Lee YH, Lin CH, Hsu LW, Hu SY, Hsiao WT, Ho YS (2003)
Jackson L, Jones DR, Scotting P, Sottile V (2007) Adult mes- Roles of ionotropic glutamate receptors in early developing
enchymal stem cells: differentiation potential and therapeutic neurons derived from the P19 mouse cell line. J Biomed Sci
applications. J Postgrad Med 53:121127. 10:199207.
Jones-Villeneuve EM, McBurney MW, Rogers KA, Kalnins VI Levin ED (1992) Nicotinic systems and cognitive function.
(1982) Retinoic acid induces embryonal carcinoma cells to Psychopharmacology (Berl) 108:417431.
differentiate into neurons and glial cells. J Cell Biol 94: Levitt P, Rakic P (1980) Immunoperoxidase localization of glial
253262. fibrillary acidic protein in radial glial cells and astrocytes
Kahan BW, Ephrussi B (1970) Developmental potentialities of of the developing rhesus monkey brain. J Comp Neurol
clonal in vitro cultures of mouse testicular teratoma. J Natl 193:815840.
Cancer Inst 44:10151036. LoTurco JJ, Owens DF, Heath MJ, Davis MB, Kriegstein
Kang SK, Lee DH, Bae YC, Kim HK, Baik SY, Jung JS (2003) AR (1995) GABA and glutamate depolarize cortical pro-
Improvement of neurological deficits by intracerebral trans- genitor cells and inhibit DNA synthesis. Neuron 15:
plantation of human adipose tissue-derived stromal cells after 12871298.
cerebral ischemia in rats. Exp Neurol 183:355366. Ma W, Liu QY, Maric D, Sathanoori R, Chang YH, Barker JL
Kang SK, Putnam LA, Ylostalo J, Popescu IR, Dufour J, (1998) Basic FGF-responsive telencephalic precursor cells
Belousov A, Bunnell BA (2004) Neurogenesis of Rhesus express functional GABA(A) receptor/Cl channels in vitro.
adipose stromal cells. J Cell Sci 117:42894299. J Neurobiol 35:277286.
Kaplan MS, Hinds JW (1977) Neurogenesis in the adult rat: elec- Ma W, Maric D, Li BS, Hu Q, Andreadis JD, Grant GM, Liu
tron microscopic analysis of light radioautographs. Science QY, Shaffer KM, Chang YH, Zhang L, Pancrazio JJ, Pant
197:10921094. HC, Stenger DA, Barker JL (2000) Acetylcholine stimulates
Kelly CM, Tyers P, Borg MT, Svendsen CN, Dunnett SB, Rosser cortical precursor cell proliferation in vitro via muscarinic
AE (2005) EGF and FGF-2 responsiveness of rat and mouse receptor activation and MAP kinase phosphorylation. Eur J
neural precursors derived from the embryonic CNS. Brain Neurosci 12:12271240.
Res Bull 68:8394. Martin GR (1981) Isolation of a pluripotent cell line from
Kinney HC, ODonnell TJ, Kriger P, White WF (1993) Early early mouse embryos cultured in medium conditioned by
developmental changes in [3H] nicotine binding in the teratocarcinoma stem cells. Proc Natl Acad Sci USA 78:
human brainstem. Neuroscience 55:11271138. 76347638.
Kitayama T, Yoneyama M, Tamaki K, Yoneda Y (2004) Mass K, Bhamra S, Eason R, Dale N, Jones EA (2007)
Regulation of neuronal differentiation by N-methyl-D- Purine-mediated signalling triggers eye development. Nature
aspartate receptors expressed in neural progenitor cells 449:10581062.
isolated from adult mouse hippocampus. J Neurosci Res Mattson MP, Dou P, Kater SB (1988b) Outgrowth-regulating
76:599612. actions of glutamate in isolated hippocampal pyramidal neu-
Komuro H, Rakic P (1993) Modulation of neuronal migration by rons. J Neurosci 8:20872100.
NMDA receptors. Science 260:9597. Mattson MP, Lee RE, Adams ME, Guthrie PB, Kater SB (1988a)
Krey JF, Dolmetsch RE (2007) Molecular mechanisms of Interactions between entorhinal axons and target hippocam-
autism: a possible role for Ca2+ signaling. Current Opin pal neurons: a role for glutamate in the development of
Neurobiol 17:112119. hippocampal circuitry. Neuron 1:865876.
132 K.K. Yuahasi et al.

McBurney MW, Jones-Villeneuve EM, Edwards MK, Anderson Narahara M, Tachibana K, Kurisu N, Kanazawa M, Miyake
PJ (1982) Control of muscle and neuronal differentiation M (2000) Immunohistochemical and chemical changes of
in a cultured embryonal carcinoma cell line. Nature 299: beta-citryl-L-glutamate in the differentiation of bovine lens
165167. epithelial cells into lens fiber cells. Biol Pharm Bull 23:
Melchiorri D, Cappuccio I, Ciceroni C, Spinsanti P, Mosillo P, 704707.
Sarichelou I, Sale P, Nicoletti F (2007) Metabotropic gluta- Neary JT, Kang Y, Shi YF (2004) Signaling from nucleotide
mate receptors in stem/progenitor cells. Neuropharmacology receptors to protein kinase cascades in astrocytes.
53:473480. Neurochem Res 29:2037 2042.
Minakami R, Iida K, Hirakawa N, Sugiyama H (1995) The Nguyen L, Rigo JM, Rocher V, Belachew S, Malgrange B,
expression of two splice variants of metabotropic glutamate Rogister B, Leprince P, Moonen G (2001) Neurotransmitters
receptor subtype 5 in the rat brain and neuronal cells during as early signals for central nervous system development. Cell
development. J Neurochem 65:15361542. Tissue Res 305:187202.
Ming GL, Song H (2005) Adult neurogenesis in the mam- Nishi R, Berg DK (1981) Effects of high K+ concentrations on
malian central nervous system. Annu Rev Neurosci 28: the growth and development of ciliary ganglion neurons in
223250. cell culture. Dev Biol 87:301307.
Miranda-Contreras L, Bentez-Diaz PR, Mendoza-Briceo RV, Nishimoto M, Furuta A, Aoki S, Kudo Y, Miyakawa H, Wada
Delgado-Saez MC, Palacios-Pr EL (1999) Levels of amino K (2007) PACAP/PAC1 autocrine system promotes pro-
acid neurotransmitters during mouse cerebellar neurogene- liferation and astrogenesis in neural progenitor cells. Glia
sis and in histotypic cerebellar cultures. Dev Neurosci 21: 55:317327.
147158. North RA (2002) Molecular physiology of P2X receptors.
Miranda-Contreras L, Mendoza-Briceo RV, Palacios-Pr EL Physiol Rev 82:10131067.
(1998) Levels of monoamine and amino acid neurotrans- Nottebohm F, Goldman SA (1983) Neuronal production, migra-
mitters in the developing male mouse hypothalamus and tion, and differentiation in a vocal control nucleus of the
in histotypic hypothalamic cultures. Int J Dev Neurosci adult female canary brain. Proc Natl Acad Sci USA 80:
16:403412. 23902394.
Miranda-Contreras L, Ramrez-Martens LM, Bentez-Diaz PR, Ostenfeld T, Svendsen CN (2004) Requirement for neuroge-
Pea-Contreras ZC, Mendoza-Briceo RV, Palacios-Pr E nesis to proceed through the division of neuronal progen-
(2000) Levels of amino acid neurotransmitters during mouse itors following differentiation of epidermal growth factor
olfactory bulb neurogenesis and in histotypic olfactory bulb and fibroblast growth factor-2-responsive human neural stem
cultures. Int J Dev Neurosci 18:8391. cells. Stem Cells 22:798811.
Miras-Portugal MT, Daz-Hernndez M, Girldez L, Hervs C, Owens DF, Boyce LH, Davis MB, Kriegstein AR (1996)
Gmez-Villafuertes R, Sen RP, Gualix J, Pintor J (2003) Excitatory GABA responses in embryonic and neonatal
P2X7 receptors in rat brain: presence in synaptic terminals cortical slices demonstrated by gramicidin perforated-patch
and granule cells. Neurochem Res 28:15971605. recordings and calcium imaging. J Neurosci 16:64146423.
Mishra SK, Braun N, Shukla V, Fllgrabe M, Schomerus Pagani F, Lauro C, Fucile S, Catalano M, Limatola C, Eusebi
C, Korf HW, Gachet C, Ikehara Y, Svigny J, Robson F, Grassi F (2006) Functional properties of neurons derived
SC, Zimmermann H (2006) Extracellular nucleotide sig- from fetal mouse neurospheres are compatible with those of
naling in adult neural stem cells: synergism with growth neuronal precursors in vivo. J Neurosci Res 83:14941501.
factor-mediated cellular proliferation. Development 33: Paraoanu LE, Steinert G, Koehler A, Wessler I, Layer PG (2007)
675684. Expression and possible functions of the cholinergic system
Miyata T, Kawaguchi A, Saito K, Kawano M, Muto T, Ogawa in a murine embryonic stem cell line. Life Sci 80:23752379.
M (2004) Asymmetric production of surface-dividing and Parnas D, Linial M (1995) Cholinergic properties of neurons dif-
non-surface-dividing cortical progenitor cells. Development ferentiated from an embryonal carcinoma cell-line (P19). Int
131:31333145. J Dev Neurosci 13:767781.
Mizuseki K, Sakamoto T, Watanabe K, Muguruma K, Ikeya Paterson D, Nordberg A (2000) Neuronal nicotinic receptors in
M, Nishiyama A, Arakawa A, Suemori H, Nakatsuji N, the human brain. Prog Neurobiol 61:75111.
Kawasaki H, Murakami F, Sasai Y (2003) Generation of neu- Pearce IA, Cambray-Deakin MA, Burgoyne RD (1987)
ral crest-derived peripheral neurons and floor plate cells from Glutamate acting on NMDA receptors stimulates neurite
mouse and primate embryonic stem cells. Proc Natl Acad Sci outgrowth from cerebellar granule cells. FEBS Lett 223:
USA 100:58285833. 143147.
Monti B, Contestabile A (2000) Blockade of the NMDA recep- Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan
tor increases developmental apoptotic elimination of granule AK, Murase N, Boggs SS, Greenberger JS, Goff JP (1999)
neurons and activates caspases in the rat cerebellum. Eur J Bone marrow as a source of hepatic oval cells. Science 284:
Neurosci 12:31173123. 11681170.
Nacher J, McEwen BS (2006) The role of N-methyl-D-asparate Phelps PE, Barber RP, Brennan LA, Maines VM, Salvaterra PM,
receptors in neurogenesis. Hippocampus 16:267270. Vaughn JE (1990) Embryonic development of four differ-
Nagase T, Matsumoto D, Nagase M, Yoshimura K, Shigeura T, ent subsets of cholinergic neurons in rat cervical spinal cord.
Inoue M, Hasegawa M, Yamagishi M, Machida M (2007) J Comp Neurol 291:926.
Neurospheres from human adipose tissue transplanted into Pin JP, Duvoisin R (1995) The metabotropic glutamate
cultured mouse embryos can contribute to craniofacial mor- receptors: structure and functions. Neuropharmacology 34:
phogenesis: a preliminary report. J Craniofac Surg 18:4953. 126.
9 Neurotransmitters as Main Players in the Neural Differentiation and Fate Determination Game 133

Poulsen FR, Blaabjerg M, Montero M, Zimmer J (2005) of murine and human adipose-derived stromal cells.
Glutamate receptor antagonists and growth factors modu- Biochem Biophys Res Commun 294:371379.
late dentate granule cell neurogenesis in organotypic, rat Sez JC, Connor JA, Spray DC, Bennett MV (1989) Hepatocyte
hippocampal slice cultures. Brain Res 1051:3549. gap junctions are permeable to the second messenger, inosi-
Pozzan T, Rizzuto R, Volpe P, Meldolesi J (1994) Molecular and tol 1,4,5-trisphosphate, and to calcium ions. Proc Natl Acad
cellular physiology of intracellular calcium stores. Physiol Sci USA 86:27082712.
Rev 74:595636. Sanchez-Ramos J, Song S, Cardozo-Pelaez F, Hazzi C,
Prockop DJ (1997) Marrow stromal cells as stem cells for non- Stedeford T, Willing A, Freeman TB, Saporta S, Janssen W,
hematopoietic tissues. Science 276:7174. Patel N, Cooper DR, Sanberg PR (2000) Adult bone mar-
Qu Y, Vadivelu S, Choi L, Liu S, Lu A, Lewis B, Girgis row stromal cells differentiate into neural cells in vitro. Exp
R, Lee CS, Snider BJ, Gottlieb DI, McDonald JW (2003) Neurol 164:247256.
Neurons derived from embryonic stem (ES) cells resemble Sarichelou I, Cappuccio I, Ferranti F, Mosillo P, Ciceroni C, Sale
normal neurons in their vulnerability to excitotoxic death. P, Stocchi F, Battaglia G, Nicoletti F, Melchiorri D (2008)
Exp Neurol 184:326336. Metabotropic glutamate receptors regulate differentiation of
Quik M, Polonskaya Y, Gillespie A, Jakowec M, Lloyd GK, embryonic stem cells into GABAergic neurons. Cell Death
Langston JW (2000) Localization of nicotinic receptor sub- Differ 15:700707.
unit mRNAs in monkey brain by in situ hybridization. Scheetz AJ, Constantine-Paton M (1994) Modulation of NMDA
J Comp Neurol 425:5869. receptor function: implications for vertebrate neural develop-
Rathbone MP, Middlemiss PJ, Gysbers JW, Andrew C, Herman ment. FASEB J 8:745752.
MA, Reed JK, Ciccarelli R, Di Iorio P, Caciagli F (1999) Schneider AS, Atluri P, Shen Q, Barnes W, Mah SJ, Stadfelt
Trophic effects of purines in neurons and glial cells. Prog D, Goderie SK, Temple S, Fleck MW (2002) Functional
Neurobiol 59:663 690. nicotinic acetylcholine receptor expression on stem and pro-
Resende RR, Alves AS, Britto LR, Ulrich H (2008b) Role genitor cells of the early embryonic nervous system. Ann N
of acetylcholine receptors in proliferation and differentia- Y Acad Sci 971:135138.
tion of P19 embryonal carcinoma cells. Exp Cell Res 314: Schubert P, Kreutzberg GW (1993) Cerebral protection by
14291443. adenosine. Acta Neurochir Suppl (Wien) 57:8088.
Resende RR, Britto LR, Ulrich H (2008c) Pharmacological Solter D, Knowles BB (1978) Monoclonal antibody defining a
properties of purinergic receptors and their effects on pro- stage-specific mouse embryonic antigen (SSEA-1). Proc Natl
liferation and induction of neuronal differentiation of P19 Acad Sci USA 75:55655569.
embryonal carcinoma cells. Int J Dev Neurosci 26:763777. Song H, Stevens CF, Gage FH (2002) Astroglia induce neuroge-
Resende RR, Gomes KN, Adhikari A, Britto LR, Ulrich nesis from adult neural stem cells. Nature 417:3944.
H (2008a) Mechanism of acetylcholine induced cal- Spitzer NC, Borodinsky LN (2008) Implications of activity-
cium signaling during neuronal differentiation of P19 dependent neurotransmitter-receptor matching. Philos Trans
embryonal carcinoma cells in vitro. Cell Calcium 43: R Soc Lond B Biol Sci 363:13931399.
107121. Spitzer NC, Debaca RC, Allen KA, Holliday J (1993) Calcium
Resende RR, Majumder P, Gomes KN, Britto LR, Ulrich H dependence of differentiation of GABA immunoreactivity in
(2007) P19 embryonal carcinoma cells as in vitro model spinal neurons. J Comp Neurol 337:168175.
for studying purinergic receptor expression and modula- Spitzer NC, Gu X, Olson E (1994) Action potentials, calcium
tion of N-methyl-D-aspartate-glutamate and acetylcholine transients and the control of differentiation of excitable cells.
receptors during neuronal differentiation. Neuroscience 146: Curr Opin Neurobiol 4:7077.
11691181. Spitzer NC, Lautermilch NJ, Smith RD, Gomez TM (2000)
Reynolds BA, Weiss S (1992) Generation of neurons and astro- Coding of neuronal differentiation by calcium transients.
cytes from isolated cells of the adult mammalian central BioEssays 22:811817.
nervous system. Science 255:17071710. Spitzer NC, Root CM, Borodinsky LN (2004) Orchestrating
Rivera C, Voipio J, Payne JA, Ruusuvuori E, Lahtinen H, Lamsa neuronal differentiation: patterns of Ca2+ spikes specify
K, Pirvola U, Saarma M, Kaila K (1999) The K+ /Cl co- transmitter choice. Trends Neurosci 27:415421.
transporter KCC2 renders GABA hyperpolarizing during Thevananther S, Rivera A, Rivkees SA (2001) A1 adenosine
neuronal maturation. Nature 397:251255. receptor activation inhibits neurite process formation by Rho
Rizzuto R, Brini M, Murgia M, Pozzan T (1993) Microdomains kinase-mediated pathways. NeuroReport 12:30573063.
with high Ca2+ close to IP3-sensitive channels that are sensed Tropepe V, Hitoshi S, Sirard C, Mak TW, Rossant J, van
by neighboring mitochondria. Science 262:744747. der Kooy D (2001) Direct neural fate specification from
Root CM, Velzquez-Ulloa NA, Monsalve GC, Minakova E, embryonic stem cells: A primitive mammalian neural stem
Spitzer NC (2008) Embryonically expressed GABA and cell stage acquired through a default mechanism. Neuron
glutamate drive electrical activity regulating neurotransmitter 30:6578.
specification. J Neurosci 28:47774784. Tropepe V, Sibilia M, Ciruna BG, Rossant J, Wagner EF, van
Sadikot AF, Burhan AM, Blanger MC, Sasseville R (1998) der Kooy D (1999) Distinct neural stem cells proliferate
NMDA receptor antagonists influence early development of in response to EGF and FGF in the developing mouse
GABAergic interneurons in the mammalian striatum. Brain telencephalon. Dev Biol 208:166188.
Res Dev Brain Res 105:3542. Turner CP, Yan H, Schwartz M, Othman T, Rivkees SA (2002)
Safford KM, Hicok KC, Safford SD, Halvorsen YD, Wilkison A1 adenosine receptor activation induces ventriculomegaly
WO, Gimble JM, Rice HE (2002) Neurogenic differentiation and white matter loss. Neuroreport 13:11991204.
134 K.K. Yuahasi et al.

Ulrich H, Majumder P (2006) Neurotransmitter receptor Wichterle H, Lieberam I, Porter JA, Jessell TM (2002) Directed
expression and activity during neuronal differentiation of differentiation of embryonic stem cells into motor neurons.
embryonal carcinoma and stem cells: from basic research Cell 110:385397.
towards clinical applications. Cell Prolif 39:281300. Xu Y, Liu Z, Liu L, Zhao C, Xiong F, Zhou C, Li Y, Shan
Vaca K (1988) The development of cholinergic neurons. Brain Y, Peng F, Zhang C (2008) Neurospheres from rat adipose-
Res 472:261286. derived stem cells could be induced into functional Schwann
van den Pol AN (1997) GABA immunoreactivity in hypothala- cell-like cells in vitro. BMC Neurosci 9:2131.
mic neurons and growth cones in early development in vitro Ying QL, Stavridis M, Griffiths D, Li M, Smith A (2003)
before synapse formation. J Comp Neurol 383:178188. Conversion of embryonic stem cells into neuroectodermal
Wang C, Pralong WF, Schulz MF, Rougon G, Aubry JM, precursors in adherent monoculture. Nat Biotechnol 21:
Pagliusi S, Robert A, Kiss JZ (1996) Functional N-methyl- 183186.
D-aspartate receptors in O-2A glial precursor cells: a crit- Yoneyama M, Fukui M, Nakamichi N, Kitayama T, Taniura
ical role in regulating polysialic acid-neural cell adhe- H, Yoneda Y (2007). Activation of GABA(A) receptors
sion molecule expression and cell migration. J Cell Biol facilitates astroglial differentiation induced by ciliary neu-
135:15651581. rotrophic factor in neural progenitors isolated from fetal rat
Watanabe K, Kamiya D, Nishiyama A, Katayama T, Nozaki brain. J Neurochem 100:16671679.
S, Kawasaki H, Watanabe Y, Mizuseki K, Sasai Y (2005) Yu H-M, Wen J, Wang R (2008) Critical role of type 2 ryanodine
Directed differentiation of telencephalic precursors from receptor in mediating activity-dependent neurogenesis from
embryonic stem cells. Nat Neurosci 8:288296. embryonic stem cells. Cell Calcium 43:417431.
Weaver DR (1993) A2a adenosine receptor gene expression Zheng JQ, Poo M (2007) Calcium signaling and neuronal motil-
in developing rat brain. Brain Res Mol Brain Res 20: ity. Annu Rev Cell Dev Biol 23:375404.
313327. Zoli M, Le Novre N, Hill JA Jr, Changeux JP (1995)
Weiss ER, Maness P, Lauder JM (1998) Why do neurotrans- Developmental regulation of nicotinic ACh receptor subunit
mitters act like growth factors? Perspect Dev Neurobiol mRNAs in the rat central and peripheral nervous systems.
5:323335. J Neurosci 15:19121939.
Weissman TA, Riquelme PA, Ivic L, Flint AC, Kriegstein AR Zoli M, Picciotto MR, Ferrari R (1999) Increased neurodegen-
(2004) Calcium waves propagate through radial glial cells eration during ageing in mice lacking high-affinity nicotine
and modulate proliferation in the developing neocortex. receptors. EMBO J 18:12351244.
Neuron 43:647661. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno
Whitaker M, Smith J (2008) Introduction. Calcium signals H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH (2002)
and developmental patterning. Phil Trans R Soc B 363: Human adipose tissue is a source of multipotent stem cells.
13071310. Mol Biol Cell 13:42794295.
Chapter 10

Rhythmic Expression of Notch Signaling in Neural


Progenitor Cells

Hiromi Shimojo, Toshiyuki Ohtsuka, and Ryoichiro Kageyama

Contents Delta1 oscillations reciprocally activate Notch signal-


ing between neighboring neural progenitor cells. These
10.1 Introduction . . . . . . . . . . . . . . . . . 135 results also suggest that oscillatory expression of Ngn2
10.2 Activator-Type bHLH Genes . . . . . . . . . 136 is not sufficient but sustained up-regulation is required
10.3 Repressor-Type bHLH Genes . . . . . . . . 137 for neuronal differentiation and that Ngn2 oscillation
10.4 Notch Signaling . . . . . . . . . . . . . . . 138
is advantageous for activation of Notch signaling by
10.5 Dynamic Expression in Neural Progenitor Cells 139
10.6 Oscillatory Versus Persistent Hes1 Expression . 140 inducing Delta1 expression without promoting neu-
10.7 Conclusions . . . . . . . . . . . . . . . . . 141 ronal differentiation.
References . . . . . . . . . . . . . . . . . . . . 142
Keywords bHLH gene Hes1 Neural progenitor
cell Notch signaling Oscillatory expression
Abstract The activator-type basic helix-loop-helix
(bHLH) genes such as Mash1 and Neurogenin2 (Ngn2) Abbreviations
promote neuronal differentiation and induce expres-
sion of Notch ligands such as Delta1, which activate bHLH basic helix-loop-helix
Notch signaling of neighboring cells. Activation of GABA -aminobutyric acid
Ngn2 Neurogenin 2
Notch signaling induces expression of the repressor-
NICD Notch intracellular domain
type bHLH genes such as Hes1 and Hes5, which main-
Stat3-P Phosphorylated Stat3
tain neural progenitor cells by antagonizing activator-
type bHLH genes. Thus, differentiating neurons keep
their neighboring cells as neural progenitor cells via
Notch signaling. How, then, are neural progenitor cells 10.1 Introduction
maintained before formation of such neurons? A recent
study revealed that Hes1 expression occurs rhythmi- In the developing mammalian nervous system, neu-
cally in neural progenitor cells, and that Ngn2 and roepithelial cells undergo self-renewal repeatedly by
Delta1 are also expressed in an oscillatory manner symmetric division (Fig. 10.1). As the number of neu-
by these cells. Inhibition of Notch signaling, a condi- roepithelial cells increases, the wall of the neural tube
tion known to induce neuronal differentiation, leads to grows thicker, and neuroepithelial cells are elongated
down-regulation of Hes1 and sustained up-regulation to become radial glial cells (Fig. 10.1). Radial glial
of Ngn2 and Delta1, suggesting that Hes1 oscillation cells have a cell body in the inner side (called the
regulates Ngn2 and Delta1 oscillations. It is likely that ventricular zone) of the neural tube and a long pro-
cess (called a radial fiber) reaching the outer surface. It
was long thought that radial glial cells are specialized
glial cells that guide neuronal migration. However, it
R. Kageyama ()
Institute for Virus Research, Kyoto University, Kyoto, Japan has been shown that radial glial cells divide and gen-
e-mail: rkageyam@virus.kyoto-u.ac.jp erate neurons (Alvarez-Buylla et al., 2001; Anthony

H. Ulrich (ed.), Perspectives of Stem Cells, 135


DOI 10.1007/978-90-481-3375-8_10, Springer Science+Business Media B.V. 2010
136 H. Shimojo et al.

Fig. 10.1 Neural stem/progenitor cells and their differ- surface. Radial glial cells give rise to neurons/neuronal pre-
entiation. Initially, neuroepithelial cells are formed. These cursors. After production of neurons, some radial glial cells
cells undergo self-renewal repeatedly by symmetric division. give rise to oligodendrocytes and ependymal cells. Radial glial
As development proceeds, neuroepithelial cells are elongated cells finally differentiate into astrocytes. Both neuroepithelial
to become radial glial cells, which have a cell body in the cells and radial glial cells are considered as embryonic neural
inner side (called the ventricular zone) of the neural tube stem/progenitor cells
and a long process (called a radial fiber) reaching the outer

et al., 2004; Fujita, 2003; Malatesta et al., 2000; dynamic bHLH gene expression regulates proliferation
Noctor et al., 2001). Each radial glial cell gives rise and differentiation of neural progenitor cells.
to two radial glial cells by symmetric division, one
radial glial cell and one neuron/neuronal precursor
by asymmetric division, or two neurons/neuronal pre-
cursors by symmetric neurogenic division. Neuronal 10.2 Activator-Type bHLH Genes
precursors further divide and produce more neurons.
After production of neurons, some radial glial cells Activator-type bHLH genes include Mash1, Math and
give rise to oligodendrocytes and ependymal cells Neurogenin (Ngn), homologs of Drosophila proneu-
(Fig. 10.1). Radial glial cells finally differentiate into ral genes achaete-scute complex and atonal (Bertrand
astrocytes (Fig. 10.1). Both neuroepithelial cells and et al., 2002; Kageyama et al., 2005; Ross et al.,
radial glial cells are considered as embryonic neural 2003). They are expressed by differentiating neurons
stem/progenitor cells (here, we call neural progeni- in the developing nervous system. These bHLH factors
tor cells). Embryonic neural progenitor cells change form heterodimers through the helix-loop-helix (HLH)
their differentiation ability during development: ini- domain with a ubiquitously expressed bHLH factor,
tially undergoing self-renewal only and then differenti- E47, a product of a homolog of Drosophila proneu-
ation into neurons and other cell types, and finally into ral gene daughterless, and bind to the E box sequences
astrocytes. (CANNTG) through the basic regions, which are rich
It has been shown that neural development is antag- in basic amino acid residues (Johnson et al., 1992).
onistically regulated by activator-type and repressor- These heterodimers activate gene expression, and thus
type basic helix-loop-helix (bHLH) genes (Bertrand these bHLH factors are categorized as the activator
et al., 2002; Kageyama et al., 2005; Ross et al., 2003). type (Fig. 10.2a).
The former genes promote neuronal differentiation, Activator-type bHLH genes not only induce the
whereas the latter genes maintain neural progeni- pan-neuronal gene expression but also promote the
tor cells by antagonizing the former. Recent studies neuronal subtype specification. In Drosophila, the
revealed that both activator-type and repressor-type proneural genes achaete and atonal regulate speci-
bHLH genes are expressed in an oscillatory man- fication of different subtypes of the peripheral ner-
ner in neural progenitor cells and that this dynamic vous system: external sensory organs and chordotonal
expression is important for maintenance of these cells organs, respectively. It was previously shown that the
(Shimojo et al., 2008). In this chapter, we describe how basic regions are involved in specification of different
10 Rhythmic Expression of Notch Signaling in Neural Progenitor Cells 137

Fig. 10.2 Activator-type and repressor-type bHLH factors. a co-repressor, and repress expression of genes such as activator
(a) Activator-type bHLH factors form heterodimers and activate bHLH genes (left). Repressor-type bHLH factors also antag-
expression of neuronal-specific genes by binding to the E box. onize the activity of activator-type bHLH factors by forming
(b) Repressor-type bHLH factors recruit a homolog of Groucho, non-functional heterodimers (right)

neuronal subtypes (Chien et al., 1996). Mammalian Activator-type bHLH factors not only stimulate
homologs of achaete and atonal also regulate speci- neuronal-specific gene expression but also repress
fication of different neuronal subtypes. For example, glial-specific gene expression. Expression of glial fib-
Mash1 regulates specification of GABAergic interneu- rillary acidic protein (GFAP), an astrocyte-specific
rons in the ventral telencephalon, while Ngn2 regulates gene, is up-regulated by Stat1/3 and Smad1, which are
specification of the glutamatergic pyramidal neurons bridged by the co-activator p300, but Ngn1 sequesters
in the dorsal telencephalon (Fode et al., 2000; Parras the p300/Smad complex from the glial promoters and
et al., 2002). In the absence of Ngn2, Mash1 is ectopi- thereby inhibits glial-specific gene expression (Sun
cally expressed and generates ectopic GABAergic et al., 2001). Thus, it is likely that activator-type
interneurons in the dorsal telencephalon. However, bHLH factors reinforce the neuronal fate specifica-
it has been shown that activator-type bHLH genes tion by inhibiting the alternative fate. In agreement
alone are not sufficient to generate diverse subtypes with this notion, in Mash1;Math3 double knock-out
of neurons. For example, retinal progenitor cells give or Mash1;Ngn2 double knock-out mice, the cells that
rise to six subtypes of neurons, which are aligned in should normally differentiate into neurons adopted the
specific layers, but activator-type bHLH genes alone glial fate instead, indicating that there is a fate switch
cannot regulate specification of these retinal neurons from neurons to glia in the absence of activator-type
(Hatakeyama et al., 2001). Although Mash1 is required bHLH genes (Tomita et al., 2000; Nieto et al., 2001).
for formation of bipolar neurons in the retina, misex- Thus, activator-type bHLH genes regulate neuronal
pression of Mash1 alone does not evoke an increase versus glial cell fate determination.
in bipolar cell genesis. Similarly, although the home-
odomain gene Chx10 is required for formation of
bipolar neurons in the retina, misexpression of Chx10
alone does not generate bipolar neurons but induces 10.3 Repressor-Type bHLH Genes
formation of non-neuronal cells in the inner nuclear
layer, where bipolar neurons normally reside. In con- Repressor-type bHLH genes include Hes genes,
trast, misexpression of both Mash1 and Chx10 effi- homologs of Drosophila hairy and Enhancer of split
ciently induces bipolar cell formation (Hatakeyama (Sasai et al., 1992; Kageyama et al., 2007). Among
et al., 2001). It is likely that homeodomain genes regu- the Hes family members, Hes1, Hes3 and Hes5 are
late the layer identity while activator-type bHLH genes highly expressed in the developing nervous system.
determine the neuronal fate specific for each layer. Neuroepithelial cells initially express Hes1 and Hes3,
138 H. Shimojo et al.

but then Hes3 expression is down-regulated whereas Hes-mutant mice is associated with up-regulation of
Hes5 expression occurs (Hatakeyama et al., 2004). activator-type bHLH genes such as Mash1 and Math3
Radial glial cells express mostly Hes1 and Hes5. Hes (Hatakeyama et al., 2004). Thus, it is likely that Hes
factors form homodimers or heterodimers with Hes- genes regulate the normal timing of differentiation by
related factors such as Hey1 through the HLH domain preventing premature onset of activator-type bHLH
and bind to the N box (CACNAG) or the class C site genes.
(CACG(C/A)G) with a higher affinity than to the E box
(CANNTG), unlike other bHLH factors (Fig. 10.2b)
(Sasai et al., 1992). Hes factors contain the sequence
Trp-Arg-Pro-Trp (the WRPW domain) at the carboxy- 10.4 Notch Signaling
terminus, which recruits the co-repressor TLE/Grg, a
homolog of Drosophila Groucho (Fig. 10.2b) (Paroush In differentiating neurons, activator-type bHLH genes
et al., 1994; Fisher et al., 1996; Grbavec and Stifani, up-regulate expression of Notch ligands such as
1996). Thus, Hes factors function as transcriptional Delta1, which then activates Notch, a transmembrane
repressors. The target genes for Hes factors include protein, on neighboring cells (Fig. 10.3) (Artavanis-
activator-type bHLH genes such as Mash1. For exam- Tsakonas et al., 1999; Selkoe and Kopan, 2003). Upon
ple, Hes1 represses Mash1 expression by directly activation by Notch ligands, the Notch intracellular
binding to the Mash1 promoter (Chen et al., 1997). domain (NICD) is released from the transmembrane
Hes factors also form heterodimers through the HLH region and moves into the nucleus, where NICD forms
domain with activator-type bHLH factors, but these a complex with the DNA-binding protein RBPj. RBPj
heterodimers cannot bind to the DNA and therefore alone is a transcriptional repressor, but the NICD-
are non-functional (Fig. 10.2b) (Sasai et al., 1992). RBPj complex is a transcriptional activator and induces
Thus, Hes1 antagonizes Mash1 by two different mech- expression of the repressor-type bHLH genes Hes1
anisms: repressing the expression at the transcriptional and Hes5 (Fig. 10.3) (Jarriault et al., 1995; Ohtsuka
level and inhibiting the activity by forming a non- et al., 1999). Hes1 and Hes5 antagonize the expression
functional heterodimer. and activity of activator-type bHLH genes, thereby
Misexpression of Hes1, Hes3 or Hes5 in the embry- inhibiting neuronal differentiation (Fig. 10.3). Thus,
onic brain inhibits neuronal differentiation and main-
tains radial glial cells, whereas in Hes1;Hes5 dou-
ble knock-out mice, many radial glial cells are not
maintained and prematurely differentiate into neu-
rons (Ishibashi et al., 1994; Ohtsuka et al., 2001;
Hatakeyama et al., 2004). In these mutant embryos,
neuroepithelial cells and some radial glial cells are
still maintained, suggesting that Hes3 may compen-
sate for Hes1 and Hes5 deficiency. In agreement with
this notion, in Hes1;Hes3;Hes5 triple knock-out mice
many neuroepithelial cells prematurely differentiate
into neurons as early as on E8.5 in contrast to wild-type
embryos, where neuroepithelial cells do not differen-
tiate into neurons at this stage (Hatakeyama et al., Fig. 10.3 Notch signaling and lateral inhibition. Activator-
2004). Furthermore, in the triple knock-out mice, vir- type bHLH factors induce expression of Notch ligands such
as Delta1, which activate Notch signaling in neighboring cells.
tually all radial glial cells prematurely differentiate
Upon activation by Notch ligands, the Notch intracellular
into neurons in the spinal cord and the hindbrain domain (NICD) is released from the transmembrane region and
by E10.0 at the expense of the later-born cell types moves into the nucleus, where NICD forms a complex with
such as later born neurons and astrocytes (Hatakeyama the DNA-binding protein RBPj. RBPj alone is a transcriptional
repressor, but the NICD-RBPj complex is a transcriptional acti-
et al., 2004). Thus, Hes1, Hes3 and Hes5 are essen-
vator and induces expression of the repressor-type bHLH genes
tial to generate a sufficient number and a full spectrum Hes1 and Hes5. Hes1 and Hes5 antagonize the expression and
of cells by maintaining neural progenitor cells until activity of activator-type bHLH genes, thereby inhibiting neu-
later stages. The premature neuronal differentiation in ronal differentiation and maintaining neural progenitor cells
10 Rhythmic Expression of Notch Signaling in Neural Progenitor Cells 139

differentiating neurons inhibit neighboring cells from round of expression. In this way, Hes1 autonomously
differentiating into the same cell types by activation of starts oscillatory expression (Fig. 10.4) (Hirata et al.,
Notch signaling. This Notch signaling-mediated cell- 2002). Real-time imaging analysis indicated that Hes1
cell regulation is called lateral inhibition. This regu- expression also oscillates in each neural progenitor cell
lation is essential for maintenance of neural progenitor with a period of about 23 h (Fig. 10.5) (Masamizu
cells, because in the absence of Notch signaling all et al., 2006; Shimojo et al., 2008). Interestingly, when
neural progenitor cells prematurely differentiate into the Hes1 expression level is high, the expression lev-
neurons and are depleted without generating later-born els of the activator-type bHLH factor Ngn2 and the
cell types. These results also indicate that activator- Notch ligand Delta1 are low, and vice versa in neu-
type bHLH genes are important for maintenance of ral progenitor cells, suggesting that Ngn2 and Delta1
neural progenitor cells, because they induce expres- expression also oscillate in these cells but with an
sion of Notch ligands, which activate Notch signaling opposite phase to Hes1 oscillation (Shimojo et al.,
in neighboring cells. In agreement with this notion, 2008). Real-time imaging showed that Ngn2 and
in Mash1;Math3 double knock-out mice, expression Delta1 expression indeed oscillate in neural progen-
of a Notch ligand is lost in the hindbrain, leading to itor cells but are persistent in post-mitotic neurons,
inactivation of Notch signaling and premature loss of which lose Hes1 expression (Fig. 10.5) (Shimojo et al.,
neural progenitor cells (Ohsawa et al., 2005). Thus, 2008). Thus, Hes1 oscillation drives Ngn2 and Delta1
activator-type bHLH genes not only promote neu- oscillations in neural progenitor cells, whereas Ngn2
ronal differentiation cell-autonomously but also main- and Delta1 expression are persistently up-regulated
tain neural progenitor cells non-cell-autonomously via when Hes1 expression disappears in post-mitotic neu-
Notch signaling. rons. It is likely that Delta1 oscillation periodically
Before neurons are formed, neural progenitor cells activates Notch signaling between neighboring neural
proliferate extensively, but even at this early stage progenitor cells. This reciprocal regulation is particu-
Notch signaling seems to be important for maintenance larly important for maintenance of neural progenitor
of neural progenitor cells. Without any neurons, how cells before formation of neurons, which persistently
is Notch signaling active? It has been recently shown express Delta1.
that neural progenitor cells also express activator-type Why do neural progenitor cells that express Ngn2
bHLH genes and Notch ligands, suggesting that these in an oscillatory manner remain undifferentiated? One
cells mutually activate Notch signaling. How does likely answer is that Ngn2 oscillation can induce
the Notch signaling operate in neural progenitor cells expression of only subsets of downstream genes such
before neuronal formation? Why do neural progeni- as Delta1 while sustained Ngn2 expression is required
tor cells remain undifferentiated although they express for expression of a full spectrum of downstream genes.
activator-type bHLH genes? We discuss these issues in For example, Delta1 is expressed in the ventricular
the next section. zone, indicating that Delta1 expression occurs rapidly
after Ngn2 expression (Castro et al., 2006). In con-
trast, another downstream gene, Rnd2, is expressed
mainly in the subventricular and intermediate zones,
10.5 Dynamic Expression in Neural
suggesting that Rnd2 expression occurs rather late after
Progenitor Cells Ngn2 expression (Heng et al., 2008). Expression of
such late-responding genes may depend on sustained
In neural progenitor cells, Hes1 expression is variable: Ngn2 expression and thus may not occur when Ngn2
some cells express Hes1 at high levels while others expression oscillates.
express it at lower levels. It was previously shown Hes1 oscillation is also important for efficient cell
that Hes1 expression oscillates with a period of about proliferation. Sustained Hes1 expression represses cell
2 h in many cell types such as fibroblasts (Fig. 10.4) cycle regulators such as cyclin D1 and cyclin E2
(Hirata et al., 2002). Hes1 represses its own expression and inhibits cell cycle progression (Baek et al., 2006;
by directly binding to the Hes1 promoter (Takebayashi Strm et al., 2000; Shimojo et al., 2008). However,
et al., 1994). This negative feedback leads to dis- Hes1 also promotes G1-S transition by repressing
appearance of both hes1 mRNA and Hes1 protein, expression of cyclin-dependent kinase inhibitors (CKI)
because they are extremely unstable, allowing the next (Castella et al., 2000; Kabos et al., 2002; Murata et al.,
140 H. Shimojo et al.

Fig. 10.4 Oscillatory


expression of Hes1. Hes1
expression oscillates with a
period of about 2 h in many
cell types such as fibroblasts.
Hes1 represses its own
expression by directly binding
to the Hes1 promoter. This
negative feedback leads to
disappearance of both hes1
mRNA and Hes1 protein,
because they are extremely
unstable, allowing the next
round of expression. In this
way, Hes1 autonomously
starts oscillatory expression

2005). Thus, Hes1 both inhibits and promotes cell


cycle progression, suggesting that oscillatory expres-
sion is required for efficient cell cycle progression.
In agreement with this notion, cells in the boundaries
such as the isthmus, the roof plate and the floor plate
that express Hes1 persistently proliferate slowly, com-
pared to neural progenitor cells whose Hes1 expression
oscillates (Hirata et al., 2001; Baek et al., 2006). Thus,
different expression mode of Hes1 (oscillatory ver-
sus persistent) leads to different characteristics of cells
(rapidly versus slowly dividing).

Fig. 10.5 Dynamic expression in neural progenitor cells. 10.6 Oscillatory Versus Persistent Hes1
Hes1 expression oscillates with a period of about 23 h in Expression
neural progenitor cells. In these cells, Ngn2 and Delta1 expres-
sion also oscillate but with an opposite phase to Hes1 oscil-
lation. However, Ngn2 and Delta1 expression are persistent in Hes1 expression oscillates in actively proliferating
post-mitotic neurons, which lose Hes1 expression. It is likely
that Delta1 oscillation periodically activates Notch signaling
neural progenitor cells, whereas it is persistent in
between neighboring neural progenitor cells. This reciprocal slowly proliferating boundary cells. The precise mech-
regulation is particularly important for maintenance of neural anism of how oscillatory versus persistent Hes1
progenitor cells before formation of neurons expression is regulated remains to be determined.
10 Rhythmic Expression of Notch Signaling in Neural Progenitor Cells 141

It was found that Socs3 expression also oscillates


with a period of about 2 h in fibroblasts (Yoshiura
et al., 2007). Phosphorylated Stat3 (Stat3-P) induces
Socs3 expression, but Socs3 inhibits phosphorylation
of Stat3, forming a negative feedback loop. As a result,
formation of Stat3-P and expression of Socs3 oscil-
late in the opposite phase (Fig. 10.6) (Yoshiura et al.,
2007). Interestingly, inhibition of Stat3-Socs3 oscilla-
tions blocks Hes1 oscillation, suggesting that Stat3-
Socs3 signaling regulates oscillatory versus persistent
Hes1 expression, although the precise mechanism is
unknown (Yoshiura et al., 2007). It was previously
shown that formation of Stat3-P is inhibited in the
absence of Hes1 (Kamakura et al., 2004), suggest-
ing that Stat3-Socs3 oscillations and Hes1 oscillation
depend on each other (Fig. 10.6).
Another possible mechanism is Id, a HLH factor Fig. 10.6 The oscillator networks in neural progenitor cells.
Phosphorylated Stat3 (Stat3-P) induces Socs3 expression, but
that does not have a basic region. Id can form a het- Socs3 inhibits phosphorylation of Stat3, forming a negative feed-
erodimer with Hes1 through the HLH domains, but back loop. As a result, formation of Stat3-P and expression of
this heterodimer cannot bind to the DNA, because Socs3 oscillate in the opposite phase. It is likely that Stat3-Socs3
Id does not have a basic region. As a result, Id can oscillations and Hes1 oscillation depend on each other. Hes1
oscillation drives Ngn2 and Delta1 oscillations. These oscillators
inhibit Hes1 activity. Id is highly expressed in the are coupled and may regulate proliferation and differentiation of
dorsal midline regions of the neural tube, and it was neural progenitor cells
shown that Id prevents Hes1 from negative autoreg-
ulation, thereby persistently up-regulating Hes1 (Bai expression of only subsets of downstream genes such
et al., 2007). Thus, it is possible that Stat3-Socs3 sig- as Delta1, and persistent expression may be required
naling and Id may regulate oscillatory versus persistent for neuronal differentiation. Hes1 oscillation is also
Hes1 expression. important for efficient proliferation of neural progen-
itor cells, and cells expressing Hes1 persistently such
as boundary cells proliferate slowly.
10.7 Conclusions The adult brain also has many neural stem/
progenitor cells, and continuous neurogenesis is essen-
tial for the maintenance of the brain structure and
It was previously thought that only differentiating neu-
function (Imayoshi et al., 2008). It is known that adult
rons express activator-type bHLH genes and Notch
neural stem cells proliferate very slowly, raising the
ligands, thereby activating Notch signaling of neigh-
possibility that Hes1 could be persistently expressed
boring neural progenitor cells. However, it does not
by these cells. Induction of Hes1 oscillation could lead
explain how Notch signaling is activated in neural pro-
to more efficient expansion and neuronal differentia-
genitor cells before neuronal formation. Recent accu-
tion of adult neural stem cells. Further characterization
mulating evidence indicates that activator-type bHLH
of dynamics of gene expression will help understand
genes and Notch ligands are expressed in salt-and-
the molecular mechanism of how proliferation and dif-
pepper patterns by neural progenitor cells (Kageyama
ferentiation of embryonic and adult neural stem cells
et al., 2008). We have found that expression of the
are controlled.
activator-type bHLH gene Ngn2 and the Notch ligand
Delta1 oscillates in neural progenitor cells, suggesting Acknowledgments This work was supported by the Grants-in-
that these cells reciprocally activate Notch signaling aid from the Ministry of Education, Culture, Sports, Science
by Delta oscillation. Our results may also explain why and Technology of Japan, and the Uehara Memorial Foundation.
neural progenitor cells do not start neuronal differ- H.S. was supported by the twenty-first century Center of
Excellence Program of the Ministry of Education, Culture,
entiation at early developmental stages even though Sports, Science and Technology of Japan and Research
activator-type bHLH genes are expressed. Oscillatory Fellowships of the Japan Society for the Promotion of Science
expression of activator-type bHLH genes may induce for Young Scientists.
142 H. Shimojo et al.

References Hatakeyama J, Tomita K, Inoue T, and Kageyama R (2001)


Roles of homeobox and bHLH genes in specification of a
retinal cell type. Development 128:13131322.
Alvarez-Buylla A, Garcia-Verdugo JM, and Tramontin AD Hatakeyama J, Bessho Y, Katoh K, Ookawara S, Fujioka M,
(2001) A unified hypothesis on the lineage of neural stem cells. Guillemot F, and Kageyama R (2004) Hes genes regulate size,
Nat Rev Neurosci 2:287293. shape and histogenesis of the nervous system by control of
Anthony TE, Klein C, Fishell G, and Heintz N (2004) Radial the timing of neural stem cell differentiation. Development
glia serve as neuronal progenitors in all regions of the central 131:55395550.
nervous system. Neuron 41:881890. Heng JI, Nguyen L, Castro DS, Zimmer C, Wildner H,
Artavanis-Tsakonas S, Rand MD, and Lake RJ (1999) Notch sig- Armant O, Skowronska-Krawczyk D, Bedogni F, Matter JM,
naling: cell fate control and signal integration in development. Hevner R, and Guillemot F (2008) Neurogenin 2 controls
Science 284:770776. cortical neuron migration through regulation of Rnd2. Nature
Baek JH, Hatakeyama J, Sakamoto S, Ohtsuka T, and Kageyama 455:114118.
R (2006) Persistent and high levels of Hes1 expression regulate Hirata H, Tomita K, Bessho Y, and Kageyama R (2001)
boundary formation in the developing central nervous system. Hes1 and Hes3 regulate maintenance of the isthmic orga-
Development 133:24672476. nizer and development of the mid/hindbrain. EMBO J
Bai G, Sheng N, Xie Z, Bian W, Yokota Y, Benezra R, Kageyama 20:44544466.
R, Guillemot F, and Jing N (2007) Id sustains Hes1 expres- Hirata H, Yoshiura S, Ohtsuka T, Bessho Y, Harada T,
sion to inhibit precocious neurogenesis by releasing negative Yoshikawa K, and Kageyama R (2002) Oscillatory expression
autoregulation of Hes1. Dev Cell 13:283297. of the bHLH factor Hes1 regulated by a negative feedback
Bertrand N, Castro DS, and Guillemot F (2002) Proneural genes loop. Science 298:840843.
and the specification of neural cell types. Nat Rev Neurosci Ishibashi M, Moriyoshi K, Sasai Y, Shiota K, Nakanishi
3:517530. S, and Kageyama R (1994) Persistent expression of
Castella P, Sawai S, Nakao K, Wagner JA, and Caudy M (2000) helix-loop-helix factor HES-1 prevents mammalian neu-
HES-1 repression of differentiation and proliferation in PC12 ral differentiation in the central nervous system. EMBO J
cells: role for the helix 3-helix 4 domain in transcription 13:17991805.
repression. Mol Cell Biol 20:61706183. Imayoshi I, Sakamoto M, Ohtsuka T et al. (2008) Roles
Castro DS, Skowronska-Krawczyk D, Armant O, Donaldson of continuous neurogenesis in the structural and func-
IJ, Parras C, Hunt C, Critchley JA, Nguyen L, Gossler A, tional integrity of the adult forebrain. Nat Neurosci
Gttgens B, Matter J-M, and Guillemot F (2006) Proneural 11:11531161.
bHLH and Brn proteins coregulate a neurogenic program Jarriault S, Brou C, Logeat F, Schroeter EH, Kopan R, and Israel
through cooperative binding to a conserved DNA motif. Dev A (1995) Signalling downstream of activated mammalian
Cell 11:831844. Notch. Nature 377:355358.
Chen H, Thiagalingam A, Chopra H, Borges MW, Feder JN, Johnson JE, Birren SJ, Saito T, and Anderson DJ (1992) DNA
Nelkin BD, Baylin SB, and Ball DW (1997) Conservation binding and transcriptional regulatory activities of mammalian
of the Drosophila lateral inhibition pathway in human lung achaete-scute homologous (MASH) proteins revealed by inter-
cancer: a hairy-related protein (HES-1) directly represses action with a muscle-specific enhancer. Proc Natl Acad Sci
achaete-scute homolog-1 expression. Proc Natl Acad Sci USA USA 89:35963600.
94:53555360. Kabos P, Kabosova A, and Neuman A (2002) Blocking HES1
Chien C, Hsiao C-D, Jan LY, and Jan YN (1996) Neuronal expression initiates GABAergic differentiation and induces the
type information encoded in the basic-helix-loop-helix expression of p21(CIP1/WAF1) in human neural stem cells. J
domain of proneural genes. Proc Natl Acad Sci USA Biol Chem 277:87638766.
93:1323913244. Kageyama R, Ohtsuka T, Hatakeyama J, and Ohsawa R (2005)
Fisher AL, Ohsako S, and Caudy M (1996) The WRPW Roles of bHLH genes in neural stem cell differentiation. Exp
motif of the Hairy-related basic helix-loop-helix repres- Cell Res 306:343348.
sor proteins acts as a 4-amino-acid transcription repres- Kageyama R, Ohtsuka T, and Kobayashi T (2007) The Hes gene
sion and proteinprotein interaction domain. Mol Cell Biol family: repressors and oscillators that orchestrate embryogen-
16:26702677. esis. Development 134:12431251.
Fode C, Ma Q, Casarosa S, Ang S-L, Anderson DJ, and Kageyama R, Ohtsuka T, Shimojo H, and Imayoshi I
Guillemot F (2000) A role for neural determination genes in (2008) Dynamic Notch signaling in neural progenitor cells
specifying the dorsoventral identity of telencephalic neurons. and a revised view of lateral inhibition. Nat Neurosci
Genes Dev 14:6780. 11:12471251.
Fujita S (2003) The discovery of the matrix cell, the identi-
Kamakura S, Oishi K, Yoshimatsu T, Nakafuku M, Masuyama
fication of the multipotent neural stem cell and the devel-
N, and Gotoh Y (2004) Hes binding to STAT3 mediates
opment of the central nervous system. Cell Struct Funct
crosstalk between Notch and JAK-STAT signaling. Nat Cell
28:205228.
Biol 6:547554.
Grbavec D and Stifani S (1996) Molecular interaction between
TLE1 and the carboxyl-terminal domain of HES-1 contain- Malatesta P, Hartfuss E, and Gtz M (2000) Isolation of radial
ing the WRPW motif. Biochem Biophys Res Commun 223: glial cells by fluorescent-activated cell sorting reveals a neu-
701705. ronal lineage. Development 127:52535263.
10 Rhythmic Expression of Notch Signaling in Neural Progenitor Cells 143

Masamizu Y, Ohtsuka T, Takashima Y, Nagahara H, Takenaka Y, Ross SE, Greenberg ME, and Stiles CD (2003) Basic
Yoshikawa K, Okamura H, and Kageyama R (2006) Real-time helix-loop-helix factors in cortical development. Neuron
imaging of the somite segmentation clock: revelation of unsta- 39:1325.
ble oscillators in the individual presomitic mesoderm cells. Sasai Y, Kageyama R, Tagawa Y, Shigemoto R, and Nakanishi
Proc Natl Acad Sci USA 103:13131318. S (1992) Two mammalian helix-loop-helix factors structurally
Murata K, Hattori M, Hirai N, Shinozuka Y, Hirata H, related to Drosophila hairy and Enhancer of split. Genes Dev
Kageyama R, Sakai T, and Minato N (2005) Hes1 directly con- 6:26202634.
trols cell proliferation through the transcriptional repression of Selkoe D and Kopan R (2003) Notch and presenilin: regulated
p27Kip1 . Mol Cell Biol 25:42624271. intramembrane proteolysis links development and degenera-
Nieto M, Schuurmans S, Britz O, and Guillemot F (2001) Neural tion. Annu Rev Neurosci 26:565597.
bHLH genes control the neuronal versus glial fate decision in Shimojo H, Ohtsuka T, and Kageyama R (2008) Oscillations
cortical progenitors. Neuron 29:401413. in notch signaling regulate maintenance of neural progenitors.
Noctor SC, Flint AC, Weissmann TA, Dammerman RS, Neuron 58:5264.
and Kriegstein AR (2001) Neurons derived from radial Strm A, Arai N, Leers J, and Gustafsson J- (2000) The Hairy
glial cells establish radial units in neocortex. Nature and Enhancer of Split homologue-1 (HES-1) mediates the pro-
109:714720. liferative effect of 17beta-estradiol on breast cancer cell lines.
Ohsawa R, Ohtsuka T, and Kageyama R (2005) Mash1 and Oncogene 19:59515953.
Math3 are required for development of branchiomotr neurons Sun Y, Nadal-Vicens M, Misono S, Lin MZ, Zubiaga A, Hua
and maintenance of neural progenitors. J Neurosci 25:5857 X, Fan G, and Greenberg ME (2001) Neurogenin promotes
5865. neurogenesis and inhibits glial differentiation by independent
Ohtsuka T, Ishibashi M, Gradwohl G, Nakanishi S, Guillemot F, mechanisms. Cell 104:365376.
and Kageyama R (1999) Hes1 and Hes5 as Notch effectors in Takebayashi K, Sasai Y, Sakai Y, Watanabe T, Nakanishi S, and
mammalian neuronal differentiation. EMBO J 18:21962207. Kageyama R (1994) Structure, chromosomal locus, and
Ohtsuka T, Sakamoto M, Guillemot F, and Kageyama R (2001) promoter analysis of the gene encoding the mouse
Roles of the basic helix-loop-helix genes Hes1 and Hes5 in helix-loop-helix factor HES-1: negative autoregula-
expansion of neural stem cells of the developing brain. J Biol tion through the multiple N box elements. J Biol Chem
Chem 276:3046730474. 269:51505156.
Paroush Z, Finley L JR, Kidd T, Wainwright SM, Ingham PW, Tomita K, Moriyoshi K, Nakanishi S, Guillemot F, and
Brent R, and Ish-Horowictz D (1994) Groucho is required for Kageyama R (2000) Mammalian achaete-scute and
Drosophila neurogenesis, segmentation, and sex determination atonal homologs regulate neuronal versus glial fate
and interacts directly with hairy-related bHLH proteins. Cell determination in the central nervous system. EMBO J
79:805815. 19:54605472.
Parras CM, Schuurmans C, Scardigli R, Kim J, Anderson DJ, Yoshiura S, Ohtsuka T, Takenaka Y, Nagahara H, Yoshikawa K,
and Guillemot F (2002) Divergent functions of the proneu- and Kageyama R (2007) Ultradian oscillations of Stat, Smad,
ral genes Mash1 and Ngn2 in the specification of neuronal and Hes1 expression in response to serum. Proc Natl Acad Sci
subtype identity. Genes Dev 16:324338. USA 104:1129211297.
Chapter 11

Neuron-Astroglial Interactions in Cell Fate Commitment


in the Central Nervous System

Joice Stipursky, Tnia Cristina Leite de Sampaio e Spohr, Luciana Ferreira Romo,
and Flvia Carvalho Alcantara Gomes

Contents actively involved in brain function than was formerly


thought. Recent evidence shows that astroglia, in addi-
11.1 Introduction. Astroglia: Old Cells, New tion to their previously known roles in neurotransmitter
Concepts . . . . . . . . . . . . . . . . . . 146 clearance, ion buffering, and neuronal trophic sup-
11.2 Astroglial Cells and Neurogenesis . . . . . . 147 port, are also involved in other functions, such as
11.2.1 Radial Glia Cells as Progenitor Cells . 147
synapse development and neurogenesis. In this review,
11.2.2 Potential Roles of Astrocytes
in Neurogenic Niches . . . . . . . . 149 we will focus on the role of astroglial cells, mainly
11.3 Role of Neuron-Glia Interactions in Astrocyte radial glia and astrocytes, in several events of nervous
Generation and Maturation . . . . . . . . . 152 system development such as cell fate commitment,
11.3.1 Neuron-Radial Glia Interactions:
neuronal and astrocyte maturation, and synapse forma-
Implications for Radial Glia Maintenance
and Astrocyte Generation . . . . . . 152 tion. We will argue that the functional architecture of
11.3.2 Role of Neuronal-Derived Molecules the brain depends on an intimate neuron-glia partner-
in Astrocyte Differentiation: Crosstalk ship. Finally, we will briefly discuss the emerging view
Between Growth Factors and
of astrocytes as essential actors in neurodegenerative
Neurotransmitters . . . . . . . . . . 156
11.4 Neuron-Astrocyte Interactions: Implications for diseases and neurological disorders, and how a better
Neuronal Differentiation and Synaptogenesis 158 understanding of glial development might open new
11.4.1 Neuron-Astrocyte Interactions avenues to develop therapeutic approaches to these
and Neuronal Differentiation . . . . . 159
pathologies.
11.4.2 Role for Glia in Synaptogenesis . . . . 161
11.5 Concluding Remarks . . . . . . . . . . . . 163
References . . . . . . . . . . . . . . . . . . . . 164 Keywords Astrocyte differentiation Neuronal
differentiation Neuron-astrocyte interactions Radial
glia Synaptogenesis
Abstract Until about 100 years ago, astroglial cells
were regarded solely as passive components of the Abbreviations
nervous system. During the past few years, however,
increasing knowledge of these cells has completely ALS Amyotrophic Lateral Sclerosis
changed this scenario: rather than mere inert brain AMPA -amino-3-hydroxy-5-methyl-4-
glue, astroglial cells are now considered active part- isoxazoleproprionic acid
ners of neurons, and they seem to be much more BLBP brain lipid-binding protein
BMP bone morphogenic protein
BrdU bromodeoxyuridine
CNTF ciliary neurotrophic factor
F.C.A. Gomes () CT-1 Cardiotrophin-1
Laboratrio de Neurobiologia Celular, Programa de Biologia E embryonic day
Celular e do Desenvolvimento, Instituto de Cincias
Biomdicas, Centro de Cincias da Sade, Universidade ECM extracellular matrix
Federal do Rio de Janeiro, Rio de Janeiro, Brazil EGF epidermal growth factor
e-mail: fgomes@anato.ufrj.br EGL external granular layer

H. Ulrich (ed.), Perspectives of Stem Cells, 145


DOI 10.1007/978-90-481-3375-8_11, Springer Science+Business Media B.V. 2010
146 J. Stipursky et al.

FGF fibroblast growth factor The above concept was developed by the German
-Gal -galactosidase pathologist Rudolf Virchow, who in the mid-nineteenth
GFAP glial fibrillary acidic protein century, by analyzing postmortem human tissues, pos-
GFP green fluorescent protein tulated that neuroglia was a connective tissue, acellular
GLAST astrocyte-specific glutamate-aspartate in nature (Virchow, 1846, quoted from Somjen, 1988).
transporter In 1851, the first neuroglial cells would be described by
GLT-1 glutamate transporter-1 Heinrich Mller, in the retina of several species (fish,
IGL internal granular layer amphibian, bird, and human): the radial cells, known
iGlu receptor ionotrophic glutamate receptor today as the Mller cells. Also in the mid-nineteenth
IL-6 interleukin-6 century, Otto Deiters provided illustrations of a cell
LGE lateral ganglionic eminence
type that resembles our modern notion of an astrocyte.
LIF leukemia inhibitor factor
Further contributions in the field of the cellular origin
LIFR leukemia inhibitor factor receptor beta
of glial cells resulted from the efforts of several histol-
LPA lysophosphatidic acid
ogists, in particular Camillo Golgi, Santiago Ramn y
MAPK mitogen-activated protein kinase
MGE medial ganglionic eminence Cajal and Po Del Ro Hortega (Somjen, 1988). Using
mGlu receptor metabotropic glutamate receptor a variety of microscopy techniques, they described a
NGF nerve growth factor wide diversity of glial cells in the brain. Although
NMDA N-methyl-d-aspartate observations of the intimate relationship between these
NMJ neuromuscular junction cells and blood vessels or nerve fibers might suggest a
NPC neural progenitor cell(s) functional role for glia, these cells were regarded as
NRG neuregulin(s) merely supportive and passive elements, completely
NS nervous system devoid of any role in brain function, for at least a
NSC neural stem cell(s) hundred years after their description. The past decade,
PI3-K phosphatidylinositol 3-K however, has been marked by a thorough revisitation of
PNS peripherical nervous system the role of glial cells in healthy brains, and especially
RA retinoic acid in brain disease.
RG radial glia Glial cells are classified into two main groups:
RGC retinal ganglion cells microglia, which is the subject of Chapter 12 of
SGZ subgranular zone this book; and macroglia, which are discussed here.
SVZ subventricular zone Macroglia are subdivided into four main specialized
T3 thyroid hormone cell types: ependymal cells, Schwann cells, oligo-
TGF- transforming growth factor- dendroglia, and astroglia. The last type includes the
TGF- transforming growth factor- astrocytes, marginal glia, radial glia in the developing
TGFRI transforming growth factor- brain and spinal cord, Bergmann cells in the cerebel-
receptor I
lar cortex, Mller cells in the retina, pituicytes in the
TGFRII transforming growth factor-
neurohypophysis, and tanycytes in the hypothalamus
receptor II
(Kettenmann and Ransom, 2005). Here we will focus
TSP thrombospondin(s)
on two subpopulations of astroglial cells: astrocytes
VZ ventricular zone
Wnt wingless and radial glia (RG) cells.
Evidence accumulating over the past decade has
revealed that neuron-astroglia interactions play key
roles in several events of brain development, such as
11.1 Introduction. Astroglia: Old Cells, the proliferation and differentiation of neuronal pre-
New Concepts cursors (Lim and Alvarez-Buylla, 1999; Song et al.,
2002; Krnyei et al., 2005; Lie et al., 2005; de
Sampaio e Spohr et al., 2008), neuronal migration
. . . this connective substance forms in the brain, in (Hatten, 2002), axonal guidance (Garcia-Abreu et al.,
the spinal cord, and in the higher sensory nerves a sort
of Nervenkitt (neuroglia), in which the nervous system
1995; Martinez and Gomes, 2002, 2005), synapse for-
elements are embedded Rudolf Virchow mation (Christopherson et al., 2005; Stevens et al.,
11 Neuron-Astroglial Interactions in Cell Fate Commitment 147

2007), and glial maturation (Gomes et al., 1999a; the ventricular surface of the neural tube, the neurons
de Sampaio e Spohr et al., 2002; Schmid et al., must migrate to their final destination. The success of
2003; Sousa et al., 2004; Barnab-Heider et al., 2005; their migration is partly dependent on the interaction
Stipursky and Gomes, 2007). Recently, RG cells and of postmitotic neurons and the system of RG fibers.
astrocytes were described as neural stem cells in the Although RG cells were originally described as guides
developing and adult brain, respectively (Noctor et al., for neuronal migration, the prevailing view is that RG
2001; Alvarez-Buylla and Lim, 2004). constitute the main NPC for neurons and astrocytes
Astroglial cells are emerging as key mediators in most regions of the nervous system (Gtz et al.,
of brain development, function, and plasticity, high- 2002).
lighting the critical need to better characterize the Until the mid-twentieth century, it was assumed
mechanisms underlying their development and inter- that no new neurons are generated after brain forma-
action with neurons. Although our understanding of tion is completed. This idea, however, gave place to
these cells has expanded dramatically during the past the description of neurogenic areas in the adult brain
decade, a great deal of mystery still surrounds the role of different species. The recent identification of the
of astrocytes and RG cells in brain development and astroglial origin of these progenitor cells assigned a
injury. new attribute to astrocytes, not previously discussed.
In this chapter, we will discuss the conceptual shift In this section, we will discuss the role of astroglia,
of the role of astroglia-neuron interactions in brain mainly RG cells and adult astrocytes, in neurogenesis,
development. We argue that astroglial cells should not both as progenitor cells themselves, and as a source of
be viewed primarily as support cells, but rather as cells trophic support in neurogenic areas.
that actively control the structural and functional orga-
nization of the developing and mature brain. We begin
by reviewing in vitro and in vivo studies that have
demonstrated a relatively novel attribute of RG cells 11.2.1 Radial Glia Cells as Progenitor Cells
and astrocytes, as neural progenitor cells (NPC) in the
developing and adult nervous system, respectively. We The radial glia cells were described in the mammal
then discuss the role of neuron-astroglia interactions in fetal encephalon at the end of the nineteenth century by
cell fate commitment, with a special focus on the tim- Giuseppe Maggini (1888) (for review see Bentivoglio
ing of neurogenesis versus gliogenesis and astrocyte and Mazzarello, 1999). These cells have a bipolar
maturation. Finally, we provide evidence of the role morphology, with the cell body located in the ventric-
of astrocytes in synapse development, and the conse- ular zone (VZ), and cell processes extending from the
quences of astroglia dysfunction for neurodegenerative ventricular through the pial surfaces.
diseases and neurological disorders. Classical studies in several species, including
humans (Choi and Lapham, 1978; de Azevedo
et al., 2003), non-human primates (Schmechel and
Rakic, 1979), carnivores (Voigt, 1989), and rodents
11.2 Astroglial Cells and Neurogenesis (Takahashi, 1990), have suggested that RG cells trans-
form into astrocytes during cerebral cortical develop-
One of the greatest challenges of neuroscience is to ment, as we will further address in Section 11.3.1 (for
understand how the widely diverse cell types of the review see Bentivoglio and Mazzarello, 1999). The
nervous system (NS) are generated. The vertebrate use of electron microscopy and immunohistochemistry
NS originates as a flat sheet of neuroepithelial cells, applied to primate embryos has confirmed the glial
which gives rise to the great variety of glial and neu- phenotype of radial cells (for review see Rakic, 2003).
ronal types that form the adult brain (Kandel et al., At the end of neurulation, the CNS is made up of
2000). Shortly after the appearance of the first neurons, a pseudostratified epithelium, where radially arranged
neuroepithelial cells undergo a change in their charac- bipolar cells span the entire thickness of the neural
teristics and acquire molecular and cytological features tube. These neuroepithelial cells, like earlier progen-
typical of the astroglial lineage, as they give rise to the itors, undergo a characteristic alternate movement of
RG cells (Malatesta et al., 2008). After their birth in the nucleus (interkinetic nuclear migration) between
148 J. Stipursky et al.

the basal and the apical surface of the neural tube. This them to be stained with a lipophilic dye applied to
migration is synchronized with the cell cycle, and these the pial surface (Malatesta et al., 2000). The analysis
cells undergo symmetrical cell division on the luminal demonstrated that the majority of RG cells generate
surface of the neural tube. From this stage, however, homogeneous clones, composed either of neurons or
an increasing number of cells begin to divide asym- of non-neuronal cells (glia and/or progenitor cells)
metrically, giving rise to another neuroepithelial cell exclusively. The generation of neurons from RG in
and to either a neuron, or, alternatively, to a progenitor the cerebral cortex was corroborated by video time-
cell that will undergo mitosis at a significant distance lapse analyses showing the generation of a radial
from the ventricular surface, the basal progenitors, unit by the asymmetric division of a single RG cell
which will generate only neurons via symmetrical divi- (Noctor et al., 2001). Such radial units were com-
sion. However, shortly after the appearance of the posed of the RG cell itself plus multiple postmitotic
first neurons, neuroepithelial cells give rise to the RG neurons. Subsequently, in vivo lineage-tracing studies
cells. Although RG cells maintain the morphology and based on transgenic-mice technology showed that RG
intermediate filament nestin expression similar to the is the main source of postmitotic neurons in the cere-
neuroepithelial cells, some of the features that define bral cortex (Malatesta et al., 2003), and possibly in the
the neuroepithelial-to-RG transition are the expres- entire telencephalon (Anthony et al., 2004). Both stud-
sion of genes that are typical of astrocytes, such as ies were based on transgenic mouse lines where Cre-
the brain lipid-binding protein (BLBP), the astrocytic recombinase expression was driven by RG-specific
glutamate-aspartate transporter (GLAST), the adhe- promoters: the hGFAP- (Malatesta et al., 2003) and
sion molecule TN-C, the enzyme glutamine synthase, the BLBP-promoter (Anthony et al., 2004). Although
the calcium-binding protein S100, the intermediate both studies agreed that RG cells are responsible for
filament vimentin, and, in some primates (but not in the generation of all projection neurons of the cerebral
rodents), the intermediate filament the glial fibrillary cortex, discrepancies were found regarding the role of
acidic protein, GFAP. Also, glycogen granules begin RG in the generation of ventral telencephalic neurons
to accumulate in the RG cytoplasm. Notably, apart (Malatesta et al., 2008).
from the peculiar shape, in many species no markers RG as well as their neurogenic potential appear to
are known that allow discrimination of RG from astro- be highly heterogeneous, both within and across differ-
cytes. This is the case, for instance, in primates, where ent brain regions (Kriegstein and Gtz, 2003). Cell-fate
the immunoreactivity for GFAP is shown by both cell mapping studies suggested that this diversity might
types; whereas in rodents it is limited to astrocytes be associated with the expression of different tran-
(Malatesta et al., 2008). scription factors such as Pax6, Olig2, and Gsh2 (Gtz
Initially described as only related to neuronal migra- et al., 1998; Malatesta et al., 2003). In the dorsal telen-
tion and cerebral cortex lamination (Hatten, 1999; cephalon, for example, RG cells contain the transcrip-
Rakic, 1971), RG cells are today well recognized for tion factor Pax6 (Gtz et al., 1998), whereas those from
their progenitor potential in all regions of the CNS the lateral ganglionic eminence (LGE) contain Gsh2,
(Anthony et al., 2004). They are considered a major and those in the medial ganglionic eminence (MGE)
intermediate between neural stem cells and cerebral contain Olig2 (Malatesta et al., 2003). All these tran-
cortex neurons (Gtz and Barde, 2005). Several obser- scription factors have been implicated as potent cues in
vations, both in vitro and in vivo, have shown that, patterning the developing neural tube (Sun et al., 2001;
during neurogenesis, RG cells generate mostly neurons Carney et al., 2008). Recent fate mapping of the entire
(Malatesta et al., 2000; Noctor et al., 2001; Anthony progeny of RG cells has demonstrated pronounced dif-
et al., 2004). The first direct observation of this event ferences in the ventral and dorsal telencephalon: while
came from the analysis of the differentiation of RG iso- cortical RG cells generate the vast majority of neu-
lated by fluorescence-activated cell sorting (Malatesta rons (more than 90%) in the cerebral cortex, RG in
et al., 2000). Cells were labeled by exploiting two inde- the ventral telencephalon generates very few neurons
pendent characteristics of RG: the ability to express (Malatesta et al., 2003). Besides RG heterogeneity
green fluorescent protein (GFP) under the control of within the brain, other RG-like cells also exist through-
the human GFAP (hGFAP) promoter, and the pres- out the CNS, such as the Mller glia in the retina and
ence of a contact with the basal membrane, allowing the Bergmann glia in the cerebellum (Pinto and Gtz,
11 Neuron-Astroglial Interactions in Cell Fate Commitment 149

2007). These cells share some morphological and func- features. As we will address in more detail in the fol-
tional features with cerebral cortical RG, such as their lowing sections (11.2.2 and 11.3.1), identification of
radial morphology and role as neuronal guides. the molecular cues that regulate RG identity might rep-
During cerebellar development, Bergmann glial resent a crucial step in understanding neurogenesis in
cells support the migration of neurons from their birth- both the developing and adult brain.
place in the external granular layer (EGL) to their
final position in the internal granular layer (IGL)
(Nicholson and Altman, 1972). As cortical RG, they
11.2.2 Potential Roles of Astrocytes
express BLBP, GLAST, and vimentin. In contrast to
rodent cortical RG, Bergmann glia cells express GFAP. in Neurogenic Niches
Recently, it was observed that Bergmann glia express
stem cell markers of the Sox family of transcrip- A classical dogma of developmental neuroscience is
tion factors (Sox/Sox2) (Sottile et al., 2006); however, that, once neurons in the adult CNS are terminally
a progenitor potential has not yet been reported for differentiated, they persist through the life of the organ-
these cells. In the retina, the Mller glia represents ism, and are not replaced when they die. This idea,
the main glial cell type, which crosses the outer and however, began to change in the 1960s with the obser-
inner layers of the retina with a radial characteristic vation of thymidine-incorporating cells with neuronal
morphology. This cell expresses cell markers such as characteristics in the postnatal brain (Altman and Das,
vimentin, GLAST, and the astrocyte-specific markers 1966). Later, a series of experiments in several dif-
GFAP and glutamine synthase (Okada et al., 1990; ferent species, including birds, mice, and humans,
Vardimon et al., 1993). In addition to their role in neu- demonstrated ongoing neurogenesis in the adult verte-
ronal migration, Mller glial cells were indicated as brate brain (Goldman and Nottebohm, 1983; Galileo
the retina potential stem cells, implicated in generation et al., 1990; Alvarez-Buylla et al., 2001; Temple,
of new neurons after injury (Raymond and Hitchcock, 2001). This phenomenon occurs through the persis-
1997). Recent in vivo and in vitro works identified tence of niches that harbor neural stem cells (NSC)
stem-cell-like properties, including self-renewal and and constitute a microenvironment that constantly
multipotency, in mammalian Mller glia (Das et al., supports and regulates neurogenic activity (Alvarez-
2006; Bernardos et al., 2007) and immortalized human Buylla and Lim, 2004). Surprisingly, evidence accu-
Mller cells (Lawrence et al., 2007). mulating in the last 10 years has supported an essential
Self-renewal and neuronal generation capacity are role for glial cells in NSC adult niches. In this section,
key features that are lost during the transformation we will discuss the role of astroglial cells in adult neu-
of RG into astrocytes at the end of neurogenesis in rogenesis, either by acting as primary precursors, or
the mammalian brain. However, in lower vertebrates, by exerting influence on other cell types as supporting
where neurogenesis persists in a rather widespread cells.
fashion in the adult brain, RG with access to the Two germinal regions within the adult mammalian
ventricle also persists into adulthood. Interestingly, brain have been shown to contain active neurogenesis
the persistence of RG may also be responsible for throughout life: the subventricular zone (SVZ) of the
the success of axonal regeneration in the CNS of lateral ventricles (Alvarez-Buylla and Lim, 2004), and
non-mammalian vertebrates, as they provide a good the subgranular zone (SGZ) of the dentate gyrus in the
substrate for axonal growth consistent with their nor- hippocampus (Song et al., 2002). Neurogenesis out-
mal role during development (for review see Pinto and side these two regions appears to be extremely limited,
Gtz, 2007). or nonexistent, in the intact adult mammalian CNS
Taken together, the maintenance of RG cells or their (Alvarez-Buylla and Lim, 2004).
key aspects of embryonic development seems to be a Strikingly, in both SVZ and SGZ, a subset of
crucial feature to allow neuronal repair. Not only is the astrocytes that is classically associated with support
persistence of RG cells in many non-mammalian ver- functions in the brain was identified as the in vivo
tebrates highly correlated with the presence of stem primary precursors for adult neurogenesis (Doetsch
cells and adult neurogenesis, but in addition, neural et al., 1999; Seri et al., 2001). These cells have been
stem cells in the adult mammalian brain retain RG defined as astrocytes based on their ultrastructural
150 J. Stipursky et al.

features, markers that they express, and electrophysi- growth factor (EGF) family, transforming growth fac-
ological properties. Supporting the idea of astrocytic tor (TGF), and neuregulins (NRG); fibroblast growth
stem cells, SVZ and SGZ astrocytes remain labeled factor (FGF), nerve growth factor (NGF), and ciliary
with thymidine or bromodeoxyuridine (BrdU) after neurotrophic factor (CNTF). By providing trophic sup-
long survival times, indicating their stem-cell property port, astrocytes might affect several events of brain
(Doetsch et al., 1999; Seri et al., 2001). Further, they development, such as neuronal precursor proliferation,
are capable of repopulating the niche after the elimina- cell fate commitment, and synaptogenesis, as we will
tion of other cell types by the anti-mitotic compound address later in this chapter.
Ara-C (Doetsch et al., 1999). An additional support We demonstrated that EGF secreted by cultured
for astrocytes as the NSC in SVZ and SGZ was pro- cerebellar astrocytes in response to thyroid hormone
vided by genetic ablation in vivo. Removal of GFAP- (T3) induces neuronal cerebellar proliferation in vitro
positive cells eliminated the ability of the germinal (Gomes et al., 1999b). Later, we demonstrated that
zone to reconstitute itself over time (Morshead et al., cocultures of astrocytes and neurons potentiate this
1994). effect, suggesting that neuron-astrocyte interaction is
In addition to astrocytes, some workers have pre- a key element in cerebellar morphogenesis (Martinez
sented data suggesting that ependymocytes might also and Gomes, 2005).
represent neural stem cells (Johansson et al., 1999). Several growth factors and signaling pathways that
However, this view is not generally accepted, because are present in astrocytes have a crucial role in adult
other reports have demonstrated that although ependy- neurogenesis, suggesting that these cells might orches-
mal cells can proliferate in vitro, they do not generate trate the proliferation in neurogenic niches. This is the
neurons (Chiasson et al., 1999), and do not proliferate case of FGF, which is important for maintenance of
in vivo (Spassky et al., 2005). the NSC pool (Zheng et al., 2004); Notch1, which is a
The NSC of the SVZ, also referred to as Type crucial regulator of NSC maintenance and self-renewal
B cells, are embedded in a peculiar niche that also within the neurogenic SVZ niche (Alvarez-Buylla
contains rapid-cycling transit-amplifying progenitor and Lim, 2004); CNTF, which promotes self-renewal
cells (Type C cells) and migrating neuroblasts (Type of neural precursors in vitro by increasing Notch1
A cells), which are guided towards the olfactory expression (Chojnacki et al., 2003); and transforming
bulb, where they become functional neurons (Doetsch, growth factor- (TGF-), the endogenous ligand of the
2003). Type B cells have astroglial characteristics and EGF receptor, which controls the pool of progenitors
express GFAP, nestin, vimentin, GLAST, and Sox2, by modulating the proliferation of transit-amplifying
but not S100 (another marker for astrocytes); accu- cells, the intermediate stage between the neural stem
mulate glycogen granules in their cytoplasm; and cells and neuroblasts (Doetsch et al., 2002).
are relatively quiescent (for review see Malatesta Astrocytes might also influence neuronal fate com-
et al., 2008). There is evidence that stem cells in mitment rather than precursor proliferation. Neuronal
the hippocampus SGZ may correspond to the type differentiation from adult NPC in the neurogenic niche
B cells that were described in the SVZ (Seri et al., proceeds partly because of the local presence of bone
2001). morphogenic protein (BMP) antagonists. BMP was
Adult SVZ stem cells (Type B cells) have been shown to instruct adult NPC to adopt a glial fate (Lim
shown to be related to embryonic ventricular RG. The et al., 2000; Zheng et al., 2004). Ependymal cells in
conversion of RG into Type B cells involves retraction the SVZ secrete Noggin (Lim et al., 2000), and astro-
of the RG basal processes, loss of the radial morphol- cytes in the SGZ secrete neurogenesin-1 (Ueki et al.,
ogy, and a slowing of the cell cycle (Merkle et al., 2003), both BMP antagonists, in the SVZ and SGZ
2004). Supporting this morphological feature, RG and respectively. Another growth factor that was recently
Type B cells share molecular regulatory mechanisms, identified as being produced by astrocytes, at least in
such as expression of the neurogenic transcription fac- vitro, and induces NSC differentiation is retinoic acid
tor Pax 6 and activation of the Notch signaling pathway (RA). RA is one of the most powerful morphogenic
(Malatesta et al., 2008). molecular regulators of neuronal cell-fate commit-
Astrocytes constitute the main source of trophic fac- ment. Recently, it was demonstrated that RA can be
tors in CNS. These include members of the epidermal produced by astrocytes in vitro; however, in vivo it
11 Neuron-Astroglial Interactions in Cell Fate Commitment 151

was demonstrated that exposure to exogenous retinoids of the EGF family, such as TGF- and NRG, are key
or blocking RA signaling interferes with prolifera- elements in this event. Sharif and colleagues (2006)
tion and differentiation within the neurogenic zones reported that TGF- is an inducer of adult astrocyte
of the mature brain, such as the higher vocal center dedifferentiation in vitro. TGF- treatment induced
in songbirds, or the SVZ and hippocampus in mice. the appearance of RG phenotype features in astro-
Endogenous RA has recently been shown to be active cytes, such as expression of BLBP and RC2, radial
within the adult stem-cell niches, including the SVZ, morphology, support of neuronal migration, and, more
the rostral migratory stream, and the hippocampus. It surprisingly, neuron generation.
was also demonstrated that there are RA-responsive As we will discuss in Section 11.3.1 of this chapter,
astrocytes in the SVZ and a population of RA-activated Eva Antons group has shown that the NRG-ErbB2 sig-
astroglial cells within the hippocampus of RA reporter naling is essential for RG maintenance (Schmid et al.,
mice (Krniey et al., 2007). 2003). More recently, the same group has shown that,
Another astrocyte-derived signal that modulates in fact, the reinduction of the ErbB2 receptor expres-
adult neurogenesis is the Wnt (wingless) pathway. sion in adult astrocytes promotes their dedifferentiation
The Wnt family of secreted glycoproteins is known into RG cells (Ghashghaei et al., 2007). Similarly, rein-
to play crucial roles in regulating other somatic stem duction of the transcription factor Pax6, by Magdalena
cells in vivo (Lobo et al., 2007). Interestingly, Wnt3 Gtzs group, in postnatal astrocytes derived from
is expressed by astrocytes of the adult hippocam- non-neurogenic regions enables these cells to generate
pus (Lie et al., 2005). Overexpression of Wnt3 leads neurons (Berninger et al., 2007). Together, these data
to increased neuronal fate specification of adult hip- open the possibility that manipulation of astrocyte dif-
pocampal NSC and increased proliferation of neurob- ferentiation may represent a potential tool for neuronal
lasts, while blockade of Wnt signaling results in reduc- replacement in injury.
tions in the generation of new neurons both in vitro and What are the differences between neurogenic and
in vivo. These data point to Wnt signalling as a prin- non-neurogenic astrocytes? What makes the SVZ and
cipal regulator of adult hippocampal neurogenesis (Lie SGZ special in supporting the proliferation and neu-
et al., 2005). On the other hand, it has been shown that ronal differentiation of multipotent neural progenitors?
the transforming growth factor beta (TGF-) signal- Answers to these questions will lead to better under-
ing pathway might also participate in the neurogenic standing of adult neurogenesis in both healthy and
process in the adult hippocampal region (Wachs et al., diseased individuals.
2006). It has been shown that TGF-1, which is also Recent evidence indicates that astrocytes of the
known to be secreted by astrocytes as we will discuss SVZ and SGZ are strongly influenced by their local
further, promotes a decrease in the proliferation in the microenvironment. However, the environment alone
hippocampus. These data are strengthened by a recent does not seem to be sufficient to induce non-germinal
paper showing that the expression of Noggin, a BMP astrocytes to behave as neural stem cells, suggest-
inhibitor, enhances neurogenesis in the hippocampus, ing that specific cell-intrinsic differences between
indicating that the TGF- family of proteins is crucial germinal-center astrocytes and other astrocytes may
for these events in the adult telencephalon (Bonaguidi exist. The S100 protein known to be expressed by
et al., 2008). many mature astrocytes is not expressed by most SVZ
Within this scenario, a question arises: To what astrocytes or by the radial astrocytes of the SGZ (Seri
extent is an astrocyte considered a cell at a termi- et al., 2004; Raponi et al., 2007). The astrocytes in
nal stage of differentiation? In the last few years, the SVZ that express both S100 and GFAP gen-
several studies have indicated that the astrocytes are erate significantly fewer neurospheres than do astro-
extremely plastic cells, in terms of their dedifferentia- cytes that express GFAP alone, indicating that S100
tion capacity. Since the 1990s, evidence has shown that expression denotes mature astrocytes that do not
the embryonic encephalon contains soluble factors that act as stem cells (Raponi et al., 2007). Interestingly,
promote astrocyte differentiation, and has clarified the horizontal astrocytes of the SGZ divide and express
discussion of astrocyte differentiation as a reversible S100, and they are thought to function as local oligo-
event (Hutter and Hatten, 1995). Growing evidence dendroglial progenitors within the hilus (Seri et al.,
from the past few years has indicated that the members 2004). It was also shown recently that the LeX-positive
152 J. Stipursky et al.

GFAP-positive subpopulation of germinal astrocytes 11.3.1 Neuron-Radial Glia Interactions:


expressing GFAP, contains the majority of multipo- Implications for Radial Glia
tent neural stem cells that are able to form multi-
Maintenance and Astrocyte
potent neurospheres; whereas LeX-negative GFAP-
expressing astrocytes are unable to do this (Imura
Generation
et al., 2006). One important point is that astrocytes
derived from the cerebral cortex are LeX-negative and Development of the vertebrate CNS is achieved
non-neurogenic, suggesting the existence of significant through a common pool of precursor cells that sequen-
phenotypic and functional differences between these tially generate neurons and glial cells (Bayer and
two types of astrocytes (Ihrie and Alvarez-Buylla, Altman, 1991). In rodents, neurons are generated
2007). In addition to S100 and LeX, other proteins from embryonic day (E) 12 to E18; astrocytes appear
are also emerging as potential markers for germinal- around E18, with their numbers peaking in the neona-
zone astrocytes, including brain-lipid binding protein, tal period, and mature oligodendrocytes appear later.
nestin, and Sox2 (Filippov et al., 2003; Steiner et al., Thus, multipotent precursors change their competence
2006). over time to generate different cell types. The ability of
Therefore, it will become extremely important to embryonic cortical precursors to make neurons when
collect more information on the characteristics of cultured on embryonic cortical sections, but to make
astrocytes in the neurogenic regions in normal or dis- astrocytes when cultured on postnatal cortical sections
ease conditions. This will be a key to developing (Morrow et al., 2001), suggests that the neurogenic-
means of effectively modulating neurogenesis in dis- to-gliogenic switch might depend on environmental
ease states. Understanding the molecular regulators signals rather than only on an intrinsic developmental
that normally control the fate of neural stem cells program.
and their progeny in the adult CNS may open new As discussed previously, one of the main neural
avenues to develop alternative therapeutic approaches precursors of the nervous system is the RG cells.
to nervous-system injury. Whereas in most vertebrates (fish, amphibians, rep-
tiles, birds) these cells persist into adulthood, in most
CNS regions of adult mammals they transform into
11.3 Role of Neuron-Glia Interactions astrocytes (for review see Bentivoglio and Mazzarello,
1999). The most direct evidence for this transformation
in Astrocyte Generation was obtained in the cerebral cortex of ferrets by detec-
and Maturation tion of fluorescent tracers in astrocytes after labeling
RG pial processes (Voigt, 1989). Later, this event was
The Neuron Doctrine, which was created in the late demonstrated for several vertebrate species including
nineteenth century and which considered neurons as humans (for review see Bentivoglio and Mazzarello,
the only cells with physiological relevance for NS 1999).
function, kept us in the dark concerning glial devel- The most prevalent view is that RG-astrocyte trans-
opment. While there is compelling evidence of the formation is initiated by detachment of the ventricular
effect of astrocyte factors on neurons, there is still a endfeet; the cell body moves upwards to a certain
lack of data on their effects on astrocytes. However, depth in the cortex, maintaining its pial attachment.
although this subject has been less addressed, evidence After extending a few processes, the RG detaches
has accumulated that indicates that neurons are modu- from the pia and progressively assumes the typical
lators of astrocyte differentiation. In this section, we astrocytic morphology (Voigt, 1989; Takahashi et al.,
will describe the emerging evidence for the role of 1990). The RG-astrocyte transition is a hallmark of
neuron-glia interactions in cell fate determination, and the neurogenesis-to-gliogenesis switch in the cerebral
the implications for the timing of neurogenesis and gli- cortex. However, although RG-astrocyte transforma-
ogenesis. Further, we will discuss recent studies on the tion is well recognized, the molecular mechanisms
role of neuronal-derived factors such as neurotransmit- underlying this process are little understood.
ters and growth factors in RG identity and astrocyte The fact that gliogenesis coincides with the end of
maturation. neurogenesis, a period of generation of a large number
11 Neuron-Astroglial Interactions in Cell Fate Commitment 153

of neurons, suggests that neurons might signal for gli- where it converts the CBF1 repressor complex into
ogenesis. Further, the double function of RG during an activator gene-transcription complex (Yoon and
CNS development, both as neuronal/astrocyte pro- Gaiano, 2005). The signaling pathway initiated by
genitors and as a scaffolding supporting neuronal Notch 1, which is present in NPC, promotes the RG
migration (Campbell and Gtz, 2002) suggests that phenotype in the embryonic telencephalon (Gaiano
there may be intimate links between the signaling and Fishell, 2002) and the cerebellum (Eiraku et al.,
pathways that control RG development, neurogene- 2005). This event implies cooperation between Notch
sis/astrocytogenesis, and migration. and other growth factors, such as FGF and NRG
One of the first evidences of neuronal modula- (Patten et al., 2003; Schmid et al., 2003; Yoon et al.,
tion of the RG phenotype came in 1985, with the 2004).
demonstration that cultures of cerebellar neurons and The NRG are a family of proteins containing an
astrocytes induce a morphological, functional, and EGF-like motif that activates membrane-associated
molecular differentiation into RG (Hatten, 1985). tyrosine kinases related to the EGF receptor (known as
Later, the same group showed that expression of ErbB-1). It has been reported that the NRG 1-ErbB2
RG cell identity is regulated in the mammalian fore- signaling is essential to the establishment of RG in the
brain by the availability of diffusible inducing signals cerebellum (Patten et al., 2003). On the other hand, the
that are present in the embryonic brain (Hunter and negative modulation of ErbB2 promotes its transfor-
Hatten, 1995). mation into astrocytes in the cerebral cortex (Schmid
The idea that the neuron-RG interaction is a crit- et al., 2003), and the introduction of the constitu-
ical event for the establishment of RG identity and tive active form of the ErbB2 receptor into mature
morphology was further supported by several in vivo astrocytes promotes its dedifferentiation into RG cells
and in vitro lines of evidence (Feng and Heintz, (Ghashghaei et al., 2007). Similarly, the ErbB4 recep-
1995; Anton et al., 1997; Soriano et al., 1997; Supr tor is also involved in the regulation of the timing
et al., 2000). Ablation of Cajal-Retzius cells in the of gliogenesis in the cerebral cortex, modulating the
cerebral cortex leads to a dramatic decrease in the transcription of glial genes during development (Sardi
number of RG apical processes, inducing prema- et al., 2006).
ture astrocyte differentiation (Supr et al., 2000). The Wnt/catenin signaling pathway has been indi-
Complementary transplantation experiments showed cated as inducing the progenitor phenotype in the
that embryonic Cajal-Retzius cells grafted to cerebel- mammal brain. Wnts are secreted glycoprotein sig-
lar tissue induce a rejuvenation of Bergmann astro- naling molecules that regulate many developmental
cytes into an RG phenotype (Soriano et al., 1997). events in vertebrates and invertebrates. In mammals,
Further compelling evidence has demonstrated that, previous studies showed that Wnt signaling regu-
in addition to modulating RG morphology, neurons lates the development of the dorsal neural tube (Zhou
also modulate the expression of RG molecular mark- et al., 2004). It has been shown that in LRP6 mutant
ers such as BLBP (Feng and Heintz, 1995; Patten et al., mice, a co-receptor for the Wnt/catenin signaling
2003). pathway shows several abnormalities in the produc-
Although the radializing soluble factor has not tion of granular cells in the dentate gyrus of the
been identified, several molecules and signaling path- hippocampus and the formation of RG fibers (Zhou
ways activated by neurons have been indicated as et al., 2004). Moreover, it seems that the synergistic
important for maintenance of RG identity, including interaction of Wnt and Notch confers neural stem-
Notch, NRG-ErbB, Wnt, meteorin, and others (Zhou cell properties on the retinal Mller glia (Das et al.,
et al., 2004; Nishino et al., 2004; Ever and Gaiano, 2006).
2005). During encephalic development, several molecules
The Notch signaling pathway is best character- interact synergistically or antagonistically to deter-
ized as mediating cellcell signaling between adja- mine the correct time of cell generation. As mentioned
cent cells. Both the ligands, members of the Delta above, the onset of gliogenesis coincides with the
and Jagged families, are able to bind the membrane- end of neurogenesis, suggesting a possible role for
associated Notch molecule and activate the Notch the growing pool of newly generated neurons as a
intracellular domain, which translocates to the nucleus source of soluble factors that promote gliogenesis. In
154 J. Stipursky et al.

fact some studies indicate that cytokine signaling is (Stipursky and Gomes, 2007). The TGF- superfamily
important, and the appropriate timing of astrogene- is constituted by multifunctional polypeptide mem-
sis can be mimicked in isolated clones of precursor bers, which perform critical functions in tissue repair
cells, which seems to argue that a cell-intrinsic mech- and development (Shi and Massague, 2003). In the
anism regulates the onset of gliogenesis (Qian et al., NS, TGF-1 has been implicated in organization of
2000). Because such clones contain both precursors the glial scar in response to injury and in several
and newly born neurons, the hypothesis is that newly neurodegenerative diseases (Zhu et al., 2002; Vivien
born neurons secrete gliogenic factors that feed back and Ali, 2006). Emerging evidence, however, has
to instruct multipotent precursors to generate astro- suggested a key role for this factor in several NS
cytes. Within this idea, several studies have suggested developmental processes such as cell adhesion, neu-
that neurons participate in glial differentiation in the ronal migration and differentiation, synaptogenesis,
telencephalon. and blood-brain barrier formation (de Sampaio e Spohr
Some of the best-studied soluble factors are the et al., 2002; Brionne et al., 2003; Miller 2003; Garcia
leukemia inhibitor factors of the interleukin-6 (IL-6) et al., 2004; Stipursky and Gomes, 2007; Feng and Ko,
family, including CNTF, leukemia inhibitor fac- 2008).
tor (LIF), and Cardiotrophin-1 (CT-1) (Qian, 2000; TGF- signaling is mediated mainly by two ser-
Barnab-Heider et al., 2005). These factors sig- ine threonine kinase receptors, transforming growth
nal through heterodimerization of coreceptors LIFR factor-beta receptor I (TGFRI) and II (TGFRII), which
(leukemia inhibitor factor receptor beta) and gp130, activate Smad 2/3 and Smad 4 transcription factors.
and activation of the intracellular Jak-STAT (for review Phosphorylation and activation of these proteins is
see Miller and Gauthier, 2007). followed by formation of the Smads 2/3-4 complex,
Several lines of evidence strongly support a role which translocates to the nucleus regulating transcrip-
for the Jak/STAT pathway in astrocytogenesis. First, tional responses to TGF- (Shi and Massagu, 2003).
the STAT transcriptional factors have been shown to We showed that embryonic cortical NPC cultures
cause direct transcriptional activation of two astro- enriched in RG-like cells, which are characterized by
cytic genes, GFAP and S100, via STAT binding sites their radial morphology and also by nestin and BLBP
within their promoters (Bonni et al., 1997; Nakashima expression, are direct targets of TGF-1, because these
et al., 1999b; Miller and Gauthier, 2007). Further, cells express TGFRII. In that study we observed a
gp130 or LIFR deficiency causes profound impair- specific upregulation of glial markers, such as GFAP
ment in astrocyte generation, both in vitro (Ware and GLAST, and downregulation of RG markers,
et al., 1995; Nakashima et al., 1999a) and in vivo such as nestin and BLBP, in the presence of TGF-
(Barnab-Heider et al., 2005). In the same context, 1 or a conditioned medium derived from neurons.
other groups have demonstrated that CNTF and LIF All of these events were followed by activation of
are sufficient to induce astrogenesis, and that this the SMADs2/3 proteins, with subsequent transloca-
action requires LIFR and gp130 in vitro (Bonni et al., tion to the nucleus. Inhibition of TGF-1 pathways by
1997; Nakashima et al., 1999a, 1999b). In addition, it the neutralizing antibody against this cytokine impairs
has been shown that cortical neurons synthesize and neuronal effects (Fig. 11.1, Stipursky and Gomes,
secrete the neurotrophic cytokine CT-1, which acti- 2007). These data and the observation that in the
vates the gp130-Jak-STAT pathway and is essential for adult rodent brain, TGF-1 seems to negatively mod-
the timed genesis of astrocytes in vitro and in vivo. ulate neurogenesis and stem-cell proliferation (Wachs
Through the use of cortical precursor cultures that et al., 2006), together with the recent identification
temporally mimic the in-vivo differentiation pattern, of TGFs isoforms in the germinative layers of the
it was demonstrated that CT-1 causes premature gli- telencephalon and spinal cord, strongly support a role
ogenesis. Moreover, the neonatal CT-1-deficient cor- for TGF- in the biology of RG cells (Mecha et al.,
tex contains fewer astrocytes (Barnab-Heider et al., 2008).
2005). Thus, during CNS development, several molecular
Recently, our group showed that cortical neurons cues instruct neural precursors as to which differen-
induce astrocyte differentiation from radial glia-like tiation pathway, neuronal or glial, they should follow
cells by activating the TGF-1 pathway in vitro (Fig. 11.2). Although several of these cues have been
11 Neuron-Astroglial Interactions in Cell Fate Commitment 155

Fig. 11.1 TGF-1 role in RG-astrocyte transformation. (a) the entire thickness of neural tube wall. During cerebral cor-
Treatment of E14.5 cerebral cortex-derived progenitors with tex development, these cells generate neuroblasts that migrate
TGF-1 decreases the number of nestin/GLAST positive cells over radial fibers (migrating neurons); (2) Postmitotic neurons
(RG-like cells) and increases that of GFAP/GLAST positive form the cortical plate and differentiate terminally into layer-
cells (Astrocytes). (b) RG cell in vitro shows a typical bipolar specific neurons; (3) At the end of the neuronal migratory phase,
morphology. (c) Schematic view of cerebral cortex develop- RG cells differentiate into astrocytes under the influence of
ment. (1) RG cells show a bipolar morphology that spans TGF-1

thoroughly investigated during the last few years, there


is still great interest in understanding how they interact.
In addition, there is a lack of knowledge concerning
what makes these progenitors lose their transient neu-
rogenic potential and differentiate towards a neuronal
or glial phenotype.
As mentioned before, it is known that early in devel-
opment, the signaling initiated by Neuregulins and its
receptor ErbBs, as well as Notch1 and FGF, for exam-
ple, play an important role in maintaining the undif-
ferentiated RG phenotype. On the other hand, later in
development the activation of the Jak-STAT pathway
by factors of the ILs family, as well as SMADs by
TGF-, promotes glial specification. Although these
are apparently temporally segregated events, in fact
the idea is that all of these pathways interact at a
Fig. 11.2 Schematic model of neuron-RG interaction sig- key moment during CNS development, promoting the
naling pathways. RG cell is a direct target of neuron-derived switch of progenitor-neuronal-glial phases. It has been
radializing factors (Neuregulin (NRG), Delta/Notch, FGF) that shown that, apparently, the balance of these pathways,
promote RG phenotype maintenance. However, growing evi-
transcription factors, as well as epigenetic silencing
dence suggests that neurons secrete astrocytogenic-inducing
growth factors (TGF-1, CT-1, ILs, CNTF). The balance gene mechanisms such as methylation and acetyla-
between these factors controls RG self-renewal (white curved tion, control the correct timing of neuron and glial-
arrows) and its transformation into astrocytes (black curved cell generation (for review see Miller and Gauthier,
arrow)
2007).
156 J. Stipursky et al.

11.3.2 Role of Neuronal-Derived Two main subtypes of glutamate transporters have


Molecules in Astrocyte been described in glia: GLAST, with expression pre-
dominating at early stages; and glutamate transporter-
Differentiation: Crosstalk Between
1 (GLT-1), the expression of which progressively
Growth Factors and increases with maturity. Recently, Regan and collab-
Neurotransmitters orators demonstrated a differential cellular expression
of glutamate transporters in the developing and mature
Neuron-astrocyte interactions in the synaptic cleft play CNS, and also showed that glutamate signaling is
a pivotal role in synapse development. The intimate under local CNS region-specific modulation. These
relationship between astrocytes and synaptic termi- results suggest the concept that astroglia is formed by
nals in vivo (Ventura and Harris, 1999) suggests that a heterogeneous population of cells that markedly vary
besides affecting synapse formation, astrocytes are in their responsiveness to several agents such as neuro-
also potential targets for neuronal-derived molecules transmitters (Regan et al., 2007). Swanson and collab-
such as neurotransmitters. This idea was further sup- orators have reported that neurons can modulate astro-
ported by the accumulating evidence that astrocytes cyte glutamate transporter expression and astrocyte
express a wide range of neurotransmitter receptors, differentiation in vitro. In the absence of neurons, cor-
both in vitro as well as in vivo (Porter and Mccarthy, tical astrocytes maintain polygonal shapes and express
1997; Volterra and Meldolesi, 2005). Synaptic trans- only the GLAST transporter. When co-cultured with a
mission is controlled by the astrocytic coverage of neuronal layer, many of the astrocytes maintain a stel-
synapses. Morphological adaptations in the astrocytes late shape and express GLT-1, suggesting that neurons
are associated with changes in the peri-synaptic space can modulate astrocyte differentiation (Swanson et al.,
and in the localization of glutamate transporters, which 1997). Although the nature of this neuronal signal
results in modified action of synaptically released glu- remains to be identified, reports have clearly demon-
tamate (Oliet et al., 2001). Glutamate clearance and, strated that astrocyte differentiation is modulated by
as a consequence, glutamate concentration and diffu- neuronal soluble factors (Perego et al., 2000) or neuro-
sion in the extracellular space, are associated with the transmitters (Runquist and Alonso, 2003; Romo et al.,
degree of astrocytic coverage of its neurons. Failure 2008).
of glutamate removal can lead to neuronal death due How can neurotransmitters affect the morphology
to its well-known neurotoxic properties (Camacho and and differentiation of an astrocyte? The morphology
Massieu, 2006). of these cells depends on the phosphorylation and
It is now clear that astrocytes respond to a vari- polymerization status of GFAP. It has been shown
ety of synaptically released transmitters in addition that different neurotransmitters including glutamate
to glutamate. For example, noradrenaline (Duffy and (Kommers et al., 1999, 2002), GABA (Runquist and
MacVicar, 1995), histamine, acetylcholine (Shelton Alonso, 2003), and serotonin (Chang et al., 2005)
and McCarthy, 2000), ATP (Wang et al., 2000), and influence the phosphorylation of GFAP, and thus in
GABA (Kang et al., 1998) can induce elevations of turn its degree of polymerization (Inagaki et al., 1994).
astrocytic Ca2+ . Indeed, the list of transmitters that can A secreted glial protein, S100, inhibits polymeriza-
mobilize astrocytic Ca2+ is almost as long as the list of tion of GFAP (Bianchi et al., 1995; Ziegler et al.,
molecules that activate neuronal receptors. 1998) and may therefore regulate astrocyte morphol-
Astrocytes express high levels of GABAA recep- ogy in an autocrine fashion. There is evidence that
tors (Tateishi et al., 2006), and differentiation of serotonin induces the release of S100 from astro-
immature astrocytes is at least partly mediated by cytes (Chang et al., 2005), suggesting a link between
the inhibitory neurotransmitter GABA. In vitro and neuronal (serotoninergic) activity, release of S100,
in vivo studies have provided evidence that GABA and astroglial morphology. Additional support for a
affects immature astrocytes by increasing both their crosstalk between GFAP/astrocyte differentiation and
GFAP expression and their stellation (Mong et al., glutamate signaling emerged from the recent demon-
2002; Runquist and Alonso, 2003), strongly sug- stration that expression of GFAP is essential to anchor
gesting a role for GABAergic signaling in astrocyte the glutamate transporter GLAST in the astrocyte
differentiation. plasma membrane, thus enhancing GLAST-mediated
11 Neuron-Astroglial Interactions in Cell Fate Commitment 157

transport (Sullivan et al., 2007). This is consistent 2001), NMDA receptors (Verkhratsky and Kirchhoff,
with the observation that GFAP knockout mice exhibit 2007), GABAA receptors (Tateishi et al., 2006), and
reduced glutamate clearance (Hughes et al., 2004), and purinergic receptors (Abbracchio and Ceruti, 2006).
loss of GLT-1 leads to elevated glutamate levels and Metabotropic glutamate receptors have been con-
subsequent neurodegeneration (Rothstein et al., 1996). sidered to be potential targets for neuroprotective
These studies suggest that changes in GFAP gene drugs since their earliest characterization. Studies by
expression and glutamate homeostasis might mutually Nicolettis group demonstrated that neuroprotection
influence each other. mediated by group-II mGlu receptors involves a novel
Glutamate is the major excitatory neurotrans- form of glial-neuronal interaction, which is promoted
mitter in the CNS, and its responses are mediated by the activation of mGlu3 receptors present in astro-
by ionotropic (iGlu) and metabotropic glutamate cytes (Bruno et al., 1997; Battaglia et al., 1998). That
(mGlu) receptors. iGlu receptors are cation-specific neuroprotection by glial mGlu3 receptors is mediated
ion channels defined by a specific pharmacology in by TGF-1 and TGF-2, which are released from
-amino-3-hydroxy-5-methyl-4-isoxazoleproprionic astrocytes and exert a potent neuroprotective activity in
acid (AMPA), kainate, and N-methyl-d-aspartate in vitro and in vivo models of excitotoxic death (Bruno
(NMDA) receptors. mGlu receptors are a family of et al., 1998). This activation of glial group-II mGlu
G-protein-coupled, seven transmembrane domain receptors enhances the de novo synthesis of TGF-1
receptors that exert a variety of effects on second through the activation of the mitogen-activated protein
messenger systems and ion channels. They are divided (MAP) kinase and phosphatidylinositol (PI)-3-K path-
into eight different subtypes (mGlu18) and three ways. Pharmacological inhibition of these two path-
sub-groupings based on agonist pharmacology: Group ways prevents the neuroprotective activity of group-II
I (mGlu1 and mGlu5 receptors), Group II (mGlu2 and mGlu receptor agonists against excitotoxic neuronal
mGlu3), and group III (mGlu4, mGlu6, mGlu7, and death (DOnofrio et al., 2001). Recently, Corti and col-
mGlu8) (Pin and Duvoisin, 1995). mGlu receptors laborators used mGlu2/Glu3 receptor knockout mice
modulate synaptic transmission, and are involved in to examine whether these two receptor subtypes have
activity-dependent modification of synaptic transmis- distinct roles in the processes of neurodegeneration
sion such as long-term potentiation and long-term and neuroprotection. They demonstrated that neuro-
depression. Each receptor subtype exhibits a well- protection by the dual mGlu2/3 receptor is entirely
defined expression pattern in several brain regions. In mediated by mGlu3 receptors (Corti et al., 2007). This
neurons, mGlu1 and mGlu5 receptors are generally suggests the existence of a novel mechanism of neuro-
found in postsynaptic densities and modulate post- protection that is mediated by the secretion of TGF-
synaptic efficacy; whereas mGlu2, mGlu3, mGlu4, species from astrocytes in response to group-II mGlu
mGlu7, and mGlu8 receptors are mainly (but not receptor activation. The importance of this mecha-
exclusively) presynaptic and regulate neurotransmitter nism is strengthened by the evidence that TGF- pro-
release (Ferraguti and Shigemoto, 2006). mGlu recep- tects against neuronal death induced by excitotoxins,
tors are also found in glial cells, where their activation ischemia, or aggregates of -amyloid peptide (Copani
exerts a variety of effects that are crucial for glial func- et al., 1995; Pratt and McPherson, 1997; Bruno et al.,
tion and glial-neuronal interaction under physiological 1998; Flanders et al., 1998; Buisson et al., 2003).
and pathological conditions (Bruno et al., 2001; Flor TGF-1 has several effects on astrocytes, includ-
et al., 2002). The presence of mGlu receptors in glial ing inhibition of proliferation (Baghdassarian et al.,
cells was initially inferred by a number of actions of 1993; Krieglstein et al., 1998), change in morphol-
glutamate in cultured astrocytes, including the increase ogy (Flanders et al., 1993) and motility (Gagelin et al.,
in intracellular Ca2+ (Glaum et al., 1990). Both mGlu3 1995), modulation of the cytoskeleton (Laping et al.,
and mGlu5 receptors have been detected by RT-PCR 1994), and the protein content of the extracellular
in hippocampal astrocytes acutely isolated from young matrix (ECM) (Baghdassarian et al., 1993; Wyss-
rats (Schools and Kimelberg, 1999), adult rats (Cai Coray et al., 1995). More recently, the role of TGF-1
et al., 2000), reactive astrocytes, human astrocytes, and in ECM production was highlighted by the demon-
glioma cells (Aronica et al., 2000, 2003). Astrocytes stration that TGF-1 knockout mice show reduced
also express AMPA receptors (Zhou and Kimelberg, expression of laminin (Brionne et al., 2003).
158 J. Stipursky et al.

Fig. 11.3 Activation of GFAP gene by glutamate involves for Smad 4 revealed a cytoplasmic location in control cultures
the TGF-1/Smad pathway. Cerebral cortex astrocytes from (b) and a nuclear distribution in TGF-1 (c) and glutamate-
newborn transgenic mice were cultured alone (a, b); in the treated (d) cultures. Glutamate and TGF-1 increased -Gal
presence of TGF-1 (10 ng/mL, (c, d)) or glutamate (100 astrocyte number, followed by activation of the Smad pathway.
M, (e, f)). (a, c and e) GFAP promoter-directed expression Scale bars correspond to 50 m
of lac-Z was revealed by X-Gal (blue nuclei) prior to anti-
GFAP immunocytochemistry (gray staining). Immunolabeling

In order to gain insight into astrocyte differentiation the mGlu2/3 receptor antagonist (Romo et al., 2008;
induced by neurons, we have focused on GFAP expres- Fig. 11.3).
sion, which is the major component of the astrocytic These data suggest a key role for TGF-1 as a
intermediate filaments (Eng et al., 1971; Bignami et al., downstream mediator of the effect of glutamate and
1972). In the rodent CNS, astrocyte maturation is fol- neurons on the GFAP gene promoter. The associa-
lowed by a replacement in the expression of vimentin tion of dysfunctional glial glutamate transporters and
by GFAP (Dahl, 1981; Pixley and De Vellis, 1984). By receptors with several neurological disorders (Sheldon
using transgenic mice bearing 2 kbp of the 5 flanking and Robinson, 2007), together with the observation of
region of the GFAP gene linked to the -galactosidase high levels of TGF-1 in several diseases where gluta-
(-Gal) reporter gene, we demonstrated that cortical mate metabolism dysfunctions are described (Tesseur
neurons activate the GFAP gene promoter, followed by and Wyss-Coray, 2006), support the hypothesis that
transgenic astrocyte differentiation in vitro. Addition glutamate/TGF-1 cross-talk might have a key role,
of a conditioned medium derived from cortical neurons not only in development but in brain pathology.
had a similar effect, suggesting that a soluble fac-
tor derived from neurons might be responsible for the
induction of the GFAP gene promoter (Gomes et al., 11.4 Neuron-Astrocyte Interactions:
1999a). Later, we identified TGF-1 as the major medi- Implications for Neuronal
ator of this event (de Sampaio e Spohr et al., 2002).
Differentiation and Synaptogenesis
Although both cell types, neurons and astrocytes, syn-
thesize and secrete this factor, addition of neurons to
astrocyte monolayers greatly increased TGF-1 syn- It has been estimated that individual astrocytes in
thesis and secretion by astrocytes (de Sampaio e Spohr the adult rodent brain ensheath and interact with as
et al., 2002; Sousa et al., 2004). More recently, we many as 140,000 synapses (Bushong et al., 2002).
demonstrated that glutamate activates the GFAP gene Traditionally, the function of astrocytes in synapses
promoter of cerebral cortical astrocytes through induc- has been associated with the regulation of ion concen-
tion of the TGF-1 signaling pathway. We reported trations, neurotransmitter clearance, and providing of
that glutamate induced Smad 4 nuclear translocation, substrates for energy metabolism (Carmignoto, 2000).
followed by activation of the GFAP gene, in cortical Accumulated data from the last 7 years, however, sug-
astrocytes. Both events were inhibited by addition of gest that astrocytes are an integral part of synaptic
11 Neuron-Astroglial Interactions in Cell Fate Commitment 159

connections, either by (1) inducing neuronal differen- et al., 2002), which are not permissive for neurite out-
tiation; or by (2) promoting synapse formation and growth. Together, these events suggest that Ca2+ oscil-
elimination; and even by (3) actively secreting neu- lations in astrocytes might be an important mechanism
romodulators or gliotransmitters. In this section, we to control neuronal development and regeneration.
will focus on the role of glial cells in synaptic plastic- Another suggested mechanism by which astrocytes
ity, with special emphasis on items (1) and (2). First, are proposed to modulate neurite outgrowth is by their
we will briefly discuss the role of astrocytes in neu- pattern of expression of extracellular-matrix (ECM)
ronal maturation, and axonal and dendritic growth, proteins such as laminin, fibronectin, and proteogly-
essential steps for synapse establishment. Then, we cans (Garcia-Abreu et al., 2000; Martinez and Gomes,
will present in vitro and in vivo evidence that astro- 2002). Astrocytes have been recognized as the major
cytes and Schwann cells can induce synaptogenesis, source of these ECM components, both in vivo and
and are involved in the maintenance of newly formed in vitro (Garcia-Abreu et al., 2000). The pattern of
synapses, in addition to ongoing structural support of these ECM proteins on the astrocyte surface, which
synapses. is highly modulated by thyroid hormone, provides
directional cues for neurite outgrowth (Garcia-Abreu
et al., 2000; Martinez and Gomes, 2002). Our group
demonstrated in 2002 that EGF secreted by T3-treated
astrocytes induces EGL neurons to undergo differ-
11.4.1 Neuron-Astrocyte Interactions
entiation initiated by the outgrowth of neurites, and
and Neuronal Differentiation this event is mediated by EGF modulation of laminin
and fibronectin astrocytic expression through MAPK
During CNS development, neurons migrate, and as and PI3K pathways. We demonstrated that EGF could
soon as they reach their appropriate positions they have a binary role for EGF in cerebellar ontogene-
begin to extend axons in order to establish connections sis, directly on the proliferation of granular precursors,
with other neurons. This event is characterized by a and indirectly through ECM components in neurite
dramatic change in cell morphology that is of funda- outgrowth.
mental importance in brain development, as well as in However, the ability of astrocytes to induce neurite
the regeneration of damaged nervous tissue (Guizzetti outgrowth depends on the region where the astrocytes
et al., 2008). originate. It was demonstrated that astrocytes derived
Growing axons navigate toward their targets in from different sections of the embryonic mouse mid-
response to a variety of guidance signals in their sur- brain show different permissivity to support neurite
rounding environment. These cues include diffusible outgrowth. This was related to differences in the
attractive or repellent molecules that are secreted by content of the ECM elements, laminin and sulfated
the intermediate or final cellular targets of the axons. glycosaminoglycans (s-GAGs) in the astrocytes from
Astrocytes are known as a potent source of cues that different sections: the punctuate pattern of laminin
influence neuronal fate and growth-cone navigation, expression was not permissive for neurite outgrowth
not only by secreting soluble factors, but also by (Garcia-Abreu et al., 1995, 2000; Mendes et al., 2003).
expressing extracellular matrix proteins (Garcia-Abreu Together, these differences support the concept that
et al., 2000; de Sampaio e Spohr et al., 2008). astrocytes display heterogeneous specializations, with
Recently, Kanemaru and co-workers (2007) sug- reports of region-specific, species-specific, and func-
gested a novel intracellular signaling mechanism by tionally distinct subpopulations. It is interesting that
which astrocytes mediate neurite outgrowth. These astrocytes lose their permissivity to neurite outgrowth
authors showed that modulation of spontaneous Ca2+ with age (Bahr et al., 1995), indicating that this prop-
oscillations in astrocytes leads to a reduction in the erty depends not only on the region of origin of
expression of N-cadherin, which correlates with inhi- the astrocytes, but also on their developmental stage.
bition of their neurite permissivity (Kanemaru et al., However, the underlying mechanism that controls this
2007). Interestingly, it was reported that spontaneous property remains unclarified.
Ca2+ oscillations become less frequent with age (Parri Thus, astroglial-derived soluble and membrane-
et al., 2001) and are lost in reactive astrocytes (Aguado bound factors could promote neuronal fate and
160 J. Stipursky et al.

arborization of neuronal precursors; however, the is committed to become an axon or dendrites, which
molecules that may instruct the astrocytes to secrete are two morphologically and functionally distinct seg-
soluble factors or membrane-bound factors remain ments? Neuronal polarity is important for neuronal
unknown. Recently, we demonstrated that astrocytes specialization. The understanding of the mechanisms
primed by lysophosphatidic acid (LPA) increase neu- and molecules that may modify axonal or dendritic
ronal differentiation, likely through a soluble factor, elongation or morphology during neuronal develop-
and that this activity is dependent on activation of ment might yield some clues for a better understand-
defined LPA receptors expressed in astrocytes (de ing of higher brain functions, such as learning and
Sampaio e Spohr et al., 2008). LPA is a lysophospho- memory. However, these mechanisms involved in the
lipid that may regulate diverse biological processes; creation and maintenance of neuronal polarity are still
however, little is known about its role in NS devel- the subject of study. What is known is that astrocytes
opment. Our group demonstrated that LPA-primed play important roles in this event: neuronal polarity
astrocytes induce neuronal differentiation of neuronal is regulated by several molecules, such as NGF and
precursors. Analysis of neuronal morphology revealed BMP (a subclass of the TGF- superfamily), and these
a marked increase in the number of processes per neu- molecules may be secreted by astrocytes (Prochiantz,
ron plated onto LPA-primed astrocytes, suggesting that 1995). It was also demonstrated that dendrite initi-
LPA-primed astrocytes may influence cell-fate specifi- ation is regulated separately from that of the axon,
cation and neuronal maturation in vitro (de Sampaio e and that local, and thus region-specific, astrocyte-
Spohr et al., 2008) (Fig. 11.4). derived factors are responsible for this phenomenon,
One of the major specializations that neurons and this can be mediated by ECM (Chamak et al.,
undergo is that related to polarization: which branch 1987).

Fig. 11.4 LPA-primed astrocytes increase neuronal differ- (c) Schematic model of LPA effect on neurons mediated by
entiation. Cerebral cortical progenitors obtained from embry- astrocytes. Extracellular LPA produced by postmitotic neurons
onic mice were cultured for 24 h on control (a) and LPA-primed directly interacts with high-affinity LPA receptors present on
(b) astrocyte monolayers. Cells were immunostained for the glial cells, which secrete a soluble factor in response to LPA.
neuronal marker, -tubulin III. There was a significant enhance- Indirectly, LPA induces maturation of postmitotic neurons and
ment in the arborization and length of neurites of neurons plated neuronal commitment of progenitors
onto LPA-primed astrocytes. Scale bar corresponds to 30 m.
11 Neuron-Astroglial Interactions in Cell Fate Commitment 161

Neuronal polarity defines the fine tuning of an integral and essential component of the tripartite
responses to numerous types of signal inputs, inte- synapse in both the peripheral nervous system (PNS)
grating and propagating them in the form of elec- and the CNS (Feng and Ko, 2008), as we will better
trical impulses over long distances to different tar- address in the next section.
gets. In addition to signal processing and propagation,
the axonal and dendritic surfaces also play house-
keeping roles such as receiving and releasing different 11.4.2 Role for Glia in Synaptogenesis
molecules from and into their environment (Bradke
and Dotti, 2000). In the CNS, astrocytes envelop the pre- and postsy-
Astrocytes modify the number of synaptic con- naptic neuronal components. This cellular architecture
nections, by releasing a variety of factors that influ- yields the concept of the tripartite synapse, consti-
ence axonal and dendritic growth and thus the poten- tuted by neurons and peri-synaptic glial cells, which
tial for synapse formation, such as laminins (Liesi is regarded today as a third integral component of
et al., 1983), neurotrophins (Althaus and Richter- synapses (Araque et al., 1999). Peri-synaptic glia have
Landsberg, 2000), S100 (Nishiyama et al., 2002), and been observed in several central and peripheral NS,
the activity-dependent neurotrophic factor (Blondel including astrocytes in the cerebral cortex (Ventura
et al., 2000). and Harris, 1999), Bergmann glia in the cerebellum
During development, inappropriate synapses must (Grosche et al., 2002), Mller cells in the retina,
be eliminated, and appropriate synaptic connections and Schwann cells at the neuromuscular junction
must be maintained and strengthened. This dynamic (Robitaille, 1998).
period of synaptic refinement coincides with the The first evidence of the role of astrocytes in
appearance of astrocytes in the postnatal brain, and synaptogenesis came from Ben Barres group at the
recent evidence indicates a role for astrocyte-derived end of the twentieth century. By using glia-free cul-
signals in synapse development (Ullian et al., 2001; tures of purified retinal ganglion cells (RGC), Pfrieger
Christopherson et al., 2005). Astrocyte contacts to and Barres (1997) found that astrocytes and oligo-
neurons may have different roles in the formation dendrocytes strongly enhanced spontaneous synaptic
of initial synapses, and the astrocytes may be able activity and the reliability of synaptic transmission.
to induce local structural and functional modifica- These effects were attributed to an increase in the effi-
tions of dendritic segments or individual synapses. cacy of existing synapses (Pfrieger and Barres, 1997).
These contacts may also induce global maturation of They found that astrocytes increased the total num-
neurons and subsequently up-regulation of synapse ber of synapses on each neuron sevenfold. Moreover,
formation, promoting the global maturation of the astrocytes were required to maintain these synapses,
neuronal network. Recently, Nishida and Okabe because most synapses that had formed in the presence
(2007) demonstrated that astrocytic contacts have of astrocytes were quickly lost when these cells were
instructive roles in the subsequent stabilization and removed (Ngler et al., 2001; Ullian et al., 2001, 2004).
maturation of immature dendritic protrusions, and that Later, the idea that glial cells induce synapse forma-
these astrocytic contacts are selective: some dendritic tion was validated, in the central and PNC, by a number
protrusions received astrocytic contacts and others of studies in different neuronal cell lines (murine and
did not, indicating that the astrocytes are capable of human) (Mauch et al., 2001; Ngler et al., 2001; Ullian
identifying the appropriate target. et al., 2001), and neuromuscular junction (NMJ) neu-
All the lines of evidence discussed here increase rons (Ullian et al., 2004). More recently, Johnson and
the support for the hypothesis that astrocytes play a collaborators demonstrated that synaptic transmission
dynamic role in the maintenance of connections in in human embryonic stem cell (hES cell) derived
the nervous system proper, either by controlling neu- neuronal cultures is enhanced by exogenous astrocytes
ronal specification and polarization or by controlling (Johnson et al., 2007). These data suggest that glia-
the numbers and establishment of the synapse con- induced synaptogenesis might be a general mechanism
nections. Therefore, astrocytes, which were formerly that is used by organisms ranging from lower inverte-
considered to be passive supporters of the chemical brates, as recently shown for C. elegans (Bacaj et al.,
synapse, have recently gained much recognition as 2008), to humans.
162 J. Stipursky et al.

If there is compelling evidence for the role of of synapses formed during postnatal stages. TSP-1
astrocytes in synaptogenesis, these findings raise a and -2 are both expressed in the developing brain
vital question of how glial cells enhance synaptoge- during the peak period of synaptogenesis, but are
nesis and regulate synaptic transmission. In vitro or shut off by adulthood. These findings suggest that
in vivo studies have revealed a variety of putative immature astrocytes provide a developmental window
synaptogenic secreted and contact glia-derived fac- during which synaptogenesis can occur, by producing
tors. These include glutamate (Parpura and Haydon, a permissive environment through the secretion
2000), cholesterol (Mauch et al., 2001; Goritz et al., of TSP (Allen and Barres, 2005; Christopherson
2005), the extracellular matrix protein thrombospondin et al., 2005).
(Christopherson et al., 2005), TGF-1 (Feng and Ko, Similar glia-enhanced synaptogenesis has also been
2008), adenosine triphosphate (Koizumi et al., 2003), observed at PNS synapses (Feng and Ko, 2008). Feng
the neuromodulator D-serine (Yang et al., 2003), and and Ko demonstrated that TGF-1 is a Schwann
the glial protein S100 (Nishiyama et al., 2002). cell-derived molecule that promotes synaptogene-
It is conceivable that astrocytes secrete other fac- sis at nerve-muscle contacts in vitro. This event
tors, not yet identified, with synapse-promoting activ- is mainly due to induction of expression of neu-
ity (Parpura and Haydon, 2000; Yang et al., 2003; ronal agrin, a large proteoglycan, which leads to
Elmariah et al., 2005). a global increase in the formation of acetylcholine
Recent studies showed that cultured neurons from receptor clusters on muscle cells. This expression
the mammalian CNS require glia-derived cholesterol induced by TGF-1 acts directly on presynaptic neu-
to form numerous and efficient synapses. Neurons rons rather than through muscle cells (Feng and Ko,
appear to produce enough cholesterol to survive, 2008).
to differentiate axons and dendrites, and to form a During development, neurons acquire competence
few inefficient synapses. The massive formation of to receive synapses. Two distinct mechanisms are
synapses, however, may require additional choles- essential to the success of this process: dendrite devel-
terol, which must be delivered by astrocytes via opment, as discussed in the previous section; and
ApoE-containing lipoproteins (Mauch et al., 2001; synaptic receptivity. Although it is generally assumed
Pfrieger, 2003; Goritz et al., 2005). So far, how- that neurons are generated with the ability to both
ever, it remains unclear how cholesterol enhances the form and receive synapses, Fletcher et al. (1994) found
formation and function of synapses. Goritz and col- that embryonic hippocampal neurons are capable of
laborators demonstrated that dendrite differentiation is forming but not receiving synapses. Recently, it was
the rate-limiting step for glia-induced synaptogenesis shown that astrocyte contact induces the acquisition
in RGC, and that this process requires cholesterol. In of competence for synaptic receptivity (Barker et al.,
addition, they demonstrated that cholesterol directly 2008).
enhances presynaptic differentiation, supports contin- The formation of mature neural circuits requires
uous synapse development, and is essential for the the selective elimination of inappropriate synaptic con-
stability of evoked synaptic transmission (Goritz et al., nections. Several lines of evidence suggest that astro-
2005). cytes may play a key role in this process, either by
Christopherson and collaborators identified the activating specific signaling pathways, such as the
ECM proteins thrombospondins (TSPs), TSP-1-2, integrin-dependent pathway (Hama et al., 2004), or by
as the synaptogenic astrocyte-derived molecule physically eliminating the synapses, through release of
(Christopherson et al., 2005). Addition of TSP-1 proteases, protease inhibitors (Nelson et al., 1995), and
or TSP-2 to cultured RGC caused a large increase components of the complement cascade (Stevens et al.,
in the number of synapses, similar to that induced 2007).
by astrocytes. Immunodepletion of TSP-2 from Murai and coworkers revealed an influence of
astrocyte-conditioned medium reduced the number ephrin signaling on the regulation of spine morphology
of induced synapses to control levels. Moreover, the in mouse hippocampal pyramidal cells in situ (Murai
TSP-1 and TSP-2 functions are essential for pro- et al., 2003). They showed that the interaction between
moting synaptogenesis: TSP-1/TSP-2 double-mutant ephrin-A3, located on astrocytic processes, and its
mice exhibit a dramatic reduction in the number EphA4 receptor on dendritic spines leads to retraction
11 Neuron-Astroglial Interactions in Cell Fate Commitment 163

of spines. This raises the question of whether contact disturbances of neuronal function that are found in
between astrocytic processes and dendritic spines dur- several neurological disorders such as epilepsy and
ing development is responsible for the elimination of schizophrenia.
synapses. One key issue in developmental neurobiology is to
Stevens and collaborators showed that a protein understand how the brain orchestrates the differentia-
in the classical complement cascade is expressed by tion of various cell types. A range of epigenetic signals
postnatal neurons in response to immature astrocytes, are involved in neural-fate potential, lineage specifica-
and is located on synapses throughout the postna- tion, and cellular differentiation in the CNS and PNS.
tal CNS and retina. They identified an unexpected The identification of RG and astrocytes as neural stem
role for astrocytes and the classical complement cas- cells in the developing and mature nervous system,
cade in mediating CNS synapse elimination in the respectively, shed light on the importance of identify-
retinogeniculate pathway. The finding that the classical ing the molecules that control astroglia differentiation.
complement cascade helps to mediate developmental Although several molecules involved in this event have
CNS synapse elimination adds to the growing evidence already been identified, we are only at the threshold
that components of the immune system contribute of understanding fully the nature and consequences
to brain development and function (Stevens et al., of these newly discovered astroglial functions. The
2007). scenario is still more rudimentary concerning brain
This evidence, together with the emerging char- pathology.
acterization of gliotransmitters (Bezzi and Volterra, Emerging evidence indicates that glial cells are
2001) and several neurotransmitters and receptors in essential elements in the establishment of several
astrocytes (which are not within the scope of this chap- neurological disorders, such as epilepsy, schizophre-
ter), argues that astrocytes are active partners of neu- nia, and depression; and neurodegenerative diseases,
rons, not only in synapse development but in function such as Alzheimers, Parkinsons, and Huntingtons
as well. diseases and Amyotrophic Lateral Sclerosis (ALS)
(for review see Seifert et al., 2006; Barres, 2008).
Whether glial dysfunction is the primary deficit or
a consequence of neuronal damage, remains to be
11.5 Concluding Remarks investigated in each pathology. The question is even
more complex when we consider that astroglia are not
The relative number of astrocytes, expressed both as unique, but a heterogeneous group of cells that express
a proportion of the total brain cell number and as a different growth factors and neurotransmitter recep-
ratio to the neuronal number, increases dramatically tors, molecular markers, different stem-cell potentials,
with phylogeny and brain complexity (Nedergaard and synaptogenic properties. This evidence leads to
et al., 2003). It has been proposed that human cor- specific questions such as What is the difference
tical evolution is accompanied by increasing com- between neurogenic and non-neurogenic astrocytes?
plexity in the form and function of astrocytes, which or Why do some synapses not require astrocyte sig-
reflects an expansion of their functional roles in synap- nals? or What signals might reprogram astrocytes
tic modulation and cortical circuitry. This is exem- to a RG phenotype in the adult brain? or Which
plified by the observation that each domain of a pathways lead to astrocyte activation or dysfunction?
single rodent astrocyte covers approximately 90,000 Answers to these questions would certainly provide
synapses, whereas in the human cortex each astrocyte a better understanding of glial biology and neuron-
supports and modulates the function of roughly two glia interaction during brain development and aging.
million synapses. Further, in addition to the increase Further, the key roles of glial cells in CNS pathol-
in number and complexity of astrocytes in evolution, ogy, together with the accumulating evidence that
the growing association of glia in synapses, which RG cells and astrocytes (and/or neurons) are con-
may be related to NS evolution, is even more striking. vertible cell types, open the possibility that manipu-
Although one cannot completely attribute the intellec- lation of glia differentiation signaling pathways may
tual capacity of humans to astrocyte complexity, the be a promising therapeutic target for brain insults
loss of astrocytic domains has been associated with (Fig. 11.5).
164 J. Stipursky et al.

Fig. 11.5 Schematic view of neuron-astroglial interactions. several neurodegenerative diseases or neurological disorders (3).
Radial glia cells and astrocytes are involved in brain develop- Manipulation of signaling pathways that modulate astrocyte
ment, either acting as neuronal progenitors (1) or secreting sol- commitment lineage (black and gray dashed lines); this func-
uble and ECM molecules that modulate neuronal fate commit- tion might be crucial to use glial cells as potential therapeutic
ment, neuronal differentiation, and synaptogenesis (2). Deficits targets
in astrocyte functions play a key role in the establishment of

Acknowledgment For financial support: FAPERJ, CNPq, Anthony TE, Klein C, Fishell G, Heintz N (2004) Radial glia
CAPES (Brazil). serves as neuronal progenitors in all regions of the central
nervous system. Neuron 25:881890.
Anton ES, Marchionni MA, Lee KF, Rakic P (1997) Role of
GGF/neuregulin signaling in interactions between migrating
neurons and radial glia in the developing cerebral cortex.
References Development 124:35013510.
Araque A, Parpura V, Sanzgiri RP, Haydon PG (1999) Tripartite
Abbracchio MP, Ceruti S (2006) Roles of P2 receptors in glial synapses: glia, the unacknowledged partner. Trends Neurosci
cells: focus on astrocytes. Purinergic Signal 4:595604. 5:208215.
Aguado F, Espinosa-Parrilla JF, Carmona MA, Soriano E (2002) Aronica E, Gorter JA, Ijlst-Keizers H, Rozemuller AJ, Yankaya
Neuronal activity regulates correlated network properties B, Leenstra S, Troost D (2003) Expression and functional
of spontaneous calcium transients in astrocytes in situ. J role of mGluR3 and mGluR5 in human astrocytes and glioma
Neurosci 22:94309444. cells: opposite regulation of glutamate transporter proteins.
Allen NJ, Barres BA (2005) Signaling between glia and neurons: Eur J Neurosci 10:21062118.
focus on synaptic plasticity. Curr Opin Neurobiol 5:542548. Aronica E, van Vliet EA, Mayboroda OA, Troost D, da Silva FH,
Althaus HH, Richter-Landsberg C (2000) Glial cells as targets Gorter JA (2000) Upregulation of metabotropic glutamate
and producers of neurotrophins. Int Rev Cytol 197:203277. receptor subtype mGluR3 and mGluR5 in reactive astro-
Altman J, Das GD (1966) Autoradiographic and histological cytes in a rat model of mesial temporal lobe epilepsy. Eur
studies of postnatal neurogenesis. I. A longitudinal investi- J Neurosci 7:23332344.
gation of the kinetics, migration and transformation of cells Bacaj T, Tevlin M, Lu Y, Shaham S (2008) Glia are essential for
incorporating tritiated thymidine in neonate rats, with special sensory organ function in C. elegans. Science 322:744747.
reference to postnatal neurogenesis in some brain regions. J Baghdassarian D, Toru-Delbauffe D, Gavaret JM, Pierre M
Comp Neurol 126:337389. (1993) Effects of transforming growth factor-beta 1 on the
Alvarez-Buylla A, Garca-Verdugo JM, Tramontin AD (2001) A extracellular matrix and cytoskeleton of cultured astrocytes.
unified hypothesis on the lineage of neural stem cells. Nat Glia 7:193202.
Rev Neurosci 2:287293. Bahr M, Przyrembel C, Bastmeyer M (1995) Astrocytes from
Alvarez-Buylla A, Lim DA (2004) For the long run: maintaining adult rat optic nerves are nonpermissive for regenerating
germinal niches in the adult brain. Neuron 41:683686. retinal ganglion cell axons. Exp Neurol 131:211220.
11 Neuron-Astroglial Interactions in Cell Fate Commitment 165

Barker AJ, Koch SM, Reed J, Barres BA, Ullian EM (2008) neuroprotective drugs. J Cereb Blood Flow Metab 9:
Developmental control of synaptic receptivity. J Neurosci 10131033.
33:81508160. Bruno V, Sureda FX, Storto M, Casabona G, Caruso A, Knopfel
Barnab-Heider F, Wasylnka JA, Fernandes KJ, Porsche C, T, Kuhn R, Nicoletti F (1997) The neuroprotective activity
Sendtner M, Kaplan DR, Miller FD (2005) Evidence that of group-II metabotropic glutamate receptors requires new
embryonic neurons regulate the onset of cortical gliogenesis protein synthesis and involves a glial-neuronal signaling. J
via cardiotrophin-1. Neuron 48:253265. Neurosci 17:18911897.
Barres B (2008) The mystery and magic of glia: a perspective on Buisson A, Lesne S, Docagne F, Ali C, Nicole O, MacKenzie
their roles in health and disease. Neuron 60:430440. ET, Vivien D (2003) Transforming growth factor-beta and
Battaglia G, Bruno V, Ngomba RT, Di Grezia R, Copani ischemic brain injury. Cell Mol Neurobiol 23:539550.
A, Nicoletti F (1998) Selective activation of group-II Bushong EA, Martone ME, Jones YZ, Ellisman MH (2002)
metabotropic glutamate receptors is protective against exci- Protoplasmic astrocytes in CA1 stratum radiatum occupy
totoxic neuronal death. Eur J Pharmacol 356:271274. separate anatomical domains. J Neurosci 1:183192.
Bayer SA, Altman J (1991) Development of the endopiriform Cai Z, Schools GP, Kimelberg HK (2000) Metabotropic gluta-
nucleus and the claustrum in the rat brain. Neuroscience mate receptors in acutely isolated hippocampal astrocytes:
45:391412. developmental changes of mGluR5 mRNA and functional
Bentivoglio M, Mazzarello P (1999) The history of radial glia. expression. Glia 1:7080.
Brain Res Bull 49:305315. Camacho A, Massieu L (2006) Role of glutamate transporters in
Bernardos RL, Barthel LK, Meyers JR, Raymond PA (2007) the clearance and release of glutamate during ischemia and
Late-stage neuronal progenitors in the retina are radial its relation to neuronal death. Arch Med Res 1:1118.
Mller glia that function as retinal stem cells. J Neurosci Campbell K, Gtz M (2002) Radial glia: multi-purpose cells for
26:70287040. vertebrate brain development. Trends Neurosci 25:235238.
Berninger B, Costa MR, Koch U, Schroeder T, Sutor B, Grothe Carmignoto G (2000) Reciprocal communication systems
B, Gtz M (2007) Functional properties of neurons derived between astrocytes and neurones. Prog Neurobiol 6:
from in vitro reprogrammed postnatal astroglia. J Neurosci 561581.
32:86548664. Carney RS, Cocas LA, Hirata T, Mansfield K, Corbin JG (2008)
Bezzi P, Volterra A (2001) A neuron-glia signalling network in Differential regulation of telencephalic pallial-subpallial
the active brain. Curr Opin Neurobiol 11:387394. boundary patterning by Pax6 and Gsh2. Cereb Cortex
Bianchi R, Garbuglia M, Verzini M, Giambanco I, Spreca A, 19:745759.
Donato R (1995) S-100 protein and annexin II2-p11(2) Chamak B, Fellous A, Glowinski J, Prochiantz A (1987) MAP2
(calpactin I) act in concert to regulate the state of assem- expression and neuritic outgrowth and branching are coregu-
bly of GFAP intermediate filaments. Biochem Biophys Res lated through region-specific neuro-astroglial interactions. J
Commun 208:910918. Neurosci 7:31633170.
Bignami A, Eng LF, Dahl D, Uyeda CT (1972) Localization of Chang MS, Ariah LM, Marks A, Azmitia EC (2005) Chronic
glial fibrillary acidic protein in astrocyte by immunofluores- gliosis induced by loss of S-100B: knockout mice have
cence. Brain Res 43:429435. enhanced GFAP-immunoreactivity but blunted response to a
Blondel O, Collin C, McCarran WJ, Zhu S, Zamostiano serotonin challenge. Brain Res 1031:19.
R, Gozes I, Brenneman DE, McKay RD (2000) A glia- Chiasson BJ, Tropepe V, Morshead CM, van der Kooy D (1999)
derived signal regulating neuronal differentiation. J Neurosci Adult mammalian forebrain ependymal and subependymal
20:80128020. cells demonstrate proliferative potential, but only subependy-
Bonaguidi MA, Peng CY, McGuire T, Falciglia G, Gobeske KT, mal cells have neural stem cell characteristics. J Neurosci
Czeisler C, Kessler JA (2008) Noggin expands neural stem 19:44624471.
cells in the adult hippocampus. J Neurosci 37:91949204. Choi BH, Lapham LW (1978) Radial glia in the human fetal
Bonni A, Sun Y, Nadal-Vicens M, Bhatt A, Frank DA, cerebrum: a combined Golgi, immunofluorescent and elec-
Rozovsky I, Stahl N, Yancopoulos GD, Greenberg ME tron microscopic study. Brain Res 148:295311.
(1997) Regulation of gliogenesis in the central nervous Chojnacki A, Shimazaki T, Gregg C, Weinmaster G, Weiss S
system by the JAK-STAT signaling pathway. Science 278: (2003) Glycoprotein 130 signaling regulates Notch1 expres-
477483. sion and activation in the self-renewal of mammalian fore-
Bradke F, Dotti CG (2000) Establishment of neuronal polar- brain neural stem cells. J Neurosci 23:17301741.
ity: lessons from cultured hippocampal neurons. Cur Opin Christopherson KS, Ullian EM, Stokes CC, Mullowney CE, Hell
Neurobiol 10:574581. JW, Agah A, Lawler J, Mosher DF, Bornstein P, Barres BA
Brionne TC, Tesseur I, Masliah E, Wyss-Coray T (2003) Loss (2005) Thrombospondins are astrocyte-secreted proteins that
of TGF-beta1 leads to increased neuronal cell death and promote CNS synaptogenesis. Cell 120:421433.
microgliosis in mouse brain. Neuron 40:11331145. Copani A, Bruno V, Battaglia G, Leanza G, Pellitteri R, Russo
Bruno V, Battaglia G, Casabona G, Copani A, Caciagli F, A, Stanzani S, Nicoletti F (1995) Activation of metabotropic
Nicoletti F (1998) Neuroprotection by glial metabotropic glutamate receptors protects cultured neurons against apop-
glutamate receptors is mediated by transforming growth tosis induced by beta-amyloid peptide. Mol Pharmacol
factor-beta. J Neurosci 18:95949600. 47:890897.
Bruno V, Battaglia G, Copani A, DOnofrio M, Di Iorio P, Corti C, Battaglia G, Molinaro G, Riozzi B, Pittaluga A, Corsi
De Blasi A, Melchiorri D, Flor PJ, Nicoletti F (2001) M, Mugnaini M, Nicoletti F, Bruno V (2007) The use of
Metabotropic glutamate receptor subtypes as targets for knock-out mice unravels distinct roles for mGlu2 and mGlu3
166 J. Stipursky et al.

metabotropic glutamate receptors in mechanisms of neurode- Ferraguti F, Shigemoto R (2006) Metabotropic glutamate recep-
generation/neuroprotection. J Neurosci 27:82978308. tors. Cell Tissue Res 326:483504.
Dahl D (1981) The vimentin-GFA protein transition in rat neu- Filippov V, Kronenberg G, Pivneva T, Reuter K, Steiner B,
roglia cytoskeleton occurs at the time of myelination. J Wang LP, Yamaguchi M, Kettenmann H, Kempermann G
Neurosci Res 6:741748. (2003) Subpopulation of nestin-expressing progenitor cells
Das AV, Mallya KB, Zhao X, Ahmad F, Bhattacharya S, in the adult murine hippocampus shows electrophysiologi-
Thoreson WB, Hegde GV, Ahmad I (2006) Neural stem cell cal and morphological characteristics of astrocytes. Mol Cell
properties of Mller glia in the mammalian retina: regulation Neurosci 23:373382.
by Notch and Wnt signaling. Dev Biol 299:283302. Flanders KC, Ldecke G, Renzig J, Hamm C, Cissel DS,
Davis AA, Temple S (2000) Timing of CNS cell generation: a Unsicker K (1993) Effect of TGF-s and bFGF on astroglial
programmed sequence of neuron and glial cell production cell growth and gene expression in vitro. Mol Cell Neurosci
from isolated murine cortical stem cells. Neuron 28:6980. 4:406417.
de Azevedo LC, Fallet C, Moura-Neto V, Daumas-Duport C, Flanders KC, Ren RF, Lippa CF (1998) Transforming growth
Hedin-Pereira C, Lent R (2003) Cortical radial glial cells factor-betas in neurodegenerative disease. Prog Neurobiol
in human fetuses: depth-correlated transformation into astro- 54:7185.
cytes. J Neurobiol 55:288298. Fletcher TL, De Camilli P, Banker G (1994) Synaptogenesis
de Sampaio e Spohr TCL, Martinez R, da Silva EF, Moura in hippocampal cultures: evidence indicating that axons and
Neto V, Gomes FC (2002) Neuro-glia interactions effects on dendrites become competent to form synapses at different
GFAP gene: a novel role for transforming growth factor beta stages of neuronal development. J Neurosci 11:66956706.
1. Eur J Neurosci 16:20592069. Flor PJ, Battaglia G, Nicoletti F, Gasparini F, Bruno V (2002)
de Sousa OV, Romo L, Neto VM, Gomes FC (2004) Glial fibril- Neuroprotective activity of metabotropic glutamate receptor
lary acidic protein gene promoter is differently modulated by ligands. Adv Exp Med Biol 513:197223.
transforming growth factor-beta 1 in astrocytes from distinct Gagelin C, Pierre M, Gavaret JM, Toru-Delbauffe D (1995)
brain regions. Eur J Neurosci 19:17211730. Rapid TGF beta 1 effects on actin cytoskeleton of astro-
Doetsch F (2003) A niche for adult neural stem cells. Curr Opin cytes: comparison with other factors and implications for cell
Genet Dev 13:543550. motility. Glia 13:283293.
Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez- Gaiano N, Fishell G (2002) The role of notch in promoting glial
Buylla A (1999) Subventricular zone astrocytes are neural and neural stem cell fates. Annu Rev Neurosci 25:471490.
stem cells in the adult mammalian brain. Cell 97:703716. Galileo DS, Gray GE, Owens GC, Majors J, Sanes JR (1990)
Doetsch F, Petreanu L, Caille I, Garcia-Verdugo JM, Alvarez- Neurons and glia arise from a common progenitor in chicken
Buylla A (2002) EGF converts transit-amplifying neurogenic optic tectum: demonstration with two retroviruses and cell
precursors in the adult brain into multipotent stem cells. type-specific antibodies. Proc Natl Acad Sci USA 87:
Neuron 36:10211034. 458462.
Duffy S, MacVicar BA (1995) Adrenergic calcium signaling in Garcia CM, Darland DC, Massingham LJ, DAmore PA (2004)
astrocyte networks within the hippocampal slice. J Neurosci Endothelial cell-astrocyte interactions and TGF beta are
15:55355550. required for induction of blood-neural barrier properties. Dev
DOnofrio M, Cuomo L, Battaglia G, Ngomba RT, Storto M, Brain Res 152:2538.
Kingston AE, Orzi F, De Blasi A, Di Iorio P, Nicoletti F, Garcia-Abreu J, Mendes FA, Onofre GR, Freitas MS, Silva LCF,
Bruno V (2001) Neuroprotection mediated by glial group- Moura Neto V, Cavalcante L (2000) Contribution of heparin
II metabotropic glutamate receptors requires the activation sulfate to the non-permissive role of the midline glia to the
of the MAP kinase and the phosphatidylinositol-3-kinase growth of midbrain neurites. Glia 29:260272.
pathways. J Neurochem 78:435445. Garcia-Abreu J, Moura-Neto V, Carvalho SL, Cavalcante LA
Eiraku M, Tohgo A, Ono K, Kaneko M, Fujishima K, Hirano T, (1995) Regionally specific properties of midbrain glia. I.
Kengaku M (2005) DNER acts as a neuron-specific Notch Interactions with midbrain neurons. J Neurosci Res 40:
ligand during Bergmann glial development. Nat Neurosci 417477.
8:873880. Ghashghaei HT, Weimer JM, Schmid RS, Yokota Y, McCarthy
Elmariah SB, Oh EJ, Hughes EG, Balice-Gordon RJ (2005) KD, Popko B, Anton ES (2007) Reinduction of ErbB2 in
Astrocytes regulate inhibitory synapse formation via Trk- astrocytes promotes radial glial progenitor identity in adult
mediated modulation of postsynaptic GABAA receptors. J cerebral cortex. Genes Dev 21:32583271.
Neurosci 25:36383650. Glaum SR, Holzwarth JA, Miller RJ (1990) Glutamate receptors
Eng LF, Vanderhaeghen JJ, Bignami A, Gerste B (1971) An activate Ca2+ mobilization and Ca2+ influx into astrocytes.
acidic protein isolated from fibrous astrocytes. Brain Res Proc Natl Acad Sci USA 87:34543458.
28:351354. Goldman SA, Nottebohm F (1983) Neuronal production, migra-
Ever L, Gaiano N (2005) Radial glial progenitors: neurogenesis tion, and differentiation in a vocal control nucleus of
and signaling. Curr Opin Neurobiol 1:2933. the adult female canary brain. Proc Natl Acad Sci USA
Feng L, Heintz N (1995) Differentiating neurons activate tran- 80:23902394.
scription of the brain lipid-binding protein gene in radial Gomes FCA, Garcia-Abreu J, Galou M, Paulin D, Moura Neto
glia through a novel regulatory element. Development 121: V (1999a) Neurons induce GFAP gene promoter of cultured
17191730. astrocytes from transgenic mice. Glia 26:97108.
Feng Z, Ko CP (2008) Schwann cells promote synaptogenesis at Gomes FCA, Maia CG, Menezes JRL, Moura Neto V (1999b)
the neuromuscular junction via transforming growth factor- Cerebellar astrocytes treated by thyroid hormone modulate
beta1. J Neurosci 39:95999609. neuronal proliferation. Glia 25:247255.
11 Neuron-Astroglial Interactions in Cell Fate Commitment 167

Goritz C, Mauch D, Pfrieger FW (2005) Multiple mechanisms Kettenmann H, Ransom BR (2005) Neuroglia, 2nd ed. Oxford
mediate cholesterol-induced synaptogenesis in a CNS neu- University Press, USA.
ron. Mol Cell Neurosci 29:190201. Koizumi S, Fujishita K, Tsuda M, Shigemoto-Mogami Y, Inoue
Grosche J, Kettenmann H, Reichenbach A (2002) Bergmann K (2003) Dynamic inhibition of excitatory synaptic transmis-
glial cells form distinct morphological structures to interact sion by astrocyte-derived ATP in hippocampal cultures. Proc
with cerebellar neurons. J Neurosci Res 2:138149. Natl Acad Sci USA 19:1102311028.
Guizzetti M, Moore N, Giordano G, Costa LG (2008) Kommers T, Rodnight R, Boeck C, Vendite D, Oliveira D, Horn
Modulation of neuritogenesis by astrocyte muscarinic recep- J, Oppelt D, Wofchuk S (2002) Phosphorylation of glial fib-
tors. J Biol Chem 283:3188431897. rillary acidic protein is stimulated by glutamate via NMDA
Gtz M, Barde YA (2005) Radial glial cells defined and major receptors in cortical microslices and in mixed neuronal/glial
intermediates between embryonic stem cells and CNS neu- cell cultures prepared from the cerebellum. Dev Brain Res
rons. Neuron 46:369372. 137:139148.
Gtz M, Hartfuss E, Malatesta R (2002) Radial glia as neuronal Kommers T, Rodnight R, Oppelt D, Oliveira D, Wofchuk S
precursors: a new perspective on the correlation of morphol- (1999) The mGluR stimulating GFAP phosphorylation in
ogy and lineage restriction in the developing cerebral cortex immature hippocampal slices has some properties of a group
of mice. Dev Brain Res Bull 57:777788. II receptor. Neuroreport 10:21192123.
Gtz M, Stoykova A, Gruss P (1998) Pax6 controls radial glia Krieglstein K, Henheik P, Farkas L, Jaszai J, Galter D, Krohn
differentiation in the cerebral cortex. Neuron 21:10311044. K, Unsicker K (1998) Glial cell line-derived neurotrophic
Hama H, Hara C, Yamaguchi K, Miyawaki A (2004) PKC sig- factor requires transforming growth factor- for exerting its
naling mediates global enhancement of excitatory synapto- full neurotrophic potential on peripheral and CNS neurons. J
genesis in neurons triggered by local contact with astrocytes. Neurosci 18:98229834.
Neuron 41:405415. Kriegstein AR, Gtz M (2003) Radial glia diversity: a matter of
Hatten ME (1985) Neuronal regulation of astroglial morphology cell fate. Glia 43:3743.
and proliferation in vitro. J Cell Biol 2:384396. Krnyei Z, Gcza E, Rhl R, Orsolits B, Vrs E, Szab B,
Hatten ME (1999) Central nervous system neuronal migration. Vgovits B, Madarsz E (2007) Astroglia-derived retinoic
Annu Rev Neurosci 22:511539. acid is a key factor in glia-induced neurogenesis. FASEB J
Hatten ME (2002) New directions in neuronal migration. 21:114.
Science 297:16601663. Krnyei Z, Szlvik V, Szab B, Gcza E, Czirk A, Madarsz
Hughes EG, Maguire JL, McMinn MT, Scholz RE, Sutherland E (2005) Humoral and contact interactions in astroglia/stem
ML (2004) Loss of glial fibrillary acidic protein results cell co-cultures in the course of glia-induced neurogenesis.
in decreased glutamate transport and inhibition of PKA- Glia 49:430444.
induced EAAT2 cell surface trafficking. Mol Brain Res Laping NJ, Morgan TE, Nichols NR, Rozovsky I, Young-Chan
124:114123. CS, Zarow C, Finch CE (1994) Transforming growth factor-
Hunter KE, Hatten ME (1995) Radial glial cell transformation beta 1 induces neuronal and astrocyte genes: tubulin alpha
to astrocytes is bidirectional: regulation by a diffusible fac- 1, glial fibrillary acidic protein and clusterin. Neuroscience
tor in embryonic forebrain. Proc Natl Acad Sci USA 92: 58:563572.
20612065. Lawrence JM, Singhal S, Bhatia B, Keegan DJ, Reh TA,
Ihrie RA, Alvarez-Buylla A (2007) Cells in the astroglial lineage Luthert PJ, Khaw PT, Limb GA (2007) MIO-M1 cells and
are neural stem cells. Cell Tissue Res 331:179191. similar muller glial cell lines derived from adult human
Imura T, Nakano I, Kornblum HI, Sofroniew MV (2006) retina exhibit neural stem cell characteristics. Stem Cells
Phenotypic and functional heterogeneity of GFAP- 25:20332043.
expressing cells in vitro: differential expression of Lie DC, Colamarino SA, Song H-J, Dsir L, Mira H, Consiglio
LeX/CD15 by GFAP-expressing multipotent neural stem A, Lein ES, Jessberger S, Lansford H, Dearie AR, Gage FH
cells and non-neurogenic astrocytes. Glia 53:277293. (2005) Wnt signalling regulates adult hippocampal neuroge-
Inagaki M, Nakamura Y, Takeda M, Nishimura T, Inagaki N nesis. Nature 437:13701375.
(1994) Glial fibrillary acidic protein: dynamic property and Liesi P, Dahl D, Vaheri A (1983) Laminin is produced by early
regulation by phosphorylation. Brain Pathol 4:239243. rat astrocytes in primary culture. J Cell Biol 96:920924.
Johansson CB, Svensson M, Wallstedt L, Janson AM, Frisen J Lim DA, Alvarez-Buylla A (1999) Interaction between astro-
(1999) Neural stem cells in the adult human brain. Exp Cell cytes and adult subventricular zone precursors stimulates
Res 253:733736. neurogenesis. Proc Natl Acad Sci USA 96:75267531.
Johnson MA, Weick JP, Pearce RA, Zhang SC (2007) Functional Lim DA, Tramontin AD, Trevejo JM, Herrera DG, Garcia-
neural development from human embryonic stem cells: Verdugo JM, Alvarez-Buylla A (2000) Noggin antagonizes
accelerated synaptic activity via astrocyte coculture. J BMP signaling to create a niche for adult neurogenesis.
Neurosci 12:30693077. Neuron 28:713726.
Kandel ER, Schwartz JH, Jessell TM (2000) Principles of Lobo NA, Shimono Y, Qian D, Clarke MF (2007) The biology
Neural Science, 4th ed. McGraw-Hill, New York. of cancer stem cells. Annu Rev Cell Dev Biol 23:675699.
Kanemaru K, Okubo Y, Hirose K, Iino M (2007) Regulation Malatesta P, Appolloni I, Calzolari F (2008) Radial glia and
of neurite growth by spontaneous Ca2+ oscillations in astro- neural stem cells. Cell Tissue Res 331:165178.
cytes. J Neurosci 27:89578966. Malatesta P, Hack MA, Hartfuss E, Kettenmann H, Klinkert
Kang J, Jiang L, Goldman SA, Nedergaard M (1998) Astrocyte- W, Kirchhoff F, Gtz M (2003) Neuronal or glial
mediated potentiation of inhibitory synaptic transmission. progeny: regional differences in radial glia fate. Neuron
Nature Neurosci 1:683692. 37:751764.
168 J. Stipursky et al.

Malatesta P, Hartfuss E, Gtz M (2000) Isolation of radial glial Nelson PG, Fields RD, Liu Y (1995) Neural activity, neuron-
cells by fluorescent-activated cell sorting reveals a neuronal glia relationships, and synapse development. Perspect Dev
lineage. Development 127:52535263. Neurobiol 2:399407.
Martinez R, Gomes FC (2002) Neuritogenesis induced by Nicholson JL, Altman J (1972) The effects of early hypo- and
thyroid hormone-treated astrocytes is mediated by epi- hyperthyroidism on the development of rat cerebellar cor-
dermal growth factor/mitogen-activated protein kinase- tex. I. Cell proliferation and differentiation. Brain Res 44:
phosphatidylinositol 3-kinase pathways and involves mod- 1323.
ulation of extracellular matrix proteins. J Biol Chem Nishida H, Okabe S (2007) Direct astrocytic contacts regulate
51:4931149318. local maturation of dendritic spines. J Neurosci 2:331340.
Martinez R, Gomes FC (2005) Proliferation of cerebellar neu- Nishino J, Yamashita K, Hashiguchi H, Fujii H, Shimazaki T,
rons induced by astrocytes treated with thyroid hormone is Hamada H (2004) Meteorin: a secreted protein that regu-
mediated by a cooperation between cell contact and solu- lates glial cell differentiation and promotes axonal extension.
ble factors and involves the epidermal growth factor-protein EMBO J 23:19982008.
kinase a pathway. J Neurosci Res 3:341349. Nishiyama H, Knopfel T, Endo S, Itohara S (2002) Glial pro-
Mauch DH, Nagler K, Schumacher S, Goritz C, Muller EC, Otto tein S100 modulates long-term neuronal synaptic plasticity.
A, Pfrieger FW (2001) CNS synaptogenesis promoted by glia Proc Natl Acad Sci USA 99:40374042.
derived cholesterol. Science 294:13541357. Noctor SC, Flint AC, Weissman TA, Dammerman RS,
Mecha M, Rabadn MA, Pea-Melin A, Valencia M, Mondjar Kriegstein AR (2001) Neurons derived from radial glial cells
T, Blanco MJ (2008) Expression of TGF-betas in the embry- establish radial units in neocortex. Nature 409:714720.
onic nervous system: analysis of interbalance between iso- Ngler K, Mauch DH, Pfrieger FW (2001) Glia-derived sig-
forms. Dev Dyn 237:17091717. nals induce synapse formation in neurones of the rat central
Mendes FA, Onofre GR, Silva LCF, Cavalcante LA, Garcia- nervous system. J Physiol 533:665679.
Abreu J (2003) Concentration-dependent actions of glial Okada M, Matsumura M, Ogino N, Honda Y (1990) Mller
chondroitin sulfate on the neuritic growth of midbrain neu- cells in detached human retina express glial fibrillary acidic
rons. Dev Brain Res 142:111119. protein and vimentin. Graefes Arch Clin Exp Ophthalmol
Merkle FT, Tramontin AD, Garcia-Verdugo JM, Alvarez-Buylla 228:467474.
A (2004) Radial glia give rise to adult neural stem cells Oliet SH, Piet R, Poulain DA (2001) Control of glutamate clear-
in the subventricular zone. Proc Natl Acad Sci USA 101: ance and synaptic efficacy by glial coverage of neurons.
1752817535. Science 5518:923926.
Miller MW (2003) Expression of transforming growth factor- Parpura V, Haydon PG (2000) Physiological astrocytic cal-
beta in developing rat cerebral cortex: effects of prenatal cium levels stimulate glutamate release to modulate adjacent
exposure to ethanol. J Comp Neurol 460:410424. neurons. Proc Natl Acad Sci USA 15:86298634.
Miller FD, Gauthier AS (2007) Timing is everything: making Parri HR, Gould TM, Crunelli V (2001) Spontaneous astrocytic
neurons versus glia in the developing cortex. Neuron 54: Ca2+ oscillations in situ drive NMDAR mediated neuronal
357369. excitation. Nature Neurosci 4:803812.
Mong JA, Nuez JL, McCarthy MM (2002) GABA mediates Patten BA, Peyrin JM, Weinmaster G, Corfas G (2003)
steroid-induced astrocyte differentiation in the neonatal rat Sequential signaling through Notch1 and erbB recep-
hypothalamus. J Neuroendocrinol 14:4555. tors mediates radial glia differentiation. J Neurosci
Morrow T, Song MR, Ghosh A (2001) Sequential specification 23:61326140.
of neurons and glia by developmentally regulated extracellu- Perego C, Vanoni C, Bossi M, Massari S, Basudev H, Longhi R,
lar factors. Development 128:35853594. Pietrini G (2000) The GLT-1 and GLAST glutamate trans-
Morshead CM, Reynolds BA, Craig CG, McBurney MW, porters are expressed on morphologically distinct astrocytes
Staines WA, Morassutti D, Weiss S, van der Kooy D (1994) and regulated by neuronal activity in primary hippocampal
Neural stem cells in the adult mammalian forebrain: a cocultures. J Neurochem 3:10761084.
relatively quiescent subpopulation of subependymal cells. Pfrieger FW (2003) Role of cholesterol in synapse formation and
Neuron 13:10711082. function. Biochim Biophys Acta 2:271280.
Murai KK, Nguyen LN, Irie F, Yamaguchi Y, Pasquale EB Pfrieger FW, Barres BA (1997) Synaptic efficacy enhanced by
(2003) Control of hippocampal dendritic spine morphol- glial cells in vitro. Science 277:16841687.
ogy through ephrin-A3/EphA4 signaling. Nat Neurosci 6: Pin JP, Duvoisin R (1995) The metabotropic glutamate recep-
153160. tors: structure and functions. Neuropharmacology 34:126.
Nakashima K, Nakashima K, Wiese S, Yanagisawa M, Arakawa Pinto L, Gtz M (2007) Radial glial cell heterogeneity the
H, Kimura N, Hisatsune T, Yoshida K, Kishimoto T, Sendtner source of diverse progeny in the CNS. Prog Neurobiol
M, Taga T (1999a) Developmental requirement of gp130 sig- 83:223.
naling in neuronal survival and astrocyte differentiation. J Pixley SKR, De Vellis J (1984) Transition between immature
Neurosci 19:54295434. radial glia and mature astrocytes studied with a monoclonal
Nakashima K, Yanagisawa M, Arakawa H, Kimura N, Hisatsune antibody to vimentin. Dev Brain Res 15:201209.
T, Kawabata M, Miyazono K, Taga T (1999b) Synergistic Porter JT, McCarthy KD (1997) Astrocytic neurotrans-
signaling in fetal brain by STAT3-Smad1 complex bridged mitter receptors in situ and in vivo. Prog Neurobiol
by p300. Science 284:479482. 51:439455.
Nedergaard M, Ransom B, Goldman SA (2003) New roles for Pratt BM, McPherson JM (1997) TGF-beta in the central ner-
astrocytes: redefining the functional architecture of the brain. vous system: potential roles in ischemic injury and neurode-
Trends Neurosci 26:523530. generative diseases. Cytokine Growth Factor Rev 8:267292.
11 Neuron-Astroglial Interactions in Cell Fate Commitment 169

Prochiantz A (1995) Neuronal polarity: giving neurons heads of the germinal zone in the adult dentate gyrus. J Comp
and tails. Neuron 15:743746. Neurol 478:359378.
Qian X, Shen Q, Goderie SK, He W, Capela A, Davis Seri B, Garca-Verdugo JM, McEwen BS, Alvarez-
AA, Temple S (2000) Timing of CNS cell generation: Buylla A (2001) Astrocytes give rise to new neurons
a programmed sequence of neuron and glial cell pro- in the adult mammalian hippocampus. J Neurosci
duction from isolated murine cortical stem cells. Neuron 21:71537160.
28:6980. Sharif A, Legendre P, Prvot V, Allet C, Romao L, Studler J-
Rakic P (1971) Guidance of neurons migrating to the fetal M, Chneiweiss H, Junier M-P (2006) Transforming growth
monkey neocortex. Brain Res 33:471476. factor promotes sequential conversion of mature astro-
Rakic P (2003) Developmental and evolutionary adaptations of cytes into neural progenitors and stem cells. Oncogene 26:
cortical radial glia. Cereb Cortex 13:541549. 26952706.
Raponi E, Agenes F, Delphin C, Assard N, Baudier J, Sheldon AL, Robinson MB (2007) The role of gluta-
Legraverend C, Deloulme JC (2007) S100 expression mate transporters in neurodegenerative diseases and poten-
defines a state in which GFAP-expressing cells lose their tial opportunities for intervention. Neurochem Int 51:
neural stem cell potential and acquire a more mature devel- 333355.
opmental stage. Glia 55:165177. Shelton MK, McCarthy KD (2000) Hippocampal astrocytes
Raymond PA, Hitchcock PF (1997) Retinal regeneration: com- exhibit Ca2+ -elevating muscarinic cholinergic and histamin-
mon principles but a diversity of mechanisms. Adv Neurol ergic receptors in situ. J Neurochem 74:555563.
72:171184. Shi Y, Massagu J (2003) Mechanisms of TGF-beta signaling
Regan MR, Huang YH, Kim YS, Dykes-Hoberg MI, Jin L, from cell membrane to the nucleus. Cell 11:685700.
Watkins AM, Bergles DE, Rothstein JD (2007) Variations in Somjen GG (1988) Nervenkitt: notes on the history of the
promoter activity reveal a differential expression and physi- concept of neuroglia. Glia 1:29.
ology of glutamate transporters by glia in the developing and Song HJ, Stevens CF, Gage FH (2002) Astroglia induce neuro-
mature CNS. J Neurosci 25:66076619. genesis from adult neural stem cells. Nature 417:3944.
Robitaille R (1998) Modulation of synaptic efficacy and synaptic Soriano E, Alvarado-Mallart RM, Dumesnil N, Del Ro J,
depression by glial cells at the frog neuromuscular junction. Sotelo C (1997) Cajal-Retzius cells regulate the radial glia
Neuron 4:847855. phenotype in the adult and developing cerebellum and alter
Romo LF, de Sousa OV, Neto VM, Gomes FC (2008) granule cell migration. Neuron 18:563577.
Glutamate activates GFAP gene promoter from cultured Sottile V, Li M, Scotting PJ (2006) Stem cell marker expression
astrocytes through TGF-beta1 pathways. J Neurochem in the Bergmann glia population of the adult mouse brain.
2:746756. Brain Res 1099:817.
Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, Spassky N, Merkle FT, Flames N, Tramontin AD, Garca-
Kuncl RW, Kanai Y, Hediger MA, Wang Y, Schielke JP, Verdugo JM, Alvarez-Buylla A (2005) Adult ependymal
Welty DF (1996) Knockout of glutamate transporters reveals cells are postmitotic and are derived from radial glial cells
a major role for astroglial transport in excitotoxicity and during embryogenesis. J Neurosci 25:1018.
clearance of glutamate. Neuron 16:675686. de Sampaio e Spohr TCL, Choi JW, Gardell SE, Herr DR,
Runquist M, Alonso G (2003) Gabaergic signaling mediates Rehen SK, Gomes FC, Chun J (2008) Lysophosphatidic acid
the morphological organization of astrocytes in the adult rat receptor-dependent secondary effects via astrocytes promote
forebrain. Glia 41:137151. neuronal differentiation. J Biol Chem 283:74707479.
Sardi SP, Murtie J, Koirala S, Patten BA, Corfas G (2006) Steiner B, Klempin F, Wang L, Kott M, Kettenmann H,
Presenilin-dependent ErbB4 nuclear signaling regulates the Kempermann G (2006) Type-2 cells as link between glial
timing of astrogenesis in the developing brain. Cell 127: and neuronal lineagein adult hippocampal neurogenesis. Glia
185197. 54:805814.
Schmechel DE, Rakic P (1979) A Golgi study of radial glial Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson
cells in developing monkey telencephalon: morphogene- KS, Nouri N, Micheva KD, Mehalow AK, Huberman
sis and transformation into astrocytes. Anat Embryol 156: AD, Stafford B, Sher A, Litke AM, Lambris JD, Smith
115152. SJ, John SW, Barres BA (2007) The classical comple-
Schmid RS, Mcgrath B, Berechid BE, Boyles B, Marchionni M, ment cascade mediates CNS synapse elimination. Cell
Sestan N, Anton ES (2003) Neuregulin 1-erbB2 signaling is 6:11641178.
required for the establishment of radial glia and their trans- Stipursky J, Gomes FC (2007) TGF-beta1/SMAD signaling
formation into astrocytes in cerebral cortex. Proc Natl Acad induces astrocyte fate commitment in vitro: implications for
Sci USA 7:42514256. radial glia development. Glia 55:10231033.
Schools GP, Kimelberg HK (1999) mGluR3 and mGluR5 are Sullivan SM, Lee A, Bjorkman ST, Miller SM, Sullivan RK,
the predominant metabotropic glutamate receptor mRNAs Poronnik P, Colditz PB, Pow DV (2007) Cytoskeletal
expressed in hippocampal astrocytes acutely isolated from anchoring of GLAST determines susceptibility to brain
young rats. J Neurosci Res 58:533543. damage: an identified role for GFAP. J Biol Chem
Seifert G, Schilling K, Steinhauser C (2006) Astrocyte dysfunc- 282:2941429423.
tion in neurological disorders: a molecular perspective. Nat Sun Y, Nadal-Vicens M, Misono S, Lin MZ, Zubiaga A, Hua
Rev Neurosci 7:194206. X, Fan G, Greenberg ME (2001) Neurogenin promotes neu-
Seri B, Garca-Verdugo JM, Collado-Morente L, McEwen BS, rogenesis and inhibits glial differentiation by independent
Alvarez-Buylla A (2004) Cell types, lineage, and architecture mechanisms. Cell 104:365376.
170 J. Stipursky et al.

Supr H, Del Ro JA, Martnez A, Prez-Sust P, Soriano E Wachs FP, Winner B, Couillard-Despres S, Schiller T, Aigner
(2000) Disruption of neuronal migration and radial glia in R, Winkler J, Bogdahn U, Aigner L (2006) Transforming
the developing cerebral cortex following ablation of Cajal- growth factor-beta1 is a negative modulator of adult
Retzius cells. Cereb Cortex 10:602613. neurogenesis. J Neuropathol Exp Neurol 65:358370.
Swanson RA, Liu J, Miller JW, Rothstein JD, Farrell K, Stein Wang Z, Haydon PG, Yeung ES (2000) Direct observation of
BA, Longuemare MC (1997) Neuronal regulation of gluta- calcium-independent intercellular ATP signaling in astro-
mate transporter subtype expression in astrocytes. J Neurosci cytes. Analyt Chem 72:20012007.
3:932940. Ware CB, Horowitz MC, Renshaw BR, Hunt JS, Liggitt D,
Takahashi T, Misson JP, Caviness VS Jr (1990) Glial process Koblar SA, Gliniak BC, McKenna HJ, Papayannopoulou
elongation and branching in the developing murine neo- T, Thoma B (1995) Targeted disruption of the low-affinity
cortex: a qualitative and quantitative immunohistochemical leukemia inhibitory factor receptor gene causes placental,
analysis. J Comp Neurol 302:1528. skeletal, neural and metabolic defects and results in perinatal
Tateishi N, Shimoda T, Manako J, Katsumata S, Shinagawa death. Development 121:12831299.
R, Ohno H (2006) Relevance of astrocytic activation Wyss-Coray T, Feng L, Masliah E, Ruppe MD, Lee HS, Toggas
to reductions of astrocytic GABAA receptors. Brain Res SM, Rockenstein EM, Mucke L (1995) Increased central ner-
1:7991. vous system production of extracellular matrix components
Temple S (2001) The development of neural stem cells. Nature and development of hydrocephalus in transgenic mice over-
414:112117. expressing transforming growth factor-beta1. Am J Pathol
Tesseur I, Wyss-Coray T (2006) A role for TGF-beta signaling 147:5367.
in neurodegeneration: evidence from genetically engineered Yang Y, Ge W, Chen Y, Zhang Z, Shen W, Wu C, Poo M, Duan S
models. Curr Alzheimer Res 3:505513. (2003) Contribution of astrocytes to hippocampal long-term
Ueki T, Tanaka M, Yamashita K, Mikawa S, Qiu Z, Maragakis potentiation through release of D-serine. Proc Natl Acad Sci
NJ, Hevner RF, Miura N, Sugimura H, Sato K (2003) USA 100:1519415199.
A novel secretory factor, Neurogenesin-1, provides neuro- Yoon K, Gaiano N (2005) Notch signaling in the mammalian
genic environmental cues for neural stem cells in the adult central nervous system: insights from mouse mutants. Nat
hippocampus. J Neurosci 23:1173211740. Neurosci 8:709715.
Ullian EM, Christopherson KS, Barres BA (2004) Role for glia Yoon K, Nery S, Rutlin ML, Radtke F, Fishell G, Gaiano N
in synaptogenesis. Glia 47:209216. (2004) Fibroblast growth factor receptor signaling promotes
Ullian EM, Sapperstein SK, Christopherson KS, Barres BA radial glial identity and interacts with Notch1 signaling in
(2001) Control of synapse number by glia. Science 291: telencephalic progenitors. J Neurosci 24:94979506.
657661. Zheng W, Nowakowski RS, Vaccarino FM (2004) Fibroblast
Vardimon L, Ben-Dror I, Havazelet N, Fox LE (1993) Molecular growth factor 2 is required for maintaining the neural stem
control of glutamine synthetase expression in the developing cell pool in the mouse brain subventricular zone. Dev
retina tissue. Dev Dyn 196:276282. Neurosci 26:181196.
Ventura R, Harris KM (1999) Three-dimensional relationships Zhou M, Kimelberg HK (2001) Freshly isolated hippocampal
between hippocampal synapses and astrocytes. J Neurosci CA1 astrocytes comprise two populations differing in gluta-
16:68976906. mate transporter and AMPA receptor expression. J Neurosci
Verkhratsky A, Kirchhoff F (2007) NMDA receptors in glia. 21:79017908.
Neuroscientist 1:2837. Zhou CJ, Zhao C, Pleasure SJ (2004) Wnt signaling mutants
Vivien D, Ali C (2006) Transforming growth factor-beta sig- have decreased dentate granule cell production and radial
nalling in brain disorders. Cytokine Growth Factor Rev glial scaffolding abnormalities. J Neurosci 24:121126.
17:121128. Zhu Y, Yang GY, Ahlemeyer B, Pang L, Che XM, Culmsee C,
Voigt T (1989) Development of glial cells in the cere- Klumpp S, Krieglstein J (2002) Transforming growth factor-
bral wall of ferrets: direct tracing of their transforma- beta 1 increases bad phosphorylation and protects neurons
tion from radial glia into astrocytes. J Comp Neurol against damage. J Neurosci 22:38983909.
289:7488. Ziegler DR, Innocente CE, Leal RB, Rodnight R, Gonalves CA
Volterra A, Meldolesi J (2005) Astrocytes, from brain glue to (1998) The S100 protein inhibits phosphorylation of GFAP
communication elements: the revolution continues. Nat Rev and vimentin in a cytoskeletal fraction from immature rat
Neurosci 6:626640. hippocampus. Neurochem Res 23:12591263.
Chapter 12

The Origin of Microglia and the Development of the Brain

Flavia R. S. Lima, Anna Carolina C. da Fonseca, Giselle P. Faria, Luiz Gustavo F. Dubois,
Trcia R. Alves, Jane Faria, and Vivaldo Moura Neto

Contents proliferation, and differentiation of microglial cells are


not completely known. These important events may
12.1 Microglia: Origin and Development . . . . . 172 depend on environmental factors such as soluble or
12.1.1 Origin of Microglia . . . . . . . . . . 173 cell-surface-bound molecules and components of the
12.1.2 Invasion of the CNS by Microglial extracellular matrix. In the developing CNS, microglial
Precursors During Development . . . . . 174
cells are involved in clearing cell debris and withdraw-
12.1.3 Expansion of Microglial Population within
CNS . . . . . . . . . . . . . . . . . 175 ing transitory or misdirected axons, and presumably
12.1.4 Microglial Development and Thyroid support neurogenesis, cell survival, and neurite growth.
Hormones . . . . . . . . . . . . . . 177 In the adult brain, activated microglia occur mostly
12.1.5 Adult CNS: Ramified Microglia . . . . . 178
in response to neuronal injuries, when, they destroy
12.2 Microglia and Regressive Processes During Brain
Development: Phagocytosis and Neurotoxic invading microorganisms, remove harmful debris, and
Factors . . . . . . . . . . . . . . . . . . . 179 promote tissue repair, as well as partaking in the
12.3 Microglial Secreted Neurotrophic Factors: Role immune response by secreting cytokines, facilitat-
in Neural Development . . . . . . . . . . . . 181
ing the return to homeostasis. Microglial activation
12.3.1 Microglia and Neural Progenitor Cells . . 182
12.4 The Future . . . . . . . . . . . . . . . . . 183 is an important event in the defense of the nervous
References . . . . . . . . . . . . . . . . . . . . 184 parenchyma against infectious, neurodegenerative, and
inflammatory diseases. The majority of data in the
literature describes microglial cells in a neuropatho-
Abstract We discuss the origin and development logical context. Little is known of the development
of microglial cells and their influence on neural of these cells in the nervous parenchyma and their
development. Unlike astrocytes, oligodendrocytes, and effective role in neurogenesis.
neurons, which are derived from neuroectoderm,
microglial cells originate from mesoderm. Microglial Keywords Microglia Development Progenitor
hematopoietic precursors enter the developing CNS Growth factor Neurogenesis
from the bloodstream, ventricles, and meninges. In
the brain, these cells migrate and proliferate, as Abbreviations
ameboid microglia, and become distributed through-
out the nervous parenchyma. Ameboid microglial b-FGF basic fibroblast growth factor
cells differentiate into ramified microglia when they CSF colony stimulating factor
reach their definitive location. The factors that con- GDNF glial-derived neurotrophic factor
trol the invasion of the nervous parenchyma, migration, GFAP glial fibrillary acidic protein
GM granulocyte macrophage
gw gestational weeks
F.R.S. Lima ()
Programa de Anatomia, Instituto de Cincias Biomdicas, HGF hepatocyte growth factor
Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil HPC hippocampal progenitor cell(s)
e-mail: flima@anato.ufrj.br ICAM-1 intercellular adhesion molecule-1

H. Ulrich (ed.), Perspectives of Stem Cells, 171


DOI 10.1007/978-90-481-3375-8_12, Springer Science+Business Media B.V. 2010
172 F.R.S. Lima et al.

IFN- interferon gamma These latter cell types can also appear in the adult brain
IL interleukin in pathological conditions.
iNOS inducible NO synthase Macrophages correspond to the terminal stages of
LFA-1 lymphocyte function-associated antigen-1 mononuclear phagocyte lineage differentiation. In gen-
LIF leukemia inhibitory factor eral, macrophages do not constitute a homogeneous
LPS lypopolysaccharide population, but rather are heterogeneous and have spe-
MAP2 microtubule-associated protein 2 cific structure and functions depending on the tissue.
MAPK mitogen-activated protein kinase In the CNS, macrophages can be present in different
MHC II class II major histocompatibility complex forms, according to their location, the stage of devel-
MIP macrophage inflammatory protein opment, and tissue integrity (Perry et al., 1994; Streit,
NGF nerve growth factor
2001). Microglial cells are considered a subpopula-
NSC neural stem cell(s)
tion of intraparenchymal cells, which are distinguished
NT neurotrophin
from phagocytes located in the meninges and in the
OPC oligodendrocyte precursor cell(s)
coroid plexus stroma, or from macrophages situated in
PAF platelet-activating factor
PI3K phosphoinositol-3-kinase the cerebral capillary walls (Fig. 12.1).
PKC protein kinase C In the immature CNS, microglial cells show a het-
PS phosphatidylserine erogeneous morphology, either a dilated cell body
RCA-1 Ricinus communed agglutinin-1 tending to several forms for developing pseudopods
ROS reactive-oxygen species and filopods, or a rounded cell body without cytoplas-
TGF 1 transforming growth factor 1 matic expansions. These variations may be associated
TH thyroid hormones with the origin and cerebral location of these cells
TNF tumor necrosis factor (Perry and Gordon, 1988; Streit, 2001). From the
ultrastructural viewpoint, ameboid microglia have a
cytoplasm rich in organelles, a developed Golgi appa-
ratus, with vacuoles and lysosomes in abundance, indi-
cating intense activity of synthesis and phagocytosis
12.1 Microglia: Origin and Development (Murabe and Sano, 1982). The expression of non-
specific esterases, acid phosphatases, and 5nucleoti-
Together with astrocytes and oligodendrocytes, dases activity by ameboid microglia also distinguish
microglial cells comprise the principal glial popu- them from the ramified microglia (Ling, 1981; Hanish
lations in the CNS. Microglial cells were initially and Kettenmann, 2007).
described by Del Ro Hortega in 1932; however, only There are two extreme microglial phenotypes: ram-
in the 1980s did they begin to receive more attention ified microglia, which are at first not very active func-
and study. Using silver-carbonate stains in nervous- tionally; and ameboid microglia, which are involved
tissue preparations, Del Ro Hortega (1932) described in CNS growth and repair. However, the situation is
cells constituted by a small cell body and numerous
ramified cell process. Because of their morphology,
this cell type was denominated microglia, in contrast
to macroglia (astrocytes and oligodendrocytes).
In the cerebral parenchyma, microglial cells are
distinguished from the other cell types by their mor-
phology, and are recognized by the expression of
specific mononuclear phagocyte markers. According
to the maturation stage of the CNS or to nerve-tissue
integrity, these cells can acquire different phenotypes.
In the absence of injury, the ramified microglial cells Fig. 12.1 Purified microglia cultures from neonatal mice
cortex. Cells stained by Isolectin B4 derived from Griffonia sim-
are the typical microglia of the adult nervous tissue. plicifolia conjugated with FITC. Nuclei stained by DAPI. Left
In the immature CNS, brain macrophages, also known panel, bar 50 m. Right panel, bar 10 m. Photomicrographs
as ameboid microglial cells, accumulate transitorily. taken by Anna Carolina Fonseca (UFRJ)
12 The Origin of Microglia and the Development of the Brain 173

more complex than it seems. It has been suggested that and before monocytes are produced in hemopoietic
intermediate microglial phenotypes exist, modifying tissues (Hurley and Streit, 1996; Navascus et al.,
morphology and function during brain development 2000). Thus, it is possible that not all microglial cells
in particular, and also after injury. These intermediate originate from circulating monocytes during develop-
microglial cell forms appear during differentiation pro- ment.
cesses or activation of the microglia (Perry et al., 1994; In the adult CNS, discrimination between infiltrat-
Kreutzberg, 1996; Streit, 2001; Schwartz et al., 2006). ing mononuclear cell and activated resident microglia
is difficult, as well as their corresponding tissue of
origin or precursor cell lineages, mostly due to the
lack of cell surface specific markers to distinguish
12.1.1 Origin of Microglia these cells (reviewed by Chan et al., 2007). Expression
of the myeloid-specific transcription factor PU.1 by
The origin of microglia has been discussed for many microglia (Walton et al., 2000) indicates a possible
years. However, current information indicates that myeloid origin, but not necessarily from blood mono-
they derive from mesenchymal precursor infiltration. cyte progenitors (Asheuer et al., 2004; Hess et al.,
During brain development, precursors generated in 2004; Vallieres and Sawchenko, 2003; Vitry et al.,
the bone marrow invade the nervous parenchyma and 2003).
differentiate into microglial cells (Ling and Wong, Monocytes ingress into the brain and seem to be
1993; Cuadros and Navascus, 1998; Kaur et al., 2001; the direct circulating precursors of postnatal microglia
Chan et al., 2007). The first studies that supported only under defined conditions, such as in whole-body
this hypothesis were done in the 1980s. Attempting irradiated chimeric mice (Mildner et al., 2007) or
to show that microglial cells derive from monocytic during post inflammatory period of bacterial menin-
lineages, Ling and cols. (1980) injected monocytes gitis (Djukic et al., 2006). Despite having a strong
labeled with carbon into the bloodstream of new- potential for proliferation and self-renewal in the adult
born rats. Subsequently, they detected the appearance CNS, microglial proliferation normally occurs at very
of microglial cells containing carbon particles in the low levels, increasing significantly after CNS injury
CNS, particularly in the corpus callosum, in the first (Graeber et al., 1988; Ajami et al., 2007).
days of postnatal life. Using antibodies against the Recently, new concepts about the origin of
F4/80 antigen, a highly specific marker for mononu- microglia during development were proposed in a
clear phagocytes in rodents, immunohistochemical review paper (Chan et al., 2007). The authors pro-
studies revealed positive labeling for circulating mono- posed that the CNS invasion by microglial precursors
cyte invaders in the nervous tissue in embryonic stages, occurs in two waves (Fig. 12.2). The first occurs during
ameboid microglia in the prenatal period, and rami- fetal development, and cells are derived mainly from
fied microglia in the adult brain (Hume et al., 1983; extravascular progenitors (of myeloid/mesenchymal
Perry et al., 1985). Later, Giulian et al. (1995) demon- but not monocytic origin). The second population
strated that, in vitro, monocytes isolated from the invades the brain during perinatal and postnatal devel-
newborn brain give rise to cells with microglial fea- opment. These cells are derived from circulating pro-
tures. However, these cells do not appear in cultures genitors (probably monocytes), and give rise to so-
from monocytes derived from other sources. In this called ameboid microglia (Rezaie and Male, 2002;
context, it was also reported that a cellular subpopula- Simard and Rivest, 2004; Rezaie et al., 2005; Tsuchiya
tion derived from bone marrow showed the same ionic et al., 2005; Chan et al., 2007).
channel model as the microglial cells (Banati et al., In accordance with this idea, Razaie and cols.
1991; Chan et al., 2007). These pioneering studies sug- (2005) reported that during human brain develop-
gested that bone marrow produces specific precursors ment, the cerebral wall of the human telencephalon
for microglia, which are different from those that will is colonized by at least two putative populations
originate macrophages for other tissues. of cells that originate microglia mainly during the
However, microglial cells appear within the CNS second trimester of gestation. These subpopulations
before brain vascularization (Ashwell, 1991; Sorokin can be discriminated based on differences in pheno-
et al., 1992; Cuadros et al., 1993; Wang et al., 1996) type, morphological characteristics, and distribution
174 F.R.S. Lima et al.

Fig. 12.2 Simplified


scheme of the theory
of mesodermal/myeloid
origin for microglia during
development. Hematopoietic
stem cells give rise to the
lymphoid and myeloid
blood-cell lineages.
A committed myeloid
precursor may give rise to
monocyte precursors or to
granulocyte precursors, and
both may originate microglia.
Adapted from Chan et al.
(2007)

patterns within the developing nervous system. The Microglial progenitors undergo homing to the ner-
first progenitor population is RCA-1(++; Ricinus com- vous system, so they could be usefull tools for ther-
muned agglutinin-1), CD68(), class II major histo- apeutic purposes (Dobrenis, 1998; Cucchiarini et al.,
compatibility complex [MHCII()], underlies the mes- 2003; Watanabe et al., 2002; Chan et al., 2007).
enchyme and meninges, and resides in the subplate. Upon genetic manipulation, microglia progenitors
These cells differentiate early within the subplate. could locally deliver specific gene products for the
The second progenitor population is CD68(++), RCA- treatment of several disorders, such as metachromatic
1(+) and MHCII(), and adopts the typical ameboid leukodystrophy, gliomas and stroke (Chan et al., 2007).
macrophage-like forms. These progenitor cells reside Nevertheless, genetic or cell therapy will only be pos-
in the deep marginal layer and at the boundary between sible when tissue of origin or cell lineage of resident
the cortical plate and the subplate at 1417 gestational microglia can be precisely identified.
weeks (gw), where they also appear to differentiate and
progressively populate the upper part of this region by
24 gw. At this time, microglia can be found all over 12.1.2 Invasion of the CNS by Microglial
the developing human fetal telencephalon, and differ- Precursors During Development
entiation into ramified forms characteristic of adult
microglial cells begins also be seen, probably consti- Considering that microglial precursors originate out-
tuting a merger of the two populations of progenitor side the nervous parenchyma, studies have demon-
cells (Rezaie et al., 2005). strated, in rodents, that these cells appear in the
12 The Origin of Microglia and the Development of the Brain 175

nervous parenchyma especially at the end of embryo- suggested that microglial precursors cross the pial bor-
genesis and in the first days of postnatal life, although der to reach the nervous parenchyma (Boya et al.,
their presence is already observed from the 12th day 1991; Navascus et al., 1996; Dalmau et al., 1997;
of embryonic life (E12; Perry et al., 1985; Ashwell, Monier et al., 2007). Navascus group reported that
1991; Milligan et al., 1991; Navascus et al., 2000; most microglial cells in the optic tectum and cerebel-
Chan et al., 2007). In humans, precursors invade the lum seem to derive from precursors that have entered
nervous tissue during the first two trimesters of ges- the nervous parenchyma by this route (Cuadros et al.,
tation (Billiards et al., 2006; Monier et al., 2007). 1994; 1997; Navascus et al., 2000).
The entry of microglial precursors during this period, It appears that the microglial precursors can use all
added to the proliferation of these cells within the three of these invasion routes. It has been suggested
CNS, must provide enough precursors to produce the that these cells enter the nervous tissue via different
mature microglial population, because the turnover of pathways, and generate different microglial subpop-
microglia in the adult brain is slow (Lawson et al., ulations (Provis et al., 1996; Navascus et al., 2000;
1992; Navascus et al., 2000). Chan et al., 2007).
Three invasion routes have been proposed. The first
route is from the bloodstream. In this route, blood cir-
culating cells may enter the CNS, adhering to vessel
12.1.3 Expansion of Microglial
walls and gaining access to the nervous parenchyma
through a mechanism similar to that described for the Population within CNS
invasion of other tissues by macrophages and mono-
cytes (Cramer, 1992). The LFA-1/ICAM-1 (lympho- 12.1.3.1 Proliferation
cyte function-associated antigen-1/lymphocytes/ inter-
cellular adhesion molecule-1) adhesion system, which The growth of the microglial population does not result
is involved in the entry of leukocytes into the injured exclusively from infiltration of the mesodermal pre-
brain parenchyma, must also be involved in this mech- cursors. Different studies have shown that microglial
anism (Akiyama et al., 1994; Cuadros and Navascus, cells proliferate actively during brain development. In
1998). Because the blood-brain barrier is formed later rodents, the number of microglia detected in the cor-
in development, many microglial precursors use this pus callosum grows significantly between the 5th day
route to invade the CNS and to reach their final location and 2nd week of postnatal life. The high proliferation
without needing to migrate alone for long distances. capacity of these cells is attested by a high rate of
However, microglial cells also appear in brain regions incorporation of tritiated thymidine, which reaches a
that are not vascularized at some stage of devel- peak of almost 80% of labeled cells on the 12th post-
opment, such as in the mammal retina, where they natal day (Imamoto and Leblond, 1978; Mallat et al.,
appear before this region is vascularized (Schnitzer, 1997; Dalmau et al., 2003).
1989; Diaz-Araya et al., 1995; Chen et al., 2002). In vitro studies have characterized certain Colony
Therefore other routes should be considered, such as Stimulating Factors (CSFs) as mitogenes for microglia
the entry of microglial precursors from the ventricles. (Suzumura et al., 1990; Elkabes et al., 1996), such
The idea that microglial precursors may reach the ner- as colony-stimulating factors-1 (CSF-1), granulo-
vous parenchyma from the ventricles is based on the cyte macrophage (GM)-CSF and interleukin (IL)-3
presence of monocyte and macrophage lineages in the (Giulian and Ingeman, 1988; Hao et al., 1990; Lee
ventricular lumen (Jordan and Thomas, 1988; Monier et al., 1993; Imai and Kohsaka, 2002). In particu-
et al., 2007). Moreover, microglial cells apparently lar, CSF-1 specifically stimulates mononuclear phago-
cross the ventricular surface during brain development cyte production, and it is expressed in several tissues,
(Cuadros et al., 1994; Cuadros and Navascus, 1998). including the brain.
The third route that has been documented is from Mitogenic effects of vitamin E, glial-derived neu-
the meninges. This entry route of microglial precur- rotrophic factor (GDNF), transforming growth fac-
sors was first proposed by Del Ro-Hortega (1932). tor (TGF) 1, and neurotrophin 3 (NT-3) were also
Studies by later investigators supported this idea, and observed in microglial cells in vitro (Elkabes et al.,
from studies using electron microscopic images, they 1996; Salimi et al., 2003; Flanary and Streit, 2006).
176 F.R.S. Lima et al.

In vivo, a decrease in the number of microglial cells in et al., 2003; Rozenfeld et al., 2005). Indeed, analyses
the subventricular zone was seen in knockout mice for using recombinant CSF-1 on microglial cultures have
NT-3 and its trkC receptor (Kahn et al., 1999). shown that this factor, which is secreted essentially by
astrocytes, may induce microglial migration in vitro
(Calvo et al., 1998). Recently, it was demonstrated that
12.1.3.2 Migration treatment with ATP increased the migration of cultured
rat microglia by about 4-fold, and that prostaglandin E
Navascus group has been studying the microglial (PGE)-2 effectively reduced this effect (Nagano et al.,
migration in some regions of the quail brain, such 2008).
as the optic tectum (Cuadros et al., 1994), the cere-
bellum (Cuadros et al., 1997), and, mostly, the retina
(Navascus et al., 1995; Marn-Teva et al., 1999). They 12.1.3.3 Differentiation
established a model according to which the migration
in these regions occurs in two different steps. During The macrophage phenotype of microglia disappears
the first step, termed tangential migration, microglial progressively during CNS maturation. In the rat cor-
precursors move together from their entry point within pus callosum, this alteration is observed from the 2nd
the nervous parenchyma, traveling over long distances week of postnatal life (Imamoto and Leblond, 1978;
on oriented substrates. These substrates (axon fasci- Dalmau et al., 2003). Part of these ameboid microglia
cles) may act as highways, leading the precursors into that are present in the embryonic brain may degen-
the interior of the nervous parenchyma. In the sec- erate, as occurs with activated microglia in the adult
ond stage, termed radial migration, microglial cells brain (Gehrmann and Banati, 1995; Jones et al., 1997).
change direction and move towards the pial surface, However, many of them develop narrow processes,
reaching all parts of the nervous parenchyma (Monier becoming progressively ramified. This microglial dif-
et al., 2007). Moreover, analyses during the retina ferentiation idea has been suggested by several workers
development suggest that microglial cells migrate (Perry and Gordon, 1988; Ling and Wong, 1993; Davis
along the radial processes of Mller cells (Navascus et al., 1994; Cuadros and Navascues, 2001), and it was
et al., 1996). Thus, microglial precursors leave high- first demonstrated in situ with different markers, such
ways and enter secondary roads to reach their final as tritiated thymidine (Imamoto and Leblond, 1978),
destination within the nervous parenchyma (Cuadros carbon (Ling et al., 1980), and rhodamine isothyocy-
and Navascus, 1998, 2001). In vitro morphological anate (Leong and Ling, 1992). This morphological
and immunocytochemical analyses indicated the pos- transformation is accompanied by loss of the phago-
sibility that microglia may alternate migration and cytic function (Kaur and Ling, 1991), decreased
proliferation cycles, contributing to the expansion of expression of some macrophagic markers such as CR3
the microglial population during retina development and CMH I, and the disappearance of antigens such
(Marn-Teva et al., 1999; Navascus et al., 2000). as ED1 in the rat (Ling et al., 1991). Retinoic acid
The migration model described above refers to can induce loss of phagocytic activity, favoring dif-
regions of the immature quail brain, which has a lam- ferentiation in vitro (Giulian and Baker, 1986). Many
inar organization. Although it is not yet clear whether immunohistochemical features that are present in ame-
this model applies to all regions of the quail brain boid microglia but are lost in ramified microglia,
or whether similar steps occur in other species, some later reappear in the activated microglia (Streit and
studies in immature CNS of mammals have indicated Kreutzberg, 1988; Davis et al., 1994; Hanisch and
this possibility (Perry et al., 1985; Milligan et al., 1991; Kettenmann, 2007). The distribution of microtubules
Monier et al., 2007). can also change in cultured microglial cells. Ramified
Ameboid microglia as well as astrocytes produce microglia show more stable microtubules, with a
cytokines that are involved in invasion and migration higher rate of acetylation and detyrosination than in
of microglial cells, such as TGF, CSF-1, and the ameboid microglia (Ilschner and Brandt, 1996). It has
extracellular matrix protein thrombospondin 1 (Wang been suggested that the microglial cell variability in
et al., 1988; Constam et al., 1992; Mansfield and different regions of the brain depends on environmen-
Suchard, 1994; Bartholdi and Schwab, 1997; Salimi tal factors (Lawson et al., 1990; Sievers et al., 1995;
12 The Origin of Microglia and the Development of the Brain 177

Streit, 2001; Schwartz et al., 2006). Microglial cells astrocytes, can also promote microglial transformation
in brain regions that are exposed to plasma proteins to the ramified phenotype. This process can be
are less ramified than are microglia in regions where reversed in vitro by another extracellular matrix pro-
the blood-brain barrier is complete (Perry and Gordon, tein, laminin, suggesting that these two molecules
1991). In the immature CNS, environmental alterations must be involved in the regulation of the microglial
as well as specific properties of the CNS at each devel- differentiation in the CNS (Chamak and Mallat, 1991).
opmental stage and in each region might influence
microglial differentiation.
Astrocytes appear to play an important role in deter-
12.1.4 Microglial Development and
mining the mature microglial phenotype, since the
co-culturing of microglial cells with astrocytes con- Thyroid Hormones
sistently yields high numbers of ramified microglia
(Tanaka and Maeda, 1996; Navascus et al., 2000). Many questions regarding microglial recruitment,
Astrocytes are the main source of CSF-1 in the CNS, expansion, and differentiation still remain without
and they can produce this factor in culture constitu- answers. The endocrine regulation of the microglial
tively (Thry et al., 1990; Lee et al., 1993). In addition, development is not well understood. Thyroid hor-
ameboid microglia stimulated by LPS can also pro- mones (THs) are involved in the regulation of impor-
duce CSF-1 (Thry et al., 1992; Lee et al., 1993). The tant events that occur during the development of the
spontaneous growth of microglial cells in astrocyte CNS (Lima et al., 1997; Gomes et al., 2001; Trentin,
monolayers is blocked by the addition of anti-CSF-1 2006). The postnatal development of rat microglia is
antibodies to the cultures (Thry and Mallat, 1993). marked by a large increase in the number of microglial
Several studies have demonstrated that CSF-1 prevents cells and the growth of their ramified processes. Our
apoptosis of cultured ameboid microglia (Tomozawa group, in collaboration with Mallats team, demon-
et al., 1996), and stimulates their proliferation (Giulian strated that THs play a crucial role in the development
and Ingeman, 1988; Hao et al., 1990; Lee et al., 1993; of microglia (Lima et al., 2001; Mallat et al., 2002).
Imai and Kohsaka, 2002) as well as the growth of cell Microglial processes were markedly less abundant in
processes, transforming these macrophagic cells into hypothyroid rat pups than in age-matched normal ani-
cells with a morphology that is reminiscent of rami- mals, from post-natal day 4 up to the end of the third
fied microglia (Sawada et al., 1990). Through analyses postnatal week of life. A delay in the extension of pro-
using neutralizing antibodies for TGF, M. Mallats cesses and a decrease in the density of microglial cell
group demonstrated that neurons secrete TGF, which bodies in the developing cortex of normal and hypothy-
potentiates the microglial proliferation promoted by roid animals were responsible for these differences.
CSF-1 (Dobbertin et al., 1997). Conversely, neonatal rat hyperthyroidism accelerated
Op (osteopetrosis) mice have a recessive muta- the extension of microglial processes and increased
tion in the region of the gene that codes for CSF-1. the density of cortical microglial cell bodies above
Consequently, these animals do not produce biologi- physiological levels, during the first postnatal week
cally active CSF-1 (Raivich et al., 1994; Berezovskaya of life (Fig. 12.3). Consistent with the effects of THs
et al., 1995). Immunohistochemical studies have observed in vivo, thyroid hormone (T3) favored the
shown that the microglial density decreased 47% in survival of cultured purified microglial cells and the
the corpus callosum, 37% in the parietal cortex, and growth of their processes (Lima et al., 2001).
34% in the frontal cortex in op mice. Moreover, in Proliferation, migration, and differentiation of
the frontal cortical region, microglial cells are smaller microglia are closely regulated by intracellular
and have less-developed cell processes, in compari- signaling cascades. Protein kinase C (PKC) and
son to normal animals (Wegiel et al., 1998; Kondo phosphoinositol-3-kinase (PI3K) mediate the prolifer-
et al., 2007). These studies suggest that CSF-1 plays ation and differentiation of rat microglia from organ-
an important role in the maturation of the microglia in otypic cortex brain sections (Zassler et al., 2003).
vivo. Using a human microglial cell line (C13NJ), Martin
Treatment with fibronectin, an extracellular matrix et al. (2003) have shown in a wound-healing model
protein secreted by different cells and particularly by and a chemotaxis assay that microglial migration can
178 F.R.S. Lima et al.

Fig. 12.3 Microglial cells in the parietal cortex of hypothy- processes. a, b: Arrows point to microglial cells, which are
roid (a), euthyroid (b), and hyperthyroid (c) rats treated with shown at higher magnification in the insets. Microglial cell bod-
TH at postnatal day 4. Peroxidase staining with isolectin B4 ies and processes appear more numerous from a to c. Bar, 100
reveals blood vessels ( arrowheads ) and branched microglial m. Figure from Lima et al. (2001)

be induced by the neuropeptide neurotensin via a basal ganglia are regions with higher microglial den-
mechanism that is dependent on both the PI3K and sity, whereas the cortex, thalamus, and hypothalamus
mitogen-activated protein kinase (MAPK) pathways. have moderate microglial density, and the cerebellum
and brainstem have lower microglial density (Perry
and Gordon, 1988; Lawson et al., 1990). In the retina
12.1.5 Adult CNS: Ramified Microglia and certain cortical regions, studies have revealed that
microglial cells are arranged to form a cellular layer.
In the absence of injury, the ramified phenotype of In this layer, each cell covers with its ramifications a
microglia is typical in the adult vertebrate CNS. In certain territory, which other microglial cells do not
the mouse, microglial cells comprise 512% of the appear to invade (Hume et al., 1983; Schnitzer, 1989).
total of CNS cells. Although they are present in all In the adult brain, the role of ramified microglia,
mature brain regions, their density varies among areas also termed resting microglia, is still unclear. Although
of the nervous parenchyma. Ramified microglial cells these cells have certain macrophagic markers, such
are more numerous in the gray matter than in the as the Fc fragment receptors of immunoglobulins or
white matter. The hippocampus, olfactory bulb, and the CR3 receptors, they do not show properties of

Fig. 12.4 Schematic


representation of
(1) molecules involved with
the regulation of the
microglial development,
(2) phagocytosis and
microglial neurotoxic
factors related with
regressive processes during
brain development,
(3) neurotrophic factors
secreted by microglia that
can be participating
of the neural development
and, (4) microglia as
possible multipotential stem
cells. The majority of studies
related with microglia
and development was
performed in vitro and
deserves further investigation
12 The Origin of Microglia and the Development of the Brain 179

phagocytosis, and moreover appear to be in a qui- addition to proliferation, when they are confronted
escent state. However, ramified microglial cells are with this kind of insult, microglial cells may also pro-
highly sensitive to environmental conditions and can duce and release toxic reactive-oxygen species (ROS)
transform rapidly into activated cells, in the case of dis- into the neural environment, leading to oxidative injury
order in the homeostasis of nervous tissue. Therefore, (Garden and Moller, 2006). Cultured rodent microglial
they may have an important role in microglial survival, cells express inducible NO synthase (iNOS) (Kim and
constituting an endogenous macrophage reservoir of de Vellis, 2005) and NADPH oxidase (Sankarapandi
the CNS (Perry and Gordon, 1988). For this reason, et al., 1998), which are enzymes that generate NO and
Hanisch and Kettenmann (2007) suggested that rest- produce superoxide anions and hydrogen peroxide,
ing microglia should be renamed surveying microglia respectively (Garden and Moller, 2006). For exam-
in the mature brain. ple, it has been demonstrated that NO induces the
Microglial cells develop and react according death of rat cerebral cortical neurons through apopto-
to changes in their environment (Fig. 12.4). The sis, with the involvement of activated caspase-3 (Wang
CNS constitutes a particular protected environment, et al., 2003). Another example is that microglial cells
where macrophage recruitment tends to be maximally recognize mouse Purkinje cells expressing activated
limited, in comparison to those observed in other tis- caspase-3, engulf these cells, and produce high levels
sues, where inflammatory reactions are much more of superoxide ions, promoting neuronal death during
marked (Perry et al., 1993). A better understanding of brain development (Marin-Teva et al., 2004).
the interactions between microglia and their microen- During the development of the CNS, microglial
vironment in the CNS will benefit the comprehension cells participate in the formation of the complex net-
of the fundamental mechanisms for macrophagic regu- work of connections that are present in the adult brain.
lation in vivo, as well as neuropathologies that are still In fact, these cells are capable of phagocytosing tran-
incurable (Perry et al., 1993; Streit, 2001; Hanisch and sient or aberrant axons, apoptotic cells, and cellular
Kettenmann, 2007). debris (Ashwell, 1990; Ferrer et al., 1990; Innocenti
et al., 1983). For example, it has been demonstrated
in a rodent model that neurohypophysial microglial
cells engulf axonal terminations without commitment
12.2 Microglia and Regressive Processes of neuronal viability, suggesting that microglial cells
During Brain Development: play a role in the remodeling of terminal arborizations
of neurosecretory neurons and in processing or degrad-
Phagocytosis and Neurotoxic
ing hormones and peptides that they contain (Pow
Factors et al., 1989). Moreover, microglial cells engulf apop-
totic retinal cells during development. Through con-
As described here, microglial cells exhibit an ame- focal microscopy, Egensperger et al. (1996) observed
boid phenotype that is typical of macrophages in the the presence of cells in the retina whose nuclei were
developing brain (Vilhardt, 2005). These cells have a TUNEL- (marker for apoptotic nucleus) negative, but
wide spectrum of important biological responses dur- the cytoplasm was TUNEL-positive. This observation
ing brain development: migration, proliferation, nitric- strongly suggests that these cells are microglial cells,
oxide (NO) production and respiratory burst, phagocy- because their cytoplasmic labeling probably results
tosis, antigen presentation, and secretion of diffusible from the ingestion of the fragmenting DNA of a dying
factors (Garden and Moller, 2006). Some of these are retinal cell. The importance of the role of microglia
discussed here in more detail. in phagocytosing apoptotic cells is that this process
Neuronal or axonal degeneration induces microglial eliminates dead cells safely by preventing leakage
proliferation. Cytokines such as IL-1, IL-4, inter- of potentially cytotoxic or antigenic substances from
feron gamma (IFN-), CSF-1 (Kim and de Vellis, dying cells and by suppressing the proinflammatory
2005) and GM-CSF (Suh et al., 2005), in addi- effects of engulfing phagocytes (Savill et al., 2002),
tion to neurotrophic factors such as brain-derived leaving the CNS undamaged.
neurotrophic factor (BDNF) and NT-3 (Elkabes Among the signals for recognition of these cellular
et al., 1996), strongly promote this proliferation. In elements are the externalization of phosphatidylserine
180 F.R.S. Lima et al.

(PS) on the surface of apoptotic cells, and the interac- injury, microglial cells express and release IL-1/,
tions with the microglial vitronectin receptor and the IL-3, IL-6, IL-8, IL-12, IL-15, tumor necrosis factor
CD36 scavenger receptor (Mallat et al., 2005; Stolzing (TNF) , and interferons (IFNs) as anti-inflammatory
and Grune, 2004; Witting et al., 2000). More specifi- cytokines such as IL-4, IL-10, IL-13, and TGF
cally, it has been demonstrated that in a co-culture of (Hanisch, 2002; Kim and de Vellis, 2005). In this situa-
induced PC12 apoptotic cells with rat microglial cells, tion, microglia also express receptors for most of these
blockage of the CD36 scavenger receptor using spe- cytokines, resulting in autocrine feedback loops and
cific antibodies results in a significant reduction of the efficient interaction with astrocytes that are crucial for
degradation of apoptotic bodies by microglia (Stolzing the homeostasis of the CNS (Hanisch, 2002; Rozenfeld
and Grune, 2004). Moreover, in a similar model utiliz- et al., 2003, 2005; Garden and Moller, 2006).
ing a co-culture of induced cerebellar apoptotic cells The role of the well-known proinflammatory
with cerebral cortical microglial cells, the presence of cytokines IL-1, IL-6, and TNF is not always detri-
ligands that potentially interfere with the PS recep- mental to the CNS. For example, many hippocam-
tor and the vitronectin receptor diminishes the ratio of pal progenitor cells (HPC) die shortly after birth.
engulfment of apoptotic bodies by microglia (Witting Investigating what cell or molecule could cause this
et al., 2000). death, Cacci et al. (2005) demonstrated that the addi-
Still in the context of phagocytosis, it has recently tion of TNF to the medium of a cultured HPC
been demonstrated that microglial cells have the capac- line shortly after the cells stopped division promotes
ity to not only phagocyte apoptotic cell bodies, but apoptosis of these cells. Once the microglial cells syn-
also to promote cell death by engulfment, during thesized and released this factor, they cultured the
the normal CNS development (Mallat et al., 2005; HPC line with conditioned medium from a microglial
Marin-Teva et al., 2004). For example, as previ- cell line and found a similar effect, which suggests
ously mentioned, microglial cells induce the death that, during development, microglial TNF seems
of developing Purkinje cells by engulfing cerebel- to control the number of HPC by promoting the
lar neurons expressing activated caspase-3 (Marin- death of these cells (Cacci et al., 2005). Still during
Teva et al., 2004). Thus, in addition to phagocytosing development, it has been demonstrated in vitro that
cells undergoing apoptosis, pathological proteins, and microglial cells synthesize and secrete IL-1, which, in
microbes (Garden and Moller, 2006), microglial cells turn, stimulates proliferation of cerebral cortical astro-
are also actively involved in promoting the elimination cytes. Because the primitive astrocytes that are found
of developing neurons in the healthy brain (Vilhardt, in radial patterns provide routes along which neurons
2005). migrate from germinal zones to cortical regions of the
In response to specific extracellular signals, a sub- CNS (Bignami and Dahl, 1974; Schmechel and Rakic,
set of microglia cells may function as an antigen- 1979; Levitt and Rakic, 1980), and, at later periods
presenting cell (APC) to T cells. For this, they up- of brain development, astrocytes secrete extracellular
regulate the expression of dendritic cell markers such matrix constituents, such as laminin, which promote
as MHC-II and CD11c, co-stimulatory molecules such selective axonal growth (Letourneau, 1975; Lander
as B7.1 and B7.2, and the cathepsin protease required et al., 1982; Hopkins et al., 1985; Garcia-Abreu et al.,
for generation of antigen peptides (Aloisi et al., 2000). 1995; Lima et al., 2007), this proliferation induced by
There are four major classes of molecules that microglial IL-1 may help to direct pathways of axonal
are responsible for the communication signals from outgrowth or to stabilize newly formed circuits, play-
microglia to CNS cells and invading leukocytes, espe- ing an important role in the maturation of the CNS
cially during brain injury; however, many of them also (Giulian et al., 1988).
appear to be present in brain development, as dis- Chemokines are a class of chemoattractant
cussed below. The molecular classes are cytokines, molecules that act through G-protein-coupled recep-
chemokines, trophic factors, and small molecule medi- tors. Cultured microglia that are exposed to microbes,
ators of inflammation. cytokines, or pathological proteins express and secrete
Cytokines are small proteins that regulate the chemokines such as macrophage inflammatory protein
growth, survival, differentiation, and activities of cells. (MIP)-2, CCL2, CCL3, CCL4, CCL5, CXCL1,
Interleukins are included in this class. During brain CXCL8, CXCL10, and fractalkine (Hanisch, 2002).
12 The Origin of Microglia and the Development of the Brain 181

Microglia also express receptors for many of the As discussed here, microglial cells aim to promote
chemokines, suggesting that one important function of the correct ontogenesis and protection of the CNS.
chemokine secretion is to attract additional microglial Even events that might seem harmful, such as the
cells to the site of the insult (Garden and Moller, promotion of neuronal death or the release of proin-
2006). flammatory cytokines, are beneficial at some point,
Studies of the role of chemokines in the CNS have proving the fundamental role of microglial cells in the
been increasingly active in recent years, and appear to development of the healthy brain.
be very promising. For example, it has been demon-
strated that fractalkine secreted from adjacent neurons,
microglial cells, or astrocytes promotes the activation 12.3 Microglial Secreted Neurotrophic
of neuronal survival pathways in hippocampal neu-
Factors: Role in Neural
rons expressing the receptor for fractalkine (CX3 CR1)
(Meucci et al., 2000). Development
Trophic factors that promote neuronal survival are
frequently synthesized in and secreted from microglia. The majority of studies describe microglia in a patho-
Microglial cells, in vitro and in vivo, synthesize classi- logical context. There is still little information about
cal neurotrophins such as nerve growth factor (NGF), these cells and their effective participation in the devel-
BDNF, NT-3, basic fibroblast growth factor (b-FGF), opment of cells of the nervous system.
and GDNF (Elkabes et al., 1996; Honda et al., 1999; In the developing brain, microglial cells take part
Presta et al., 1995). in many morpho- and histogenetic events, helping to
The role of trophic factors may be indirectly bene- establish the complex network of connections that
ficial. For example, it has been demonstrated in vivo is present in the adult brain. Several studies suggest
that, during development, microglial NGF promotes that microglial cells participate in neuritogenesis and
the death of retinal neurons that specifically express the guidance of axons during development (Nagata et al.,
neurotrophin receptor p75 (Frade and Barde, 1998). It 1993; Chamak et al., 1994; Navascus et al., 2000;
has also been observed that both the volume occupied Streit, 2001; Rochefort et al., 2002). In fact, Stolz and
by the area of cell death and the density of its pyknotic cols. (1991) have shown that ameboid microglial cells
fragments undergo considerable variation during the guide the neurite growth of motoneurons in culture.
development of the retina, and this is thought to occur These cells are able to transform CNS regions into
in order to create space to accommodate the incoming permissive substrates for axonal growth (David et al.,
axons of the retinal ganglion cells, converging toward 1990; Rabchevsky and Strait, 1997; Rochefort et al.,
the optic disk, for the correct formation of the optic 2002; Batchelor et al., 2002). Moreover, microglial
nerve, showing that microglia has an essential role in cells increase myelination and modulate neuronal sur-
this well-regulated process (Cuadros and Rios, 1988; vival and synaptogenesis (Hamilton and Rome, 1994;
Martin-Partido et al., 1988). Shin et al., 2004; Bessis et al., 2007). In addition, they
Small-molecule lipid mediators of inflammation stimulate CNS vascularization, and also promote astro-
such as arachidonic acid, prostaglandins D2 , E2, cyte differentiation and astrocyte proliferation, both
and F2 , platelet-activating factor (PAF), tromboxane during development and after injury, favoring the for-
B2, and leukotriene B4 (Minghetti and Levi, 1998) mation of glial scars (Giulian et al., 1988; Navascus
also play a role in the communication signals from et al., 2000; Checchin et al., 2006; Nakanishi et al.,
microglia to CNS cells and invading leukocytes. 2007). Trophic factors secreted by microglia partic-
During brain injury, microglial cells secrete large ipate in these events (Mallat and Chamak, 1994;
amounts of prostaglandins and express several recep- Nakajima et al., 2007). In the developing CNS, IL-
tors for them (Minghetti and Levi, 1998). Besides 1, which is involved in astrocyte proliferation, is only
mediating inflammation, prostaglandins may function detected in the brain after the appearance of the ame-
as neuroprotector molecules (Rozenfeld et al., 2003). boid microglia (Giulian et al., 1988). In fact, in vitro
It has been demonstrated that prostaglandins amelio- microglial cells synthesize many interleukins, such
rate the glutamate-induced toxicity of cortical neurons as IL-1, IL-4, and IL-6, besides TNF, in response
(Akaike et al., 1994; Cazevieille et al., 1994). to several stimuli (Giulian et al., 1988; Frei et al.,
182 F.R.S. Lima et al.

1989; Htier et al., 1990, Park et al., 2005; Nakanishi NSC of the subventricular neuraxis have the abil-
et al., 2007). Additionally, microglial cells secrete in ity to self-renew and form multipotent neurospheres in
culture, spontaneously, FGF2 (Shimojo et al., 1991), vitro. This property to generate committed neuroblasts
GDNF (Matsushita et al., 2008), NGF after stimula- is progressively lost with continued culture. Walton
tion with LPS (Mallat et al., 1989), NT-3 (Elkabes and cols. (2006) showed that when NSC were co-
et al., 1996; Nakajima et al., 2001), and BDNF (Bessis cultured with microglial cells or with their conditioned
et al., 2007), as well as others. All these factors are medium, neurogenesis was greatly expanded, indicat-
involved in the regulation of growth of the CNS. In par- ing that microglia secrete factors that are essential for
ticular, the role of microglia in neuronal network for- neurogenesis. In fact, it was reported that microglial
mation has been specifically studied. Microglial cells cells can interact with tenascin-R and secrete NGF,
isolated from the developing rat brain spontaneously BDNF, and TGF, promoting proliferation of NSC and
secrete factors that stimulate neurite growth in differ- differentiation into neurons but not oligodendrocytes
ent regions of the CNS (Nagata et al., 1993; Chamak (Liao et al., 2008). Cacci and cols. (2008) also demon-
et al., 1994; Streit, 2001). One of these microglial strated in microglia that the induction of the proin-
factors, thrombospondin, was identified both in vitro flammatory cytokines IL-1, IL-1, IL-6, and TNF
and in vivo (Chamak et al., 1994, 1995). Recently, was sharply reduced after chronic lypopolysaccharide
Nakajima and cols. (2007) have demonstrated that neu- (LPS) stimulation, compared with acute LPS stimu-
rons stimulate microglia to secrete NGF and enhance lation (24 h). Conversely, the production of the anti-
the microglial secretion of other trophic factors such as inflammatory cytokine IL-10 and the immunomodula-
GDNF, FGF2, neurotrophin 4/5 (NT-4/5), TGF1, IL- tory PGE2 was still increased after chronic LPS expo-
3, and IL-10. These factors induced neuronal survival sure. Acutely activated microglia reduced survival of
and maturation in culture (Nakajima et al., 2007). neural precursor cells, prevented neuronal differentia-
tion, and strongly increased glial differentiation, likely
through the release of proinflammatory cytokines;
12.3.1 Microglia and Neural Progenitor whereas chronically activated microglia were permis-
Cells sive to neuronal differentiation and cell survival, and
still supported glial differentiation. These events indi-
Another important event that has been widely debated cate the complexity of the microglial activation process
in recent years, regarding the role of microglial cells during neural differentiation (Cacci et al., 2008).
during CNS development, is their involvement in regu- Evidence suggests that microglia have the capacity
lating neural induction and determining the fate of neu- to influence the differentiation, not only of embryonic
ral progenitors (Fig. 12.4). The differentiation and pro- neural precursors, but also of adult neural precursors
liferation of neural stem cells (NSC) are regulated by toward a neuronal phenotype (Aarum et al., 2003;
a combination of their intrinsic properties (e.g., tran- Butovsky et al., 2006; Ziv et al., 2006). Indeed, Battista
scription factors, epigenetic factors) and cell-extrinsic and cols. (2006), using adrenalectomized animals,
properties from the microenvironment around NSC have reported that activated microglia may secrete
(e.g., cytokines, growth factors, and cell-cell contact). TGF, promoting neurogenesis in adult NSC from the
Recently, there has been great interest in clarifying the dentate gyrus of the hippocampus.
mechanism of the influence of the microenvironment Microglia are also implicated in the regulation of
on NSC, especially cell-cell contact between NSC and gliogenesis, in both the developing and adult brain.
other nearby types of cells (Zhu et al., 2008). It has In vitro studies have shown that microglia induce
been reported that astrocytes promote the differenti- oligodendrocyte differentiation with involvement of
ation of NSC, suggesting the importance of cell-cell the transcription factor NF-kappa B (Nicholas et al.,
interactions between glial cells and NSC. However, the 2001). Additionally, Golli proteins are expressed in
contribution of microglia to the modulation of neuro- both the nervous and immune systems. LPS-treated
genesis is still unclear. Both pro- and anti-neurogenic microglia, but not LPS-treated astrocytes, secrete these
effects have been demonstrated (Walton et al., 2006; proteins and induce proliferation of oligodendrocyte
Cacci et al., 2008; Ideguchi et al., 2008). precursor cells (OPC) from the cortical subventricular
12 The Origin of Microglia and the Development of the Brain 183

zone (Filipovi and Zecevi, 2005). Lalive and cols. GFAP-positive cells. They also investigated the possi-
(2005) have also described TGF as a factor involved bility of microglia to differentiate into functional neu-
in oligodendrocyte differentiation. They suggested rons. Electrophysiological studies demonstrated that
that, in the mature CNS after injury, TGF may play microglia-derived MAP2-positive cells have a sim-
a pivotal role in remyelination by inducing microglia ilar pattern to cultured cortical neurons, suggesting
to release HGF (hepatocyte growth factor), which is that these cells possess properties of actual functional
both a chemotactic and differentiation factor for OPC neurons (Matsuda et al., 2008). Moreover, western
(Lalive et al., 2005). blot and immunocytochemical analyses revealed that
The promotion of astrocyte differentiation of neural activation of bone morphogenetic protein (BMP) sig-
progenitor cells by microglia has also been investi- naling through Smad and Id2 proteins is one of the
gated. Using NSC and microglial cultures obtained molecular pathways involved in the generation of
from rat embryonic day 16 of the subventricular zone microglia-derived MAP2-positive and GFAP-positive
and rat postnatal day 1 of the cortex, respectively, cells (Niidome et al., 2008). These reports are very
Nakanishi and cols (2007) showed that the microglial- recent, and need to be confirmed. We cannot discard
conditioned medium had no significant effect on the the possibility that microglia are really multipotential
proliferation of NSC. However, this medium increased stem cells, since they originate from hematopoietic
the percentage of cells that were positive for GFAP, precursors. However there is still little information
a marker of astrocytes, during differentiation. In addi- available on this issue, which deserves further inves-
tion, Nakanishi and cols (2007) reported that at least tigation.
IL-6 and LIF (leukemia inhibitory factor) released
by activated microglia promote astrocytic differentia-
tion of NSC via the activation of the JAK/STAT and 12.4 The Future
mitogen-activated protein kinase (MAPK) pathways.
Another study, using co-cultures of microglia-NSC, In this chapter, we have exploited the data on
demonstrated that ramified microglia promote astrogli- microglial development looking to the interactions
ogenesis of NSC by activating the STAT3 function and with parenchyma. The majority of data in the litera-
via the notch and sox9 signaling pathways (Zhu et al., ture describes microglial cells in a neuropathological
2008). context. Little is known about the development of these
Although microglial cells have a mesodermal ori- cells in the nervous parenchyma and their effective role
gin, recent studies have suggested a novel role for in neurogenesis. The interactions of microglia with
microglia as multipotential stem cells to give rise others glial cells and neurons contribute importantly
to neurons, astrocytes, or oligodendrocytes. Tanakas to the harmony of the brain. These interactions are
group demonstrated that rat microglial cells express based on exchange of secreted factors as ILs, growth
nestin, A2B5, and O4 antigens, which are markers factors or the noticed chemokines. However these
of OPC. When microglial cells were cultured in the exchanges are a very complex system of cell interac-
presence of 10% fetal bovine serum for 3 days, fol- tions, as it is suggested in the Fig. 12.4. Microglial
lowed by culture in the presence of 70% serum for 2 cells are considered a subpopulation of intraparenchy-
days, they became highly proliferative and dediffer- mal cells, which are distinguished from phagocytes
entiated. Then, dedifferentiated cells were transferred located in the meninges and in the coroid plexus
into serum-free medium, and a substantial number stroma, or from macrophages situated in the cerebral
of the cells rapidly turned into long-process-bearing capillary walls. More recently, attention is done to neu-
cells, which expressed microtubule-associated pro- ral progenitor cells and its implications on therapy.
tein 2, synapsin I, neurofilament proteins, glial fib- Microglial progenitors undergo homing to the nervous
rillary acidic protein (GFAP), or galactocerebroside system, could they be usefull tools for therapeutic pur-
(Yokovama et al., 2004, 2006). In accordance with poses? Upon genetic manipulation, microglia progen-
these data, Sugimotos group has recently shown that itors could locally deliver specific gene products for
microglia are multipotential stem cells that give rise to the treatment of several disorders, such as metachro-
microtubule-associated protein (MAP) 2 -positive and matic leukodystrophy, gliomas and stroke, as well as
184 F.R.S. Lima et al.

comment by Chan et al., 2007. Nevertheless, genetic or growth factor beta increases neurogenesis in the adult dentate
cell therapy will only be possible when tissue of origin gyrus. Eur J Neurosci 23:8393.
Berezovskaya O, Maysinger D, Fedoroff S (1995) The
or cell lineage of resident microglia can be precisely
hematopoetic cytokine colony stimulating factor in the CNS:
identified. Congenital absence of CSF-1 in mice results in abnormal
A better understanding of the interactions between microglial response and increased neuron vulnerability to
microglia and their microenvironment in the CNS injury. Int J Devl Neurosci 13:285299.
Bessis A, Bchade C, Bernard D, Roumier A (2007) Microglial
will benefit the comprehension of the fundamental control of neuronal death and synaptic properties. Glia
mechanisms for macrophagic regulation in vivo, devel- 55:233238.
opment, as well as neuropathologies that are still Bignami A, Dahl D (1974) Astrocyte-specific protein and radial
incurable. glia in the cerebral cortex of newborn rat. Nature 252:
5556.
Billiards SS, Haynes RL, Folkerth RD, Trachtenberg FL, Liu
LG, Volpe JJ, Kinney HC (2006) Development of microglia
in the cerebral white matter of the human fetus and infant.
References J Comp Neurol 497:199208.
Boya J, Calvo JL, Carbonell AL, Borregon A (1991) A lectin
histochemistry study on the development of rat microglial
Aarum J, Sandberg K, Haeberlein SL, Persson MA (2003) cells. J Anat 175: 229236.
Migration and differentiation of neural precursor cells can Butovsky O, Ziv Y, Schwartz A, Landa G, Talpalar AE, Pluchino
be directed by microglia. Proc Natl Acad Sci USA 100: S, Martino G, Schwartz M (2006) Microglia activated by
1598315988. IL-4 or IFN-gamma differentially induce neurogenesis and
Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM (2007) oligodendrogenesis from adult stem/progenitor cells. Mol
Local self-renewal can sustain CNS microglia mainte- Cell Neurosci 31:149160.
nance and function throughout adult life. Nat Neurosci 10: Cacci E, Ajmone-Cat MA, Anelli T, Biagioni S, Minghetti L
15381543. (2008) In vitro neuronal and glial differentiation from embry-
Akaike A, Kaneko S, Tamura Y, Nakata N, Shiomi H, Ushikubi onic or adult neural precursor cells are differently affected by
F, Narumiya S (1994) Prostaglandin E2 protects cultured chronic or acute activation of microglia. Glia 56:412425.
cortical neurons against N-methyl-D-aspartate receptor- Cacci E, Claasen JH, Kokaia Z (2005) Microglia-derived tumor
mediated glutamate cytotoxicity. Brain Res 663:237243. necrosis factor-alpha exaggerates death of newborn hip-
Akiyama H, Tooyama I, Kondo H, Ikeda K, Kimura H, McGreer pocampal progenitor cells in vitro. J Neurosci Res 80:
EG, McGreer PL (1994) Early response of brain resident 789797.
microglia to kainic acid-induced hippocampal lesions. Brain Calvo CF, Dobbertin A, Gelman M, Glowinski J, Mallat M
Res 635:257268. (1998) Identification of CSF-1 as a ameboid microglia migra-
Aloisi F, Serafini B, Adorini L (2000) Glia-T cell dialogue. tory activity produced by astrocytes. Glia 24:180186.
J Neuroimmunol 107:111117. Cazevieille C, Muller A, Meynier F, Dutrait N, Bonne C (1994)
Asheuer M, Pflumio F, Benhamida S, Dubart-Kupperschmitt Protection by prostaglandins from glutamate toxicity in cor-
A, Fouquet F, Imai Y, Aubourg P, Cartier N (2004) tical neurons. Neurochem Int 24:395398.
Human CD34+ cells differentiate into microglia and express Chamak B, Dobbertin A, Mallat M (1995) Immunochemical
recombinat therapeutic protein. Proc Natl Acad Sci USA detection of thrombospondin in microglia in the developing
101:35573562. rat brain. Neuroscience 69:177187.
Ashwell K (1990) Microglia and cell death in the developing Chamak B, Mallat M (1991) Fibronectin and laminin regulate
mouse cerebellum. Brain Res Dev Brain Res 55:219230. the in vitro differentiation of microglial cells. Neuroscience
Ashwell K (1991) The distribution of microglia and cell death in 45:513527.
the fetal rat forebrain. Dev Brain Res 58:112. Chamak B, Morandi V, Mallat M (1994) Brain macrophages
Banati RB, Rothe G, Valet G, Kreutzberg GW (1991) stimulate neurite growth and regeneration by secreting
Respiratory burst in the ameboid microglia: a flow cytometric thrombospondin. J Neuros Res 38:221233.
study on cultured rat microglia. Neuropathol Appl Neurobiol Chan WY, Kohsaka S, Razaie P (2007) The origin and cell
17:223230. lineage of microglia new concepts. Brain Res Rev 53:
Bartholdi D, Schwab ME (1997) Expression of pro- 344354.
inflammatory cytokine and chemokine mRNA upon Checchin D, Sennlaub F, Levavasseur E, Leduc M, Chemtob
experimental spinal cord injury in mouse: an in situ S (2006) Potential role of microglia in retinal blood vessel
hybridization study. Eur J Neurosci 9:14221438. formation. Invest Ophthalmol Vis Sci 47:35953602.
Batchelor PE, Porritt MJ, Martinello P, Parish CL, Liberatore Chen L, Yang P, Kijlstra A (2002) Distribution, markers,
GT, Donnan GA, Howells DW (2002) Macrophages and and functions of retinal microglia. Ocul Immunol Inflamm
microglia produce local trophic gradients that stimulate 10:2739.
axonal sprouting toward but not beyond the wound edge. Mol Constam DB, Philipp J, Malipiero UV, Ten-Dijke P, Schachner
Cell Neurosci 21:436453. M, Fontana A (1992) Differential expression of TGF1,
Battista D, Ferrari CC, Gage FH, Pitossi FJ (2006) Neurogenic -2, -3 by glioblastoma cells, astrocytes and microglia.
niche modulation by activated microglia: transforming J Immunol 148:14041410.
12 The Origin of Microglia and the Development of the Brain 185

Cramer EB (1992) Cell biology of phagocyte migration from uptake by microglia and macroglia (Muller cells). Brain Res
the bone marrow, out of the bloodstream, and across organ Dev Brain Res 97:18.
epithelia. In: Inflammation: Basic Principles and Clinical Elkabes ST, Di Cicco-Bloom EM, Black IB (1996) Brain
Correlates, 2nd ed., (Gallin JI, Goldstein IM and Snyderman microglia/macrophages express neurotrophins that selec-
R, eds.), pp. 341351. Raven Press, New York. tively regulate microglial proliferation and function.
Cuadros MA, Martin C, Coltey P, Almendros A, Navascues J Neurosci 16:25082521.
J (1993) First appearance, distribution and origin pf Ferrer I, Bernet E, Soriano E, del Rio T, Fonseca M (1990)
macrophages in the early development of the avian central Naturally occurring cell death in the cerebral cortex of the
nervous system. J Comp Neurol 330:113129. rat and removal of dead cells by transitory phagocytes.
Cuadros MA, Moujahid A, Quesada A, Navascus J (1994) Neuroscience 39:451458.
Development of microglia in the quail optic tectum. J Comp Filipovic R, Zecevic N (2005) Interaction between microglia and
Neurol 348:207224. oligodendrocyte cell progenitors involves Golli proteins. Ann
Cuadros MA, Navascus J (1998) The origin and differentita- N Y Acad Sci 1048:166174.
tion of microglial cells during development. Prog Neurobiol Flanary BE, Streit WJ (2006) Alpha-tocopherol (vitamin E)
56:173189. induces rapid, nonsustained proliferation in cultured rat
Cuadros MA, Navascus J (2001) Early origin and coloniza- microglia. Glia 53:669674.
tion of the developing central nervous system by microglial Frade JM, Barde YA (1998) Microglia-derived nerve growth
precursors. Prog Brain Res 132:5159. factor causes cell death in the developing retina. Neuron
Cuadros MA, Rios A (1988) Spatial and temporal correla- 20:3541.
tion between early nerve fiber growth and neuroepithelial Frei K, Malipiero UV, Leist TP, Zinkernagel RM, Schwab ME,
cell death in the chick embryo retina. Anat Embryol 178: Fontana A (1989) On the cellular source and function of
543551. interleukin 6 produced in the central nervous system in viral
Cuadros MA, Rodriguez-Ruiz J, Calvente R, Almendros A, diseases. Eur J Immunol 19:689694.
Marin-Teva JL, Navascus J (1997) Microglia development Garcia-Abreu J, Cavalcante LA, Moura Neto V (1995)
in the quail cerebellum. J Comp Neurol 389:390401. Differential patterns of laminin expression in lateral and
Cucchiarini M, Ren XL, Perides G, Terwilliger EF (2003) medial midbrain glia. Neuroreport 6:761764.
Selective gene expression in brain microglia mediated via Garden GA, Moller T (2006) Microglia biology in health and
adeno-associated virus type2 and type 5 vectors. Gene Ther disease. J Neuroimmune Pharmacol 1:127137.
10:657667. Gehrmann J, Banati RB (1995) Microglial turnover in the injured
Dalmau I, Finsen B, Tonder N, Zimmer J, Gonzalez CNS: activated microglia undergo delayed DNA fragmen-
B, Castellano B (1997) Development of microglia in tation following peripheral nerve injury. J Neuropathol Exp
the prenatal rat hippocampus. J Comp Neurol 377: Morphol 54:680688.
7084. Giulian D, Baker TJ (1986) Characterization of ameboid
Dalmau I, Vela JM, Gonzlez B, Finsen B, Castellano B (2003) microglia isolated from developing mammalian brain.
Dynamics of microglia in the developing rat brain. J Comp J Neurosci 6:21632178.
Neurol 458:144157. Giulian D, Haverkamp LJ, Li J, Karshin WL, Yu J, Tom D, Li X,
David S, Bouchard C, Tsatas O, Giftochristos N (1990) Kirkpatrick JB (1995) Senile plaques stimulate microglia to
Macrophages can modify the nonpermissive nature of the release a neurotoxin found in Alzheimer brain. Neurochem
adult central nervous system. Neuron 5:463469. Int 27:119137.
Davis EJ, Foster TD, Thomas WE (1994) Cellular forms and Giulian D, Ingeman GE (1988a) Colony-stimulating fac-
functions of brain microglia: Brain Res Bull 34:7378. tors as promoters of ameboid microglia. J Neurosci 8:
Del Rio Hortega P (1932) Microglia. Section X. In: Cytology 47074717.
and Cellular Pathology of Nervous System (Penfield W ed.), Giulian D, Young DG, Woodward J, Brown DC, Lachman LB
pp. 481534. Paul B. Hoeber, New York. (1988b) Interleukin-1 is an astroglial growth factor in the
Diaz-Araya CM, Provis JM, Penfold PL, Billson FA (1995) developing brain. J Neurosci 8:709714.
Development of microglia topography in human retina Gomes FA, Lima FRS, Trentin AG and Moura Neto V (2001)
J Comp Neurol 363:5368. Thyroid hormone role on nervous system morphogenesis.
Djukic M, Mildner A, Schmidt H, Czesnik D, Bruck W, Priller Progr Brain Res (Rev), 132:4150.
J, Nau R, Prinz M (2006) Circulating monocytes engraft Graeber MB, Tetzlaff W, Streit WJ, Kreutzberg GW (1988)
in the brain, differentiate into microglia and contribute to Microglial cells but not astrocytes undergo mitosis following
the pathology following meningitis in mice. Brain 129: facial nerve axotomy. Neurosci Lett 85:317321.
23942403. Hamilton SP, Rome LH (1994) Stimulation of in vitro myelin
Dobbertin A, Schmid P, Gelman M, Glowinski J, Mallat synthesis by microglia. Glia 11:326335.
M (1997) Neurons promote macrophage proliferation by Hanisch UK (2002) Microglia as a source and target of
producing transforming growth factor-beta2. J Neurosci cytokines. Glia 40:140155.
17:53055315. Hanisch UK, Kettenmann H (2007) Microglia: active sensor and
Dobrenis K (1998) Microglia in cell culture and in transplan- versatile effector cells in the normal and pathologic brain.
tation therapy for central nervous system disease. Methods Nat Neurosci 10:13871394.
16:320344. Hao C, Guilbert LJ, Fedoroff S (1990) Production of colony-
Egensperger R, Maslim J, Bisti S, Hollander H, Stone J (1996) stimulating factor-1 (CSF-1) by mouse astroglia in vitro.
Fate of DNA from retinal cells dying during development: J Neurosci Res 27:314323.
186 F.R.S. Lima et al.

Hess DC, Abe T, Hill WD, Studdard AM, Carothers J, Masuya Kreutzberg G (1996) Microglia: a sensor for pathological events
M, Fleming PA, Drake CJ, Ogawa M (2004) Hematopoietic in the CNS. Trends Neurosci 19:312319.
origin of microglial and perivascular cells in brain. Exp Lalive PH, Paglinawan R, Biollaz G, Kappos EA, Leone DP,
Neurol 186:134144. Malipiero U, Relvas JB, Moransard M, Suter T, Fontana A
Honda S, Nakajima K, Nakamura Y, Imai Y, Kohsaka S (1999) (2005) TGF-beta-treated microglia induce oligodendrocyte
Rat primary cultured microglia express glial cell line-derived precursor cell chemotaxis through the HGF-c-Met pathway.
neurotrophic factor receptors. Neurosci Lett 275:203206. Eur J Immunol 35:727737.
Hopkins JM, Ford-Holevinski TS, McCoy JP, Agranoff BW Lander AD, Fujii DK, Gospodarowicz D, Reichardt LF (1982)
(1985) Laminin and optic nerve regeneration in the goldfish. Characterization of a factor that promotes neurite outgrowth:
J Neurosci 5:30303038. evidence linking activity to a heparan sulfate proteoglycan.
Hume DA, Perry H, Gordon S (1983) Immunohistochemical J Cell Biol 94:574585.
localization of a macrophage-specific antigen in developing Lawson LJ, Perry VH, Dri P, Gordon S (1990) Heterogeneity in
mouse retina: phagocytosis of dying neurons and differen- the distribution and morphology of microglia in the normal
tiation of microglial cells to form a regular array in the adult mouse brain. Neurosci 39:151170.
plexiform layers. J Cell Biol 97:253257. Lawson LJ, Perry VH, Gordon S (1992) Turnover of resident
Hurley SD, Streit WJ (1996) Microglia and the mononuclear microglia in the normal adult mouse brain. Neurosci 48:
phagocytic system. In: Topical Issues in Microglia Research 405415.
(Ling EA, Tan CK and Tan CBC, eds.), pp. 119. Singapore Lee SC, Liu W, Roth P, Dickson DW, Berman JW, Brosnan
Neuroscience Association, Singapore. CF (1993) Macrophage colony stimulating factor in fetal
Htier E, Ayala J, Bousseau A, Denfle P, Prochiantz A (1990) astrocytes and microglia: differential regulation by citokines
Amoeboid microglial cells and not astrocytes synthesize and lipopolysaccharide and modulation of class II MHC on
TNF in Swiss mouse brain cell cultures. Eur J Neurosci microglia. J Immunol 150:594604.
2:762768. Leong SK, Ling EA (1992) Amoeboid and ramified microglia:
Ideguchi M, Shinoyama M, Gomi M, Hayashi H, Hashimoto their interrelationship and response to brain injury. Glia
N, Takahashi J (2008) Immune or inflammatory response by 7:3947.
the host brain suppresses neuronal differentiation of trans- Letourneau PC (1975) Possible roles for cell-to-substratum
planted ES cell-derived neural precursor cells. J Neurosci adhesion in neuronal morphogenesis. Dev Biol 44:7791.
Res 86:19361943. Levitt P, Rakic P (1980) Immunoperoxidase localization of glial
Ilschner S, Brandt R (1996) The transition of microglia to a ram- fibrillary acidic protein in radial glial cells and astrocytes
ified phenotype is associated with the formation of stable of the developing rhesus monkey brain. J Comp Neurol
acetylated and detyrosinated microtubules. Glia 18:129140. 193:815840.
Imai Y, Kohsaka S (2002) Intracellular signaling in M- Liao H, Huang W, Niu R, Sun L, Zhang L (2008) Cross-talk
CSF-induced microglia activation: role of Iba1. Glia 40: between the epidermal growth factor-like repeats/fibronectin
164174. 6-8 repeats domains of Tenascin-R and microglia modulates
Imamoto K, Leblond CP (1978) Radioautographic investigation neural stem/progenitor cell proliferation and differentiation.
of gliogenesis in the corpus callosum of young rats. J Comp J Neurosci Res 86:2734.
Neurol 180:139164. Lima FR, Arantes CP, Muras AG, Nomizo R, Brentani RR,
Innocenti GM, Clarke S, Koppel H (1983) Transitory Martins VR (2007) Cellular prion protein expression in
macrophages in the white matter of the developing visual astrocytes modulates neuronal survival and differentiation.
cortex. II. Development and relations with axonal pathways. J Neurochem 103:21642176.
Brain Res 313:5566. Lima FRS, Gervais A, Colin C, Izambar M, Moura Neto V,
Jones LL, Banati RB, Graeber MB, Bonfanti L, Raivich G, Mallat M (2001) Regulation of microglial development: a
Kreutzberg GW (1997) Population control of microglia: does novel role for thyroid hormone. J Neurosci 21:20282038.
apoptosis play a role? J Neurocytol 26:755770. Lima FRS, Trentin AG, Rosenthal D, Chagas C, Moura Neto
Jordan FL, Thomas WE (1988) Brain macrophage: question of V (1997) Thyroid hormone induces protein secretion and
origin and interrelationship. Brain Res Rev 13:164178. morphological changes in astroglial cells with an increase
Kahn MA, Kumar S, Liebl D, Chang R, Parada LF, De Vellis in expression of glial fibrillary acidic protein. J Endocrinol
J (1999) Mice lacking NT-3, and its receptor TrkC, exhibit 154:167175.
profound deficiencies in CNS glial cells. Glia 26:153165. Ling EA (1981) The origin and nature of microglia. In: Advances
Kaur C, Hao AJ, Wu CH, Ling EA (2001) Origin of microglia. in Cell Neurobiology. Vol. 2 (Fedoroff S and Hertz L, eds.),
Microsc Res Tech 54:29. pp. 3382. Academic Press. London.
Kaur C, Ling EA (1991) A study of the transformation of Ling EA, Kaur C, Wong WC (1991) Expression of major his-
amoeboid microglia cells into microglia labeled with the tocompatibility complex and leukocyte common antigens in
isolectin Griffonia simplicifolia in postnatal rats. Acta Anat amoeboid microglia in postnatal rats. J Anat 177:117126.
142:118125. Ling EA, Penney D, Leblond CP (1980) Use of carbon labeling
Kim SU, de Vellis J (2005) Microglia in health and disease. to demonstrate the role of blood monocytes as precursors of
J Neurosci Res 81:302313. the ameboid cells present in the corpus callosum of postnatal
Kondo Y, Lemere CA, Seabrook TJ (2007) Osteopetrotic (op/op) rats. J Comp Neur 193:631657.
mice have reduced microglia, no Abeta deposition, and Ling EA, Wong WC (1993) The origin and nature of rami-
no changes in dopaminergic neurons. J Neuroinflammation fied and amoeboid microglia: a historical review and current
4:3141. concepts. Glia 7:918.
12 The Origin of Microglia and the Development of the Brain 187

Mallat M, Calvo CF, Dobbertin A (1997) Migration and pro- Murabe Y, Sano Y (1982) Microglial cells in the cerebral cortex
liferation of mononuclear phagocytes in the central nervous of the rat, with special reference to their possible involvement
system. Adv Exp Med Biol 429:99108. in synaptic function. Cell Tissue Res 223:493506.
Mallat M, Chamak B (1994) Brain macrophage: neurotoxic or Nagano T, Kimura SH, Takemura M (2008) Prostaglandin E2
neurotrophic effector cells? J Leukoc Biol 56:416422. reduces extracellular ATP-induced migration in cultured rat
Mallat M, Houlagatte R, Brachet P, Prochiantz A (1989) microglia. Brain Res 1221:15.
Lipopolysaccharide-stimulated rat brain macrophages Nagata K, Nakajima K, Kohsaka S (1993) Plasminogen pro-
release NGF in vitro. Dev Biol 133:309311. motes the development of rat mesencephalic dopaminergic
Mallat M, Lima FRS, Gervais A, Colin C, Moura Neto V (2002) neurons in vitro. Dev Brain Res 75:3137.
New insights in the role of thyroid hormone in the CNS: the Nakajima K, Honda S, Tohyama Y, Imai Y, Kohsaka S, Kurihara
microglial track. Mol Psychiatry 7:78. T (2001) Neurotrophin secretion from cultured microglia.
Mallat M, Marin-Teva JL, Cheret C (2005) Phagocytosis in the J Neurosci Res 65:322331.
developing CNS: more than clearing the corpses. Curr Opin Nakajima K, Tohyama Y, Maeda S, Kohsaka S, Kurihara T
Neurobiol 15:101107. (2007) Neuronal regulation by which microglia enhance
Mansfield PJ, Suchard SJ (1994) Thrombospondin promotes the production of neurotrophic factors for GABAergic, cat-
chemotaxis and haptotaxis of peripheral blood monocytes. echolaminergic, and cholinergic neurons. Neurochem Int
J Immunol 153:42194229. 50:807820.
Marin-Teva JL, Dusart I, Colin C, Gervais A, van Rooijen N, Nakanishi M, Niidome T, Matsuda S, Akaike A, Kihara T,
Mallat M (2004) Microglia promote the death of developing Sugimoto H (2007) Microglia-derived interleukin-6 and
Purkinje cells. Neuron 41:535547. leukaemia inhibitory factor promote astrocytic differentia-
Martin S, Vincent JP, Mazella J (2003) Involvement of the neu- tion of neural stem/progenitor cells. Eur J Neurosci 25:
rotensin receptor-3 in the neurotensin-induced migration of 649658.
human microglia. J Neurosci 23:11981205. Navascus J, Calvente R, Marn-Teva JL, Cuadros MA (2000)
Martin-Partido G, Rodriguez-Gallardo L, Alvarez IS, Navascues Entry, dispersion and differentiation of microglia in the
J (1988) Cell death in the ventral region of the neural retina developing central nervous system. An Acad Bras C 72:
during the early development of the chick embryo eye. Anat 91102.
Rec 222:272281. Navascus J, Cuadros MA, Almendros A (1996) Development
Marn-Teva JL, Almendros A, Calvente R, Cuadros MA, of microglia: evidences from studies in the avian central
Navascus J (1999) Proliferation of actively migrating ame- nervous system. In: Topical Issues in Microglia Research
boid microglia in the developing quail retina. Anat Embryol (Ling EA, Tan CK and Tan CBC, eds.), pp. 4364. Singapore
200:289300. Neuroscience Association, Singapore.
Matsuda S, Niidome T, Nonaka H, Goto Y, Fujimura K, Kato Navascus J, Moujahid A, Almendros A, Marin-Teva JL,
M, Nakanishi M, Akaike A, Kihara T, Sugimoto H (2008) Cuadros J (1995) Origin of microglia in the quail retina:
Microtubule-associated protein 2-positive cells derived from central-to-peripheral and vitreal-to-scleral migration of
microglia possess properties of functional neurons. Biochem microglial precursors during development. J Comp Neurol
Biophys Res Commun 368:971976. 354:209228.
Matsushita Y, Nakajima K, Tohyama Y, Kurihara T, Kohsaka Nicholas RS, Wing MG, Compston A (2001) Nonactivated
S (2008) Activation of microglia by endotoxin suppresses microglia promote oligodendrocyte precursor survival and
the secretion of glial cell line-derived neurotrophic fac- maturation through the transcription factor NF-kappa B. Eur
tor (GDNF) through the action of protein kinase C J Neurosci13:959967.
alpha (PKCalpha) and mitogen-activated protein kinases Niidome T, Matsuda S, Nonaka H, Akaike A, Kihara T,
(MAPKS). J Neurosci Res 86:19591971. Sugimoto H (2008) A molecular pathway involved in
Meucci O, Fatatis A, Simen AA, Miller RJ (2000) Expression of the generation of microtubule-associated protein 2-positive
CX3CR1 chemokine receptors on neurons and their role in cells from microglia. Biochem Biophys Res Commun 370:
neuronal survival. Proc Natl Acad Sci USA 97:80758080. 184188.
Mildner A, Schmid H, Nitsche M, Merkler D, Hanisch UK, Park KW, Lee DY, Joe EH, Kim SU, Jin BK (2005)
Mack M, Heikenwalder M, Bruck W, Priller J, Prinz Neuroprotective role of microglia expressing interleukin-4.
M (2007) Microglia in the adult brain arise from Ly- J Neurosci Res 81:397402.
6Chi CCR2+ monocytes only under defined host conditions. Perry VH, Anderson PB, Gordon S (1993) Macrophages and
Nature Neurosci 12:15441553. inflammation in the central nervous system. Trends Neurosci
Milligan CE, Cunningham J, Levitt P (1991) Differential 16:268273.
immunochemical markers reveal the normal distribution of Perry VH, Gordon S (1988) Macrophages and microglia in the
brain macrophages and microglia in the developing rat brain. nervous system. Trends Neurosci 11:273277.
J Comp Neurol 314:125135. Perry VH, Gordon S (1991) Macrophages and the nervous
Minghetti L, Levi G (1998) Microglia as effector cells in brain system. Int Rev Cytol 125:203244.
damage and repair: focus on prostanoids and nitric oxide. Perry VH, Hume DA, Gordon S (1985) Immunohistochemical
Prog Neurobiol 54:99125. localization of macrophages and microglia in the adult and
Monier A, Adle-Biassette H, Delezoide AL, Evrard P, Gressens developing mouse brain. Neurosci 15:313326.
P, Verney C (2007) Entry and distribution of microglial cells Perry VH, Lawson LJ, Reid DM (1994) Biology of the mononu-
in human embryonic and fetal cerebral cortex. J Neuropathol clear phagocyte system of the central nervous system and
Exp Neurol 66:372382. HIV infection. J Leukoc Biol 56:399406.
188 F.R.S. Lima et al.

Pow DV, Perry VH, Morris JF, Gordon S (1989) Microglia in Schnitzer J (1989) Enzyme-histochemical demonstration of
the neurohypophysis associate with and endocytose terminal microglial cells in the adult and postnatal rabbit retina.
portions of neurosecretory neurons. Neurosci 33:567578. J Comp Neurol 282:249263.
Presta M, Urbinati C, Dellera P, Lauro GM, Sogos V, Balaci L, Schwartz M, Butovsky O, Brck W, Hanisch UK (2006)
Ennas MG, Gremo F (1995) Expression of basic fibroblast Microglial phenotype: is the commitment reversible? Trends
growth factor and its receptors in human fetal microglia cells. Neurosci 29:6874.
Int J Dev Neurosci 13:2939. Shimojo M, Nakajima K, Takei N, Hamanoue M, Kohsaka
Provis JM, Diaz CM, Penfold PL (1996) Microglia in human S (1991) Production of basic fibroblast growth factor in
retina: a heterogenous population with distinct ontogenies. cultured rat brain microglia. Neurosci Lett 123:229231.
Persp Dev Neurobiol 3:213222. Shin WH, Lee DY, Park KW, Kim SU, Yang MS, Joe EH,
Rabchevsky AV, Streit WJ (1997) Grafting of cultured microglial Jin BK (2004) Microglia expressing interleukin-13 undergo
cells into the lesioned spinal cord of adult rats enhances cell death and contribute to neuronal survival in vivo. Glia
neurite outgrowth. J Neurosci Res 47:3448. 46:142152.
Raivich G, Gehrmann J, Kreutzberg GW (1994) Inhibition of Sievers J, Bamberger C, Debus OM, Lucius R (1995)
posttraumatic microglial proliferation in a genetic model Regeneration in the optic nerve of adult rats: influences
of macrophage colony-stimulating factor deficiency in the of cultured astrocytes and optic nerve grafts of different
mouse. Eur J Neurosci 6:16151618. ontogenetic stages. J Neurocytol 10:783793.
Rezaie P, Dean A, Male D, Ulfig N (2005) Microglia in the cere- Simard AR, Rivest S (2004) Bone marrow stem cells have
bral wall of the human telencephalon at second trimester. the ability to populate the entire central nervous system
Cereb Cortex 15:938949. into fully differentiated parenchymal microglia. FASEB
Rezaie P, Male D (2002) Mesoglia and microglia a historical J 18:9981000.
review of the concept of mononuclear phagocytes within the Sorokin SJ, Hoyt RF, Blunt DG, McNelly NA (1992)
central nervous system. J Hist Neurosci 11:325374. Macrophage development: II. Early ontogeny of macrophage
Rochefort N, Quenechdu N, Watroba L, Mallat M, Giaume C, populations in brain, liver, and lungs of rat embryos as
Milleret C (2002) Microglia and astrocytes may participate revealed by a lectin marker. Anat Record 232:527550.
in the shaping of visual callosal projections during postnatal Stolz B, Erulkar S, Kuffler DP (1991) Macrophages direct pro-
development. J Physiol Paris 96:183192. cess elongation from adult frog motoneurons in culture. Proc
Rozenfeld C, Martinez R, Figueiredo RT, Bozza MT, Lima R Soc Lond B 244:227231.
FR, Pires AL, Silva PM, Bonomo A, Lannes-Vieira J, De Stolzing A, Grune T (2004) Neuronal apoptotic bodies: phago-
Souza W, Moura-Neto V (2003) Soluble factors released by cytosis and degradation by primary microglial cells. FASEB
Toxoplasma gondii-infected astrocytes down-modulate nitric J 18:743745.
oxide production by gamma interferon-activated microglia Streit WJ (2001) Microglia and macrophages in the developing
and prevent neuronal degeneration. Infect Immun 71: CNS. Neurotoxicol 22:619624.
20472057. Streit WJ, Kreutzberg GW (1988) Response of endogenous glial
Rozenfeld C, Martinez R, Seabra S, Santanna C, Gonalves JG, cells to motor neuron degeneration induced by toxic ricin.
Bozza M, Moura-Neto V, De Souza W (2005) Toxoplasma J Comp Neurol 268:248263.
gondii prevents neuron degeneration by interferon-gamma- Suh HS, Kim MO, Lee SC (2005) Inhibition of granulocyte-
activated microglia in a mechanism involving inhibition of macrophage colony-stimulating factor signaling and
inducible nitric oxide synthase and transforming growth microglial proliferation by anti-CD45RO: role of Hck
factor-beta1 production by infected microglia. Am J Pathol tyrosine kinase and phosphatidylinositol 3-kinase/Akt.
167:10211031. J Immunol 174:27122719.
Salimi K, Moser KV, Marksteiner J, Reindl M, Humpel C (2003) Suzumura A, Sawada M, Yamamoto H, Marunouchi T (1990)
GDNF and TGF-beta1 promote cell survival in serum-free Effects of colony stimulating factors on isolated microglia in
cultures of primary rat microglia. Cell Tissue Res 312: vitro. J Neuroimmunol 30:237244.
135139. Tanaka J, Maeda N (1996) Microglia ramification requires
Sankarapandi S, Zweier JL, Mukherjee G, Quinn MT, Huso nondiffusible factors derived from astrocytes. Exp Neurol
DL (1998) Measurement and characterization of superox- 137:367375.
ide generation in microglial cells: evidence for an NADPH Thry C, Htier E, Evrard C, Mallat M (1990) Expression of
oxidase-dependent pathway. Arch Biochem Biophys 353: macrophage colony-stimulating factor gene in the mouse
312321. brain during development. J Neurosci Res 26:129133.
Savill J, Dransfield I, Gregory C, Haslett C (2002) A blast Thry C, Mallat M (1993). Influences of interleukin 1 and
from the past: clearance of apoptotic cells regulates immune tumor necrosis factor-a on the growth of microglial cells in
responses. Nat Rev Immunol 2:965975. primary cultures of mouse cerebral cortex: involvement of
Sawada M, Suzumura A, Yamamoto H, Marunouchi T (1990) colony-stimulating factor 1. Neurosci Lett 150:195199.
Activation and proliferation of the isolated microglia by Thry C, Stanley ER, Mallat M (1992) Interleukin 1 and
CSF-1 and possible involvement of protein kinase C. Brain tumor necrosis factor-a stimulate the production of colony-
Res 509:119124. stimulating factor 1 by murine astrocytes. J Neurochem
Schmechel DE, Rakic P (1979) A Golgi study of radial glial 59:11831186.
cells in developing monkey telencephalon: morphogene- Tomozawa Y, Inoue T, Takahashi M, Adachi M, Satoh M
sis and transformation into astrocytes. Anat Embryol 156: (1996) Apoptosis of cultured microglia by the deprivation of
115152. macrophage. Neurosci Res 25:715.
12 The Origin of Microglia and the Development of the Brain 189

Trentin AG. (2006) Thyroid hormone and astrocyte morphogen- Watanabe R, Takase-Yoden S, Fukumitsu H, Nakajima K (2002)
esis. J Endocrinol 189:189197. Cell transplantation to the brain with microglia labeled by
Tsuchiya T, Park KC, Toyonaga S, Yamada SM, Nakabayashi neuropathogenic retroviral vector system. Cell Transplant
H, Nakai E, Ikawa N, Furuya M, Tominaga A, Shimizu K 11:471473.
(2005) Characterization of microglia induced from mouse Wegiel J, Wisniewski HM, Dziewiatkowski J, Tarnawski M,
embryonic stem cells and their migration into the brain Kozielski R, Trenknert E, Wiktor-Jedrzejczak W (1998)
parenchyma. J Neuroimmunol 160:210218. Reduced number and altered morphology of microglial cells
Vallieres L, Sawchenko PE (2003) Bone marrow-derived cells in colony stimulating factor-1-deficient osteopetrotic op/op
that populate the adult mouse brain preserve their hematopoi- mice. Brain Res 804:135139.
etic identity. J Neurosci 23:51975207. Witting A, Muller P, Herrmann A, Kettenmann H, Nolte
Vilhardt F (2005) Microglia: phagocyte and glia cell. Int C (2000) Phagocytic clearance of apoptotic neurons by
J Biochem Cell Biol 37:1721. Microglia/Brain macrophages in vitro: involvement of lectin-
Vitry S, Bertrand JY, Cumano A, Dubois-Dalcq M (2003) , integrin-, and phosphatidylserine-mediated recognition.
Primordial hematopoietic stem cells generate microglia but J Neurochem 75:10601070.
not myelin-forming cells in a neural environment. J Neurosci Yokoyama A, Sakamoto A, Kameda K, Imai Y, Tanaka J
23:1072410731. (2006) NG2 proteoglycan-expressing microglia as multipo-
Walton MR, Gibbons H, MasGibbo GA, Sirimanne E, Saura tent neural progenitors in normal and pathologic brains. Glia
J, Gluckman PD, Dragunow M (2000) PU.1 expression in 53:754768.
microglia. J Neuroimmunol 104:109115. Yokoyama A, Yang L, Itoh S, Mori K, Tanaka J (2004)
Walton NM, Sutter BM, Laywell ED, Levkoff LH, Kearns Microglia, a potential source of neurons, astrocytes and
SM, Marshall GP 2nd; Scheffler B, Steindler DA (2006) oligodendrocytes. Glia 45:96104.
Microglia instruct subventricular zone neurogenesis. Glia Zassler B, Schermer C, Humpel C (2003) Protein kinase C
54:815825. and phosphoinositol-3-kinase mediate differentiation or pro-
Wang JM, Chen ZG, Colella S, Bonilla MA, Welte K, Bordignon liferation of slice-derived rat microglia Pharmacology 67:
C, Mantovani A (1988) Chemotactic activity of recombi- 211215.
nant human granulocyte colony-stimulating factor. Blood Zhu P, Hata R, Cao F, Gu F, Hanakawa Y, Hashimoto
72:14561460. K, Sakanaka M (2008) Ramified microglial cells pro-
Wang JY, Shum AY, Ho YJ (2003) Oxidative neurotoxicity mote astrogliogenesis and maintenance of neural stem
in rat cerebral cortex neurons: synergistic effects of H2 O2 cells through activation of Stat3 function. FASEB J 22:
and NO on apoptosis involving activation of p38 mitogen- 38663877.
activated protein kinase and caspase-3. J Neurosci Res Ziv Y, Avidan H, Pluchino S, Martino G, Schwartz M
72:508519. (2006) Synergy between immune cells and adult neu-
Wang CC, Wu CG, Shieh JY, Wen CY, Ling EA (1996) ral stem/progenitor cells promotes functional recovery
Immunohistochemical study of ameboid microglial cells in from spinal cord injury. Proc Natl Acad Sci USA 103:
fetal rat brain. J Anat 189:567574. 1317413179.
Chapter 13

Tissue Biology of Proliferation and Cell Death Among


Retinal Progenitor Cells

Rafael Linden, Rodrigo A.P. Martins, Mariana S. Silveira, Helena L. Borges, Alfred
Sholl-Franco, Lucianne Fragel-Madeira, and Ana Carolina Dudenhoeffer-Carneiro

Contents Abstract The retina is a complex network of various


molecularly and neurochemically distinct cell types.
13.1 Introduction . . . . . . . . . . . . . . . . . 192 These heterogeneous and highly interactive neurons
13.1.1 Retinal Progenitor Cells . . . . . . . . 193 and glia are stratified in rather precisely organized
13.1.2 Cell Proliferation in the Retina: On-the-fly layers, and often distributed in regular arrays, sur-
Restriction of Phenotype . . . . . . . . 194
rounded by rich, laminated extracellular matrix, as well
13.1.3 Retinal Tissue and Microenvironment
Around Progenitor Cells . . . . . . . . 194 as a dual vascular system. All components of retinal
13.2 The Cell Cycle Among Retinal Progenitor Cells 195 tissue affect the proliferation and survival of retinal
13.2.1 Morphology of Retinal Progenitor Cells . 195 progenitor cells through a combination of tightly reg-
13.2.2 Interkinetic Nuclear Migration and the Cell
ulated genetic and microenvironmental mechanisms.
Cycle in the Developing Retina . . . . . 196
13.2.3 The Cell Cycle Machinery in Retinal The latter are provided for by a variety of extrin-
Progenitor Cells . . . . . . . . . . . . 197 sic modulators, including neurotrophins, interleukins,
13.2.4 Checkpoint Control of the Cell Cycle . . 198 neurotransmitters and neuropeptides, acting through
13.3 Control of Retinal Progenitor Cell Proliferation
numerous types of plasma membrane receptors, and
by Growth Factors and Cytokines . . . . . . 199
13.3.1 Growth Factors . . . . . . . . . . . . 199 cross-talking intracellular signaling pathways. This
13.3.2 Interleukins . . . . . . . . . . . . . . 200 chapter reviews some of the major determinants of reti-
13.3.3 Neurotrophins . . . . . . . . . . . . . 200 nal cell population dynamics, which are a pre-requisite
13.3.4 Hedgehog, Notch and Wnt . . . . . . . 201
for the design of tissue engineering applied to retinal
13.3.5 Platelet Activating Factor . . . . . . . . 202
13.4 Control of the Retinal Cell Cycle degenerations.
by Neurotransmitters and Neuromodulators . 203
13.4.1 Classical Neurotransmitters . . . . . . . 203 Keywords Cell cycle Growth factors
13.4.2 Neuropeptides . . . . . . . . . . . . . 209 Neurotransmitters Programmed cell death Stem
13.5 Signal Transduction in the Extrinsic Control of
the Retinal Cell Cycle . . . . . . . . . . . . 210 cells
13.6 Death and Survival of Retinal Progenitor Cells 212
13.6.1 Mechanisms of Cell Death . . . . . . . 212
Abbreviations
13.6.2 Sensitivity to Cell Death Within the Retinal
Cell Cycle . . . . . . . . . . . . . . 215
13.6.3 Molecular Mechanisms of Cell Death 5-HT 5-hydroxytryptamine, serotonin
Among Retinal Progenitor Cells . . . . . 216 AC adenylyl cyclase
13.7 Conclusion and Future Directions . . . . . . 217
References . . . . . . . . . . . . . . . . . . . . 218
ACh acetylcholine
AIF apoptosis-inducing factor
AMPA -amino-3-hydroxy-5-methyl-4-
isoxazole propionic acid
Apaf-1 apoptotic protease-activating factor-1
R. Linden ()
Instituto de Biofsica, Universidade Federal do Rio de Janeiro, ATM ataxia and teleangiectasia mutated
Cidade Universitaria, Rio de Janeiro, Brazil ATP adenosine triphosphate
e-mail: rlinden@biof.ufrj.br ATR ATM and Rad3-related

H. Ulrich (ed.), Perspectives of Stem Cells, 191


DOI 10.1007/978-90-481-3375-8_13, Springer Science+Business Media B.V. 2010
192 R. Linden et al.

Bcl-2 B-cell lymphoma protein-2 OLO olomoucine


BDNF brain-derived growth factor PACAP Pituitary Adenylyl Cyclase-activating
BMP bone morphogenetic protein Polypeptide
BrdU bromo-deoxyuridine PAF platelet activating factor
CAK CDK activating kinase PCD programmed cell death
cAMP cyclic adenosine-3 -5 -monophosphate PDTC pyrrolidinedithiocarbamate
CDC cell division cycle PI3K phosphoinositide-3-kinase
CDK cyclin-dependent protein kinase PKA cAMP-activated protein kinase
CE cilliary epithelium PKC protein kinase C
CK2 casein kinase 2 PLC phospholipase C
Cl- chloride ion PNMT phenylethanolamine N-methyltransferase
CMZ cilliary marginal zone Rb retinoblastoma protein
CNS central nervous system RPC retinal progenitor cell(s)
CNTF ciliary neurotrophic factor Shh sonic hedgehog
CREB cAMP Response Element Binding protein STAT signal transducer and activator
DDC DOPA decarboxylase of transcription
DFO deferoxamine TGF transforming growth factor
Dhh desert hedgehog TH tyrosine hydroxylase
DNA-PK DNA protein kinase TrK tyrosine kinase receptor
DOPA dihydroxyphenylalanine twhh tiggy-winkle hedgehog
DSB double strand break Wnt wingless-int oncogene
EGF epidermal growth factor
ERK extracellular signal-related kinase
FasL Fas ligand
FGF fibroblast growth factor
GABA gamma amino butiric acid 13.1 Introduction
GAD glutamic acid decarboxylase
GCL ganglion cell layer Recent discoveries concerning stem cells in both the
GTP guanosine triphosphate developing and adult nervous system, and current
Gy gray experimental assays of cell therapies have spread a
Hh hedgehog proteins wave of excitement concerning the possibility of devel-
IFN interferon oping restorative treatments for degenerative diseases
IGF insulin-related growth factor of the nervous system (Muotri and Gage, 2006). The
Ihh Indian hedgehog case of the retina includes both retinal dystrophies
IL interleukin (Vugler et al., 2007; MacLaren and Pearson, 2007), as
INL inner nuclear layer well as glaucomatous neurodegeneration (Bull et al.,
INM interkinetic nuclear migration
2008).
IP3 inositol triphosphate
As a necessary step for the establishment of new
IR ionizing radiation
technologies of regenerative neurology and neuro-
mAChR muscarinic acetylcholine receptor(s)
ophthalmology, a thorough understanding of develop-
MAPK mitogen activated kinase
MG132 carbobenzoxyl-leucinyl-leucinyl-leucinal mental mechanisms depends heavily upon unraveling
mGluR metabotropic glutamate receptor the basic biology of stem/progenitor cells. A large
MIMO mimosine body of work has been directed at the behavior of stem
MOP-R opioid receptor cells, mostly in vitro, and a number of both genetic and
nAChR nicotinic acetylcholine receptor(s) epigenetic events involved in either the maintenance
NBL neuroblastic layer or the exit from an undifferentiated state have been
NMDA N-methyl-D-aspartate uncovered in recent years (Yeo et al., 2008; Lunyak
NO nitrous oxide and Rosenfeld, 2008).
NPY neuropeptide Y Nonetheless, stem/progenitor cells reside among
NT neurotrophin a complex environment, and just like any other
ODC ornitine decarboxylase issue concerning developmental biology, would greatly
13 Tissue Biology of Proliferation and Cell Death 193

benefit from studying their dependence on the struc- A


ture, cellular and extracellular matrix components, GCL
cellcell communication and signaling systems in the INL
context of the tissue (e.g. Raff, 1992; Linden, 2000;
Rutishauer, 2008). The present chapter aims at review- NBL
ing some of the cellular and molecular interactions, and
elements of their signal transduction pathways, that M G1 S G2 M G1
affect two of the major events of the life cycle of retinal
progenitor cells, namely the cell cycle and consequent B C
proliferation rates, and the sensitivity to programmed
cell death.

13.1.1 Retinal Progenitor Cells

The mammalian neural retina is derived from retinal Fig. 13.1 Retinal progenitor cells and interkinetic nuclear
progenitor cells (RPC) of neuroectodermal origin, ini- migration in the newborn rat retina. (a) Drawing of a cross-
tially found in the protrusive optic vesicles on both section of the neural retina from a newborn rat, showing the
differentiating cell body and plexiform layers found at birth in
sides of the neural tube. RPC proliferate repeatedly,
this species. Basal (inner) is up, apical (outer) is down. Round
and daughter cells eventually exit the cell cycle and grey profiles depict differentiating ganglion cells in the gan-
differentiate into several dozen types, which form reg- glion cell layer (GCL), amacrine cells in the inner nuclear layer
ular mosaics distributed in alternating layers of cell (INL), and the inner plexiform layer (horizontal lines) between
GCL and INL. Occasional differentiating photoreceptors are
bodies and synaptic contacts (Galli-Resta et al., 2008).
shown at the lower rim, and horizontal cells in the middle of
Therein, individual retinal cell types can be identified the neuroblastic layer (NBL). The white profiles straddling the
by their position, morphology, biochemical properties retinal thickness represent proliferating cells undergoing interki-
and function (Adler, 1986; Kolb et al., 1992; Pourcho, netic nuclear migration at successive phases of the cell cycle
(M-G1-S-G2-M, from left to right), amidst a population of
1996; Kolb, 1997; Jeon et al., 1998; Cayouette et al.,
unsynchronized proliferating cells (light grey-lined profiles in
2006). the background). (b, c). The S-phase (b) and M-phase (c) strata
Mature retinal cells are grouped in 6 distinct classes of the NBL in the newborn rat. Both photomicrographs are lim-
of neurons ganglion cells, amacrine (inclusive of ited to the NBL (bracket in a), and show immunohistochemical
staining of bromo-deoxy-uridine (BrdU) injected 30 min before
interplexiform) cells, horizontal cells, bipolar cells,
fixation (b), and of phosphorylated Histone H3 (c), markers of
cone photoreceptors and rod photoreceptors and one DNA synthesis and of the G2/M transition, respectively. The
type of glia, the Mller cell (Livesey and Cepko, pigment epithelium was removed when retinae were explanted
2001). These cell classes are produced according to an (Mod from Carneiro et al., 2008)
evolutionary conserved birth order (Finlay, 2008), in
which ganglion cells, cone photoreceptors, horizontal
cells and roughly half of the amacrine cells are gener- some mitotic figures have been observed within the
ated first during embryogenesis, followed by bipolar inner nuclear layer of the developing vertebrate retina,
cells, Mller glia and the remaining amacrine cells. which give rise exclusively to horizontal cells that
Rod photoreceptors are generated throughout retinal remain in the same laminar location as their mother
development (Young, 1985a,b; Wallace, 2008). cell (Rapaport et al., 1985; Rapaport and Vietri, 1991;
Similar to other layered structures of the CNS, Godinho et al., 2007; Rompani and Cepko, 2008).
the RPC are located in a pseudostratified proliferative Neurogenesis in the mammalian retina ceases at
epithelium (Fig. 13.1, see Section 2.3), analogous to the end of the developmental period and is essentially
the ventricular zone of the developing brain (Miyata, absent in the adult. Exception to this rule is the reac-
2008), and here referred to as the neuroblastic layer tive proliferation of Mller glial cells after retinal
(NBL). The majority of RPC cell division occur at the injury, which may be followed by neuronal differen-
apical tip of the NBL (Baye and Link, 2008), although tiation, thus indicating potential neurogenic properties
194 R. Linden et al.

of reactive Mller cells in adult mammalian retina to the generation of the so-called late born cell
(Fischer and Reh, 2001; Ooto et al., 2004; Bringmann classes.
et al., 2006; Osakada et al., 2007; Florian et al., 2008). The mechanisms underlying progressive lineage
In addition, retinal cell differentiation has also been restriction in RPC are still poorly understood, but
reportedly derived in vitro from quiescent progeni- evidence has accumulated that both intrinsic factors,
tor cells located in both the ciliary body and iris as well as responses to extrinsic factors regulate the
pigment epithelium (Tropepe et al., 2000; Ohta et behavior of RPC, to ensure that appropriate numbers of
al., 2008), which would suggest similarities with the the various retinal cell types are generated throughout
ciliary marginal zone and retinal pigment epithelium retinal development (Reh and Levine, 1998; Cepko,
of teleost fish, amphibia and birds (Kubota et al., 2002; 1999; Marquardt and Gruss, 2002; Marquardt, 2003;
Reh and Fischer, 2006). Nevertheless, recent studies Donovan and Dyer, 2005; Cayouette et al., 2006).
suggest that the ciliary epithelium (CE) of mammals
does not contain retinal stem cells, since all prolifer-
ating cells of CE-derived spheres display features of 13.1.3 Retinal Tissue
differentiated pigmented epithelial cells. In addition,
these pigmented proliferating cells from either human
and Microenvironment Around
or mouse CE-derived spheres failed to fully differ- Progenitor Cells
entiate into retinal cell types, which was interpreted
as the activation of an incomplete transdifferentiation Notwithstanding the remarkable intrinsic factors that
program (Cicero et al., 2009). guide both RPC proliferation and phenotypic deter-
mination, their sensitivity to extrinsic control raises
fundamental questions as to the role of cellcell inter-
13.1.2 Cell Proliferation in the Retina: actions upon the behavior of RPC. Within the develop-
ing retina, similar to other areas of the central nervous
On-the-fly Restriction
system, RPC thrive amidst a rich, varied, and histo-
of Phenotype typically structured environment (Fig. 13.1). It is of
particularly importance that the morphology of prolif-
In the developing nervous system, multipotent progen- erating retinal cells, which span the full depth of the
itor cells may undergo either symmetric or asymmetric retina, allows for a changing set of partners to inter-
cell division. Symmetrical divisions may give rise act with successive generations of RPC, as postmitotic
either to multipotent daughter cells that reenter the cells progressively populate the retinal layers during
cell cycle, or to a pair of postmitotic cells. Following development.
asymmetrical divisions the daughter cells have distinct Several studies have shown that changes in the time
fates: Usually one cell reenters the cell cycle, while course and spatial patterns of expression or distribu-
the other differentiates into a post-mitotic cell (Chenn tion of growth factors and cytokines are critical for the
and McConnell, 1995; Mione et al., 1997; Yoshikawa, control of not only the proliferative state and the spec-
2000; Cayouette and Raff, 2003; Roegiers and Jan, ification of cell fate in the retina (Gaur et al., 1992;
2004; Cayouette et al., 2006). Zhao and Barnstable, 1996; Zhao and Lemke, 1998;
Lineage analysis has shown that RPC are multi- McFarlane et al., 1998; von Bartheld, 1998; Jaynes
potent and individual RPC can give rise to a variety and Turner, 2000; Patel and McFarlane, 2000; Dyer
of combinations of retinal neurons and glia (Price et and Cepko, 2001; Das et al., 2006; Silva et al., 2008),
al., 1987; Turner et al., 1990). However, RPC are but also of the survival of early proliferating neural
somewhat heterogeneous (Lillien, 1998), and during progenitor cells (de la Rosa and de Pablo, 2000).
retinal development, these cells undergo progressive Both the pigment epithelium and the differentiating
changes in their competence to generate distinct cell retinal cells themselves provide a continuously chang-
types (Cepko et al., 1996), such that RPC that exit the ing source of a large number of distinct cytokines
cell cycle early in development give rise to the early (Holtkamp et al., 2001), as well as neurotransmitters
born cell classes, whereas RPC born later are restricted and neuromodulators (Linden et al., 2005; Martins and
13 Tissue Biology of Proliferation and Cell Death 195

Pearson, 2007). Adding to these, adhesion molecules of the epithelium and another process that terminates
(Sharma and Johnson, 2000), and extracelular matrix at the basal surface, show typical epithelial features
components (Inatani and Tanihara, 2002; Erlich et al., and are highly polarized along their apicalbasal axis
2003) present within the developing retinal tissue, may (Turner et al., 1990; Gtz and Huttner, 2005; Godinho
interact with responsive RPC. Vascular development, et al., 2007). The proliferative NBL appears to be strat-
together with the presence of immune cells in the ified because the nuclei of RPC are distributed along
developing retina (Linden et al., 1986) contribute an the apical-basal axis, and migrate up and down during
additional set of extracellular factors that may affect the cell cycle (see below).
the developing RPC. During cell division, most RPC maintain both their
Independent of the coupling between proliferation basal and apical attachments throughout the cell cycle.
and cell differentiation, the rate of cell proliferation After division, one daughter cell inherits the basal
per se (Levine and Green, 2004), together with pro- process and its sibling cell initiates a new one. The
grammed cell death (Linden et al., 1999; de la Rosa inheritor, which can be either a neuron or a progen-
and de Pablo, 2000) rank among the major determi- itor cell, uses the elongated process for translocation
nants of the correct wiring of the retina. Therefore, the and positioning (Saito et al., 2003; Baye and Link,
understanding of RPC to a level allowing their eventual 2008). Previous findings using DiI-labeled cerebral
manipulation in therapeutic context requires a thor- wall slices, also demonstrated that the basal process of
ough analysis of the factors that control the retinal cell each cortical progenitor cell is maintained throughout
cycle and the sensitivity of retinal progenitor cells to the M phase, and is inherited by one of its daughter
programmed cell death. cells (Miyata et al., 2001, 2002). Interestingly, how-
ever, and differing from the developing cerebral cortex,
retinal progenitors either do not, or only partially
develop radial glial-cell features during neurogenesis,
13.2 The Cell Cycle Among Retinal suggesting that the migration of early generated neu-
Progenitor Cells rons is guided by the basal process (Turner et al., 1990;
Kriegstein and Gtz, 2003; Gtz and Huttner, 2005;
Kriegstein et al., 2006). However, it was additionally
13.2.1 Morphology of Retinal shown that a small number of committed precursor
Progenitor Cells cells lose their attachment to the apical surface and give
rise exclusively to two horizontal cells (Godinho et al.,
The neural retina is derived from bilateral evaginations 2007).
of the anterior neural plate, which form the bilayered The apicalbasal polarity of RPC is an important
optic cup, the inner layer of which contains the multi- basis for their symmetric versus asymmetric division
potent RPC. Retinal volume and eye size are primarily (Fig. 13.2). Some studies indicate that through the con-
determined by the number of times each progenitor trol of the orientation of the cleavage plane during cell
cell divides before its final division to generate two division, progenitors may differentially distribute cell
postmitotic cells. In mammals and birds, proliferation fate-determining factors to daughter cells, thereby pro-
of RPC ceases before retinal histogenesis is complete ducing equal or unequal outcomes (Cepko et al., 1996;
(Young, 1985a; Cepko et al., 1996; Dhomen et al., Gtz and Huttner, 2005; Cayouette et al., 2006). In
2006). Retinal cell differentiation obeys the inner-to- vivo imaging of retrovirally labeled progenitor cells
outer and central-to-periphery gradients, beginning in in the rat retina showed that cleavage planes that are
the inner layer of the central optic cup and progressing either parallel to the apical surface or oriented verti-
concentrically in a wave-like fashion until it reaches cally have distinct outcomes, as to whether the daugh-
the peripheral edges of the retina (Rapaport and Stone, ter cells will adopt either the same or distinct fates
1983; Young, 1985b; Beazley et al., 1987; Prada et al., (Chenn and McConnell, 1995; Cayouette and Raff,
1991; Marquardt and Gruss, 2002). 2003; Cayouette et al., 2006; Godinho et al., 2007).
Retinal progenitor cells are bipolar in shape, bear- Thereby, controlling the orientation of RPC division
ing one process that extends towards the apical surface could help establishing distinct lineages of RPC.
196 R. Linden et al.

basal ILM

either... or...

apical RPE
G1 S G2 M G1 M G1
Fig. 13.2 Symmetrical and asymmetrical divisions in the on the plane of cell division, daughter cells can either remain in
retinal neuroepithelium. Mitotic cells can divide either hor- the cell cycle and proceed through the G1 phase with nuclear
izontally or vertically at the apical margin of the neuroblastic migration anchored to the inner limiting membrane (ILM), or
layer, close to the retinal pigment epithelium (RPE), Depending leave the cell cycle and differentiate

13.2.2 Interkinetic Nuclear Migration the thickness of the ventricular layer (Sauer and
and the Cell Cycle in the Chittenden, 1959; Sauer and Walker, 1959; Langman
et al., 1966; Baye and Link, 2008 for review).
Developing Retina
In the developing retina, similar to the ventricu-
lar zone of other parts of the central nervous sys-
In 1935, Sauer correlated the shape and histological tem, the nuclei of cell progenitors migrate back and
appearance of the nuclei of proliferating cells in the forth through the full thickness of the NBL in syn-
ventricular layer of the cerebral cortex of vertebrate chrony with the cell cycle (Sidman, 1961; Fujita, 1962;
embryos, with their distance to the lumen of the neu- Young, 1985b; Rapaport et al., 1984). DNA is dupli-
ral tube, and established the concept of interkinetic cated in the innermost margin, while mitosis occurs
nuclear migration (INM). This event has so far been in the outer rim of the NBL (Fig. 13.1, Fujita, 1962;
described only in the nervous system, and is char- Sidman, 1961).
acterized by intracellular movement of the nuclei of The total duration of the cell cycle in the retina
the neuroblasts, as these cells pass through successive increases as development proceeds, in part due to
phases of the cell cycle (Sauer, 1935). lengthening of the S phase, which comprises at least
The nuclei originated in each mitotic division (M- half of the cell cycle during postnatal development of
phase) in the apical margin of the ventricular zone the rat retina. Therefore, roughly 50% of the asyn-
migrate towards the basal margin, where DNA repli- chronous population of proliferating cells are in S
cation (S-phase) takes place. Once DNA synthesis phase at any time in the newborn rat retina, and
is completed, the nucleus migrates back to the api- these cells can be identified by a single injection of
cal margin, where cell division occurs. The G1 and a nucleotide analogue, such as Bromo-deoxyuridine
G2 phases occur along the nuclear migration pathway (BrdU) (Alexiades and Cepko, 1996; Rehen et al.,
(Figs. 13.1, and 13.2). Thus, cell cycle phases corre- 1999). Immediately after incorporating the marker
late with nucleus position during interkinetic migration nucleotide, the labeled nuclei of cells in S phase are
(Sauer, 1935; Sidman, 1961; Fujita, 1962; Rapaport et found within the basal (inner) half to two-thirds of the
al., 1984; Young, 1985a; Jacobson, 1991; Takahashi et NBL in the newborn rat retina (Fig. 13.1).
al., 1996; Saito et al., 2003; Gtz and Huttner, 2005; Hayes and Nowakowski, using DNA labeling tech-
Cayouette, 2007). niques, correlated the distance and velocity of move-
INM was confirmed by experimental observations ment of nuclei to the phases of the cell cycle, and
in which treatment with mitotic inhibitors resulted in observed that during G1 the nuclei move for various
arrested metaphase cells only at the apical surface and distances and velocities, reaching the inner half of the
3 H-thymidine pulse-labeling experiments showed dif-
ventricular zone where they distribute along various
ferential labeling of progenitor cell nuclei throughout levels. Upon entering S phase, the nuclei stop moving
13 Tissue Biology of Proliferation and Cell Death 197

and remain still until completion of DNA synthesis. of the cell cycle (Gran and Reddy, 1995; Stewart et
The passage through G2 is fast, and the nuclei soon al., 2003). Therefore, both the transcriptional control
reach the ventricular surface, where they stay during of cyclin mRNA and the cyclic degradation cyclin
mitosis (Hayes and Nowakowski, 2000). proteins are necessary to allow exit from mitosis and
It is likely that positioning of intracellular nuclei re-entry into a new proliferative cycle (Page and Hieter,
during INM is regulated by extracellular signals 1997; Stewart et al., 2003).
present along different strata of the proliferative Distinct combinations of cyclins and CDK regulate
layer, associated with transitions of cell cycle phases. the transition between different phases of the cell cycle
Disruption of retinal cell cycle by genetic alterations by directly regulating the phosphorylation state of mul-
or pharmacological manipulations accordingly alters tiple target proteins (Sanchez and Dynlacht, 2005 for
INM (Pearson et al., 2005b; Willer et al., 2005). review). Following the completion of cell division, the
Moreover, when cell cycle progression is partially or decision to exit the cell cycle and turn into a post-
completely inhibited, INM is either slowed or stopped mitotic cell, or to re-enter in S-phase, depends on the
(Ueno et al., 2006; L. Fragel-Madeira, unpublished state of phosphorylation of the retinoblastoma (Rb)
results). An elegant study recently showed that an family proteins. Distinct cyclin/CDK holoenzymes can
apical-basal gradient of notch signaling regulates the phosphorylate the Rb and/or its family members (p107
interplay of INM and the exit from the cell cycle (Del and p130) (Sherr, 1996; Weinberg, 1995). Ample evi-
Bene et al., 2008). Notwithstanding, the causal rela- dence support a model of sequential phosphorylation
tionships between nuclear migration and progression of the Rb proteins by distinct cyclin/CDK complexes.
of the cell cycle are yet to be uncovered. Indeed, the The three D-type cyclins (D1, D2 and D3), which are
inextricable link between the INM and the succession expressed in a tissue-specific manner, associate with
of phases of the cell cycle still defies investigators in either one of two CDK (CDK4 and CDK6). Cyclin
this field. D/CDK complexes are believed to act early during
G1, while the cyclin E/CDK2 complex is believed to
be the main Rb/p107/p130 kinase later in G1. The
phosphorylation of the Rb family proteins leads to
13.2.3 The Cell Cycle Machinery in Retinal
their dissociation from the transcription factor E2F.
Progenitor Cells When dissociated from Rb proteins, E2Fs regulate
the transcription of genes required for the transition
The molecular mechanisms involved in the control and progression through S-phase. Conversely, if Rb is
of the cell cycle have been usually viewed as evo- unphosphorylated, the cell will exit the cell cycle and
lutionarily conserved. However, cloning of the genes become post-mitotic (Morgan, 1997).
that encode components of the cell-cycle machinery Lack of Rb causes ectopic mitoses and increased
revealed multiple, closely related family members for cell death in the mouse retina at E16.5 (Maandag et
almost all major regulators, and current evidence sup- al., 1994), and the loss of both Rb and p107 leads to
ports the notion that related family members play severe retinal dysplasia and the formation of tumors
highly specialized roles during development, which (Robanus-Maandag et al., 1998, Zhang et al., 2004).
may vary among distinct species (Dyer and Cepko, Studies of the regulation of retinal development by
2001). Rb proteins have demonstrated a crucial role of these
Progression through the distinct phases of the cell in the control of RPC proliferation (MacPherson et
cycle depends upon the activity of a family of ser- al., 2004; Zhang et al., 2004). Importantly, Rb fam-
ine/threonine kinases, called CDK (cyclin-dependent ily members are not interchangeable in the developing
kinases). Cyclins are regulatory proteins that bind and retina, therefore, when Rb gene is inactivated in the
activate the kinase activity of the CDK. In general, the developing mouse retina, a compensatory increase in
CDKs are constitutive, while the expression of cyclins p107 expression was observed. In addition, Rb and
changes during the different phases of the cell cycle p130 are redundantly expressed in a subset of reti-
(Sherr, 1993). The formation of specific cyclin/CDK nal cells (Donovan et al., 2006). The development of
complexes, and the consequent kinase activity, is fun- tumors in response to the inactivation of Rb family
damental for progression through the distinct phases genes, as well as the redundancy and compensation
198 R. Linden et al.

described above, emphasize the importance of the Rb to CDK activation (Kaldis, 1999; Abbas and Dutta,
pathway for control of the cell cycle during retinal 2006 for reviews).
development (MacPherson et al., 2004; Zhang et al., Retinal cells appear to be critically dependent on
2004). both cyclin D1 and p27Kip1 , as shown in transgenic
Rb family proteins bind and regulate various mem- mice. The retinae of cyclin D1-knockout mice show
bers of the E2F transcription factor family. E2F1- reduced cell number (Sicinski et al., 1995), lower rates
3 are believed to function as transcriptional activa- of proliferation and an unique pattern of photorecep-
tors, while E2F4-5 act as repressors (for reviews: tor cell death (Ma et al., 1998), while p27Kip1 -deficient
Cam and Dynlacht, 2003; Korenjak and Brehm, animals show disturbances in retinal organization with
2005). Expression of E2F transcription factors in the an increased cell number which is consistent with
developing retina has been previously demonstrated. a requirement for p27Kip1 in RPC cell cycle exit
Specifically, both E2F1 and E2F2 are expressed in the (Nakayama et al., 1996). The p27Kip1 protein was
NBL, while expression of E2F3-4 may be confined reportedly upregulated during late G2 or early G1 in
to postmitotic cells of the developing retina (Dagnino most cells that leave the cell cycle at any stage of devel-
et al., 1997). Ectopic expression of E2F1 driven by a opment (Dyer and Cepko, 2001). In turn, p57Kip2 is
photoreceptor-specific promoter induces the prolifera- expressed in only a subset of progenitor cells that leave
tion of differentiating photoreceptors, indicating that the cell cycle during a brief period of retinal develop-
E2F1 activity must be downregulated to ensure cell ment in mouse embryos. RPC that upregulate p57Kip2
cycle exit (Lin et al., 2001). Further evidence for the differ from those that upregulate p27Kip1 , and these
regulation of RPC proliferation by E2Fs was shown proteins are upregulated at distinct times during the
in a knock-in mouse. Developing retinas expressing a cell cycle (Dyer and Cepko, 2001). Little is known
mutant Rb protein incapable of binding E2F1-3, dis- about the role of CAK, WEE or CDC25 in the regu-
played similar defects as described for Rb-deficient lation of progenitor cell cycle in developing vertebrate
retinas, including ectopic proliferation of RPC. These retina.
findings reinforce the importance of the Rb/E2F path- Cyclin D1 was shown to be genetically upstream of
way in the regulation of the RPC cell cycle (Sun et al., cyclin E and p27Kip1 function. Nonetheless, prolifera-
2006). tion still occurs in the cyclin D1-null retina, which may
In addition to the association to the cyclins, the be due to either or both persistent expression of cyclin
activity of CDK complexes is also regulated in D3 or to a D-cyclin-independent mechanism. This sug-
other ways. Two families of cyclin-dependent kinase gests that multiple mechanisms may exist to support at
inhibitors (CKIs) have been identified that can inhibit least a basal level of G1 progression in RPC (Levine
the activity of the CDK (Sherr and Roberts, 1995; and Green, 2004).
Vidal and Koff, 2000; Stewart et al., 2003, Harper and In the retina, p57Kip2 is both necessary and suffi-
Brooks, 2005). The INK family (p15Ink4a , p16Ink4b , cient for appropriate cell cycle exit. However, p57Kip2
p18Ink4c and p19Ink4d ) is believed to bind prefer- is not expressed in RPC exiting the cell cycle dur-
entially cyclin-DCDK complexes and lead to dis- ing the postnatal period. RPC deficient in p19Ink4d
ruption of the holoenzyme subunits. Cip/Kip CKIs also proliferate beyond the normal period, although the
(p21Cip1 , p27Kip1 and p57Kip2 ) can form stable com- phenotype of p19Ink4d -null retinas is less severe than
plexes with either cyclin-DCDK or cyclin-ECDK. that of p27Kip1 knockout (Levine and Green, 2004).
In addition, several transcriptional mechanisms, as The available data, therefore, indicate that certain
well as post-translational regulation (e.g., phospho- regulators of the cell cycle, such as cyclin D family
rylation and ubiquitination) of cyclins, CDKs and members and several CKIs, have major roles in the
CKIs have been reported. For example, CAKs (Cdk control of the proliferation of RPC.
Activating Kinases) are known to phosphorylate Thr-
160, which promotes structural changes and increases
13.2.4 Checkpoint Control
CDK2 kinase activity by several orders of magnitude.
Similarly, CDKs are inhibited by phosphorylation by of the Cell Cycle
the WEE1 family of kinases, which may be counter-
acted by the action of CDC25 phosphatases, which Together with the cell cycle progression machinery,
dephosphorylate WEE1 sites (Thr-14, Tyr-15) leading cells are equipped with checkpoints that guarantee the
13 Tissue Biology of Proliferation and Cell Death 199

correct ordering of cell cycle events. Cell cycle check- (ataxia-telangiectasia mutated), ATR (ATM and Rad3-
points are mechanisms capable to arrest or block phase related) and DNA protein kinase (Bartek and Lukas,
transition until the recognition of appropriate sig- 2007). ATM and ATR kinases respond to distinct types
nals necessary to cell cycle progression (Hartwell and of DNA damage. ATM mainly responds to double-
Weinert, 1989). Checkpoints are composed of cellu- strand breaks (DSBs), whereas ATR is activated by S
lar surveillance and signaling pathways that coordinate phase damage such as single stranded (ss)DNA and
DNA repair with chromosome metabolism and cell- stalled forks (Shilloh, 2003).
cycle transitions (Branzei and Foiani, 2006; Bartek and When ATM and ATR are recruited to sites of
Lukas, 2007). damage, they target various substrates, including
For example, the genome is under constant threat checkpoint kinase-2 (CHK2) and CHK1, respectively
of damage from both exogenous agents and intrinsic (Abraham, 2001; Matsuoka et al., 2007 for review).
mechanical events that may damage DNA, including ATM or ATR phosphorylate and activates CHK1/2,
the unwinding of chromatin during the normal cell which phosphorylate, for example, distinct members
cycle (Branzei and Foiani, 2008 for review). DNA of CDC25 phosphatase family depending of cell cycle
lesions activate checkpoint pathways that regulate spe- phase. This phosphorylation reduces CDC25 activ-
cific DNA-repair mechanisms in the various phases ity and prevents removal of inhibitory phosphates
of the cell cycle, and result in a reversible delay of of cyclin-CDK complexes required for cell cycle
cell cycle progression (Walworth, 2000; Sampath and progression (Branzei and Foiani, 2008). DSB resec-
Plunkett, 2001; Bartek and Lukas, 2001; Borges et al., tion and activation of ATR requires both ATM and
2004). cyclin-dependent kinase (CDK) activity (Jazayeri et
Post-translational modifications, such as phospho- al., 2006), whereas in other circumstances, ATR may
rylation, ubiquitylation and sumoylation, are crucial be required for activation of ATM (Burdak-Rothkamm
for the regulation of the checkpoint machinery, thereby et al., 2008).
regulating important cell cycle events (Gutierrez and Notwithstanding the classical responses to DNA
Ronai, 2006). These post-translational modifications damage, the possibility cannot be discarded that other
may affect the recruitment of repair proteins to dam- pathways of activation of checkpoint kinases such as
aged DNA, or direct the repair machinery towards a ATM/ATR, or CHK1/CHK2 may exist, and may be
certain type of lesion, often to allow repair in a specific subject to extrinsic control in proliferating cells.
cell-cycle phase.
The checkpoint proteins are recruited to DNA
lesions, transducer complexes transmit and amplify the
signals to downstream targets such as the DNA-repair 13.3 Control of Retinal Progenitor Cell
apparatus and the cell-cycle machinery (Branzei and Proliferation by Growth Factors
Foiani, 2006). The transmission of the signal or acti- and Cytokines
vation of these targets is often achieved by phosphory-
lation events that affect either the transcription level
or activity of repair genes, and modulate cell-cycle 13.3.1 Growth Factors
transitions.
There are three major checkpoints, depending on Experiments in vitro showed that acid fibroblast
cell cycle phase and all related to DNA damage (Bartek growth factor (aFGF, or FGF-1), basic FGF (bFGF, or
and Lukas, 2001; Cortez, 2003; Xiao et al., 2003). FGF-2), insulin, or insulin-like growth factors (IGF-1
G1 checkpoint results in that DNA synthesis will not and IGF-2) promote cell proliferation in rat (E15-E18),
occur if the replication machinery is not ready; intra-S chicken (E-6), and teleost fish (Carassius auratus,
checkpoint monitors DNA integrity during its dupli- 315 cm in standard) retina (Frade et al., 1996a;
cation, and G2/M checkpoint verifies DNA damage Hernandez-Sanchez et al., 1995; Lillien and Cepko,
before cell enters M-phase (Walworth, 2000; Sampath 1992; Otteson et al., 2002). The dependence on stage
and Plunkett, 2001; Bartek and Lukas, 2001). of development observed for these proliferative effects
Central components of the checkpoint machinery may reflect important stage-dependent factors associ-
are the phosphoinositide 3-kinase related kinases ATM ated with cell responsiveness. Other studies showed an
200 R. Linden et al.

effect of epidermal growth factor (EGF) upon prolif- by the co-administration of insulin with EGF or bFGF
eration, but not self-renewal of embryonic (E18.5) and (Fischer et al., 2002; Fischer and Reh, 2003), indicat-
early postnatal (P1) mouse RPC (Ahmad et al., 2004; ing that these growth factors have similar effects during
James et al., 2004), although mouse adult retinal cells development of the retina. Basic FGF, EGF, and TGF
proliferate and self-renew in response to both FGF- stimulate the proliferation of embryonic RPC (Anchan
2 and EGF (Tropepe et al., 2000), depending on the and Reh, 1995; Anchan et al., 1991; Lillien and Cepko,
presence of laminin (Otaegi et al., 2007). 1992), and induce the differentiation of neurofilament-
Basic fibroblastic growth factor has important roles expressing retinal ganglion cells in embryonic chick
in the CNS (Hicks, 1998), inclusive of RPC. FGF- retina in vivo (McCabe et al., 1999) as well as at
2 is abundantly expressed in both embryonic and the retinal margin of the retina of postnatal chickens
adult RPC, and regulates retinal growth, organiza- (Fischer et al., 2002).
tion, and differentiation (Hicks, 1998). The prolifer- IGF-1 is expressed at high levels in the teleost
ation of rodent retinal cells was stimulated by bFGF retina, and was shown to induce RPC to stop prolif-
(Guilarducci-Ferraz et al., 2008; Hicks and Courtois, eration and produce new rod photoreceptors (Otteson
1992) in monolayer cultures, coupled with a sig- et al., 2002). Corroborating these data, the blockade of
nificant increase in the number of photoreceptors IGF receptors reduced the number of dividing rod pro-
(Hicks and Courtois, 1992). Both the proliferation and genitor cells in the outer nuclear layer (Zygar et al.,
differentiation induced by bFGF depend on an uncom- 2005).
mitted stage of retinal precursors, as well as of the
presence of laminin (Otaegi et al., 2007).
It has also been shown that earlier RPC express 13.3.2 Interleukins
lower levels of EGF receptors (EGF-R) than late pro-
genitors (Lillien and Wancio, 1998), suggesting that
Cytokines and chemokines have often been recognized
EGF contributes to the timing of progenitor cell dif-
as relevant players in neuroimmune interactions (Bauer
ferentiation (Lillien and Wancio, 1998). These data
et al., 2007; Schafers and Sorkin, 2008; Reale et al.,
are consistent with the previous observation of age-
2008; Fontaine et al., 2008; Zhang et al., 2008), but lit-
dependent differences in mitotic responses to tumor
tle is known of their roles in the control of the cell cycle
growth factor alfa (TGF), and TGF-3 in vitro
of neural progenitor cells (Vela et al., 2002; Vallieres
(Anchan and Reh, 1995; Lillien and Cepko, 1992;
et al., 2002; Wang et al., 2007; Nakanishi et al., 2007;
Lillien and Wancio, 1998). Immature RPC were main-
Kang and Kang, 2008).
tained in vitro as long as 6 months in serum free
We have, however, recently shown that interleukin
medium with both bFGF and EGF (Akagi et al.,
(IL)-4 has an anti-proliferative effect upon RPC of
2003), and could be stimulated to form neurospheres
newborn mice, an effect mediated by activation of
(Liljekvist-Soltic et al., 2008). Both EGF and bFGF
protein kinase A. This effect was associated with
were shown to establish Mller cell spheres (Das et al.,
blockade of the transition from G1 to S phase, since
2006). Either withdrawal of both factors or the addi-
IL-4 enhanced expression of p27Kip1 and decreased the
tion of TGF were optimal to establish changes in the
expression of cyclin D1 (Fig. 13.3, Silva et al., 2008).
proliferative state of RPC at the time of birth in rats
(Lillien, 1995; Lillien and Cepko, 1992).
Intraocular injections of bFGF, EGF, or insulin
stimulated the proliferation of nonpigmented cells in
13.3.3 Neurotrophins
distinct regions of the ciliary body. This led to dif-
ferentiation of some of these cells into various types Brain-derived neurotrophic factor (BDNF) also
of retinal neurons, in a regional specific way, which increased the number of neuronal progeny from
indicates differential responsiveness to growth fac- the RPC population at the expense of Mller glia
tors (Fischer and Reh, 2003). The same properties (Ahmed et al., 1995). Reports indicated that Mller
were observed in some retinal pigmented cells in mice glia express both the high affinity TrkB and the low
(Ahmad et al., 2000; Tropepe et al., 2000). This effect affinity p75 receptors for neurotrophins (Garcia et
can be obtained by either insulin or EGF, as well as al., 2003; Oku et al., 2002), and that the loss of p75
13 Tissue Biology of Proliferation and Cell Death 201

A B C

Fig. 13.3 Modulation of the retinal cell cycle by interleukin- kinase A (H-89) prevents the effect of IL-4. (b, c) IL-4 induces
4. (a) Incubation of retinal explants from neonatal rats with IL-4 a decrease in content of cyclin D1 and an increase in the CDK
reduces the incorporation of tritiated thymide, and indication inhibitor p27Kip1 , which are likely to underlie the blockade of
of partial inhibition of the cell cycle. An inhibitor of protein the cell cycle (Mod. from Silva et al., 2008)

is implicated in the progression of retinoblastoma produced by retinal amacrine and RGC acts on central
(Dimaras et al., 2006). The influence of treatment with retinal targets (von Bartheld et al., 1996). The mecha-
BDNF and erythropoietin on the differentiation and nisms of control of the cell cycle by NT-3 were studied
on de-differentiation of mammalian Mller glia into in oligodendroglial progenitor cells. NT-3 regulates
RPC was analyzed in vitro (Nickerson et al., 2008). entry into S-phase through the expression of c-myc
Treatment with BDNF induced Mller glia prolifera- and cdc2 (Kumar et al., 1998). Consequently, blockade
tion, RPC phenotype up-regulation, and development of NT-3 signaling, by either antibodies or overexpres-
of sphere-like colonies, similar to treatment with both sion of truncated TrkC (Bovolenta et al., 1996; Das et
EGF and bFGF (Das et al., 2006). al., 2000), inhibits the transition to S phase, and thus
Neurotrophin-3 (NT-3) regulates neural prolifera- lengthens the cell cycle.
tion, differentiation, and survival in the nervous system
(Chalazonitis, 2004; Frade et al., 1999). In the retina,
the expression and localization of both NT-3 and its
high affinity receptor TrkC were characterized at early 13.3.4 Hedgehog, Notch and Wnt
developmental stages, when RPC predominate (Das et
al., 2000). During retinal development, the disruption The first member of the hedgehog (Hh) family was
of NT-3 signaling by overexpression of a truncated described in Drosophila, but now highly conserved Hh
TrkC isoform produced a significant reduction in the proteins have been identified in various organisms. The
numbers of all retinal cell types, as well as decreased mammalian homologues are: Sonic hedgehog (Shh),
cell proliferation, suggesting that NT-3 targets RPC Indian hedgehog (Ihh) and Desert hedgehog (Dhh);
rather than differentiated cells (Das et al., 2000). and in zebrafish besides sonic hedgehog (shh), tiggy-
Similar to other growth factors, the inhibition of RPC winkle hedgehog (twhh) was also described. These
proliferation is not complete, consistent with in vitro signaling molecules are well known to regulate various
studies showing that RPC change their responsiveness phases of the development of different tissues in a vari-
to mitogenic signals in a spatial and temporal man- ety of organisms and are also related to the arousal of
ner during development, and that growth factors may tumors (Ingham and McMahon, 2001; Agathocleous et
act in combination (Anchan and Reh, 1995; Anchan al., 2007; Amato et al., 2004; Burke and Basler, 1996).
et al., 1991; Fischer et al., 2002; Fischer and Reh, In the retina, hedgehog proteins (Hh) are required
2003; Lillien and Cepko, 1992). At later stages, NT-3 for progression of the neurogenic wave, RPC
202 R. Linden et al.

proliferation, photoreceptor differentiation, and RGC not only maintains the progenitor state, but is required
axon growth (Russell, 2003). Hh acts as a mitogen in to inhibit the photoreceptor fate (Jadhav et al., 2006;
the developing mammalian retina (Jensen and Wallace, Yaron et al., 2006). Jadhav and colleagues (2006))
1997; Levine et al., 1997). It is expressed in mouse also showed that prolonged Notch activity in progeni-
retina by cells in the ganglion cell layer and inner tor cells maintains cells in the progenitor state without
nuclear layer, and produces an increased number of perturbing temporal identity, promoting early progen-
rod photoreceptors, amacrine cells, and Mller glial itor characteristics early in development and late pro-
cells (Jensen and Wallace, 1997). In fact, it was pro- genitor characteristics later in development. However,
posed that Hh in the retina accelerates both the G1 eventually, constitutive Notch activation led these cells
and G2 phases of the cell cycle, and then pushes these to acquire characteristics of glial and stem cells. And
rapidly dividing cells out of the cell cycle prematurely reactivating the Notch pathway in newly postmitotic
(Agathocleous et al., 2007), such that quiescent RPC retinal cells promoted mature glial cell formation in a
are converted into fast-cycling, transient amplifying subset of cells.
progenitors that are closer to both cell cycle exit and The role of Wnt in the regulation of the prolifer-
differentiation, similar to that observed in other tissues ation of progenitors in the marginal ciliary zone has
(Amato et al., 2004; Ingham and McMahon, 2001). also been discussed (Kubo et al., 2003; Kubo and
Mutations in the Hh and bone morphogenetic Nakagawa, 2008). In addition, activation the Wnt-
protein-4 (BMP4) signaling pathways can give rise to Frizzled signaling pathway leads to neurosphere for-
severe developmental defects, including anophthalmia- mation of adult ciliary margin stem cells (Ahmad et
microphthalmia (AM), retinal dystrophy, myopia, al., 2004). In adult mammalian retina, the presence of
brain anomalies, and polydactyly (Bakrania et al., injury could activate quiescent properties of RPC in
2008). Using in situ hybridization in human embryos, Mller cells in a process that involves activation of
expression of BMP4 was shown in the optic vesicle, both Wnt and Notch (Das et al., 2006).
developing retina and lens, pituitary region, and dig-
its. Reduced Hh signaling from the ventral forebrain is
likely to be the cause of cyclopia (Chan et al., 2007;
Muller et al., 2000). Moreover, it was demonstrated 13.3.5 Platelet Activating Factor
that knockdown of Hh expression slows or arrests rod
and cone photoreceptor differentiation (Stenkamp et Platelet-activating factor (PAF) is the trivial
al., 2000). Thus, Hh seems to be required for prolif- name for the 1-O-alcyl-2-acetyl-sn-glycero-3-
eration and differentiation of several cell classes in the phosphorylcholine phospholipid, which elicits a wide
retina. range of physiological responses in a variety of cell
Activation of the Notch-Delta pathway may also be types, such as proliferation, differentiation, inflamma-
important for the maintenance of the undifferentiated tion and allergy (Izumi and Shimizu, 1995). Almost
state in RPC. These cells from both rodent embryo all signaling pathways triggered by PAF are mediated
(E14 and E18) and adult ciliary margin express Notch, by a G protein-coupled receptor and tyrosine kinases
whereas the expression of its ligand Delta increases (Dhar and Shukla, 1991, 1994; Thurston et al., 1993;
from E14 to adult (Ahmad et al., 2004; Bao and Cepko, Zhu and Shukla, 1993; Honda et al., 1994; Kuruvilla
1997), particularly in the outer neuroblastic layer, sim- et al., 1994; Liu et al., 1994).
ilar to the distribution of STAT3 (Shao-Min Zhang et PAF induced proliferation in human bone marrow,
al., 2005). Moreover, it was shown that conditional fibroblasts and cell lines such as endometrial adenocar-
removal of Notch1 early in development led to a reduc- cinome (HEC1A) and immature erythrocytes (K562)
tion in the size of the retina due to a decrease in (Ishii and Shimizu, 2000). Tumor cell lines both syn-
the number of progenitor cells and premature neu- thesize PAF and express its membrane receptor, thus
rogenesis. Additionally, ablation of Notch1 in early producing an autocrine proliferative effect together
progenitor cells led to enhanced cone photoreceptor with a paracrine effect in endothelial cells, which pro-
production, and ablation of Notch1 at later points led motes angiogenesis required for tumor establishment
to an almost exclusive production of rod photorecep- and development (Dupuis et al., 1997; Bussolati et al.,
tor cells. Altogether, these data suggest that Notch1 2000; Denizot et al., 2001; Montruchio et al., 2000).
13 Tissue Biology of Proliferation and Cell Death 203

PAF was shown to accumulate in the retina in days of neurogenesis (Prada et al., 1991), are the pre-
response to injury, such as ischemic proliferative dominant subpopulation of retinal cholinergic cells
retinopathy or diabetic retinopathy, and was impli- (Baughman and Bader, 1977), in addition to immature
cated in vincristine-induced experimental retinopathy horizontal cells that may transiently release ACh prior
and other retinal impairments (Hardy et al., 2005; de to synaptogenesis (Kim et al., 1998, 2000).
La Cruz et al., 1998; Doly et al., 1995; Cluzel et al., Acetylcholine (Ach) signals both ligand-gated
1995). It was also shown both that acetylcholine and ionotropic receptors and metabotropic G protein-
dopamine promote the production of PAF in immature coupled receptors, respectively known as nico-
cells of the embryonic chick retina (Bussolino et al., tinic (nAChR) and muscarinic receptors (mAChR).
1988, 1989). Activated nicotinic receptors allow sodium influx,
Both a PAF-like lipid and PAF surface receptor while activated mAChR lead either to the release
were found in the developing rat retina, and treatment of Ca2+ from IP3 -sensitive intracellular stores or to
with exogenous PAF partially inhibited interkinetic the production of cAMP by adenylyl cyclase (AC)
nuclear migration of proliferating cells pulse-labeled (Albuquerque et al., 1995; van Koppen and Kaiser,
with BrdU, whereas both the total number of prolif- 2003). Both nAChR and mAChR are expressed in the
erating cells and the incorporation of nucleotide were developing retina of various species. In chick retina,
preserved. The reduced number of cell nuclei that nAChR mRNA is present by E4.5 (Hamassaki-Britto
reached the outer stratum following treatment with et al., 1994), but both the expression and functional
PAF had a heterochromatic pattern of labeling for studies performed in mammals indicate that expres-
BrdU, suggesting incorporation at the end of S phase, sion of nicotinic receptors may be restricted to dif-
at which time the dividing cells may have escaped ferentiating cells (Wong, 1995; Pearson et al., 2002).
a period of sensitivity to the lipid. The velocity of Muscarinic AChRs are present in both the avian and
interkinetic nuclear migration in cells that passed the mammalian retina at the start of neurogenesis (Wong,
S/G2 transition was, however, the same in either PAF- 1995; McKinnon and Nathanson, 1995; Pearson et al.,
treated or control retinal tissue. The results strongly 2002). Imaging studies have shown that, as early as E3
suggest the induction by PAF of an S/G2 checkpoint, in the chick and E20 in the rabbit, the activation of
resulting in blockade of interkinetic nuclear migration functional mAChR in retinal progenitor cells induces
in the developing retina. The finding that Mller cells changes in the concentration of intracellular calcium
produce abundant PAF suggest that glial cells may con- ([Ca2+ ]i ) (Yamashita and Fukuda, 1993; Yamashita
trol the cell cycle through lipid mediators, inclusive of et al., 1994; Wong, 1995; Pearson et al., 2002).
PAF (Fragel-Madeira et al., manuscript in preparation). Regulation of cell proliferation by ACh through the
activation of mAChR has been reported in a num-
ber of biological systems. Some studies suggested that
13.4 Control of the Retinal Cell Cycle muscarinic agonists induce cell proliferation in cul-
by Neurotransmitters tured glial cells (Cohen et al., 1996, Guizzetti et al.,
1996, 1998). However, the activation of mAChR in the
and Neuromodulators
developing chick retina negatively regulates the pro-
liferation of progenitor cells, by increasing the time
13.4.1 Classical Neurotransmitters necessary for a progenitor cell to divide during mitosis
(Pearson et al., 2002). Regulation of RPC proliferation
13.4.1.1 Acetylcholine by endogenous cholinergic activity was also observed
in cultures of immature rat retina. While muscarinic
Markers of cholinergic activity (e.g. choline acetyl antagonists led to a small increase in cell proliferation,
transferase and acetylcholine esterase) are expressed agonists reduced DNA synthesis and cell proliferation,
in the neuroblastic layer of the chick retina (Layer, corroborating previous evidence of a negative regula-
1991), and evidence have also been reported of the tion of cellular proliferation after activation of mAChR
expression of the cholinergic system around birthdate (dos Santos et al., 2003).
in mouse and ferret (Feller et al., 1996). Starburst Interestingly, exposure to either muscarinic ago-
amacrine cells, which are born during the first few nists or antagonists leads to smaller or larger eyes,
204 R. Linden et al.

respectively, in agreement with reported changes in the (Pellegrini-Giampietro et al., 1992). Metabotropic glu-
number of proliferating cells (Pearson et al., 2002). tamate receptors (mGluR) are grouped in three fami-
Also consistent with the demonstrated roles of ACh in lies differentially coupled to G-proteins: mGluR1 and
the regulation of RPC proliferation through mAChR, mGluR5 receptors are linked to the IP3 /Ca2+ signal-
the proportion of progenitor cells responding to mus- ing cascade (Abe et al., 1992; Aramori and Nakanishi,
carinic agonists, which is maximal during the peak 1992), while other classes of metabotropic receptors
of neurogenesis, declines rapidly, and is maintained inhibit the production of cAMP (Prezeau et al., 1994).
only in differentiating neurons of the inner retina Pioneering studies in the embryonic rabbit retina
(Yamashita et al., 1994; Wong, 1995; Pearson et al., demonstrated the expression of functional glutamate
2002). Although a growing number of studies demon- receptors during retinal development (Wong, 1995),
strate a role of muscarinic signaling in retinal develop- which was later confirmed in other species (Allcorn et
ment, it is still hard to distinguish between effects on al., 1996; Grunder et al., 2000; Martins et al., 2006).
either cell proliferation or cell differentiation, since the Wong showed that activation of glutamate receptors
molecular mechanisms that underlie the effects of ACh leads to an increase in [Ca2+ ]i in cells located in
in the regulation of retinal progenitor cell proliferation the neuroblastic layer, possibly retinal progenitors
are not completely understood. or recent post-mitotic cells. Interestingly, NMDA-
induced responses were restricted to differentiating
cells (Wong, 1995). In the chick retina, expression
of both AMPA/kainate subunits and the NR1 subunit
13.4.1.2 Glutamate of the NMDA receptor was reported as early as E5,
mostly in immature ganglion cells (Silveira dos Santos
In adult retinas, the excitatory neurotransmitter glu- and Hamassaki-Britto, 2001). Imaging analysis con-
tamate is the primary neurotransmitter of rod pho- firmed the expression studies, by showing that the
toreceptors, bipolar cells and ganglion cells, which induction of calcium influx by glutamate was restricted
comprise the vertical visual pathway (Thoreson and to post-mitotic cells (Pearson et al., 2002). In the rat
Witkovsky, 1999; Yang, 2004; Shen et al., 2006). retina, the NR1 subunit of the NMDA receptor was
Glutamate is found in several areas of the developing first detected in ganglion cells at E21 and strong label-
CNS, including the retina (Fletcher and Kalloniatis, ing was detected in ganglion, amacrine and horizontal
1997; Miranda-Contreras et al., 1998, 1999, 2000), cells by P3 (Grunder et al., 2000). Other NMDA recep-
where glutamate was detected by immunohistochem- tor subunits (NR2A, NR2B and NR3A) have been
istry in cells of the neuroblastic layer in both rat detected with similar patterns (Hartveit et al., 1994;
and rabbit. Immunoreactivity was first detected around Sucher et al., 2003). All AMPA/ Kainate receptor sub-
birth in the rat, and during late embryonic development units (GluR1, 2, 3 and 4) are expressed in ganglion and
(E20) in rabbit retinas (Fletcher and Kalloniatis, 1997; amacrine cells by late embryonic development (E20)
Pow et al., 1994). It is possible that progenitor cells in the rat retina (Grunder et al., 2000). Expression of
may release glutamate into the retinal environment, but NMDA receptor subunit NR1 and AMPA receptor sub-
since glutamate can be synthesized via several bio- units GluR1 and GluR5 was also reported in retinal
chemical pathways, it is not trivial to determine the progenitor cells, as verified by co-labeling in cells that
precise source of this neurotransmitter. incorporated 3 [H]-thymidine, in both embryonic and
Glutamate signals through both ionotropic and postnatal mouse retina (Martins et al., 2006).
metabotropic receptors. Ionotropic receptors are clas- Activation of glutamatergic receptors has been
sified in either NMDA receptors, permeable to Na+ , shown to regulate cell proliferation in various areas
K+ and Ca2+ , or AMPA/Kainate receptors, perme- of the developing CNS. Interestingly, both stimulatory
able to Na+ and K+ , but with low permeability to and inhibitory effects of glutamate receptor activa-
Ca2+ . However, an edited version of the subunit tion upon proliferation of neuronal progenitor cells
GluR2 confers Ca2+ permeability to AMPA recep- were demonstrated. While in the cerebral cortex, glu-
tors (Murphy and Miller, 1989; Pruss et al., 1991; tamate has an anti-proliferative effect through the
Burnashev et al., 1992), and this Ca2+ -permeable AMPA/kainate receptors (LoTurco et al., 1995; Haydar
AMPA receptor is prevalent during early development et al., 2000), in developing striatum, activation of
13 Tissue Biology of Proliferation and Cell Death 205

NMDA receptor induced proliferation of progenitor CDK2/4 was observed, a decrease in the kinase activ-
cells (Luk et al., 2003). ity of CDK2 was demonstrated after activation of
Glutamatergic regulation of proliferation of retinal glutamate receptors (Martins et al., 2006). In agree-
progenitor cells was studied in various species. In the ment with previous studies reporting induction of cell
developing chick retina, activation of glutamate recep- cycle exit by cell-extrinsic factors (Nagahara et al.,
tors did not change the time retinal progenitors spent 1999), a decrease in the phosphorylation of CDK2
to complete mitosis, and no change in the eye size threonine 160 was observed. These results indicate
was observed after in vivo modulation of glutamatergic a post-transcriptional mechanism for the glutamate-
signaling (Pearson et al., 2002). An anti-proliferative induced cell cycle exit.
role for glutamate upon mouse retinal progenitor cells Modulation of cell proliferation in response to
was recently described. It was shown that retinal pro- glutamate-induced neuronal injury has been demon-
genitor cells express AMPA/Kainate receptor subunits, strated in adult retina (Dyer and Cepko, 2000; Fischer
and that activation of these receptors induced cell and Reh, 2001). These studies demonstrated the induc-
cycle exit with no effect upon cell death. Lineage tion of proliferation of Mller and microglia cells, in
analysis studies after pharmacological blockade of response to in vivo overstimulation of glutamatergic
AMPA/kainate receptors, and overexpression of domi- receptors in the chick and mouse eyes. Expression
nant negative isoforms of GluR1 receptor, suggested a of glutamatergic receptors was previously reported in
cell-autonomous role for glutamatergic signaling in the Mller glial cells (Grunder et al., 2000) and recent
regulation of proliferation of progenitor cells during studies suggest that direct activation of these recep-
development of the mouse retina (Fig. 13.4, Martins et tors by sub toxic concentrations of glutamate would
al., 2006). However, it cannot be ruled out that activa- directly trigger cell proliferation of Mller glial cells
tion of glutamate receptors may regulate the expression (Takeda et al., 2008). However, it cannot be ruled out
of other anti-proliferative extrinsic factors. The molec- that glial cell proliferation in response to excitotoxic
ular mechanisms underlying glutamate-induced cell insults in the adult retina could be a consequence of
cycle exit were also examined in this study. Even secondary events that may take place in retinal tissue
though no difference in the expression of various cell after neuronal injury.
cycle regulators, such as cyclin D1, p27Kip1 , p57Kip2 ,

A C 50 untreated D 50 untreated

IRE S nuclear LacZ


glutamate NBQX
LTR LTR
40 40
p=7x10
% clones

% clones

B 30 30
1
2
4 3
5 20 20
1 6
7
8
10 10
9

0 0
1 2 3 >4 1 2 3 >4
clone size (cells/clone) clone size (cells/clone)

Fig. 13.4 Modulation of neurotransmitter signaling and Graphs show the proportion of clones containing the indicated
its effects upon clonal expansion of retinal progenitor cells number of cells. Pharmacological activation (c) or blockade
(RPC). (a) Retinal explants were infected with a replication- (d) of glutamate receptors had opposite effects upon RPC
incompetent retrovirus NIN-E, that encodes nuclear-localized clonal expansion. Treatment with the AMPA/Kainate recep-
-galactosidase. (b) After 10 days in culture, E14.5 mouse tor antagonist NBQX (100 M) decreased the proportion of
retinal explants were stained with X-gal and sectioned. The one-cell clones. Consistently, the proportion of clones con-
clonal expansion of RPC after the modulation of glutamater- taining four or more cells was increased after inhibition of
gic signaling was determined by quantifying the number non-NMDA receptor signaling (Mod from Martins et al.,
of clones and the number of cells in each clone. (c, d) 2006)
206 R. Linden et al.

13.4.1.3 GABA and Glycine is also indirectly coupled to Ca2+ and K+ channels
(North et al., 1987; Hill, 1985)
The classical pathway of synthesis of GABA is through Expression of functional GABA receptors in the
decarboxylation of glutamate by the enzyme glutamic developing retina has been extensively reported in
acid decarboxylase (GAD). However, an alternative a number of species (Huang and Redburn, 1996;
pathway of GABA synthesis was demonstrated, in Mitchell and Redburn, 1996; Mitchell et al., 1999;
which ornithine decarboxylase (ODC) is the limiting Greka et al., 2000; Catsicas and Mobbs, 2001; Wu
synthesizing enzyme and putrescine is used as a sub- and Cutting, 2001; Pearson et al., 2002). However, it
strate (Yamasaki et al., 1999). In the developing chick is still not clear if these receptors are expressed in
retina, GABA uptake and immunostaining for GABA- retinal progenitor cells or in cells that have already
positive neuroblast-like cells have been reported as left the cell cycle. In the mouse retina, mRNA for
early as E6 (Frederick, 1987; Hokoc et al., 1990), the GABAA receptor subunit 6 was detected in reti-
which confirmed the presence of this neurotransmitter nal progenitor cells during embryonic development,
in the embryonic retinal environment. Elegant stud- but the expression of a functional receptor in these
ies of thymidine incorporation revealed that GAD- cells was not determined (Young and Cepko, 2004).
expressing neurons (amacrine and horizontal cells), In the chick retina, it has been suggested that GABA-
first detected around E10 (Hokoc et al., 1990), are pre- evoked Ca2+ response is largely restricted either to
dominantly born between E6 and E9 (da Costa Calaza immature neurons in the ganglion cell layer (GCL) and
et al., 2000). In rats, GABA is detected in the devel- inner nuclear layer (INL) or to migrating post-mitotic
oping retinal tissue around birth (Versaux-Botteri et cells, rather than progenitor cells (Catsicas and Mobbs,
al., 1989; Fletcher and Kalloniatis, 1997). Although 2001; Pearson et al., 2002).
the precise onset of GAD immunoreactivity in the Glycine receptors are ligand-gated channels that
developing rat retina is controversial (see: Yamasaki form both homo- and heteropentamers comprised of
et al., 1999 and Dkhissi et al., 2001), the presence of multiple combinations of alpha (1-4) and beta ()
GABA in the retinal environment before the detection subunits. Activation of glycine receptors in the mature
of GAD-immunopositive interneurons in the chick, nervous system leads to Cl- influx. However, similar to
and most likely in the rat retina, indicates that the GABA, activation of glycinergic receptors also induces
ODC-mediated pathway of GABA synthesis plays an membrane Ca2+ influx in the developing nervous sys-
important role in vertebrate retinal development. tem. In addition to glycine, other amino acids, such as
GABA signals through both ionotropic and -alanine, L-alanine, L-serine, proline and taurine can
metabotropic receptors. Ligand-gated ion channels also activate glycinergic receptors.
include GABAA and GABAC , while GABAB receptor Expression of glycine receptors in developing
is a G-protein coupled metabotropic receptor (see retina has been shown in both rat and mouse.
Rudolph et al., 2001; Yang, 2004). GABA is widely Immunostaining for glycine receptors appears as dif-
accepted as an inhibitory neurotransmitter, because fuse staining in the neuroblastic layer during early
in the mature nervous system, activation of GABAA postnatal development of the rat retina. However, this
receptors leads to an influx of chloride ions (Cl- ) observation does not directly demonstrate expression
through the receptor channel. However, during devel- of glycine receptors in retinal progenitor cells. Later in
opment, GABA acts as an excitatory neurotransmitter development, glycine receptor expression is restricted
(see Ben-Ari, 2002 for review). A delay in the matura- to the INL (Sassoe-Pognetto and Wassle, 1997).In the
tion of the Cl- extrusion system relative to Cl- uptake mouse, transcripts for the alpha2 subunit (GlyR2)
mechanisms results in a developmental window in were first detected at birth in the outer half of the
which the concentration of [Cl- ]i is higher than that neuroblastic layer (Young and Cepko, 2004).
found in the extracellular environment (Nishi et al., A pioneering study by Altshuler and colleagues
1974; Zhang et al., 1991). During this period, GABAA demonstrated that taurine was the factor within retinal-
receptor activation and the consequent increase in Cl conditioned medium responsible for inducing rod
conductance and Cl efflux result in an increase in cell photoreceptor fate (Altshuler et al., 1993). Taurine
excitability. On the other hand, activation of GABAB is present at high levels in developing nervous
receptors inhibits cAMP production. This receptor system (Sturman, 1988; Lombardini, 1991) and may
13 Tissue Biology of Proliferation and Cell Death 207

signal either by entering the cells through a trans- 13.4.1.5 Dopamine


porter or through the activation of glycine or GABA
receptors (for review, see: Tapaz, 2004). Recently, Dopamine is expressed by a subpopulation of amacrine
Young and Cepko reported that endogenous taurine cells, but is also present at very early stages in
present within developing retinal tissue activates both the developing retinal environment (Martin-Martinelli
GABAergic and glycinergic pathways in developing et al., 1989). The identification of dopamine-producing
mouse retina (Young and Cepko, 2004). In addition, cells may be made by the detection of the limiting
this study demonstrated that pharmacological block- enzyme for its synthesis, tyrosine hydroxylase (TH).
ade of either glycine or GABA receptors prevented the In the rat, TH immunoreactivity was detected in late
rod-promoting effect of taurine. embryonic to early postnatal development (E19-P2)
However, the regulation of retinal progenitor cell (Nguyen-Legros et al., 1983; Wu and Cepko, 1993),
proliferation by GABA and glycine during develop- by P3 in the rabbit (Osborne et al., 1984; Casini and
ment appears to differ between species. Lineage anal- Brecha, 1992) and in the chick at the equivalent period
ysis experiments in which glycine receptor GlyR2 of development (E12) (Gardino et al., 1993). In addi-
was overexpressed indicated that glycinergic signaling tion, it was recently demonstrated that dopamine may
induces retinal progenitor cells to exit the cell cycle be produced via a TH-independent pathway. In the
(Young and Cepko, 2004). In contrast, in the devel- chick retina, the enzyme DOPA decarboxylase (DDC),
oping chick retina GABA receptor activation would which was first detected as early as E8, is able to pro-
not directly affect retinal progenitor cell proliferation. duce dopamine from L-DOPA before TH expression
Even though endogenous activation of GABA recep- is detected. Only in E18, DDC was clearly identi-
tors appears to be responsible for generating sponta- fiable in dopaminergic amacrine cells. On the other
neous [Ca2+ ]i transients in cells located at the NBL hand, around E8, expression of DDC was found in
of the embryonic chick retina (Pearson et al., 2002), the immature outer plexiform layer (Kubrusly et al.,
GABA-responding cells were predominantly interpha- 2003). Recently, it was reported that DDC is expressed
sic cells that expressed the early neuronal marker, - in the neuroblastic layer and INL of P0 rat retinas
III-tubulin (Tuj1), suggesting that, in the chick, GABA (Kralj-Hans et al., 2006), suggesting that retinal pro-
only signals postmitotic cells. In addition, GABA had genitors and/or post-mitotic cells may be a source of
little effect on the time taken by retinal progenitors of dopamine synthesized from L-DOPA. It is, neverthe-
the developing chick retina to progress through mitosis less, not yet known if this TH-independent pathway for
(Pearson et al., 2002). dopamine synthesis is present at more immature stages
of mammalian retinal development.
Dopamine acts through the activation of specific
G-protein coupled receptors, classified in two sub-
13.4.1.4 Adrenergics families, D1-like (receptors D1 and D5) and D2-
like (D2, D3 and D4). The former are coupled to
The expression of adrenergic neurotransmitters and protein Gs , while the latter are usually coupled to
receptors was previously reported in both the devel- Go or Gi proteins. The distribution of D1 receptor
oping and the adult retina (Hadjiconstantinou et al., (D1R) immunoreactivity was investigated during post-
1984; Zarbin et al., 1986; Shelke et al., 1997; Lograno natal development of the rat retina and this receptor
et al., 2000; Woldemussie et al., 2007). In addition, was found already at birth (Koulen, 1999). Kralj-
the activity of the epinephrine biosynthetic enzyme, Hans and colleagues (2006) confirmed the pattern
phenylethanolamine N-methyltransferase (PNMT) observed around P0, and also detected D1R expres-
(Cohen, 1987), as well as the activation of specific sion in the neuroblastic layer, which suggests that
signaling pathways by norepinephrine have been it may be expressed in either progenitors or recent
reported during retinal development (Kubrusly et al., post-mitotic cells. D2R in rat retinas is mainly asso-
2007). However, no study has as yet demonstrated ciated with extrasynaptic membranes of dopaminergic
either the expression of functional adrenergic recep- cells, as well as in processes of non-dopaminergic
tors in retinal progenitor cells or the regulation of amacrine cells. Derouiche and Asan (1999) also sug-
proliferation of progenitor cells by manipulation of gested that D4R is the D2-like receptor expressed in
adrenergic signaling in developing retinal tissue. photoreceptors.
208 R. Linden et al.

Some studies have already suggested a role for and 5-HT7 were found in adult rat retina (Pootanakit
dopamine in the regulation of cell proliferation in and Brunken, 2000). In adult rabbit retina, mRNA for
the retinal tissue. It was reported that, in albino rats, 5-HT1a, 5-HT2a, 5-HT3a, 5-HT3b and 5-HT7 were
dopamine increased the time retinal progenitor cells detected, and immunohistochemical assays demon-
take to complete mitosis, and reduced the number of strated protein expression of 5-HT2a and 5-HT3
progenitors arriving in the proliferative zone, whereas (Pootanakit et al., 1999; Pootanakit and Brunken,
the same effects were not observed in pigmented reti- 2001).
nas (Kralj-Hans et al., 2006). The interpretation of Conversely, De Lucchini et al. (2003) reported that,
these findings may be either that dopaminergic signal- in developing Xenopus retina, 5-HT2B and 5-HT2C
ing in developing retina regulates cell-cycle length, as receptors are expressed at, comparatively, much earlier
suggested by the authors, or that the reduction in the stages in both the proliferative zone (stage 40) and in
number of proliferating cells at the proliferative zone the ciliary margin zone (stage 45), the latter of which
may also be due to regulation of migration of retinal is a region at the anterior margin of the retina in some
progenitor cells. vertebrates, where it is believed new neurons are gen-
In another study, in vivo administration of L-DOPA erate throughout life. Both transcripts were expressed
was shown to regulate cell division and the orienta- in BrdU incorporating (i.e., proliferating) cells, and
tion of cleavage in developing rat retina (Tibber et al., expression of 5-HT2B was maintained in post-mitotic
2006). L-DOPA is believed to act through its conver- cells located in the INL layer (De Lucchini et al.,
sion to dopamine by DDC. Although it was suggested 2003).
that L-DOPA may have neurotransmitter activity inde- Evidence for the role of serotonin upon the pro-
pendent of its conversion, L-DOPA-specific receptors liferation of retinal progenitor cells was obtained in
have not been found. For example, treatment with L- Xenopus. In this study, a combination of pharmaco-
DOPA in vitro decreased the number of mitotic figures logical and genetic manipulations was used to demon-
in embryonic rat retinas (Ilia and Jeffery, 1999) and strate that signaling through 5-HT2B receptors ontrols
it was not determined if this effect was dependent on both cell proliferation and cell death in developing
dopamine synthesis. Xenopus retina, with impact in morphogenesis of the
These studies suggest an antiproliferative role for eye (De Lucchini et al., 2005). Overexpression of 5-
dopamine in developing rat retina. Testing how the HT2B, followed by treatment with serotonin, increased
modulation of endogenous dopaminergic signaling the number of mitotic profiles. Conversely, pharma-
regulates the proliferation of retinal progenitor cells cological inhibition with ritanserin and morpholinos
will be important to confirm the developmental role targeting 5-HT2B receptors resulted in smaller eyes
of this neurotransmitter. An alternative proposal is that and disorganization of retinal cytoarchitecture. In addi-
some extracellular factors may otherwise specifically tion, these morpholinos reduced the expression of
regulate cell-cycle length, but not cell-cycle exit, as cyclin D1. These results strongly suggest a mitogenic
was shown for Shh in the developing Xenopus retina role for serotonin in developing Xenopus retina. Data
(Locker et al., 2006). are still lacking to determine whether cyclin D1 is
required for the mitogenic effects of serotonin in vivo,
and whether this role for serotonin is conserved among
13.4.1.6 Serotonin species.

Serotonin (5-hydroxytryptamine, 5-HT) acts through


the activation of 5-HT receptor subtypes. At least thir- 13.4.1.7 ATP
teen different receptor subtypes, grouped into seven
families (5HT1- 5HT7), have now been described Both neurons and glia are potential sources of extracel-
(Hoyer et al., 2002). lular ATP. In the adult CNS, ATP is released together
The expression of 5-HT1b was detected by in situ with several other neurotransmitters such as ACh, glu-
hybridization in both developing and adult mouse tamate and GABA, and it is reasonable to suggest
retina (Upton et al., 1999), apparently restricted to that the same may happen in the developing retina. To
the ganglion cell layer. Transcripts for both 5-HT1a date, ATP release has been demonstrated in cultures of
13 Tissue Biology of Proliferation and Cell Death 209

isolated amacrine cells (Santos et al., 1999), Mller Sanches et al. (2002) found that adenosine had no
glia (Newman, 2001) and embryonic chick RPE effect on the proliferation of embryonic chick retinal
(Pearson et al., 2005). cells in culture, although an earlier report described a
ATP and its related nucleotides activate both small reduction in [3 H]-thymidine incorporation, sug-
ionotropic and metabotropic receptors. P2X receptors gesting an inhibitory effect on proliferation, in chick
are ligand-gated ion channels, permeable to Na+ , K+ retinal explant cultures (Sugioka et al., 1999b).
and Ca2+ and P2Y receptors are G-protein coupled On the other hand evidence accumulates on the
receptors, predominantly coupled to the IP3 /Ca2+ cas- effects of adenosine in cell survival (Fredholm, 1997;
cade, although some isoforms are linked with adenylyl Dunwiddie and Masino, 2001), and it may have simi-
cyclase/cAMP. ATP receptors are among the earli- lar actions in the developing retina. For instance, when
est functional receptors (Laasberg, 1990), but due to purified cultures of immature retinal neurons are re-
limited availability of research tools, such as specific fed with fresh medium they display an intense cell
antibodies, the precise onset of the multiple purinergic death, an effect that can be greatly reduced when the
receptor subunits during development of the vertebrate cultures are pre-incubated with adenosine (reviewed in
retina is still unclear. Imaging studies provided evi- Paes-de-Carvalho, 2002).
dence of the expression of functional ATP receptors
in the embryonic chick retina. ATP-induced increase
in [Ca2+ ]i through the activation of P2Y receptors was
the largest at early developmental stages (E3), and 13.4.2 Neuropeptides
was predominantly observed in retinal progenitor cells,
rather than immature neurons (Pearson et al., 2002, Various neuropeptides and/or their receptors have
2005; Sakaki et al., 1996; Sugioka et al., 1996). already been described in the developing reti-
nal tissue of mammals, such as substance P
and other tachykinins, somatostatin, neuropeptide
13.4.1.8 Adenosine Y, corticotrophin releasing factor and related pep-
tides, angiotensin, opioid peptides, pituitary adenylyl
G-protein-coupled receptors (GPCRs) for adenosine cyclase-activating polypeptide and vasoactive intesti-
were initially classified as A1 and A2, which inhibit nal peptide (see for review Bagnoli et al., 2003).
and stimulate cAMP accumulation, respectively. Later However, evidence is scarce concerning the role of
these receptors were subclassified, and A2-adenosine neuropeptides in the control of proliferation in retinal
receptors were termed A2A, to designate the recep- progenitor cells.
tors that required low concentrations of adenosine and Several opioid peptides were detected in the mam-
adenosine analogues for the stimulation of Adenylyl malian retina. Early studies detected the expression
Cyclase. A second subtype was named A2B, which of [Met5 ]-enkephalin, preproenkephalin mRNA and
also signals via Gs . The two other receptors, called A1- [Met5 ]-enkephalin binding sites in the NBL and in
and A3-adenosine receptors, interact with pertussis- the GCL of early postnatal as well as embryonic
toxin-sensitive G proteins of the Gi and Go family rat retinas (Isayama et al., 1991, 1995), and pre-
(Zezula and Freissmuth, 2008). proenkephalin mRNA was found in the INL of adult
Both A1 and A2 adenosine receptors are present rat retinas (Isayama et al., 1996). Moreover, opioid
since early stages of chick retina development (Paes- receptors (MOP-R) were detected in ganglion cells of
de-Carvalho, 2002). In immature mammalian retina it rats and monkeys, and receptors for enkephalins were
was shown the expression of A2a receptors in ganglion described in guinea pig and developing human reti-
cells and in the INL during retinal development in dogs nas (Wamsley et al., 1981; Altschuler et al., 1982;
(Taomoto et al., 2000). The expression of purinergic Yew et al., 1991; Jotwani et al., 1994). In addition,
neurotransmitters early in retinal development raised Makman and Dvorkin (1997) also showed binding for
the possibility that they might modulate proliferation nociceptin (orphanin FQ) in the retina of adult rats.
and/or differentiation. Limited data suggested a role for [Met5 ]-enkephalin
However, the potential roles of adenosine in the in the modulation of cell proliferation (Isayama et al.,
control of proliferation have not been fully described. 1991). [Met5 ]-enkephalin presented a subtle effect of
210 R. Linden et al.

a 10% reduction in the labeling index, defined by et al., 1997, 1998), as well as in all layers in early
the number of [3 H]-thymidine positive cells 4 h after postnatal rat retina (Silveira et al., 2002).
injection in postnatal day 1 (P1) rats. Although the Interestingly, it has been shown that glucagon-
co-injection of the antagonists naloxone or naltrexone expressing neurons in the retina may have a role in
both reverted the effect of [Met5 ]-enkephalin, only nal- the regulation of the proliferation of progenitor cells
trexone induced an increase in the labeling index when in the CMZ of the postnatal chicken eye (Fischer
injected alone (Isayama et al., 1991). et al., 2005). Glucagon is a neuropeptide highly con-
Neuropeptide Y (NPY) was also shown as a poten- served among species that belong to the same family of
tial modulator of the proliferation of retinal progen- PACAP, and was reportedly expressed by a subpopula-
itors in rat retinal cultures (lvaro et al., 2008). tion of amacrine cells in the chicken retina (Tornqvist
Immunoreactivity for NPY was detected in amacrine et al., 1981; Kuwayama et al., 1982; Tornqvist and
and displaced amacrine cells in mammalian retinas. In Ehinger, 1983; Ekman and Tornqvist, 1985), although
addition, in some species, such as cat and human, NPY its roles are not precisely known. It was suggested that
was also found in ganglion cells (Oh et al., 2002). In glucagon antagonizes the role of insulin or insulin-like
the retina of the rat, NPY-positive cells appear in the growth factor in the regulation of the proliferation of
GCL at late prenatal stages, and a transient increase in retinal progenitors in the CMZ, to control de addition
the expression of this peptide was found close to the of new cells to the edge of the retina (Fischer et al.,
time of eye opening (Ferriero and Sagar, 1989). 2005).
Alvaro and collaborators (2007) showed that NPY
and NPY receptors were expressed in differentiated
cells in retinal cultures from P3-P5 rats, but the expres-
sion pattern of NPY earlier in retinal development is
13.5 Signal Transduction in the Extrinsic
not available. The same group has recently suggested
a role for NPY in the stimulation of proliferation, Control of the Retinal Cell Cycle
which involved the participation of nitric oxide, in
cultures of retinal cells from the same developmen- Extracellular mediators, including neurotransmitters,
tal stage mentioned above (Alvaro et al., 2008). In activate a number of second messengers and signal-
this study, treatment of dissociated cell cultures for ing pathways that may act upon the retinal cell cycle
48 h with NPY increased in almost 65% the num- (Martins and Pearson, 2008 for review). For the most
ber of BrdU+ -nestin+ positive cells. This prolifera- part, however, little is known of signal transduction
tive effect was reverted by NO synthase as well as triggered by extracellular modulators of the cell cycle
soluble guanylyl cyclase inhibitors, and it was sug- in the nervous system.
gested to depend on ERK1/2 activation (Alvaro et al., Transients of intracellular calcium are required for
2008). progression through several steps in the proliferative
An anti-mitogenic role was identified for pituitary cell cycle including the G1/S-phase transition, S phase,
adenylyl cyclase-activating polypeptide (PACAP) in entry into mitosis and key points within mitosis includ-
the neonatal rat retina (Njaine et al., 2009). In this ing the metaphaseanaphase transition and induction
study, we found that mRNA for PACAP precursor is of cytokinesis (Short et al., 1993; Berridge, 1995;
present in rat retina from embryonic day 19 (E19), Santella et al., 1998; Whitaker and Larman, 2001).
while mRNA for the PACAP-specific PAC1 recep- For exemple, P2Y receptor stimulation leads to the
tor was detected from E16 (Njaine et al., 2009). activation of the IP3 /Ca2+ second messenger cascade
Previously, reports of expression of PACAP were (Sugioka et al., 1999a; Pearson et al., 2002; Sanches
restricted to adult rat retinas in the ganglion cell, inner et al., 2002). This leads to the activation of phospho-
plexiform and inner nuclear layers (Seki et al., 1998), lipase C (PLC), activation of PKC and stimulation of
and in fetal human retinas at 1218 weeks of gestation the ERK/MAPK cascade (Sanches et al., 2002; Nunes
(Olianas et al., 1997). On the other hand, expres- et al., 2007). The ERK/MAPK pathway is usually
sion of receptors for PACAP was reported in ganglion associated with stimulation of cell-cycle progression,
and amacrine cells, innerplexiform layer, outer nuclear primarily by relieving cell-cycle blockades at the step
layer and outer plexiform layer in adult rat retinas (Seki between G1 and S-phase (Sherr, 1996). Noteworthy,
13 Tissue Biology of Proliferation and Cell Death 211

evidence was gathered that factors released by PI3K include protein kinases, such as PDKs, Akt and
postmitotic cells may interfere with the sensitivity of S6 kinase, and regulators of small GTPases (Rodgers
the P2 receptors (Frana et al., 2007), which suggests and Theibert, 2002; Cantrell, 2001; Wymann and
tissue constraints upon the regulation of the cell cycle Pirola, 1998 for reviews). Recently, it was reported that
of retinal progenitor cells by purinergic receptors. ATP, acting via P2YR, might promote the prolifera-
A number of extracellular mediators act via either tion of neural stem cells through the PI3K-dependent
stimulatory or inhibitory G-protein-coupled receptors pathway (Ryu et al., 2003; Heo and Han, 2006).
to modulate adenylyl cyclase and the conversion of The expression of signal transducers and activators
ATP to cAMP. An important downstream effector of transcription (STATs) were modulated specifically
of cAMP is protein kinase A (PKA), which phos- by some growth factors, and STAT3 was activated in a
phorylates a wide array of target proteins that reg- restricted spatial pattern in the presence of aFGF, cil-
ulate diverse cellular functions. For example, phos- iary neurotrophic factor (CNTF), leukemia inhibitory
phorylation of the transcription factor CREB (cAMP factor (LIF), and interferon- (IFN-), but not by
Response Element Binding protein) regulates the bFGF, EGF, IFN-, and retinoic acid (RA) (Shao-
expression of various genes involved in cell survival Min Zhang et al., 2005). Since activated STAT3 co-
and synaptic plasticity (Tao et al., 1998; Patterson localized with incorporation of BrdU, it is likely that
et al., 2001). Recent data suggest that neurotrans- STAT3 is directly involved in maintaining RPC in
mitters may act via cAMP to regulate the expres- mammalian retina. In fact, it was shown that STAT3
sion of cell cycle molecules in cultured mouse stem acts in the transition from G1 to S phase of the cell
cells. Thus, dopamine receptor activation led to a cycle, through up-regulation of cyclins D2, D3 and
decrease in DNA synthesis and reduced expression of A, and cdc25A, with simultaneous down-regulation of
cyclins and CDK (Lee et al., 2006). Activation of the p21 and p27Kip1 (Fukada et al., 1998), which agrees
cAMP/PKA pathway inhibited the cell cycle exit of with studies that indicated a pivotal role for STAT3
zebrafish retinoblasts (Masai et al., 2005). Forskolin in the maintenance of the undifferentiated state and
treatment, which increases the levels of cAMP, com- pluripotency of embryonic stem cells in the CNS
pletely inhibits the generation of new post-mitotic cells (Bauer and Patterson, 2006; Niwa et al., 1998).
in the neural retina. Thus, in developing zebrafish Regarding the effects of PAF upon the retinal cell
retina activation of PKA causes retinal progenitor cells cycle, the inhibition of either p38 or p42/44 MAPKs,
to remain in the cell cycle, although the molecular but not of PKC, prevented the inhibition of interkinetic
mechanisms that underlie this effect are not clear. One nuclear migration by PAF. PAF also induced activa-
possibility is that PKA inhibits the expression of the tion of Chk1, which is likely responsible for the cell
CDK inhibitor p27Kip1 . Alternatively, a major PKA cycle arrest at the S/G2 transition, because a pharma-
substrate, CREB, binds to the promoter and may reg- cological inhibitor of Chk1 prevented the effect of PAF
ulate the expression of cell-cycle regulators such as (Fragel-Madeira et al., unpublished).
cyclin D1 (Lonze and Ginty, 2002). In contrast with Finally, other protein kinases have been implicated
the data observed in zebrafish, cAMP/PKA antago- in the cell cycle or in interkinetic nuclear migration,
nizes the proliferative role of Shh in other areas of the although upstream pathways have not been identified.
nervous system in mammals (Waschek et al., 2006). Thus, in the developing rat retina, MAP kinases of
In addition, PKA-mediated regulation of WEE kinases the p38 family were shown to regulate the metaphase-
and cdc25 phosphatase activities was also reported (for anaphase transition within M-phase (Campos et al.,
reviews, see Shibuya, 2003; Han and Conti, 2006). We 2002), whereas CK2 was implicated in various phases
have also found that activation of PKA is involved in of the retinal cell cycle, including motor aspects of
the anti-proliferative effect of interleukin-4 upon the interkinetic nuclear migration (Fig. 13.5, Carneiro et
developing retina, associated with increased expres- al., 2008).
sion of p27kip1 and reduced cyclin D1 (Silva et al., Therefore, several signal transduction pathways
2008). have already been implicated in the control of the reti-
Another important intracellular signaling path- nal cell cycle by neuroactive substances. Nonetheless,
way activated by extracellular mitogens involves the the definition of the relevant pathways is still fragmen-
enzyme PI3-kinase (PI3K). Downstream targets of tary.
212 R. Linden et al.

mechanisms that modulate cell death among early neu-


ral progenitors. It has been shown that developing
photoreceptors undergo progressive changes in mech-
anisms of programmed cell death, associated with the
transition from the proliferative state to the stage of
phenotypic differentiation. Thus, in the developing
A B rat retina, although the degeneration of both photore-
ceptors and proliferating/immediate postmitotic cells
C 125 induced by various agents depended on protein syn-
3H-TDR INCORPORATION (% control)

100
thesis, an increase of intracellular cyclic AMP blocked
degeneration of postmitotic, but not of proliferat-
75 ing photoreceptor precursors (Chiarini et al., 2003).
Recently, it was shown that neuroprotection provided
50 by insulin in the chick retina is distinctly mediated
by either the MAP kinase or the PI3-kinase signal-
25
ing pathways in either proliferating or postmitotic
differentiating cells (Chavarria et al., 2007).
CTR 1 3 10 AraC On the other hand, despite the reports that the den-
TBB (M)
sity of pyknotic profiles in the neuroblastic layer of the
Fig. 13.5 Role of CK2 in the retinal cell cycle. (a, b) chick retina, prior to all but the genesis of ganglion
Distribution of BrdU-labeled nuclei within the NBL (see Fig. cells, is of the same order of magnitude as for post-
13.1), at 3 h of incubation of retinal explants made 30 min mitotic ganglion cells (de la Rosa and de Pablo, 2000;
after intraperitoneal injection of the nucleotide analogue, show-
ing normal interkinetic nuclear migration (a), and its blockade
Chavarria et al., 2007), it is as difficult to estimate the
by 10 M 4,5,6,7-tetrabromobenzotriazole (TBB), an inhibitor true rates of cell death among proliferating neural pro-
of CK2 activity (b). (c) TBB also inhibits incorporation of genitor cells as it is for postmitotic neurons (Linden
tritiated thymidine into retinal tissue, indicating that CK2 activ- and Reese, 2006). One of the major reasons for the
ity is required in S-phase. AraC = cytosine-arabinofuranoside.
Control experiments showed that the blockade of nuclear migra-
lack of reliable estimates of the absolute magnitude
tion is not a consequence of the blockade of DNA synthesis of developmental cell death is the multiplicity of cell
(Mod. from Carneiro et al., 2008) death programs available to the developing cells (Fig.
13.6, Guimares and Linden, 2004), which cast doubts
on the estimates usually made on the basis of counting
pyknotic profiles in sections of the developing nervous
system (Linden and Reese, 2006).
13.6 Death and Survival of Retinal
Nonetheless, some evidence is available of factors
Progenitor Cells that affect the sensitivity of RPC to programmed cell
death depending on the phase of the cell cycle. Both the
Extensive work has been reported on naturally- major cell death programs and evidence for differential
occurring cell death during development of the nervous sensitivity to the induction of cell death among distinct
system. It is well established that programmed cell phases of the cell cycle will be reviewed in this section.
death occurs not only among differentiating neurons
forming synaptic connections, but also among neural
progenitor cells (de la Rosa and de Pablo, 2000; Lossi
and Merighi, 2003; Buss et al., 2006). 13.6.1 Mechanisms of Cell Death
Notwithstanding several earlier studies that demon-
strated both the occurrence and extrinsic control of 13.6.1.1 Apoptosis
retinal progenitor cell death (Silver and Robb, 1979;
Cuadros and Rios, 1988; Martin-Partido et al., 1988; Apoptosis, or programmed cell death type I, has been
Frade et al., 1996b, 1997; Diaz et al., 1999, 2000), extensively studied due to its importance for the devel-
only recently has evidence appeared on intracellular opment of multicellular organisms, homeostasis of
13 Tissue Biology of Proliferation and Cell Death 213

PCD Type III (Necrosis)


Loss of membrane integrity
PCD Type II (Autophagy) Cell swelling
Degradation of organeles Organelle swelling
PCD Type I (Apoptosis)
No change in cell size No DNA laddering
Cell shrinkage
No DNA laddering
Blebbing
Partial chromatin condensation
Chromatin condensation
Numerous autophagic vesicles
DNA laddering
Nuclear fragmentation
Membrane integrity maintained

Fig. 13.6 Major types of programmed cell death. Expanding progenitor cells in specific circumstances. Cornification has been
the classical distinction between apoptosis, as programmed cell recognized as a fourth major type of programmed cell death by a
death, and necrosis, as accidental cell death (Kerr et al., 1972), nomenclature committee (Kroemer et al., 2008), but is exclusive
current studies support at least 3 major types of programmed of upper epidermal layers and unlikely to affect progenitor cells
cell death (Guimares and Linden, 2004), all of which have been in any tissue
detected in the nervous system, and are likely to affect neural

adult tissues, as well as human diseases. It is acknowl- Apoptosis has, nonetheless, been more recently
edged as part of a natural process to remove damaged recognized as a cell death process that could
cells during metazoan development, and to sculp- occurs through caspase-dependent or independent
ture tissues and organs, a good example of which is pathways. DNA condensation and fragmentation can
the formation of digits (Chen and Zhao, 1998; Kerr, occur by a caspase-independent pathway. The best-
2002; Borges et al., 2008, Domingos and Steller, 2007; studied mechanism include the nuclear translocation of
Degterev and Yuan, 2008). Besides physiological con- Apoptosis-Inducing Factor (AIF), a flavoprotein local-
ditions, apoptosis is one of the pathways by which ized in the mitochondrial intermembrane space that is
damaged cells are eliminated in pathological circum- conserved in eukaryotic cells from yeasts to humans
stances, or following genotoxic stress such as in cancer (Lorenzo et al., 1999; Cande et al., 2002). Cleavage
treatment. and release of AIF from mitochondria is regulated by
The term apoptosis was coined by Kerr in 1972 to calpain I. Calpains are cystein proteases that are acti-
define an active, organized cell death process involv- vated by autoprocessing due to intracellular calcium
ing membrane blebbing, shrinkage of the cytoplasm increase. Calpain I and II (micro- and milli-calpain) are
and nucleus, which generate pyknotic features. Then, expressed in the retina and are activated by micromolar
apoptotic cells disintegrate into apoptotic bodies sur- and millimolar calcium concentrations, respectively.
rounded by an intact cell membrane that are engulfed Activation of calpain has been associated with cell
by surrounded cells and macrophages. Apoptosis is death in retinal degeneration models (Marigo, 2007).
often accompanied by biochemical markers such as The caspase-dependent machinery was revealed
DNA fragmentation, mitochondrial membrane perme- first from studies of developmentally programmed cell
abilization (MMP), and activation of aspartate-specific death in the nematode C. elegans (Domingos and
cystein-dependent proteases called caspases (Kerr, Steller, 2007; Kumar, 2007) and is evolutionarily con-
2002; DSa et al., 2003; Kumar, 2007). served (Liang et al., 2008; Low et al., 2008; Low
214 R. Linden et al.

and Yang, 2008). Mice lacking either caspase-3 or regulators, including the retinoblastoma protein (Rb)
caspase-9 show an excess of cells in the develop- (Chau et al., 2002), which are substrates involved in the
ing nervous system, as a consequence of decreased regulation of apoptosis. For example, the developing
rates of apoptosis among immature post-mitotic neu- ganglion cell layer that expresses a mutant RB, resis-
rons and progenitor cells. In addition, mice deficient tant to caspase cleavage, shows less cell death induced
in molecules that are important for caspase 9 activa- by axonal damage (Chau et al., 2002). Retinal progen-
tion, such as cytochrome c or Apaf-1, also show an itors, however, do not depend on RB cleavage for cell
excess of cells in nervous tissue (Kuida et al., 1996; death induced by ionizing radiation (Chau et al., 2002).
Cecconi et al., 1998; Hakem et al., 1998; Kuida et al., Inhibition or deletion of caspases will delay or block
1998; Yoshida et al., 1998; Kuan et al., 2000; Hao cell death, showing the central role of caspases on the
et al., 2005). These data support an important role of apoptotic process. Nevertheless, cells will often find an
caspases at the core of the cell death pathways in the alternative way to die by other mechanism (Guimares
nervous system. and Linden, 2004).
How caspases are activated will depend on the apop-
totic stimuli. In summary, if cell death is triggered by
extrinsic factors, such as tumor necrosis factor (TNF) 13.6.1.2 Autophagy
or Fas ligand (FasL), transmembrane receptors will
activate caspase 8, which will activate downstream cas- Autophagy is an evolutionarily conserved, homeo-
pase 3, to execute cell death (Jin and El-Deiry, 2005; static process that allows for lysosomal-dependent
Kim, 2005). If apoptosis is triggered by an intrinsic degradation of long-lived proteins and organelles
damage, such as DNA damage, caspase activation will (Tasdemir et al., 2008). Under either starvation or
usually depend on activation of p53 (Hurley and Bunz, stress, autophagy provides a cell protective func-
2007; Lavin and Kozlov, 2007; Borges et al., 2008). tion. However, it can also induce cell death if over-
The tumor suppressor p53 is a downstream target of stimulated (Guimares and Linden, 2004; Galluzzi
the DNA damage-signaling network, which involves et al., 2008). Strategies for discriminating cell death
apical kinases such as ataxia-telangiectasia mutated despite autophagic attempts at adaptation from cell
(ATM), and downstream kinases such as Chk2. ATM death through autophagy are a current focus of discus-
and Chk2 phosphorylate p53, respectively at serines 15 sion (Tasdemir et al., 2008).
and 20, leading to its stabilization and activation of its Autophagic cell death is characterized by mas-
transcriptional function (Hurley and Bunz, 2007; Lavin sive accumulation of large autophagic vacuoles called
and Kozlov, 2007). autophagosomes, derived from part of the endoplas-
Among p53 induced genes, the pro-apoptotic mem- mic reticulum. The contents of autophagosomes are
bers of the B-cell lymphoma protein-2 (Bcl-2) family, degraded by lysosomal enzymes after autophagosomes
such as Puma, Noxa, Bax and Bak, are major players fuse with lysosomes (Guimares and Linden, 2004;
to induce cell death. Puma (Yu and Zhang, 2003; Roos Kim, 2005; Degterev and Yuan, 2008; Galluzzi et
and Kaina, 2006) and Noxa (Roos and Kaina, 2006) al., 2008; Tasdemir et al., 2008). In addition to the
will activate Bax or Bak to cause cytochrome c release presence of the double-membrane vesicular organelles,
from the mitochondria (Abu-Qare and Abou-Donia, autophagic cell death can show partial chromatin con-
2001). Cytochrome c binds to and stimulates apop- densation, but neither cell shrinkage nor DNA ladder-
totic protease-activating factor-1 (Apaf-1) to assemble ing are observed (Henriquez et al., 2008).
the apoptosome, leading to the activation of caspase Autophagic and apoptotic processes are intercon-
9 (Hajra and Liu, 2004; Riedl and Salvesen, 2007). nected in several ways. For example, molecules in
Active caspase 9 will then activate caspase 3 in a the apoptotic pathways are also related to autophagy,
feedback loop to increase caspase activation, and to i.e. the p53 protein, as stated above. Another exam-
cleave cellular substrates that will lead to morpho- ple is the autophagy-specific gene Bcn-1, which is
logical and biochemical characteristics of apoptosis identified as a novel member of the pro-apoptotic
(Enari et al., 1998; Sahara et al., 1999). Moreover, cas- BH3-only family of Bcl-2 proteins (Henriquez et al.,
pases cleave DNA repair enzymes and transcription 2008). Moreover, inducers of apoptosis can induce
13 Tissue Biology of Proliferation and Cell Death 215

autophagy and vice-versa (Ciechomska et al., 2008) 3-methyladenine (3-MA) increase light-induced
and the blockade of one type of cell death may necrosis of photoreceptors (Kunchithapautham and
increase the other (Ciechomska et al., 2008). For exam- Rohrer, 2007).
ple, the large-scale loss of differentiating cells, which These results changed the view of necrosis as
occurs during normal neural development and requires a passive form of degeneration and suggest that
classical apoptotic proteins (caspases and APF-1), it represent a backup pathway (Henriquez et al.,
can undergo caspase-independent cell death involv- 2008). Mechanisms of programmed necrosis, also
ing autophagy when apoptosis is impaired (Oppenheim known as necroptosis, are currently under investigation
et al., 2008). On the other hand, inhibition of (Degterev et al., 2005, Degterev and Yuan, 2008; Li et
autophagy sensitizes cells to caspase-dependent apop- al., 2008), but details remain to be elucidated.
tosis (Tasdemir et al., 2008).

13.6.1.3 Necrosis
13.6.2 Sensitivity to Cell Death Within
A typical necrotic cell is swollen, with enlarged the Retinal Cell Cycle
organelles and no DNA laddering is present. Moreover,
necrotic cells lose membrane integrity, which cause Sensitivity to cell death among proliferating cells
spillage of intracellular contents and triggers inflam- has been investigated in several models using ioniz-
mation. In tissue culture and in the absence of phago- ing radiation (IR). This stimulus will mostly induce
cytosis, apoptotic cells become secondarily necrotic, caspase-dependent apoptosis, which can be identified
which sometimes obscures studies of cell death. in vivo by the detection of TUNEL-positive condensed
Besides morphological criteria, however, there is no profiles. The use of ionizing radiation has additional
clear biochemical hallmark of necrotic cell death leav- advantages for the study of intrinsic sensitivity to DNA
ing characterization of negative markers for apoptosis damage during the cell cycle, since this physical agent
and autophagy (Krysko et al., 2008). penetrates tissue and even organisms independent on
Overwhelming physical (membrane damage, DNA the permeability of biologic membranes.
damage, loss of ion homeostasis) or chemical (tox- In several cell lines, the highest radioresistant stage
icity, ATP depletion) stress, incompatible with cell of the cell cycle is found in middle to late S phase
survival, causes necrosis. Consistent with this view, (Cheong et al., 1994; Biade et al., 1997). In the devel-
necrotic cell death is frequently detected under patho- oping retina, IR induced two waves of cell death in
logical conditions. For example, at the core of ischemic the NBL, and these were related with distinct phases
areas, necrotic figures are found, whereas apoptotic of the cell cycle. Approximately 60% of the apoptotic
features predominate at peripheral regions. In addi- profiles found at 6 hours after irradiation were neu-
tion to pathological conditions, necrosis occurs also roblasts, the nuclei of which are preferentially located
during development and represents a third type of at the outermost region of the proliferative zone. This
programmed cell death (Type III). corresponds to the expected location of the G2/M
Cell death has historically been subdivided into transition (Fujita, 1962), which is usually one of the
regulated (apoptosis) and unregulated mechanisms most radiosensitive phases of the cell cycle (Cheong
(necrosis) (Borges et al., 2008; Degterev and Yuan, et al., 1994). However, 40% of the early apoptotic
2008; Henriquez et al., 2008). Recent findings, cells were young post-mitotic cells, since they were
however, indicate that necrosis not only can be neither labeled with proliferation nor with cellular dif-
accidental but also programmed (Guimares and ferentiation markers (Borges and Linden, 1999). The
Linden, 2004; Degterev and Yuan, 2008; Henriquez second wave of apoptosis was observed 24 hr after
et al., 2008). Indeed, when both apoptosis and IR, close to the inner margin of the NBL, where the
autophagy are blocked, a necrotic form of cell death nuclei of cells in S phase are located. These two waves
ensues. For example, the simultaneous treatment with are likely independent, since an increase in lipid per-
pan-caspase-3 inhibitor and the autophagy inhibitor oxidation was detected at 6 hr, but not at 24 hr after
216 R. Linden et al.

-IRRADIATION

LATE APOPTOSIS (24h)

NEUROBLASTS IN S-PHASE

no increased lipid peroxidation


not rescued by PDTC
EARLY APOPTOSIS (6h)

-40% POST-MITOTIC CELLS


-60% PCNA+ CELLS (out of S-phase)

lipid peroxidadion
rescued by PDTC

Fig. 13.7 Two waves of programmed cell death induced that are in S-phase cells at the time of irradiation, either when
by gamma irradiation of retinal progenitor cells in S-phase. cells re-enter a second S-phase after irradiation, or following
An early wave of apoptosis kills both undifferentiated post- a prolonged intra-S checkpoint, and does not depend on lipid
mitotic cells and proliferating neuroblasts which were not in peroxidation. Both waves of apoptosis require ATM and phos-
S-phase at the time of irradiation, and depends on an increase phorylation of p53 in serine-18 (Mod from Borges and Linden,
in lipid peroxidation. A later wave of apoptosis kills neuroblasts 1999 and Borges et al., 2004, 2008).

IR. Accordingly, the antioxidant pyrrolidinedithiocar- 13.6.3 Molecular Mechanisms of Cell


bamate (PDTC) prevented the first, but not the second Death Among Retinal Progenitor
wave of apoptosis (Fig. 13.7, Borges and Linden,
Cells
1999). Therefore, the studies of IR-induced cell death
in the developing retina of rodents demonstrated that
the dying retinal cells could be grouped in at least Proliferating cells are preferential targets of several
two populations that exhibit spatially and temporally cytotoxic stimuli, among which blockers of cell cycle
distinct apoptotic responses, which are influenced by kinases and proteasome inhibitors. Pharmacological
developmental stage as well as by the phase of the blockers of cyclin-dependent kinases (CDK), such as
cell cycle (Borges and Linden, 1999; Borges et al., deferoxamine (DFO), mimosine (MIMO), and olo-
2004). moucine (OLO) induce cell death in retinal explants.
13 Tissue Biology of Proliferation and Cell Death 217

These cell cycle inhibitors induced cell death in the phosphorylation site of ATM in mice, comparable to
retinal neuroblastic layer (NBL), where DFO and serine 15 in humans), showed a response to IR similar
MIMO killed only proliferating cells, whereas OLO to Atm mutant mice (Borges et al., 2004). This result
affected both proliferating and undifferentiated post- suggests that major contribution of ATM to cell death
mitotic cells (Rehen et al., 2002). The inhibitor of is through direct activation of p53. Moreover, a lysine
proteasome function carbobenzoxyl-leucinyl-leucinyl- residue in mouse p53, Lys317, is also acetylated in
leucinal (MG132) induced cell death in a subset of vivo after DNA damage, and a knock-in mutation at
cells within the neuroblastic layer of the retinal tissue. this site indicated that acetylation negatively regulates
MG132-sensitive population included both proliferat- p53-induced apoptosis in the developing retina (Chao
ing cells, most likely in their last round of cell division, et al., 2006). Therefore, posttranslational modifications
and post-mitotic undifferentiated cells. These results of p53 can result in either positive or negative regula-
show that both retinal progenitors and retinal cells in tion of apoptosis. These findings further illustrate the
the transition from the cell cycle to the differentiated complex regulation of the DNA damage response.
state are particularly sensitive to cell death induced by Interestingly, despite caspase activation, in RPC
either proteasome or CDK inhibitors, thus defining a no impairment of DNA fragmentation following ion-
window of development with particular mechanism of izing radiation was observed in the presence of a
cell death (Neves et al., 2001; Rehen et al., 2002). pan-caspase inhibitor, indicating p53-dependent, but
Irradiation induces a complex death response in the caspase-independent pathways of cell death (Herzog
developing retinal tissue both in vivo and in vitro, et al., 2007).
involving progenitors at distinct phases of the cell In conclusion, cells in either distinct stage of dif-
cycle and stages of differentiation. Although these ferentiation or distinct phases of the cell cycle show
waves showed different sensitivity to lipid peroxida- diverse sensitivity to the same death stimulus. This
tion (Borges and Linden, 1999), they share similar selective sensitivity either reflects upon the timing of
mechanism of execution of apoptosis regarding the cellular demise, or defines a window of development
DNA damage-activated network. Genetic studies have with selective sensitivity to specific mechanisms of cell
shown that the ATM-p53 pathway is necessary for death. In addition, it is now clear that cell death can
both waves of programmed cell death (Borges et al., occur with a variety of morphologies and mechanisms,
2004). Genes of the ATM-p53 pathway, however, con- even when cells are exposed to the same stimulus. An
tributed differently, depending on the dose of IR used. extensive crosstalk does exist among the 3 major types
For instance, while the lack of p53 resulted in com- of programmed cell death.
plete resistance to both low and high doses (2 Gy and
14 Gy), deficiency of Atm conferred only partial resis-
tance, suggesting that other pathways converge upon
p53 to induce cell death. The defect of apoptosis in 13.7 Conclusion and Future Directions
p53 knockout mice is comparable to Puma-/- mice,
suggesting that p53-dependent cell death is mainly The data above show that the rate of prolifera-
meditated thought activation of Puma (Yu and Zhang, tion of progenitor cells in the developing retina is
2003). deeply affected by a variety of signals from neigh-
ATM can contribute to activation of p53 both boring cells. Some of these signals modulate specif-
directly and indirectly, through activation of the Chk2 ically certain cell cycle transitions, and a few com-
protein kinase. Knockout mice for Chk2 are defec- ponents of signal transduction have been identified.
tive in apoptosis induced by 5-8 Gy of IR in all areas Nevertheless, most details are still missing as to
of the developing CNS examined (Takai et al., 2002). how the cell cycle is regulated by the barrage of
Thus, similar to p53, Chk2 is required for IR-induced incoming signals produced by differentiated and pro-
apoptosis. liferating cells in their vicinity. In turn, since the
The importance of several posttranslational mod- phase of the cell cycle also affects the sensitiv-
ifications in p53 was investigated in vivo by tak- ity to cell death, both the capacity to proliferate
ing advantage of knock-in mutations. For instance, as well as the survival of stem/progenitor cells are
knock-in mice p53-mutated at serine 18 (the direct likely to depend on the tissue environment, including
218 R. Linden et al.

both the identity and the activity of the various cell Altschuler RA, Mosinger JL, Hoffman DW, Parakkal MH
types. (1982) Immunocytochemical localization of enkephalin-like
immunoreactivity in the retina of the guinea pig. Proc Natl
Understanding the life cycle and the determinants
Acad Sci U S A 79:23982400.
of cell proliferation, differentiation and survival of Altshuler D, Lo Turco JJ, Rush J, Cepko C (1993) Taurine pro-
stem/progenitor cells is required for their successful motes the differentiation of a vertebrate retinal cell type in
application in novel therapeutic procedures. Further vitro. Development 119:13171328.
Alvaro AR, Martins J, Arajo IM, Rosmaninho-Salgado J,
studies of the tissue biology of stem/progenitor cells
Ambrsio AF, Cavadas C (2008) Neuropeptide Y stimulates
are thus warranted, to provide the basic knowledge retinal neural cell proliferation involvement of nitric oxide.
necessary for the safe development of cell therapies for J Neurochem 105:25012510.
retinal, as well as other neural degenerations. Alvaro AR, Rosmaninho-Salgado J, Santiago AR, Martins J,
Aveleira C, Santos PF, Pereira T, Gouveia D, Carvalho AL,
Grouzmann E, Ambrsio AF, Cavadas C (2007) NPY in rat
retina is present in neurons, in endothelial cells and also in
References microglial and Mller cells. Neurochem Int 50:757763.
Amato MA, Boy S, Perron M (2004) Hedgehog signaling in ver-
tebrate eye development: a growing puzzle. Cell Mol Life Sci
Abbas T, Dutta A (2006) CDK2-activating kinase (CAK): more 61:899910.
questions than answers. Cell Cycle 5:11231124. Anchan RM, Reh TA (1995) Transforming growth factor-beta-
Abe T, Sugihara H, Nawa H, Shigemoto R, Mizuno N, 3 is mitogenic for rat retinal progenitor cells in vitro.
Nakanishi S (1992) Molecular characterization of a novel J Neurobiol 28:133145.
metabotropic glutamate receptor mGluR5 coupled to inos- Anchan RM, Reh TA, Angello J, Balliet A, Walker M (1991)
itol phosphate/Ca2+ signal transduction. J Biol Chem EGF and TGF-alpha stimulate retinal neuroepithelial cell
267:1336113368. proliferation in vitro. Neuron 6:923936.
Abraham RT (2001) Cell cycle checkpoint signaling through the Aramori I, Nakanishi S (1992) Signal transduction and pharma-
ATM and ATR kinases. Genes Dev 15:21772196. cological characteristics of a metabotropic glutamate recep-
Abu-Qare AW, Abou-Donia MB (2001) Biomarkers of apop- tor, mGluR1, in transfected CHO cells. Neuron 8:757765.
tosis: release of cytochrome c, activation of caspase-3, Bagnoli P, Dal Monte M, Casini G (2003) Expression of neu-
induction of 8-hydroxy-2 -deoxyguanosine, increased 3- ropeptides and their receptors in the developing retina of
nitrotyrosine, and alteration of p53 gene. J Toxicol Environ mammals. Histol Histopathol 18:12191242.
Health B Crit Rev 4:313332. Bakrania P, Efthymiou M, Klein JC, Salt A, Bunyan DJ, Wyatt
Adler R (1986) Developmental predetermination of the struc- A, Ponting CP, Martin A, Williams S, Lindley V, Gilmore
tural and molecular polarization of photoreceptor cells. Dev J, Restori M, Robson AG, Neveu MM, Holder GE, Collin
Biol 117:520527. JR, Robinson DO, Farndon P, Johansen-Berg H, Gerrelli D,
Agathocleous M, Locker M, Harris WA, Perron M (2007) A gen- Ragge NK (2008) Mutations in BMP4 cause eye, brain, and
eral role of hedgehog in the regulation of proliferation. Cell digit developmental anomalies: overlap between the BMP4
Cycle 6:156159. and hedgehog signaling pathways. Am J Hum Genet 82:
Ahmad I, Das AV, James J, Bhattacharya S, Zhao X (2004) 304319.
Neural stem cells in the mammalian eye: types and regula- Bao ZZ, Cepko CL (1997) The expression and function of
tion. Semin Cell Dev Biol 15:5362. Notch pathway genes in the developing rat eye. J Neurosci
Ahmad I, Tang L, Pham H (2000) Identification of neural pro- 17:14251434.
genitors in the adult mammalian eye. Biochem Biophys Res Bartek J, Lukas J (2001) Mammalian G1 and S-phase check-
Commun 270:517521. points in response to DNA damage. Curr Opin Cell Biol
Ahmed S, Reynolds BA, Weiss S (1995) BDNF enhances the 13:738747.
differentiation but not the survival of CNS stem cell-derived Bartek J, Lukas J (2007) DNA damage checkpoints: from
neuronal precursors. J Neurosci 15:57655778. initiation to recovery or adaptation. Curr Opin Cell Biol
Akagi T, Haruta M, Akita J, Nishida A, Honda Y, Takahashi M 19:238245.
(2003) Different characteristics of rat retinal progenitor cells Bauer S, Kerr BJ, Patterson PH (2007) The neuropoietic
from different culture periods. Neurosci Lett 341:213216. cytokine family in development, plasticity, disease and
Albuquerque EX, Pereira EF, Castro NG, Alkondon M, injury. Nat Rev Neurosci 8:221232.
Reinhardt S, Schroder H, Maelicke A (1995) Nicotinic recep- Bauer S, Patterson PH (2006) Leukemia inhibitory factor pro-
tor function in the mammalian central nervous system. Ann motes neural stem cell self-renewal in the adult brain.
N Y Acad Sci 757:4872. J Neurosci 26:1208912099.
Alexiades MR, Cepko CL (1996) Quantitative analysis of pro- Baughman RW, Bader CR (1977) Biochemical characterization
liferation and cell cycle length during development of the rat and cellular localization of the cholinergic system in the
retina. Dev Dyn 205:293307. chicken retina. Brain Res 138:469485.
Allcorn S, Catsicas M, Mobbs P (1996) Developmental expres- Baye LM, Link BA (2008) Nuclear migration during retinal
sion and self-regulation of Ca2+ entry via AMPA/KA recep- development. Brain Res 1192:2936.
tors in the embryonic chick retina. Eur J Neurosci 8: Beazley LD, Perry VH, Baker B, Darby JE (1987) An inves-
24992510. tigation into the role of ganglion cells in the regulation of
13 Tissue Biology of Proliferation and Cell Death 219

division and death of other retinal cells. Dev Brain Res Cam H, Dynlacht BD (2003) Emerging roles for E2F: beyond
33:169184. the G1/S transition and DNA replication. Cancer Cell 3:
Ben-Ari Y (2002) Excitatory actions of gaba during develop- 311316.
ment: the nature of the nurture. Nat Rev Neurosci 3(9): Campos CB, Bdard PA, Linden R (2002) Activation of p38
72839. mitogen-activated protein kinase during normal mitosis in
Berridge MJ (1995) Calcium signalling and cell proliferation. the developing retina. Neuroscience 112:583591.
Bioessays 17:491500. Cande C, Cecconi F, Dessen P, Kroemer G (2002) Apoptosis-
Biade S, Stobbe CC, Chapman JD (1997) The intrinsic radiosen- inducing factor (AIF): key to the conserved caspase-
sitivity of some human tumor cells throughout their cell independent pathways of cell death? J Cell Sci 115:
cycles. Radiat Res 147:416421. 47274734.
Borges HL, Chao C, Xu Y, Linden R, Wang JY (2004) Cantrell DA (2001) Phosphoinositide 3-kinase signalling path-
Radiation-induced apoptosis in developing mouse retina ways. J Cell Sci 114:14391445.
exhibits dose-dependent requirement for ATM phosphoryla- Carneiro AC, Fragel-Madeira L, Silva-Neto MA, Linden R
tion of p53. Cell Death Differ 11:494502. (2008) A role for CK2 upon interkinetic nuclear migration
Borges HL, Linden R (1999) Gamma irradiation leads to two in the cell cycle of retinal progenitor cells. Dev Neurobiol
waves of apoptosis in distinct cell populations of the retina 68:620631.
of newborn rats. J Cell Sci 112:43154324. Casini G, Brecha NC (1992) Postnatal development of tyro-
Borges HL, Linden R, Wang JY (2008) DNA damage-induced sine hydroxylase immunoreactive amacrine cells in the rabbit
cell death: lessons from the central nervous system. Cell Res retina: I. Morphological characterization. J Comp Neurol
18:1726. 326:283301.
Bovolenta P, Frade JM, Marti E, Rodriguez-Pena MA, Barde Catsicas M, Mobbs P (2001) GABAb receptors regulate chick
YA, Rodriguez-Tebar A (1996) Neurotrophin-3 antibod- retinal calcium waves. J Neurosci 21:897910.
ies disrupt the normal development of the chick retina. Cayouette M (2007) Expanding neuronal layers by the local
J Neurosci 16:44024410. division of committed precursors. Neuron 56:575577.
Branzei D, Foiani M (2006) The Rad53 signal transduction Cayouette M, Poggi L, Harris WA (2006) Lineage in the verte-
pathway: replication fork stabilization, DNA repair, and brate retina. Trends Neurosci 29:563570.
adaptation. Exp Cell Res 312:26542659. Cayouette M, Raff M (2003) The orientation of cell division
Branzei D, Foiani M (2008) Regulation of DNA repair through- influences cell-fate choice in the developing mammalian
out the cell cycle. Nat Rev Mol Cell Biol 9:297308. retina. Development 130:23292339.
Bringmann A, Pannicke T, Grosche J, Francke M, Wiedemann P, Cecconi F, Alvarez-Bolado G, Meyer BI, Roth KA, Gruss P
Skatchkov SN, Osborne NN, Reichenbach A (2006) Mller (1998) Apaf1 (CED-4 homolog) regulates programmed cell
cells in the healthy and diseased retina. Prog Retin Eye Res death in mammalian development. Cell 94:727737.
25:397424. Cepko CL (1999) The roles of intrinsic and extrinsic cues and
Bull ND, Johnson TV, Martin KR (2008) Stem cells bHLH genes in the determination of retinal cell fates. Curr
for neuroprotection in glaucoma. Prog Brain Res 173: Opin Neurobiol 9:3746.
511519. Cepko CL, Austin CP, Yang X, Alexiades M, Ezzeddine D
Burdak-Rothkamm S, Rothkamm K, Prise KM (2008) ATM acts (1996) Cell fate determination in the vertebrate retina. Proc
downstream of ATR in the DNA damage response signaling Natl Acad Sci U S A 93:589595.
of bystander cells. Cancer Res 68:70597065. Chalazonitis A (2004) Neurotrophin-3 in the development of the
Burke R, Basler K (1996) Hedgehog-dependent patterning in the enteric nervous system. Prog Brain Res 146:243263.
Drosophila eye can occur in the absence of Dpp signaling. Chan A, Lakshminrusimha S, Heffner R, Gonzalez-Fernandez F
Dev Biol 179:360368. (2007) Histogenesis of retinal dysplasia in trisomy 13. Diagn
Burnashev N, Monyer H, Seeburg PH, Sakmann B (1992) Pathol 2:4849.
Divalent ion permeability of AMPA receptor channels is Chao C, Wu Z, Mazur SJ, Borges H, Rossi M, Lin T, Wang
dominated by the edited form of a single subunit. Neuron JY, Anderson CW, Appella E, Xu Y (2006) Acetylation of
8:189198. mouse p53 at lysine 317 negatively regulates p53 apop-
Buss RR, Sun W, Oppenheim RW (2006) Adaptive roles of totic activities after DNA damage. Mol Cell Biol 26:
programmed cell death during nervous system development. 68596869.
Annu Rev Neurosci 29:135. Chau BN, Borges HL, Chen TT, Masselli A, Hunton
Bussolati B, Biancone L, Cassoni P, Russo S, Rola-Pleszczynski IC, Wang JY (2002) Signal-dependent protection from
M, Montrucchio G, Camussi G (2000) PAF produced by apoptosis in mice expressing caspase-resistant Rb. Nat Cell
human breast cancer cellspromotes migration and prolif- Biol 4: 757765.
eration of tumor cells and neo-angiogenesis. Am J Pathol Chavarra T, Valenciano AI, Mayordomo R, Egea J, Comella JX,
157:17131725. Hallbk F, de Pablo F, de la Rosa EJ (2007) Differential,
Bussolino F, Pescarmona GP, Camussi G, Gremo F (1988) age-dependent MEK-ERK and PI3K-Akt activation by
Acetylcholine and dopamine promote the production of insulin acting as a survival factor during embryonic retinal
Platelet-activating factor in immature cells of chick embry- development. Dev Neurobiol 67:17771788.
onic retina. J Neurochem 51:17551759. Chen Y, Zhao X (1998) Shaping limbs by apoptosis. J Exp Zool
Bussolino F, Torelli S, Stefanini E, Gremo F (1989) Platelet- 282:691702.
activating factor production occurs through stimulation of Chenn A, McConnell SK (1995) Cleavage orientation and
cholinergic and dopaminergic receptors in the chick retina. the asymmetric inheritance of Notch1 immunoreactivity in
J Lipid Mediat 1:283288. mammalian neurogenesis. Cell 82:631641.
220 R. Linden et al.

Cheong N, Wang X, Wang Y, Iliakis G (1994) Loss of S-phase- Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima
dependent radioresistance in irs-1 cells exposed to X-rays. N, Cuny GD, Mitchison TJ, Moskowitz MA, Yuan J (2005)
Mutat Res 314:7785. Chemical inhibitor of nonapoptotic cell death with thera-
Chiarini LB, Leal-Ferreira ML, de Freitas FG, Linden R (2003) peutic potential for ischemic brain injury. Nat Chem Biol
Changing sensitivity to cell death during development of 1:112119.
retinal photoreceptors. J Neurosci Res 74:875883. Degterev A, Yuan J (2008) Expansion and evolution of cell death
Cicero SA, Johnson D, Reyntjens S, Frase S, Connell S, Chow programmes. Nat Rev Mol Cell Biol 9:378390.
LM, Baker SJ, Sorrentino BP, Dyer MA (2009) cells previ- Del Bene F, Wehman AM, Link BA, Baier H (2008) Regulation
ously identified as retinal stem cells are pigmented ciliary of neurogenesis by interkinetic nuclear migration through an
epithelial cells. Proc. Natl Acad Sci USA. 106(16):6685 apical-basal notch gradient. Cell 134:10551065.
6690. Denizot Y, Desplat V, Drouet M, Bertin F, Melloni B (2001)
Ciechomska IA, Goemans CG, Tolkovsky AM (2008) Molecular Is there a role of platelet-activating factor in human lung
links between autophagy and apoptosis. Methods Mol Biol cancer? Lung Cancer 33:195202.
445:175193. Derouiche A, Asan E (1999) The dopamine D2 receptor sub-
Cluzel J, Doly M, Bazan NG, Bonhomme B, Braquet P (1995) family in rat retina: ultrastructural immunogold and in situ
Inhibition of platelet-activating factor-induced retinal impair- hybridization studies. Eur J Neurosci 11:13911402.
ments by cholera and pertussis toxins. Ophthalmic Res Dhar A, Shukla SD (1991) Involvement of pp60c-src in
27:153157. platelet-activating factor-stimulated platelets. Evidence for
Cohen J (1987) Postnatal development of phenylethanolamine- translocation from cytosol to membrane. J Biol Chem 266:
N-methyltransferase activity of rat retina. Neurosci Lett 1879718801.
83:138142. Dhar A, Shukla SD (1994) Electrotransjection of pp60v-src
Cohen RI, Molina-Holgado E, Almazan G (1996) Carbachol monoclonal antibody inhibits activation of phospholipase C
stimulates c-fos expression and proliferation in oligodendro- in platelets. A new mechanism for platelet-activating factor
cyte progenitors. Brain Res Mol Brain Res 43:193201. responses. J Biol Chem 269:91239127.
Cortez D (2003) Caffeine inhibits checkpoint responses with- Dhomen NS, Balaggan KS, Pearson RA, Bainbridge JW, Levine
out inhibiting the ataxia-telangiectasia-mutated (ATM) and ED, Ali RR, Sowden JC (2006) Absence of chx10 causes
ATM- and Rad3-related (ATR) protein kinases. J Biol Chem neural progenitors to persist in the adult retina. Invest
278:3713937145. Ophthalmol Vis Sci 47:386396.
Cuadros MA, Ros A (1988) Spatial and temporal correla- Dimaras H, Coburn B, Pajovic S, Gallie BL (2006) Loss of
tion between early nerve fiber growth and neuroepithelial p75 neurotrophin receptor expression accompanies malig-
cell death in the chick embryo retina. Anat Embryol 178: nant progression to human and murine retinoblastoma. Mol
543551. Carcinog 45:333343.
da Costa Calaza K, Hokoc JN, Gardino PF (2000) Neurogenesis Dkhissi O, Julien JF, Wasowicz M, Dalil-Thiney N, Nguyen-
of GABAergic cells in the chick retina. Int J Dev Neurosci Legros J, Versaux-Botteri C (2001) Differential expression
18:721726. of GAD(65) and GAD(67) during the development of the rat
Dagnino L, Fry CJ, Bartley SM, Farnham P, Gallie BL, Phillips retina. Brain Res 919:242249.
RA (1997) Expression patterns of the E2F family of tran- Doly M, Cluzel J, Bonhomme B, Millerin M, Braquet P (1995)
scription factors during mouse nervous system development. Protective effect of a specific PAF antagonist on vincristine-
Mech Dev 66:1325. induced experimental retinopathy. Acta Ophthalmol Scand
Das AV, Mallya KB, Zhao X, Ahmad F, Bhattacharya S, 73:155157.
Thoreson WB, Hegde GV, Ahmad I (2006) Neural stem cell Domingos PM, Steller H (2007) Pathways regulating apoptosis
properties of Mller glia in the mammalian retina: regulation during patterning and development. Curr Opin Genet Dev
by Notch and Wnt signaling. Dev Biol 299:283302. 17:294299.
Das I, Sparrow JR, Lin MI, Shih E, Mikawa T, Hempstead BL Donovan SL, Dyer MA (2005) Regulation of proliferation dur-
(2000) Trk C signaling is required for retinal progenitor cell ing central nervous system development. Semin Cell Dev
proliferation. J Neurosci 20:28872895. Biol 16:407421.
de la Cruz JP, Moreno A, Ruiz-Ruiz MI, Garcia-Campos J, Donovan SL, Schweers B, Martins R, Johnson D, Dyer MA
Sanchez de la Cuesta F (1998) Effect of WEB 2086- (2006) Compensation by tumor suppressor genes during reti-
BS, an antagonist of platelet-activating factor receptors, on nal development in mice and humans. BMC Biol 4:1415.
retinal vascularity in diabetic rats. Eur J Pharmacol 360: dos Santos AA, Medina SV, Sholl-Franco A, de Araujo EG
3742. (2003) PMA decreases the proliferation of retinal cells
de la Rosa EJ, de Pablo F (2000) Cell death in early neural devel- in vitro: the involvement of acetylcholine and BDNF.
opment: beyond the neurotrophic theory. Trends Neurosci Neurochem Int 42:7380.
23:454458. Dunwiddie TV, Masino SA (2001) The role and regulation of
De Lucchini S, Ori M, Cremisi F, Nardini M, Nardi I (2005) adenosine in the central nervous system. Annu Rev Neurosci
5-HT2B-mediated serotonin signaling is required for eye 24:3155.
morphogenesis in Xenopus. Mol Cell Neurosci 29:299 312. Dupuis F, Levasseur S, Jean-Louis F, Dulery C, Praloran V,
De Lucchini S, Ori M, Nardini M, Marracci S, Nardi I (2003) Denizot Y, Michel L (1997) Production, metabolism and
Expression of 5-HT2B and 5-HT2C receptor genes is associ- effect of platelet-activating factor on the growth of the human
ated with proliferative regions of Xenopus developing brain K562 erythroid cell line. Biochim Biophys Acta 1359:
and eye. Mol Brain Res 115:196201. 241249.
13 Tissue Biology of Proliferation and Cell Death 221

Dyer M, Cepko CL (2000a) p57(Kip2) regulates progenitor cell Florian C, Langmann T, Weber BH, Morsczeck C (2008)
proliferation and amacrine interneuron development in the Murine Mller cells are progenitor cells for neuronal cells
mouse retina. Development 127:35933605. and fibrous tissue cells. Biochem Biophys Res Commun
Dyer MA, Cepko CL (2000b) Control of Mller glial cell prolif- 374:187191.
eration and activation following retinal injury. Nat Neurosci Fontaine RH, Cases O, Lelivre V, Mespls B, Renauld JC,
3:873880. Loron G, Degos V, Dournaud P, Baud O, Gressens P (2008)
Dyer M, Cepko CL (2001a) p27Kip1 and p57Kip2 regulate IL-9/IL-9 receptor signaling selectively protects cortical neu-
proliferation in distinct retinal progenitor cell populations. rons against developmental apoptosis. Cell Death Differ
J Neurosci 21:42594271. 15:15421552.
Dyer MA, Cepko CL (2001b) Regulating proliferation during Frade JM, Bovolenta P, Martnez-Morales JR, Arribas A, Barbas
retinal development. Nat Rev Neurosci 2:333342. JA, Rodrguez-Tbar A (1997) Control of early cell death by
Daz B, Pimentel B, de Pablo F, de la Rosa EJ (1999) Apoptotic BDNF in the chick retina. Development 124:33133320.
cell death affecting proliferating neuroepithelial cells in the Frade JM, Bovolenta P, Rodriguez-Tebar A (1999)
embryonic retina is prevented by insulin. Eur J Neurosci Neurotrophins and other growth factors in the generation of
11:16241632. retinal neurons. Microsc Res Tech 45:243251.
Daz B, Serna J, De Pablo F, de la Rosa EJ (2000) In vivo regu- Frade JM, Marti E, Bovolenta P, Rodriguez-Pena MA, Perez-
lation of cell death by embryonic (pro)insulin and the insulin Garcia D, Rohrer H, Edgar D, Rodriguez-Tebar A (1996a)
receptor during early retinal neurogenesis. Development Insulin-like growth factor-I stimulates neurogenesis in chick
127:16411649. retina by regulating expression of the alpha 6 integrin sub-
DSa C, Klocke BJ, Cecconi F, Lindsten T, Thompson CB, unit. Development 122:24972506.
Korsmeyer SJ, Flavell RA, Roth KA (2003) Caspase reg- Frade JM, Rodrguez-Tbar A, Barde YA (1996b) Induction of
ulation of genotoxin-induced neural precursor cell death. cell death by endogenous nerve growth factor through its p75
J Neurosci Res 74:435445. receptor. Nature 383:166168.
Ekman R, Tornqvist K (1985) Glucagon and VIP in the retina. Frana GR, Freitas RC, Ventura AL (2007) ATP-induced pro-
Invest Ophthalmol Vis Sci 26:14051409. liferation of developing retinal cells: regulation by factors
Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, released from postmitotic cells in culture. Int J Dev Neurosci
Nagata S (1998) A caspase-activated DNase that degrades 25:283291.
DNA during apoptosis, and its inhibitor ICAD. Nature Frederick JM (1987) The emergence of GABA-accumulating
391:4350. neurons during retinal histogenesis in the embryonic chick.
Erlich RB, Werneck CC, Mouro PA, Linden R (2003) Major Exp Eye Res 45:933945.
glycosaminoglycan species in the developing retina: synthe- Fredholm BB (1997) Adenosine and neuroprotection. Int Rev
sis, tissue distribution and effects upon cell death. Exp Eye Neurobiol 40:259280.
Res 77:157165. Fujita S (1962) Kinetics of cellular proliferation. Exp Cell Res
Feller MB, Wellis DP, Stellwagen D, Werblin FS, Shatz CJ 28:5260.
(1996) Requirement for cholinergic synaptic transmission Fukada T, Ohtani T, Yoshida Y, Shirogane T, Nishida K,
in the propagation of spontaneous retinal waves. Science Nakajima K, Hibi M, Hirano T (1998) STAT3 orchestrates
272:11821187. contradictory signals in cytokine-induced G1 to S cell-cycle
Ferriero DM, Sagar SM (1989) Development of neuropeptide transition. EMBO J 17:66706677.
Y immunoreactive neurons in the rat retina. Dev Brain Res Galli-Resta L, Leone P, Bottari D, Ensini M, Rigosi E, Novelli
48:1926. E (2008) The genesis of retinal architecture: an emerg-
Finlay BL (2008) The developing and evolving retina: using time ing role for mechanical interactions? Prog Retin Eye Res
to organize form. Brain Res 1192:516. 27:260283.
Fischer AJ, Dierks BD, Reh TA (2002a) Exogenous growth fac- Galluzzi L, Vicencio JM, Kepp O, Tasdemir E, Maiuri MC,
tors induce the production of ganglion cells at the retinal Kroemer G (2008) To die or not to die: that is the autophagic
margin. Development 129:22832291. question. Curr Mol Med 8:7891.
Fischer AJ, McGuire CR, Dierks BD, Reh TA (2002b) Insulin Garcia M, Forster V, Hicks D, Vecino E (2003) In vivo expres-
and fibroblast growth factor 2 activate a neurogenic pro- sion of neurotrophins and neurotrophin receptors is con-
gram in Mller glia of the chicken retina. J Neurosci 22: served in adult porcine retina in vitro. Invest Ophthalmol Vis
93879398. Sci 44:45324541.
Fischer AJ, Omar G, Walton NA, Verrill TA, Unson CG Gardino PF, dos Santos RM, Hokoc JN (1993) Histogenesis
(2005) Glucagon-expressing neurons within the retina reg- and topographical distribution of tyrosine hydroxylase
ulate the proliferation of neural progenitors in the circum- immunoreactive amacrine cells in the developing chick
ferential marginal zone of the avian eye. J Neurosci 25: retina. Brain Res Dev Brain Res 72:226236.
1015710166. Gaur VP, Liu Y, Turner JE (1992) RPE conditioned medium
Fischer AJ, Reh TA (2001) Mller glia are a potential source stimulates photoreceptor cell survival, neurite outgrowth and
of neural regeneration in the postnatal chicken retina. Nat differentiation in vitro. Exp Eye Res 54:645659.
Neurosci 4:2472052. Godinho L, Williams PR, Claassen Y, Provost E, Leach
Fischer AJ, Reh TA (2003) Growth factors induce neurogenesis SD, Kamermans M, Wong RO (2007) Nonapical
in the ciliary body. Dev Biol 259:225240. symmetric divisions underlie horizontal cell layer formation
Fletcher EL, Kalloniatis M (1997) Localisation of amino acid in the developing retina in vivo. Neuron 56:597603.
neurotransmitters during postnatal development of the rat Gran X, Reddy EP (1995) Cell cycle control in mammalian
retina. J Comp Neurol 380:449471. cells: role of cyclins, cyclin-dependent kinases (CDK),
222 R. Linden et al.

growth supressor genes and cyclin-dependent kinases Harper JV, Brooks G (2005) The mammalian cell cycle: an
inhibitors (CDKIs). Oncogene 11:211219. overview. Methods Mol Biol 296:113153.
Greka A, Lipton SA, Zhang D (2000) Expression of GABA(C) Hartveit E, Brandstatter JH, Sassoe-Pognetto M, Laurie DJ,
receptor rho1 and rho2 subunits during development of the Seeburg PH, Wassle H (1994) Localization and devel-
mouse retina. Eur J Neurosci 12:35753582. opmental expression of the NMDA receptor subunit
Grunder T, Kohler K, Guenther E (2000a) Distribution and NR2A in the mammalian retina. J Comp Neurol 348:
developmental regulation of AMPA receptor subunit proteins 570582.
in rat retina. Invest Ophthalmol Vis Sci 41:36003606. Hartwell LH, Weinert TA (1989) Checkpoints: controls that
Grunder T, Kohler K, Kaletta A, Guenther E (2000b) The dis- ensure the order of cell cycle events. Science 246:
tribution and developmental regulation of NMDA receptor 629634.
subunit proteins in the outer and inner retina of the rat. J Haydar TF, Wang F, Schwartz ML, Rakic P (2000) Differential
Neurobiol 44:333342. modulation of proliferation in the neocortical ventricular and
Guilarducci-Ferraz CV, da Silva GM, Torres PM, Dos Santos subventricular zones. J Neurosci 20:57645774.
AA, Araujo EG (2008) The increase in retinal cells prolif- Hayes NL, Nowakowski RS (2000) Exploiting the dynamics of
eration induced by FGF2 is mediated by tyrosine and PI3 S-phase tracers in the developing brain: Interkinetic nuclear
kinases. Neurochem Res 33:754764. migration for cells entering versus leavinhg the S-phase. Dev
Guimaraes CA, Linden R (2004) Programmed cell deaths. Neurosci 22:4455.
Apoptosis and alternative deathstyles. Eur J Biochem Henriquez M, Armisen R, Stutzin A, Quest AF (2008) Cell death
271:16381650. by necrosis, a regulated way to go. Curr Mol Med 8:187206.
Guizzetti M, Costa P, Peters J, Costa LG (1996) Acetylcholine Heo JS, Han HJ (2006) ATP stimulates mouse embryonic stem
as a mitogen: muscarinic receptor-mediated proliferation of cell proliferation via PKC, PI3K/Akt, and MAPKs signaling
rat astrocytes and human astrocytoma cells. Eur J Pharmacol pathways. Stem Cells 12:26372648.
297:265273. Hernandez-Sanchez C, Lopez-Carranza A, Alarcon C, de La
Guizzetti M, Wei M, Costa LG (1998) The role of protein kinase Rosa EJ, de Pablo F (1995) Autocrine/paracrine role of
C alpha and epsilon isozymes in DNA synthesis induced by insulin-related growth factors in neurogenesis: local expres-
muscarinic receptors in a glial cell line. Eur J Pharmacol sion and effects on cell proliferation and differentiation in
359:223233. retina. Proc Natl Acad Sci U S A 92:98349838.
Gutierrez GJ, Ronai Z (2006) Ubiquitin and SUMO systems in Herzog KH, Schulz A, Buerkle C, Gromoll C, Braun JS (2007)
the regulation of mitotic checkpoints. Trends Biochem Sci Radiation-induced apoptosis in retinal progenitor cells is
31:324332. p53-dependent with caspase-independent DNA fragmenta-
Gtz M, Huttner WB (2005) The cell biology of neurogenesis. tion. Eur J Neurosci 25:13491356.
Nat Rev Mol Cell Biol 6:777788. Hicks D (1998) Putative functions of fibroblast growth factors
Hadjiconstantinou M, Mariani AP, Panula P, Joh TH, Neff in retinal development, maturation and survival. Semin Cell
NH (1984) Immunohistochemical evidence for epinephrine- Dev Biol 9:263269.
containing retinal amacrine cells. Neuroscience 13:547551. Hicks D, Courtois Y (1992) Fibroblast growth factor stimu-
Hajra KM, Liu JR (2004) Apoptosome dysfunction in human lates photoreceptor differentiation in vitro. J Neurosci 12:
cancer. Apoptosis 9:691704. 20222033.
Hakem R, Hakem A, Duncan GS, Henderson JT, Woo M, Hill DR (1985) GABAB receptor modulation of adenylate
Soengas MS, Elia A, de la Pompa JL, Kagi D, Khoo W, cyclase activity in rat brain slices. Br J Pharmacol 84:
Potter J, Yoshida R, Kaufman SA, Lowe SW, Penninger JM, 249257.
Mak TW (1998) Differential requirement for caspase 9 in Hokoc JN, Ventura AL, Gardino PF, de Mello FG (1990)
apoptotic pathways in vivo. Cell 94:339352. Developmental immunoreactivity for GABA and GAD in
Hamassaki-Britto DE, Gardino PF, Hokoc JN, Keyser KT, the avian retina: possible alternative pathway for GABA
Karten HJ, Lindstrom JM, Britto LR (1994) Differential synthesis. Brain Res 532:197202.
development of alpha-bungarotoxin-sensitive and alpha- Holtkamp GM, Kijlstra A, Peek R, de Vos AF (2001) Retinal
bungarotoxin-insensitive nicotinic acetylcholine receptors in pigment epithelium-immune system interactions: cytokine
the chick retina. J Comp Neurol 347:161170. production and cytokine-induced changes. Prog Retin Eye
Han SJ, Conti M (2006) New pathways from PKA to the Res 20:2948.
Cdc2/cyclin B complex in oocytes: Wee1B as a potential Honda Z-I, Takano T, Gotoh Y, Nishida E, Ito K, Shimizu T
PKA substrate. Cell Cycle 5:227231. (1994) Transfected platelet-activating factor receptor acti-
Hao Z, Duncan GS, Chang CC, Elia A, Fang M, Wakeham A, vates mitogen-activated protein (MAP) kinase and MAP
Okada H, Calzascia T, Jang Y, You-Ten A, Yeh WC, Ohashi kinase kinase in Chinese hamster ovary cells. J Biol Chem
P, Wang X, Mak TW (2005) Specific ablation of the apoptotic 269:23072315.
functions of cytochrome C reveals a differential require- Hoyer D, Hannon JP, Martin GR (2002) Molecular, phar-
ment for cytochrome C and Apaf-1 in apoptosis. Cell 121: macological and functional diversity of 5-HT receptors.
579591. Pharmacol Biochem Behav 71:533554.
Hardy P, Beauchamp M, Sennlaub F, Gobeil F Jr, Tremblay Huang BO, Redburn DA (1996) GABA-induced increases in
L, Mwaikambo B, Lachapelle P, Chemtob S (2005) New [Ca2+ ]i in retinal neurons of postnatal rabbits. Vis Neurosci
insights into the retinal circulation: inflammatory lipid medi- 13:441447.
ators in ischemic retinopathy. Prostaglandins. Leukot Essent Hurley PJ, Bunz F (2007) ATM and ATR: components of an
Fatty Acids 72:301325. integrated circuit. Cell Cycle 6:414417.
13 Tissue Biology of Proliferation and Cell Death 223

Ilia M, Jeffery G (1999) Retinal mitosis is regulated by dopa, a progenitors via the JAK2/STAT3 pathway with EGF-
melanin precursor that may influence the time at which cells induced MAPK phosphorylation. Cell Prolif 41:
exit the cell cycle: analysis of patterns of cell production in 377392.
pigmented and albino retinae. J Comp Neurol 405:394405. Kerr JF (2002) History of the events leading to the formulation
Inatani M, Tanihara H (2002) Proteoglycans in retina. Prog Retin of the apoptosis concept. Toxicology 181182:471474.
Eye Res 21:429447. Kerr JF, Wyllie AH, Currie AR (1972) Apoptosis: a basic bio-
Ingham PW, McMahon AP (2001) Hedgehog signaling in ani- logical phenomenon with wide-ranging implications in tissue
mal development: paradigms and principles. Genes Dev kinetics. Br J Cancer 26:23957.
15:30593087. Kim R (2005) Recent advances in understanding the cell
Isayama T, Hurst WJ, McLaughlin PJ, Zagon IS (1995) death pathways activated by anticancer therapy. Cancer 103:
Ontogeny of the opioid growth factor, [Met5]-enkephalin, 15511560.
and its binding activity in the rat retina. Visual Neurosci Kim IB, Lee EJ, Kim MK, Park DK, Chun MH (2000) Choline
12:939950. acetyltransferase-immunoreactive neurons in the developing
Isayama T, McLaughlin PJ, Zagon IS (1991) Endogenous opi- rat retina. J Comp Neurol 427:604616.
oids regulate cell proliferation in the retina of developing rat. Kim IB, Park DK, Oh SJ, Chun MH (1998) Horizontal cells
Brain Res 544:7985. of the rat retina show choline acetyltransferase- and vesicu-
Isayama T, McLaughlin PJ, Zagon IS (1996) Ontogeny of pre- lar acetylcholine transporter-like immunoreactivities during
proenkephalin mRNA expression in the rat retina. Visual early postnatal developmental stages. Neurosci Lett 253:
Neurosci 13:695704. 8386.
Ishi I, Shimizu T (2000) Platelet-activating factor receptor and Kolb H (1997) Amacrine cells of the mammalian retina: neuro-
genetically engineered PAF receptor mutant mice. Prog Lipid circuitry and functional roles. Eye 11:904923.
Res 39:4182. Kolb H, Linberg KA, Fisher SK (1992) Neurons of the human
Izumi T, Shimizu T (1995) Platelet-activating factor receptor: retina: a Golgi study. J Comp Neurol 318:147187.
gene expression and signal transduction. Biochim Biophys Korenjak M, Brehm A (2005) E2F-Rb complexes regulating
Acta 1259:317333. transcription of genes important for differentiation and devel-
Jacobson M (1991) Developmental Neurobiology. Plenum Press, opment. Curr Opin Genet Dev 15:520527.
New York, 1776. Koulen P (1999) Postnatal development of dopamine D1 recep-
Jadhav AP, Cho S, Cepko CL (2006a) Notch activity permits tor immunoreactivity in the rat retina. J Neurosci Res
retinal cells to progress through multiple progenitor states 56:397404.
and acquire a stem cell property. Proc Natl Acad Sci U S Kralj-Hans I, Tibber M, Jeffery G, Mobbs P (2006) Differential
A 103:1899819003. effect of dopamine on mitosis in early postnatal albino and
Jadhav AP, Mason HA, Cepko CL (2006b) Notch 1 inhibits pho- pigmented rat retinae. J Neurobiol 66:4755.
toreceptor production in the developing mammalian retina. Kriegstein AR, Gtz M (2003) Radial glia diversity: a matter of
Development 133:913923. cell fate. Glia 43:3743.
James J, Das AV, Rahnenfuhrer J, Ahmad I (2004) Cellular Kriegstein AR, Noctor S, Martinez-Cerdeno M (2006) Patterns
and molecular characterization of early and late retinal stem of neural stem and progenitor cell division may underlie evo-
cells/progenitors: differential regulation of proliferation and lutionary cortical expansion. Nat Rev Neurosci 7:883890.
context dependent role of Notch signaling. J Neurobiol Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES,
61:359376. Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein
Jaynes CD, Turner JE (2000) Isolation of a retinal pigment P, Green DR, Hengartner M, Knight RA, Kumar S, Lipton
epithelial cell-derived fraction which promotes Mller cell SA, Malorni W, Nuez G, Peter ME, Tschopp J, Yuan J,
proliferation. Dev Brain Res 120:267271. Piacentini M, Zhivotovsky B, Melino G (2008) Classification
Jazayeri A, Falck J, Lukas C, Bartek J, Smith GC, Lukas J, of cell death: recommendations of the Nomenclature
Jackson SP (2006) ATM- and cell cycle-dependent regula- Committee on Cell Death 2009. Cell Death Differ 16:
tion of ATR in response to DNA double-strand breaks. Nat 311.
Cell Biol 8:3745. Krysko DV, Vanden Berghe T, Parthoens E, DHerde K,
Jensen AM, Wallace VA (1997) Expression of Sonic hedge- Vandenabeele P (2008) Methods for distinguishing apoptotic
hog and its putative role as a precursor cell mitogen in the from necrotic cells and measuring their clearance. Methods
developing mouse retina. Development 124:363371. Enzymol 442:307341.
Jeon CJ, Strettoi E, Masland RH (1998) The major cell popula- Kuan CY, Roth KA, Flavell RA, Rakic P (2000) Mechanisms
tions of the mouse retina. J Neurosci 18:89368946. of programmed cell death in the developing brain. Trends
Jin Z, El-Deiry WS (2005) Overview of cell death signaling Neurosci 23:291297.
pathways. Cancer Biol Ther 4:139163. Kubo F, Takeichi M, Nakagawa S 2003. Wnt2b controls retinal
Jotwani G, Itoh K, Wadhwa S (1994) Immunohistochemical cell differentiation at the ciliary marginal zone. Development
localization of tyrosine hydroxylase, substance P, 130(3):587598.
neuropeptide-Y and leucine-enkephalin in developing Kubo F, Nakagawa S (2008) Wnt signaling in retinal stem cells
human retinal amacrine cells. Dev Brain Res 77:285289. and regeneration. Dev Growth Differ 50(4):245251.
Kaldis P (1999) The cdk-activating kinase (CAK): from yeast to Kubota R, Hokoc JN, Moshiri A, McGuire C, Reh TA (2002)
mammals. Cell Mol Life Sci 55:284296. A comparative study of neurogenesis in the retinal ciliary
Kang MK, Kang SK (2008) Interleukin-6 induces marginal zone of homeothermic vertebrates. Dev Brain Res
proliferation in adult spinal cord-derived neural 134:3141.
224 R. Linden et al.

Kubrusly RC, Guimaraes MZ, Vieira AP, Hokoc JN, Casarini Liang Q, Li W, Zhou B (2008) Caspase-independent apoptosis
DE, de Mello MC, de Mello FG (2003) L-DOPA supply in yeast. Biochim Biophys Acta 1783:13111319.
to the neuro retina activates dopaminergic communication Liljekvist-Soltic I, Olofsson J, Johansson K (2008) Progenitor
at the early stages of embryonic development. J Neurochem cell-derived factors enhance photoreceptor survival in rat
86:4554. retinal explants. Brain Res 1227:226233.
Kubrusly RC, Ventura AL, de Melo Reis RA, Serra GC, Lillien L (1995) Changes in retinal cell fate induced by overex-
Yamasaki EN, Gardino PF, de Mello MC, de Mello FG pression of EGF receptor. Nature 377:158162.
(2007) Norepinephrine acts as D1-dopaminergic agonist Lillien L (1998) Neural progenitors and stem cells: mechanisms
in the embryonic avian retina: late expression of beta1- of progenitor heterogeneity. Curr Opin Neurobiol 8:3744.
adrenergic receptor shifts norepinephrine specificity in the Lillien L, Cepko C (1992) Control of proliferation in the retina:
adult tissue. Neurochem Int 50:211218. temporal changes in responsiveness to FGF and TGF alpha.
Kuida K, Haydar TF, Kuan CY, Gu Y, Taya C, Karasuyama H, Development 115:253266.
Su MS, Rakic P, Flavell RA (1998) Reduced apoptosis and Lillien L, Wancio D (1998) Changes in epidermal growth factor
cytochrome c-mediated caspase activation in mice lacking receptor expression and competence to generate glia regulate
caspase 9. Cell 94:325337. timing and choice of differentiation in the retina. Mol Cell
Kuida K, Zheng TS, Na S, Kuan C, Yang D, Karasuyama H, Neurosci 10:296308.
Rakic P, Flavell RA (1996) Decreased apoptosis in the brain Lin SC, Skapek SX, Papermaster DS, Hankin M, Lee EY (2001)
and premature lethality in CPP32-deficient mice. Nature The proliferative and apoptotic activities of E2F1 in the
384:368372. mouse retina. Oncogene 20:70737084.
Kumar S (2007) Caspase function in programmed cell death. Linden R (2000) The anti-death league: associative control of
Cell Death Differ 14:3243. apoptosis in developing retinal tissue. Brain Res Rev 32:
Kumar S, Kahn MA, Dinh L, de Vellis J (1998) NT-3-mediated 146158.
TrkC receptor activation promotes proliferation and cell sur- Linden, R. Reese BE (2006) Programmed cell death. In: Retinal
vival of rodent progenitor oligodendrocyte cells in vitro and Development Evelyn Sernagor; Stephen Eglen; William
in vivo. J Neurosci Res 54:754765. A. Harris; Rachel Wong. (Org.), p. 208241. Cambridge
Kunchithapautham K, Rohrer B (2007) Apoptosis and University Press, Cambridge.
autophagy in photoreceptors exposed to oxidative stress. Linden R, Cavalcante LA, Barradas PC (1986) Mononuclear
Autophagy 3:433441. phagocytes in the retina of developing rats. Histochemistry
Kuruvilla A, Pielop C, Shearer WT (1994) Platelet-activating 85:335339.
factor induces the tyrosine phosphorylation and activation of Linden R, Martins RA, Silveira MS (2005) Control of pro-
phospholipase C-gamma 1, Fyn and Lyn kinases, and phos- grammed cell death by neurotransmitters and neuropeptides
phatidylinositol 3-kinase in a human B cell line. J Immunol in the developing mammalian retina. Prog Retin Eye Res
153:54335442. 24:457491.
Kuwayama Y, Ishimoto I, Fukuda M, Shimiza Y, Shiosaka Linden R, Rehen SK, Chiarini LB (1999) Apoptosis in develop-
S, Inagaki S, Senba E, Sakanaka M, Takagi H, Takatsuki ing retinal tissue. Prog Retin Eye Res 18:133165.
K, Hara Y, Kawai Y, Tohyama M (1982) Overall distri- Liu B, Nakashima S, Kanoh H, Takano T, Shimizu T, Nozawa
bution of glucagon-like immunoreactivity in the chicken Y (1994) Activation of phospholipase D in Chinese ham-
retina: an immunohistochemical study with flat-mounts. ster ovary cells expressing platelet-activating factor receptor.
Invest Ophthalmol Vis Sci 22:681 686. J Biochem 116:882891.
Laasberg T (1990) Ca2(+)-mobilizing receptors of gastrulating Livesey FJ, Cepko CL (2001) Vertebrate neural cell-fate deter-
chick embryo. Comp Biochem Physiol C 97:912. mination: lessons from the retina. Nat Rev Neurosci 2:
Langman J, Guerrant RL, Freeman BG (1966) Behavior of 109118.
neuro-epithelial cells during closure of the neural tube. J Locker M, Agathocleous M, Amato MA, Parain K, Harris WA,
Comp Neurol 127:399411. Perron M (2006) Hedgehog signaling and the retina: insights
Lavin MF, Kozlov S (2007) ATM activation and DNA damage into the mechanisms controlling the proliferative properties
response. Cell Cycle 6:931942. of neural precursors. Genes Dev 20:30363048.
Layer PG (1991) Cholinesterases during development of the Lograno MD, Tricarico D, Masciopinto V, Scuderl AC (2000)
avian nervous system. Cell Mol Neurobiol 11:733. Specific binding of nicergoline on an alpha1-like adrenore-
Lee MY, Heo JS, Han HJ (2006) Dopamine regulates cell cycle ceptor in the rat retina. J Pharm Pharmacol 52:207211.
regulatory proteins via cAMP, Ca(2+)/PKC, MAPKs, and Lombardini JB (1991) Taurine: retinal function. Brain Res Brain
NF-kappaB in mouse embryonic stem cells. J Cell Physiol Res Rev 16:151169.
208:399406. Lonze BE, Ginty D (2002) Function and regulation of CREB
Levine EM, Green ES (2004) Cell-intrinsic regulators of prolif- family transcription factors in the nervous system. Neuron
eration in vertebrate retinal progenitors. Semin Cell Dev Biol 35:605623.
15:6374. Lorenzo HK, Susin SA, Penninger J, Kroemer G (1999)
Levine EM, Roelink H, Turner J, Reh TA (1997) Sonic hedgehog Apoptosis inducing factor (AIF): a phylogenetically old,
promotes rod photoreceptor differentiation in mammalian caspase-independent effector of cell death. Cell Death Differ
retinal cells in vitro. J Neurosci 17:62776288. 6:516524.
Li, Y, Yang X, Ma C, Qiao J, Zhang C (2008) Necroptosis con- Lossi L, Merighi A (2003) In vivo cellular and molecular mech-
tributes to the NMDA-induced excitotoxicity in rats cultured anisms of neuronal apoptosis in the mammalian CNS. Prog
cortical neurons. Neurosci Lett 447:120123. Neurobiol 69:287312.
13 Tissue Biology of Proliferation and Cell Death 225

LoTurco JJ, Owens DF, Heath MJ, Davis MB, Kriegstein AR Lerenthal Y, Shiloh Y, Gygi SP, Elledge SJ (2007) ATM and
(1995) GABA and glutamate depolarize cortical progenitor ATR substrate analysis reveals extensive protein networks
cells and inhibit DNA synthesis. Neuron 15:12871298. responsive to DNA damage. Science 316:11601166.
Low CP, Shui G, Liew LP, Buttner S, Madeo F, Dawes IW, Wenk McCabe KL, Gunther EC, Reh TA (1999) The development
MR, Yang H (2008) Caspase-dependent and -independent of the pattern of retinal ganglion cells in the chick retina:
lipotoxic cell-death pathways in fission yeast. J Cell Sci mechanisms that control differentiation. Development 126:
121:26712684. 57135724.
Low CP, Yang H (2008) Programmed cell death in fission McFarlane S, Zuber ME, Holt CE (1998) A role for the fibroblast
yeast Schizosaccharomyces pombe. Biochim Biophys Acta growth factor receptor in cell fate decisions in the developing
1783:13351349. vertebrate retina. Development 125:39673975.
Luk KC, Kennedy TE, Sadikot AF (2003) Glutamate promotes McKinnon LA, Nathanson NM (1995) Tissue-specific regula-
proliferation of striatal neuronal progenitors by an NMDA tion of muscarinic acetylcholine receptor expression during
receptor-mediated mechanism. J Neurosci 23:22392250. embryonic development. J Biol Chem 270:2063620642.
Lunyak VV, Rosenfeld MG (2008) Epigenetic regulation of stem Mione MC, Cavanagh JF, Harris B, Parnavelas JG (1997)
cell fate. Hum Mol Genet 17:R28R36. Cell fate specification and symmetrical/asymmetrical divi-
Ma C, Papermaster D, Cepko CL (1998) A unique pattern of sions in the developing cerebral cortex. J Neurosci 17:
photoreceptor degeneration in cyclin D1 mutant mice. Proc 20182029.
Natl Acad Sci 95:99389943. Miranda-Contreras L, Benitez-Diaz PR, Mendoza-Briceno RV,
Maandag EC, van der Valk M, Vlaar M, Feltkamp C, OBrien J, Delgado-Saez MC, Palacios-Pru EL (1999) Levels of amino
van Roon M (1994) Developmental rescue of an embryonic- acid neurotransmitters during mouse cerebellar neurogene-
lethal mutation in the retinoblastoma gene in chimeric mice. sis and in histotypic cerebellar cultures. Dev Neurosci 21:
EMBO J 13:42604268. 147158.
MacLaren RE, Pearson RA (2007) Stem cell therapy and the Miranda-Contreras L, Mendoza-Briceno RV, Palacios-Pru EL
retina. Eye 21:13521359. (1998) Levels of monoamine and amino acid neurotrans-
MacPherson D, Sage J, Kim T, Ho D, McLaughlin ME, Jacks T mitters in the developing male mouse hypothalamus and
(2004) Cell type-specific effects of Rb deletion in the murine in histotypic hypothalamic cultures. Int J Dev Neurosci
retina. Genes Dev 18:16811694. 16:403412.
Makman MK, Dvorkin B (1997) Presence of nociceptin Miranda-Contreras L, Ramirez-Martens LM, Benitez-Diaz PR,
(orphanin FQ) receptors in rat retina: comparison with recep- Pena-Contreras ZC, Mendoza-Briceno RV, Palacios-Pru EL
tors in striatum. Eur J Pharmacol 338:171176. (2000) Levels of amino acid neurotransmitters during mouse
Marigo V (2007) Programmed cell death in retinal degeneration: olfactory bulb neurogenesis and in histotypic olfactory bulb
targeting apoptosis in photoreceptors as potential therapy for cultures. Int J Dev Neurosci 18:8391.
retinal degeneration. Cell Cycle 6:652655. Mitchell CK, Huang B, Redburn-Johnson DA (1999) GABA(A)
Marquardt T (2003) Transcriptional control of neuronal diversi- receptor immunoreactivity is transiently expressed in the
fication in the retina. Prog Retin Eye Res 22:567577. developing outer retina. Vis Neurosci 16:10831088.
Marquardt T, Gruss P (2002) Generating neuronal diversity in Mitchell CK, Redburn DA (1996) GABA and GABA-A recep-
the retina: one for nearly all. Trends Neurosci 25:3238. tors are maximally expressed in association with cone
Martins RA, Pearson RA (2008) Control of cell proliferation by synaptogenesis in neonatal rabbit retina. Dev Brain Res 95:
neurotransmitters in the developing vertebrate retina. Brain 6371.
Res 1192:3760. Miyata T (2008) Development of three-dimensional architec-
Martin-Martinelli E, Simon A, Vigny A, Nguyen-Legros J ture of the neuroepithelium: role of pseudostratification and
(1989) Postnatal development of tyrosine-hydroxylase cellular community. Dev Growth Differ 50:S105S112.
hydroxylaseimmunoreactive cells in the rat retina. Miyata T, Kawaguchi A, Okano H, Ogawa M (2001)
Morphology and distribution. Dev Neurosci 11:1125. Asymmetric inheritance of radial glial fibers by cortical
Martins RA, Linden R, Dyer MA (2006) Glutamate regulates neurons. Neuron 31:727741.
retinal progenitors cells proliferation during development. Miyata T, Kawaguchi A, Saito K, Kuramochi H, Ogawa M
Eur J Neurosci 24:969980. (2002) Visualization of cell cycling by an improvement in
Martins RA, Pearson RA (2008) Control of cell proliferation by slice culture methods. J Neurosci Res 69:861868.
neurotransmitters in the developing vertebrate retina. Brain Montrucchio G, Lupia E, Battaglia E, Del Sorbo L, Boccellino
Res 1192:3760. M, Biancone L, Emanuelli G, Camussi G (2000) Platelet-
Martn-Partido G, Rodrguez-Gallardo L, Alvarez IS, Navascus activating factor enhances vascular endothelial growth
J (1988) Cell death in the ventral region of the neural retina factor-induced endothelial cell motility and neoangiogenesis
during the early development of the chick embryo eye. Anat in a murine matrigel model. Arterioscler Thromb Vasc Biol
Rec 222:272281. 20:8088.
Masai I, Yamaguchi M, Tonou-Fujimori N, Komori A, Okamoto Morgan DO (1997) Cyclin-dependent kinases: engines, clocks,
H (2005) The hedgehog-PKA pathway regulates two and microprocessors. Annu Rev Cell Dev Biol 13:261291.
distinct steps of the differentiation of retinal ganglion cells: Muotri AR, Gage FH (2006) Generation of neuronal variability
the cell-cycle exit of retinoblasts and their neuronal matura- and complexity. Nature 441:10871093.
tion. Development 132:15391553. Murphy SN, Miller RJ (1989) Two distinct quisqualate receptors
Matsuoka S, Ballif BA, Smogorzewska A, McDonald ER 3rd, regulate Ca2+ homeostasis in hippocampal neurons in vitro.
Hurov KE, Luo J, Bakalarski CE, Zhao Z, Solimini N, Mol Pharmacol 35:671680.
226 R. Linden et al.

Mller F, Albert S, Blader P, Fischer N, Hallonet M, Strahle neurotrophins and their high-affinity Trk receptors in cul-
U (2000) Direct action of the nodal-related signal cyclops tured human Muller cells. Ophthalmic Res 34:3842.
in induction of sonic hedgehog in the ventral midline of the Olianas MC, Ingianni A, Sogos V, Onali P (1997) Expression of
CNS. Development 127:38893897. pituitary adenylate cyclase-activating polypeptide (PACAP)
Nagahara H, Ezhevsky SA, Vocero-Akbani AM, Kaldis P, receptors and PACAP in human retina. J Neurochem
Solomon MJ, Dowdy SF (1999) Transforming growth fac- 69:12131218.
tor beta targeted inactivation of cyclin E:cyclin-dependent Ooto S, Akagi T, Kageyama R, Akita J, Mandai M, Honda Y,
kinase 2 (Cdk2) complexes by inhibition of Cdk2 acti- Takahashi M (2004) Potential for neural regeneration after
vating kinase activity. Proc Natl Acad Sci U S A 96: neurotoxic injury in the adult mammalian retina. Proc Natl
1496114966. Acad Sci U S A 101:1365413659.
Nakanishi M, Niidome T, Matsuda S, Akaike A, Kihara T, Oppenheim RW, Blomgren K, Ethell DW, Koike M,
Sugimoto H (2007) Microglia-derived interleukin-6 and Komatsu M, Prevette D, Roth KA, Uchiyama Y,
leukaemia inhibitory factor promote astrocytic differentia- Vinsant S, Zhu C (2008) Developing postmitotic
tion of neural stem/progenitor cells. Eur J Neurosci 25: mammalian neurons in vivo lacking Apaf-1 undergo
649658. programmed cell death by a caspase-independent, non-
Nakayama K, Ishida N, Shirane M, Inomata A, Inoue T, apoptotic pathway involving autophagy. J Neurosci 28:
Shishido N, Horii I, Loy D, Nakayama K (1996) Mice lack- 14901497.
ing p27Kip1 display increased body side, multiple organ Osakada F, Ooto S, Akagi T, Mandai M, Akaike A, Takahashi M
hyperplasia, retinal dysplasia and pituitary tumors. Cell (2007) Wnt signaling promotes regeneration in the retina of
85:707720. adult mammals. J Neurosci 27:42104219.
Neves DD, Rehen SK, Linden R (2001) Differentiation- Osborne NN, Patel S, Vigny A (1984) Dopaminergic neu-
dependent sensitivity to cell death induced in the developing rones in various retinas and the postnatal development of
retina by inhibitors of the ubiquitin-proteasome proteolytic tyrosine-hydroxylase immunoreactivity in the rabbit retina.
pathway. Eur J Neurosci 13:19381944. Histochemistry 80:389393.
Newman EA (2001) Propagation of intercellular calcium waves Otaegi G, de Pablo F, Vicario-Abejn C, de la Rosa EJ
in retinal astrocytes and Mller cells. J Neurosci 21: (2007) Retinal and olfactory bulb precursor cells show dis-
22152223. tinct responses to FGF-2 and laminin. Cell Biol Int 31:
Nguyen-Legros J, Vigny A, Gay M (1983) Post-natal develop- 752758.
ment of TH-like immunoreactivity in the rat retina. Exp Eye Otteson DC, Cirenza PF, Hitchcock PF (2002) Persistent neuro-
Res 37:2332. genesis in the teleost retina: evidence for regulation by the
Nickerson PEB, Da Silva N, Myers T, Stevens K, Clarke DB growth-hormone/insulin-like growth factor-I axis. Mech Dev
(2008) Neural progenitor potential in cultured Mller glia: 117:137149.
effects of passaging and exogenous growth factor exposure. Paes-De-Carvalho R (2002) Adenosine as a signaling molecule
Brain Res 1230:112. in the retina: biochemical and developmental aspects. An
Nishi S, Minota S, Karczmar AG (1974) Primary afferent neu- Acad Bras Cienc 74:437451.
rones: the ionic mechanism of GABA-mediated depolariza- Page AM, Hieter P (1997) The anaphase promoting complex. In:
tion. Neuropharmacology 13:215219. Checkpoint Controls and Cancer (Kastan MB, ed.), pp. 133
Niwa H, Burdon T, Chambers I, Smith A (1998) Self-renewal of 150. Cold Spring Harbor Lab. Press, Cold Spring Harbor,
pluripotent embryonic stem cells is mediated via activation NY.
of STAT3. Genes Dev 12:20482060. Patel A, McFarlane S (2000) Overexpression of FGF-2 alters cell
Njaine B, Martins RAP, Santiago MF, Linden R, Silveira MS fate specification in the developing retina of Xenopus laevis.
(2009) Pituitary adenylyl cyclase-activating polypeptide con- Dev Biol 222:170180.
trols the proliferation of retinal progenitor cells through Patterson SL, Pittenger C, Morozov A, Martin KC, Scanlin H,
downregulation of cyclin D1. Submitted. Drake C, Kandel ER (2001) Some forms of cAMP-mediated
North RA, Williams JT, Surprenant A, Christie MJ, (1987) Mu long-lasting potentiation are associated with release of
and delta receptors belong to a family of receptors that are BDNF and nuclear translocation of phospho-MAP kinase.
coupled to potassium channels. Proc Natl Acad Sci USA Neuron 32:123140.
84:54875491. Pearson R, Catsicas M, Becker D, Mobbs P (2002) Purinergic
Nunes PH, Calaza Kda C, Albuquerque LM, Fragel-Madeira and muscarinic modulation of the cell cycle and calcium
L, Sholl-Franco A, Ventura AL (2007) Signal transduction signaling in the chick retinal ventricular zone. J Neurosci
pathways associated with ATP-induced proliferation of cell 22:75697579.
progenitors in the intact embryonic retina. Int J Dev Neurosci Pearson RA, Dale N, Llaudet E, Mobbs P (2005a) ATP released
25:499508. via gap junction hemichannels from the pigment epithe-
Oh S, Dangelo I, Lee E, Chun M, Brecha NC (2002) lium regulates neural retinal progenitor proliferation. Neuron
Distribution and synaptic connectivity of neuropeptide Y- 46:731744.
immunoreactive amacrine cells in the rat retina. J Comp Pearson RA, Lneborg NL, Becker DL, Mobbs P (2005b) Gap
Neurol 446:219234. junctions modulate interkinetic nuclear movement in retinal
Ohta K, Ito A, Tanaka H (2008) Neuronal stem/progenitor cells progenitor cells. J Neurosci 25:1080310814.
in the vertebrate eye. Dev Growth Differ 50:253259. Pellegrini-Giampietro DE, Bennett MV, Zukin RS (1992) Are
Oku H, Ikeda T, Honma Y, Sotozono C, Nishida K, Nakamura Ca(2+)-permeable kainate/AMPA receptors more abundant
Y, Kida T, Kinoshita S (2002) Gene expression of in immature brain? Neurosci Lett 144:6569.
13 Tissue Biology of Proliferation and Cell Death 227

Pootanakit K, Brunken WJ (2000) 5-HT(1A) and 5-HT(7) Robanus-Maandag E, Dekker M, van der Valk M, Carrozza ML,
receptor expression in the mammalian retina. Brain Res Jeanny JC, Dannenberg JH (1998) p107 is a suppressor of
875:152156. retinoblastoma development in pRb-deficient mice. Genes
Pootanakit K, Brunken WJ (2001) Identification of 5-HT(3A) Dev 12:15991609.
and 5-HT(3B) receptor subunits in mammalian retinae: Rodgers EE, Theibert AB (2002) Functions of PI 3-kinase in
potential pre-synaptic modulators of photoreceptors. Brain development of the nervous system. Int J Dev Neurosci
Res 896:7785. 20:187197.
Pootanakit K, Prior KJ, Hunter DD, Brunken WJ (1999) Roegiers F, Jan YN (2004) Asymmetric cell division. Curr Opin
5-HT2a receptors in the rabbit retina: potential presynaptic Cell Biol 16:195205.
modulators. Visual Neurosci 16:221230. Rompani SB, Cepko CL (2008) Retinal progenitor cells can pro-
Pourcho RG (1996) Neurotransmitters in the retina. Curr Eye duce restricted subsets of horizontal cells. Proc Natl Acad Sci
Res 15:797803. U S A 105:192197.
Pow DV, Crook DK, Wong RO (1994) Early appearance Roos WP, Kaina B (2006) DNA damage-induced cell death by
and transient expression of putative amino acid neuro- apoptosis. Trends Mol Med 12:440450.
transmitters and related molecules in the developing rab- Rudolph U, Crestani F, Mohler H (2001) GABA(A) receptor
bit retina: an immunocytochemical study. Vis Neurosci 11: subtypes: dissecting their pharmacological functions. Trends
11151134. Pharmacol Sci 22:188194.
Prada C, Puga J, Prez-Mndez L, Lpez R, Ramrez G (1991) Russell C (2003) The roles of hedgehogs and fibroblast growth
Spatial and temporal patterns of neurogenesis in the chick factors in eye development and retinal cell rescue. Vision Res
retina. Eur J Neurosci 3:559569. 43:899912.
Prezeau L, Carrette J, Helpap B, Curry K, Pin JP, Bockaert Rutishauser U (2008) Polysialic acid in the plasticity of the
J (1994) Pharmacological characterization of metabotropic developing and adult vertebrate nervous system. Nat Rev
glutamate receptors in several types of brain cells in primary Neurosci 9:2635.
cultures. Mol Pharmacol 45:570577. Ryu JK, Choi HB, Hatori K, Heisel RL, Pelech SL, McLarnon
Price J, Turner D, Cepko C (1987) Lineage analysis in the ver- JG, Kim SU (2003) Adenosine triphosphate induces pro-
tebrate nervous system by retrovirus-mediated gene transfer. liferation of human neural stem cells: role of calcium
Proc Natl Acad Sci U S A 84:156160. and p70 ribosomal protein S6 kinase. J Neurosci Res 72:
Pruss RM, Akeson RL, Racke MM, Wilburn JL (1991) Agonist- 352362.
activated cobalt uptake identifies divalent cation-permeable Sahara S, Aoto M, Eguchi Y, Imamoto N, Yoneda Y, Tsujimoto
kainate receptors on neurons and glial cells. Neuron 7: Y (1999) Acinus is a caspase-3-activated protein required for
509518. apoptotic chromatin condensation. Nature 401:168173.
Raff MC (1992) Social controls on cell survival and cell death. Saito K, Kawaguchi A, Kashiwagi S, Yasugi S, Ogawa M,
Nature 356:397400. Miyata T (2003) Morphological asymmetry in dividing reti-
Rapaport DH, Robinson SR, Stone J (1984) Cell movement and nal progenitor cells. Develop Growth Differ 45:219229.
birth in the developing cat retina. In: Development of Visual Sakaki Y, Fukuda Y, Yamashita M (1996) Muscarinic and
Pathways in Mammals (Stone J, ed.), pp. 2338. Alan R. purinergic Ca2+ mobilizations in the neural retina of early
Liss, New York. embryonic chick. Int J Dev Neurosci 14:691699.
Rapaport DH, Robinson SR, Stone J (1985) Cytogenesis in the Sampath D, and Plunkett W (2001) Design of new anticancer
developing retina of the cat. Aust N Z J Ophthalmol 13: therapies targeting cell cycle checkpoint pathways. Curr
113124. Opin Oncol 13(6):484490
Rapaport DH, Stone J (1983) The topography of cytogenesis in Sanches G, de Alencar LS, Ventura AL (2002) ATP induces pro-
the developing retina of the cat. J Neurosci 3:18241834. liferation of retinal cells in culture via activation of PKC
Rapaport DH, Vietri AJ (1991) Identity of cells produced by and extracellular signal-regulated kinase cascade. Int J Dev
two stages of cytogenesis in the postnatal cat retina. J Comp Neurosci 20:2127.
Neurol 312:341352. Sanchez I, Dynlacht BD (2005) New insights into cyclins, CDK,
Reale M, Iarlori C, Thomas A, Gambi D, Perfetti B, Di and cell cycle control. Semin Cell Dev Biol 16:311321.
Nicola M, Onofrj M (2008) Peripheral cytokines profile in Santella L, Kyozuka K, De Riso L, Carafoli E (1998) Calcium,
Parkinsons disease. Brain Behav Immun 23:5563. protease action, and the regulation of the cell cycle. Cell
Reh TA, Fischer AJ (2006) Retinal stem cells. Methods Enzymol Calcium 23:123130.
419:5273. Santos PF, Caramelo OL, Carvalho AP, Duarte CB (1999)
Reh TA, Levine EM (1998) Multipotential stem cells and pro- Characterization of ATP release from cultures enriched in
genitors in the vertebrate retina. J Neurobiol 36:206220. cholinergic amacrine-like neurons. J Neurobiol 41:340348.
Rehen SK, Cid M, Fragel-Madeira L, Linden R (2002) Sassoe-Pognetto M, Wassle H (1997) Synaptogenesis in the
Differential effects of cyclin-dependent kinase blockers upon rat retina: subcellular localization of glycine receptors,
cell death in the developing retina. Brain Res 947:7883. GABA(A) receptors, and the anchoring protein gephyrin.
Rehen SK, Diniz DM, Fragel-Madeira L, Brito LRG, Linden J Comp Neurol 381:158174.
R (1999) Selective sensitivity of early postmitotic cells to Sauer FC (1935) Mitosis in the neural tube. J Comp Neurol
apoptosis induced by inhibition of protein synthesis. Eur J 62:377405.
Neurosci 11:349356. Sauer ME, Chittenden AC (1959) Deoxyribonucleic acid content
Riedl SJ, Salvesen GS (2007) The apoptosome: signalling plat- of cell nuclei in the neural tube of the chick embryo: evidence
form of cell death. Nat Rev Mol Cell Biol 8:405413. for intermitotic migration of nuclei. Exp Cell Res 16:16.
228 R. Linden et al.

Sauer ME, Walker BE (1959) Radioautographic study of interki- Stewart ZA, Westfall MD, Pietenpol JA (2003) Cell-cycle dys-
netic nuclear migration in the neural tube. Proc Soc Exp Biol regulation and anticancer therapy. Trends Pharmacol Sci
Med 101:557560. 24:139145.
Schfers M, Sorkin L (2008) Effect of cytokines on neuronal Sturman JA (1988) Taurine in development. J Nutr 118:
excitability. Neurosci Lett 437:188193. 11691176.
Seki T, Shioda S, Nakai Y, Arimura A, Koide R (1998) Sucher NJ, Kohler K, Tenneti L, Wong HK, Grunder T, Fauser
Distribution and ultrastructural localization of pituitary S, Wheeler-Schilling T, Nakanishi N, Lipton SA, Guenther E
adenylate cyclase-activating polypeptide (PACAP) and its (2003) N-methyl-D-aspartate receptor subunit NR3A in the
receptor in the rat retina. Ann N Y Acad Sci 865:408411. retina: developmental expression, cellular localization, and
Seki T, Shioda S, Ogino D, Nakai Y, Arimura A, Koide R (1997) functional aspects. Invest Ophthalmol Vis Sci 44:44514456.
Distribution and ultrastructural localization of a receptor for Sugioka M, Fukuda Y, Yamashita M (1996) Ca2+ responses to
pituitary adenylate cyclase activating polypeptide and its ATP via purinoceptors in the early embryonic chick retina.
mRNA in the rat retina. Neurosci Lett 238:127130. J Physiol 493:855863.
Shao-Min Zhang S, Liu M-G, Kano A, Zhang C, Fu X-Y, Sugioka M, Zhou WL, Hofmann HD, Yamashita M (1999a)
Barnstable CJ (2005) STAT3 activation in response to growth Ca2+ mobilization and capacitative Ca2+ entry regulate DNA
factors or cytokines participates in retina precursor prolifera- synthesis in cultured chick retinal neuroepithelial cells. Int J
tion. Exp Eye Res 81:103115. Dev Neurosci 17:163172.
Sharma RK, Johnson DA (2000) Molecular signals for devel- Sugioka M, Zhou W, Hofmann H, Yamashita M (1999b)
opment of neuronal circuitry in the retina. Neurochem Res Involvement of P1 purinoceptors in the regulation of DNA
25:12571263. synthesis in the neural retina of chick embryo. Int J Devl
Shelke RR, Lakshmana MK, Ramamohan Y, Raju TR (1997) Neurosci 17:135144.
Levels of dopamine and noradrenaline in the developing Sun H, Chang Y, Schweers B, Dyer MA, Zhang X, Hayward SW,
of retina effect of light deprivation. Int J Dev Neurosci Goodrich DW (2006) An E2F binding-deficient Rb1 protein
15:139143. partially rescues developmental defects associated with Rb1
Shen Y, Liu XL, Yang XL (2006) N-methyl-D-aspartate recep- nullizygosity. Mol Cell Biol 26:15271537.
tors in the retina. Mol Neurobiol 34:163179. Takahashi T, Nowakowski RS, Caviness VS Jr (1996) The
Sherr CJ (1993) Mammalian G1 Cyclins. Cell 73:10591065. leaving or Q fraction of the murine cerebral proliferative
Sherr CJ (1996) Cancer cell cycles. Science 274:16721677. epithelium: a general model of neocortical neurogenesis.
Sherr CJ, Roberts JM (1995) Inhibitors of mammalian G1 J Neurosci 16:61836196.
cyclin-dependent kinases. Genes Dev 9:11491163. Takai H, Naka K, Okada Y, Watanabe M, Harada N, Saito
Shibuya EK (2003) G2 cell cycle arrest-a direct link between S, Anderson CW, Appella E, Nakanishi M, Suzuki H,
PKA and Cdc25C. Cell Cycle 2:3941. Nagashima K, Sawa H, Ikeda K, Motoyama N (2002)
Shiloh Y (2003) ATM and related protein kinases: safeguarding Chk2-deficient mice exhibit radioresistance and defective
genome integrity. Nat Rev Cancer 3:155168. p53-mediated transcription. EMBO J 21:51955205.
Short AD, Bian J, Ghosh TK, Waldron RT, Rybak SL, Gill DL Takeda M, Takamiya A, Jiao JW, Cho KS, Trevino SG, Matsuda
(1993) Intracellular Ca2+ pool content is linked to control of T, Chen DF (2008) alpha-Aminoadipate induces progen-
cell growth. Proc Natl Acad Sci U S A 90:49864990. itor cell properties of Muller glia in adult mice. Invest
Sicinski P, Donaher JL, Parker SB, Li T, Fazeli A, Gardner Ophthalmol Vis Sci 49:11421150.
H, Haslam SZ, Bronson RT, Elledge SJ, Weinberg RA Tao X, Finkbeiner S, Arnold DB, Shaywitz AJ, Greenberg ME
(1995) Cyclin D1 provides a link between development and (1998) Ca2+ influx regulates BDNF transcription by a CREB
oncogenesis in the retina and breast. Cell 82:621630. family transcription factor-dependent mechanism. Neuron
Sidman RL (1961) Histogenesis of mouse retina studied with 20:709726.
thymidine-H3. In: The Structure of the Eye (Smelser GK, Taomoto M, McLeod DS, Merges C, Lutty GA (2000)
ed.), pp. 487506. Academic Press, London. Localization of adenosine A2a receptor in retinal develop-
Silva AG, Campello-Costa P, Linden R, Sholl-Franco A (2008) ment and oxygen-induced retinopathy. Invest Ophthalmol
Interleukin-4 blocks proliferation of retinal progenitor cells Vis Sci 41:230243.
and increases rod photoreceptor differentiation through dis- Tappaz ML (2004) Taurine biosynthetic enzymes and tau-
tinct signaling pathways. J Neuroimmunol 196:8293. rine transporter: molecular identification and regulations.
Silveira dos Santos Bredariol A, Hamassaki-Britto DE (2001) Neurochem Res 29:8396.
Ionotropic glutamate receptors during the development of the Tasdemir E, Galluzzi L, Maiuri MC, Criollo A, Vitale I, Hangen
chick retina. J Comp Neurol 441:5870. E, Modjtahedi N, Kroemer G (2008) Methods for assess-
Silveira MS, Costa MR, Bozza M, Linden R (2002) Pituitary ing autophagy and autophagic cell death. Methods Mol Biol
adenylyl cyclase-activating polypeptide prevents induced 445:2976.
cell death in retinal tissue through activation of cyclic AMP- Thoreson WB, Witkovsky P (1999) Glutamate receptors and
dependent protein kinase. J Biol Chem 277:1607516080. circuits in the vertebrate retina. Prog Retin Eye Res 18:
Silver J, Robb RM (1979) Studies on the development of the 765810.
eye cup and optic nerve in normal mice and in mutants with Thurston AW Jr, Rhee SG, Shukla SD (1993) Role of guanine
congenital optic nerve aplasia. Dev Biol 68:175190. nucleotide-binding protein and tyrosine kinase in platelet-
Stenkamp DL, Frey RA, Prabhudesai SN, Raymond PA (2000) activating factor activation of phospholipase C in A431
Function for hedgehog genes in zebrafish retinal develop- cells: proposal for dual mechanisms. J Pharmacol Exp Ther
ment. Dev Biol 220:238252. 266:11061112.
13 Tissue Biology of Proliferation and Cell Death 229

Tibber MS, Whitmore AV, Jeffery G (2006) Cell division and differentiation of multipotent neural precursor cells through
cleavage orientation in the developing retina are regulated by the activation of SAPK/JNK pathway. Mol Cell Neurosci 36:
L-DOPA. J Comp Neurol 496:369381. 343354.
Tornqvist K, Ehinger B (1983) Glucagon immunoreactive neu- Waschek JA, Dicicco-Bloom E, Nicot A, Lelievre V (2006)
rons in the retina of different species. Graefes Arch Clin Exp Hedgehog signaling new targets for GPCRs coupled to
Ophthalmol 220:15. cAMP and protein kinase A. Ann N Y Acad Sci 1070:
Tornqvist K, Loren I, Hakanson R, Sundler F (1981) Peptide- 120128.
containing neurons in the chicken retina. Exp Eye Res 33: Weinberg RA (1995) The retinoblastoma protein and cell cycle
5564. control. Cell 81:323330.
Tropepe V, Coles BL, Chiasson BJ, Horsford DJ, Elia AJ, Whitaker M, Larman MG (2001) Calcium and mitosis. Semin
McInnes RR, van der Kooy D (2000) Retinal stem cells in Cell Dev Biol 12:5358.
the adult mammalian eye. Science 287:20322036. Willer GB, Lee VM, Gregg RG, Link BA (2005) Analysis of the
Turner DL, Snyder EY, Cepko CL (1990) Lineage-independent zebrafish perplexed mutation reveals tissue-specific roles for
determination of cell type in the embryonic mouse retina. de novo pyrimidine synthesis during development. Genetics
Neuron 4:833845. 170:18271837.
Ueno M, Katayama K, Yamauchi H, Nakayama H, Doi K (2006) Woldemussie E, Wijono M, Pow D (2007) Localization of alpha
Cell cycle progression is required for nuclear migration of 2 receptors in ocular tissues. Vis Neurosci 24:745756.
neural progenitor cells. Brain Res 1088:5767. Wong RO (1995a) Cholinergic regulation of [Ca2+ ]i during
Upton AL, Salichon N, Lebrand C, Ravary A, Blakely cell division and differentiation in the mammalian retina.
R, Seif I,Gaspar P (1999) Excess of serotonin (5-HT) J Neurosci 15:26962706.
alters the segregation of ipsilateral and contralat- Wong RO (1995b) Effects of glutamate and its analogs on intra-
eral retinal projections in monoamine oxidase A cellular calcium levels in the developing retina. Vis Neurosci
knock-outmice: possible role of 5-HT uptake in reti- 12:907917.
nal ganglion cells during development. J Neurosci 19: Wu DK, Cepko CL (1993) Development of dopaminergic neu-
70077024. rons is insensitive to optic nerve section in the neonatal rat
Vallires L, Campbell IL, Gage FH, Sawchenko PE (2002) retina. Dev Brain Res 74:253260.
Reduced hippocampal neurogenesis in adult transgenic Wu Y, Cutting GR (2001) Developmentally regulated expression
mice with chronic astrocytic production of interleukin-6. J of GABA receptor rho1 and rho2 subunits, L7 and cone-
Neurosci 22:486492. rod homeobox (CRX) genes in mouse retina. Brain Res 912:
van Koppen CJ, Kaiser B (2003) Regulation of mus- 18.
carinic acetylcholine receptor signaling. Pharmacol Ther 98: Wymann MP, Pirola L (1998) Structure and function of phospho-
197220. inositide 3-kinases. Biochim Biophys Acta 1436:127150.
Vela JM, Molina-Holgado E, Arvalo-Martn A, Almazn G, Xiao Z, Chen Z, Gunasekera AH, Sowin TJ, Rosenberg SH,
Guaza C (2002) Interleukin-1 regulates proliferation and dif- Fesik S, Zhang H (2003) Chk1 mediates S and G2 arrests
ferentiation of oligodendrocyte progenitor cells. Mol Cell through CDC25A degradation in response to DNA-damaging
Neurosci 20:489502. agents. J Biol Chem 278:2176721773.
Versaux-Botteri C, Pochet R, Nguyen-Legros J (1989) Yamasaki EN, Barbosa VD, De Mello FG, Hokoc JN (1999)
Immunohistochemical localization of GABA-containing GABAergic system in the developing mammalian retina:
neurons during postnatal development of the rat retina. Invest dual sources of GABA at early stages of postnatal develop-
Ophthalmol Vis Sci 30:652659. ment. Int J Dev Neurosci 17:201213.
Vidal A, Koff A (2000) Cell-cycle inhibitors: three families Yamashita M, Fukuda Y (1993a) Calcium channels and GABA
united by a common cause. Gene 247:115. receptors in the early embryonic chick retina. J Neurobiol
von Bartheld CS (1998) Neurotrophins in the developing and 24:16001614.
regenerating visual system. Histol Histopathol 13:437459. Yamashita M, Yoshimoto Y, Fukuda Y (1994) Muscarinic
von Bartheld CS, Byers MR, Williams R, Bothwell M (1996) acetylcholine responses in the early embryonic chick retina.
Anterograde transport of neurotrophins and axodendritic J Neurobiol 25:11441153.
transfer in the developing visual system. Nature 379: Yang XL (2004) Characterization of receptors for glutamate and
830833. GABA in retinal neurons. Prog Neurobiol 73:127150.
Vugler A, Lawrence J, Walsh J, Carr A, Gias C, Semo M, Yaron O, Farhy C, Marquardt T, Applebury M, Ashery-Padan R
Ahmado A, da Cruz L, Andrews P, Coffey P (2007) (2006) Notch1 functions to suppress cone-photoreceptor fate
Embryonic stem cells and retinal repair. Mech Dev 124: specification in the developing mouse retina. Development
807829. 133:13671378.
Wallace VA (2008) Proliferative and cell fate effects of hedgehog Yeo GW, Coufal N, Aigner S, Winner B, Scolnick JA, Marchetto
signaling in the vertebrate retina. Brain Res 1192:6175. MC, Muotri AR, Carson C, Gage FH (2008) Multiple lay-
Walworth NC (2000) Cell cycle checkpoint kinases: checking in ers of molecular controls modulate self-renewal and neuronal
on the cell cycle. Curr Opin Cell Biol 12:697704. lineage specification of embryonic stem cells. Hum Mol
Wamsley JK, Palacios JM, Kuhar MJ (1981) Autoradiographic Genet 17:R67R75.
localization of opioid receptors in the mammalian retina. Yew DT, Luo CB, Zheng DR, Guan YL, Tsang D, Stadlin
Neurosci Lett 27:1924. A (1991) Immunohistochemical localization of substance P,
Wang X, Fu S, Wang Y, Yu P, Hu J, Gu W, Xu XM, enkephalin and serotonin in the developing human retina. J
Lu P (2007) Interleukin-1beta mediates proliferation and Hirnforsch 32:6167.
230 R. Linden et al.

Yoshida H, Kong YY, Yoshida R, Elia AJ, Hakem A, Hakem R, cyclooxygenase-2 in the blood and brain. Neuroscience
Penninger JM, Mak TW (1998) Apaf1 is required for mito- 157:895907.
chondrial pathways of apoptosis and brain development. Cell Zhang J, Gray J, Wu L, Leone G, Rowan S, Cepko CL, Zhu
94:739750. X, Craft CM, Dyer MA (2004a) Rb regulates proliferation
Yoshikawa K (2000) Cell cycle regulators in neural stem cells and rod photoreceptor development in the mouse retina. Nat
and postmitotic neurons. Neurosci Res 37:114. Genet 36:351360.
Young RW (1985a) Cell proliferation during postnatal develop- Zhang J, Schweers B, Dyer MA (2004b) The first knockout
ment of the retina in the mouse. Brain Res 353:229239. mouse model of retinoblastoma. Cell Cycle 3:952959.
Young RW (1985b) Cell differentiation in the retina of the Zhang L, Spigelman I, Carlen PL (1991) Development of
mouse. Anat Rec 212:199205. GABA-mediated, chloride-dependent inhibition in CA1
Young TL, Cepko CL (2004) A role for ligand-gated ion chan- pyramidal neurones of immature rat hippocampal slices.
nels in rod photoreceptor development. Neuron 41:867879. J Physiol 444:2549.
Yu J, Zhang L (2003) No PUMA, no death: implications for p53- Zhao S, Barnstable CJ (1996) Differential effects of bFGF on
dependent apoptosis. Cancer Cell 4:248249. development of the rat retina. Brain Res 723:169176.
Zarbin MA, Wamsley JK, Palacios JM, Kuhar MJ (1986) Zhao JJ, Lemke G (1998) Selective disruption of neuregulin-1
Autoradiographic localization of high affinity GABA, benzo- function in vertebrate embryos using ribozyme-tRNA trans-
diazepine, dopaminergic, adrenergic and muscarinic cholin- genes. Development 125:18991907.
ergic receptors in the rat, monkey and human retina. Brain Zhu CY, Shukla SD (1993) Increased tyrosine kinase activity
Res 374:7592. in pp60c-src immunoprecipitate from platelet activating fac-
Zezula J, Freissmuth M (2008) The A2A-adenosine receptor: tor stimulated human platelets: in vitro phosphorylation of a
a GPCR with unique features? Br J Pharmacol 153:S184 synthetic peptide. Life Sci 53:175183.
S190. Zygar CA, Colbert S, Yang D, Fernald RD (2005) IGF-1
Zhang H, Ching S, Chen Q, Li Q, An Y, Quan N (2008) produced by cone photoreceptors regulates rod progenitor
Localized inflammation in peripheral tissue signals the CNS proliferation in the teleost retina. Brain Res Dev Brain Res
for sickness response in the absence of interleukin-1 and 154:91100.
Chapter 14

Potential Application of Very Small Embryonic Like (VSEL)


Stem Cells in Neural Regeneration

Mariusz Z. Ratajczak, Ewa Zuba-Surma, Magda Kucia, Przemyslaw Nowacki,


and Bogdan Machalinski

Contents mice, the stroke-related stress may trigger mobiliza-


tion of these cells into peripheral blood. We noticed
14.1 Introduction . . . . . . . . . . . . . . . . . 232 in stroke patients an increase in the number of circu-
14.2 Identification of Very Small Embryonic Like lating primitive cells expressing the VSEL phenotype.
Stem Cells (VSEL) in Adult Murine Bone Additionally, we noticed a positive correlation between
Marrow . . . . . . . . . . . . . . . . . . . 233
stroke extensiveness, SDF-1 concentration in serum,
14.3 Identification of VSEL in Adult Murine Organs
Including Adult Brain . . . . . . . . . . . . 234 and the number of VSEL circulating in the peripheral
14.4 Bone-Marrow-Derived VSEL as Population of blood. We conclude that VSEL could have a potential
Circulating Pluripotent Stem Cells . . . . . . 237 prognostic value in stroke patients, and more impor-
14.5 Biological Properties of VSEL . . . . . . . . 239
tant that their role in brain regeneration requires further
14.6 Cells that Express VSEL Markers are Mobilized
into PB in Patients After Stroke . . . . . . . 240 study.
14.7 Conclusions . . . . . . . . . . . . . . . . . 241
References . . . . . . . . . . . . . . . . . . . . 241 Keywords CXCR4 Mobilization Pluripotent stem
cells Stroke VSEL

Abstract We found that murine bone marrow con-


tains a mobile population of Oct-4+ CXCR4+ SSEA- Abbreviations
1+ Sca-1+ lin CD45 very small embryonic like stem
cells (VSEL) that are mobilized into peripheral blood 7-AAD 7-aminoactinomycin D
(PB) e.g., after pharmacological mobilization with BM bone marrow
G-CSF or in murine model of stroke. We postu- BMNC bone-marrow nucleated cells
late that, VSEL are a mobile population of epi- CB cord blood
blast/germ line derived stem cells and play an CXCR4 CXC chemokine receptor 4
important role as organ-residing reserve population ESC embryonic stem cells
of pluripotent stem cells that gives rise to stem FACS fluorescence activated cell sorting
cells committed to particular organs and tissues FSC forward scatter
G-CSF granulocyte colony stimulating factor
including neural tissue. Moreover, since a similar
GFAP glial fibrillary acidic protein
population of small CXCR4+ CD133+ CD34+ SSEA-
HGF hepatocyte growth factor
4+ Oct-4+ lin CD45 cells resides also in human bone
HGF/SF hepatocyte growth factor/scatter factor
marrow, we asked whether in humans, similarly as in
HSC hematopoietic stem cells
ISS ImageStream
LACI lacunar infarcts
LIF leukemia inhibitory factor
M.Z. Ratajczak ()
Stem Cell Institute at James Graham Brown Cancer Center, MAPC multipotent adult progenitor cells
University of Louisville, Louisville, KY, USA MSC mesenchymal stem cells
e-mail: mzrata01@louisville.edu PACI partial anterior circulation infarcts

H. Ulrich (ed.), Perspectives of Stem Cells, 231


DOI 10.1007/978-90-481-3375-8_14, Springer Science+Business Media B.V. 2010
232 M.Z. Ratajczak et al.

PB peripheral blood developed brains and brain-derived cells from these


PBNC peripheral blood nucleated cells animals are able to grow neurospheres (Walzlein et al.,
PGC primordial germ cells 2008). This suggests a potential involvement in brain
POCI posterior circulation infarcts development and contribution to neural tissue home-
PSC pluripotent stem cells(s) ostasis, stem cells that reside in other locations. These
RBC red blood cells cells could reside in other brain areas or translocate
SC stem cell to the brain using peripheral blood (PB) from other
SDF-1 stromal derived factor-1 organs/tissues most likely bone marrow (BM) (Hess
SSC side scatter and Borlongan, 2008; Shyu et al., 2006).
SSEA-1 stage specific embryonic antigen-1 To support this last notion, it has been shown that in
TACI total anterior ciculation infarcts
response to organ damage, stem cells could be mobi-
TC computer tomography
lized into PB from BM and other tissue-specific niches
VEGF vascular endothelial growth factor
as well. These circulating stem cells potentially would
VSEL very small embryonic like stem cell(s)
home to the damaged tissues and attempt to contribute
VSEL-DS very small embryonic like stem
cells-derived spheres to regeneration (Shyu et al., 2006; Quesenberry et al.,
2007). We observed that the number of circulating
stem cells expressing pluripotent and tissue-specific
markers increases in PB after granulocyte-colony
14.1 Introduction stimulating factor (G-CSF) administration, heart
infarct in humans and mice, and in murine model of
The occlusion of a cerebral artery leads to focal stroke (Kucia et al., 2004a, b, 2008b; Wojakowski
ischemia and stroke that results in damage to neu- et al., 2004; Zuba-Surma et al., 2008b). While these
rons and glial cells (Dirnagl et al., 1999; Endres cells may act somehow in regeneration from small tis-
and Dirnagl, 2002). Thus the goal of regenerative sue injuries, their contribution toward repairing more
medicine is to ameliorate irreversible destruction of extensive tissue damages requires further study. Larger
brain tissue by harnessing the power of stem cells in injuries (e.g., massive stroke) by creating highly pro-
process of neurogenesis (Kalluri and Dempsey, 2008; teolytic environment may perturb mechanisms that
Komitova et al., 2005). Neurogenesis supplies new regulate chemotaxis and homing of these cells to the
neurons and glial cells (astrocytes and oligodendro- damaged organ.
cytes) and occurs in the mammalian brain throughout Recently, we identified a population of very small
life, and so far has been clearly demonstrated at two embryonic-like (VSEL) stem cells in adult murine BM
locations in subventricular zone of the lateral ven- that express several pluripotent stem cells (PSC) mark-
tricles and olfactory bulb as well as in the subgranu- ers and purified them at the single cell level (Kucia
lar zone of the dentate gyrus in hippocampus (Duan et al., 2006a; Zuba-Surma et al., 2008a). We noticed
et al., 2008; Encinas and Enikolopov, 2008). Data from that VSEL are mobilized into PB after G-CSF admin-
mouse and rats have shown that stroke and subsequent istration, during toxic liver and skeletal muscle damage
death of neurons leads to increased proliferation of in mice (Kucia et al., 2008b) and after heart infarct and
neural precursors that are located in the subventricu- stroke both in humans and mice (Wojakowski et al.,
lar zone, olfactory bulb and hippocampus (Komitova 2004; Zuba-Surma et al., 2008b). VSEL are present in
et al., 2005; Zhang et al., 2001). Unfortunately, this versatile adult organs and interestingly, our data indi-
response is not effective enough to fully restore mor- cates that the brain contains a high number of cells that
phology and function of neural tissue damaged by display VSEL phenotype (Ratajczak et al., 2008b, d).
stroke. Thus the role of both circulating primitive VSEL as
Whether neurogenesis also occurs in areas of the well as those VSEL that reside in normal brain in
mammalian brain other than hippocampus and sub- regeneration of damaged neural tissue requires further
ventricular region remains controversial. Interestingly, studies. In this chapter we will present the concept that
mice with cyclin D2 deficiency that have severely these cells could be employed as a prognostic marker
reduced number of stem cells in subventricular zone, as well as could be harnessed in regenerative medicine
olfactory bulb and hippocampus, yet possess normally as a potential source of stem cells for neurogenesis.
14 Potential Application of VSEL Stem Cells in Neural Regeneration 233

14.2 Identification of Very Small unorganized chromatin (euchromatin) (Zuba-Surma


Embryonic Like Stem Cells (VSEL) et al., 2008a; Kucia et al., 2007b).
An example of this novel fluorescence activated cell
in Adult Murine Bone Marrow
sorting (FACS) approach controlled by size bead mark-
ers is shown at Fig. 14.1. The first step in this strategy
Based on our initial work suggesting that BM may was to gate in regions containing small events (2
contain unusually small cells (36 m in diame- 10 m) shown as region R1 on the dot plot (Fig.
ter) that are (i) negative for lineage markers (lin ), 14.1 panel A and B). This region mostly contains cell
(ii) CD45 , (iii) express Sca-1 antigen (mouse) and debris, erythrocytes and large blood platelets but also
CXC chemokine receptor 4 (CXCR4), CD133, CD34 as we expected includes some rare nucleated small
antigens (mouse and humans) and (iv) are pos- cells. The events enclosed in region R1 (Fig. 14.1
itive for markers of PSC (Oct-4, SSEA) (Kucia panel A and B), which include an average of ~50%
et al., 2006a; Zuba-Surma et al., 2008a; Kucia et al., of total events scored by cytometer, are further ana-
2007a) we develop a sorting strategy of BM- lyzed for the expression of Sca-1 and lineage markers
derived cells controlled by the beads with prede- (lin). The Sca-1+ /Lin events shown in region R2 (Fig.
fined sizes (1, 2, 4, 6, 10, and 15 m in diameter) 14.1 panel D) consist of 0.30 0.05% of total analyzed
(Zuba-Surma et al., 2008a). Generally, this strategy BM nucleated cells on average. Cells from region R2
allowed us to sort cells (i) very small in size (~3 are subsequently sorted according to the expression
5 m and 46 m mouse and human respec- of CD45 antigen as Sca-1+ /lin /CD45 (region R3)
tively); (ii) Oct-4+ CXCR4+ SSEA-1+ Sca-1+ CD45 lin and Sca-1+ /lin /CD45+ (region R4) subpopulations
(mouse) Oct-4+ CXCR4+ SSEA-4+ CD133+ CD45 lin (Fig. 14.1 panel C).
(human) (iii) possessing large nuclei containing

A B C
Lineage
SSC

SSC

Beads in size:
2-10 m

R2
0.300.05%
R1 R1 47.2 2.1%

FSC FSC Sca-1

R1 & R2 & R3 D
VSEL R3 R4
- - +
CD45 /Lin /Sca-1 0.03 0.01% 0.35 0.06%
Counts

R1 & R2 & R4
HSC
+ - +
CD45 /Lin /Sca-1
CD45

Fig. 14.1 Gating strategy for VSEL sorting by FACS. BM- region R1 were further analyzed for Sca-1 and Lin expres-
derived VSEL were isolated from immunofluorescence stained sion and only Sca-1+ /Lin events were included into region R2.
murine BM nucleated cells by FACS. Panel A: Agranular, Population from region R2 was subsequently sorted based on
small events ranging from 2 to 10 m were included into gate CD45 antigen expression into CD45 and CD45+ subpopula-
R1 after comparison with six differently sized bead particles tions visualized on histogram (Panel D, regions R3 and R4,
with standard diameters of 1, 2, 4, 6, 10, and 15 m (Flow respectively). Sca-1+ /Lin /CD45 cells (VSEL) were sorted as
Cytometry Size beads, Invitrogen; Molecular Probes, Carlsbad, events enclosed in logical gate including regions R1, R2, and
CA, USA). Panel B: BM nucleated cells were visualized by R3, while Sca-1+ /Lin /CD45+ cells (HSC) from gate including
dot plot showing forward scatter (FSC) vs. side scatter (SSC) regions R1, R2, and R4. Percentages show the average content
signals, that are parameters related to the size and granular- of each cellular subpopulation ( SEM) in total BM nucleated
ity/complexity of the cell, respectively. Panel C: Cells from cells
234 M.Z. Ratajczak et al.

The first population shown in R3 (Sca-1+ /lin / (7-AAD) cells to visualize better nucleated cells
CD45 ) contains VSEL that highly express early (Fig. 14.2 panel A). By employing this staining,
embryonic markers Oct-4 and SSEA-1 as revealed by the nuclear area was calculated by automatic masks
subsequent immunofluorescence analysis, and a sec- created on the 7-AAD nuclear images (morphology
ond one shown in R4 (Sca-1+ /lin /CD45+ ) is highly mask). Cytoplasmic area was computed by subtract-
enriched for hematopoietic stem cells (HSC). Direct ing the nuclear area from the total area of the cell.
transmission electron microscopy (TEM) analysis con- The N/C ratio was computed as the ratio between
firmed that Sca-1+ /lin /CD45 cells display several nuclear and cytoplasmic areas. Moreover, based on
features typical for embryonic stem cells such as small morphological features of acquired objects and their
size, a large nucleus surrounded by a narrow rim direct microscopic visualization by the ISS, we were
of cytoplasm, and open-type chromatin (euchromatin) able to exclude debris and artifacts and nucleated,
(Kucia et al., 2006a). In contrast Sca-1+ lin CD45+ intact cells only were selected for further analysis
cells display heterogeneous morphology and are larger. (Fig. 14.2).
They measure on average 810 m in diameter; By employing ISS analysis that allows the analysis
possess scattered chromatin and prominent nucleoli of gallery of pictures of acquired cells, we confirmed
(Kucia et al., 2006a). We found that VSEL com- with greater precision that murine BM-derived VSEL
prise ~0.03% while HSC are ~0.35% of total BM are ~3.6 m in diameter, while Sca-1+ /lin /CD45+
nucleated cells (Fig. 14.1 panel C). We also noticed HSC are larger at ~6.5 m in diameter (Zuba-Surma
that 95% of Sca-1+ /lin /CD45 (VSEL) are located et al., 2008a). We also noticed that VSEL have signif-
within the 26 m size range, while 86% of Sca- icantly higher nuclear/cytoplasmic ratio as compared
1+ /lin /CD45+ (HSC) are found in the 610 m size with HSC (1.5 0.2 and 0.8 0.03, respectively).
range (Zuba-Surma et al., 2008a). Thus, by employ- Furthermore, VSEL had significantly lower cytoplas-
ing flow cytometry and the size marker beads, we mic area as compared with HSC (5.4 0.6 and
have confirmed that the majority of Sca-1+ /lin /CD45 33.8 1.7, respectively) (Zuba-Surma et al., 2008a).
cells isolated from adult BM are unusually small cells Despite their small size, VSEL when evaluated by
(<6 m) (Zuba-Surma et al., 2008a), and as expected FACS for DNA content possess diploid DNA. They
similar to those observed in our previous chemotactic do not express MHC-I and human leukocyte antigen-
isolation experiments. D related (HLA-DR) antigens on the surface and
In the next step a novel imaging strategy called are CD90 CD105 CD29 (Kucia et al., 2008a).
ImageStream system (ISS) analysis was employed to Furthermore, as directly confirmed by ISS analysis
better enumerate and evaluate morphological features BM-derived VSEL are larger than peripheral blood
of VSEL (Zuba-Surma et al., 2008a). This novel ISS- platelets and smaller than erythrocytes (Fig. 14.3).
based analysis involves flow cytometry combined with
microscopy and allows for statistical analysis of var-
ious cellular parameters as well as visualization of
cells in suspension during flow analysis via high res- 14.3 Identification of VSEL in Adult
olution brightfield, darkfield and fluorescence images
Murine Organs Including Adult
(Basiji et al., 2007; Ortyn et al., 2006; Zuba-Surma
et al., 2007, 2008c). The high resolution of ISS imag- Brain
ing enables the identification of objects even as small
as 1 m in diameter. Thus this imaging strategy is ideal As described in previous paragraph, VSEL isolated
to analyze small cellular events in a range of 26 m from murine BM are Oct-4 positive subpopulation
on a flow cytometry cytogram, where VSEL are to be of Sca-1+ Lin CD45 cells. Thus in the next step
found (Zuba-Surma et al., 2008a). we evaluated a number of VSEL in versatile adult
Thus, we employed ISS to verify the results murine organs (Ratajczak et al., 2008d, a). In order
obtained from flow classical FACS analysis to visu- to analyze adult organs for the presence of Sca-1+
alize better events of interest. For ISS analysis, BM lin CD45 cells, mice were perfused to remove resid-
nucleated cells (BMNC) were fixed and stained with ual VSEL that as we described may also circulate
DNA-binding fluorescence dye 7-aminoactinomycin at a very low level in peripheral blood. Removed
14 Potential Application of VSEL Stem Cells in Neural Regeneration 235

A VSEL 3.63 0.27 m

10m

RBC 4.94 0.46 m Platelets 2.19 0.71 m

10m 10m

B RBC VSEL Platelets

Synthetic beads
10m 6m 4m 2m 1m

Fig. 14.2 Morphological comparison between murine show cellular structure containing nuclei. Average size of each
VSEL, HSC, erythrocytes, and platelets by ISS. Panel A population was computed as the length of the minor cellular axis
presents murine VSEL stained for Sca-1 (FITC, green), Lin based on the mask covering the brightfield image of the cell
(PE, orange), and CD45 (PE-Cy5, magenta), and blood-derived (Mean SEM). Scale bars show 10 m. Panel B shows cells
erythrocytes and platelets stained for Ter119 (PE, orange) from all three populations (VSEL, red blood cells [RBC] and
and CD41 (FITC, green), respectively. Following fixation, all platelets) when compared with size predefined beads in the same
samples were stained with 7-AAD (red) to visualize nuclei. scale
Erythrocytes and platelets do not possess nuclei, while VSEL

organs were subsequently enzymatically homogenized positive cells from falsely positive artifacts (Ratajczak
and isolated nucleated cells washed and stained with et al., 2008d, a). As expected we detected a presence of
specific antibodies for flow cytometric and ISS anal- small nucleated Oct-4+ Sca-1+ lin CD45 cells in all
ysis. Flow cytometric analyses were performed on analyzed organs and tissues including brain (Fig. 14.4).
freshly isolated or fixed cells co-stained as in case W found that pancreas, brain, skeletal muscles and kid-
of BM-derived cells with nuclear DNA-binding dye neys are the organs with the highest percent content of
7-AAD. Inclusion of 7-ADD into the staining pro- these cells (0.3300.099, 0.1100.027, 0.0820.018
tocol of fixed cells as mentioned above allowed us and 0.0560.004%, respectively), while bone mar-
to visualize real nucleated events and exclude arti- row, thymus and spleen contain the lowest percentage
facts. By employing classical flow cytometry and ISS of Oct-4+ Sca-1+ Lin CD45 cells (0.00180.0003,
we found that all analyzed organs contain popula- 0.00180.0003 and 0.0050.001%, respectively).
tion of Sca-1+ lin CD45 cells. Furthermore, inclu- Next, we calculated the absolute number of these
sion of nuclear dye (7-ADD) into staining protocol cells/organ (Table 14.1). We found that the high-
significantly improved the accuracy of our analysis est number of small nucleated Oct-4+ Sca-1+ lin
by distinguishing nucleated objects from anucleated CD45 cells/organ is in the brain, kidneys, skeletal
7-ADD-negative debris. ISS allowing visualization of muscles, pancreas and bone marrow (43.9712.38,
acquired objects by showing their real images gave us 19.872.03, 15.186.79, 9.414.71 and 8.392.00
opportunity to not only distinguish nucleated objects 103 , respectively). On the other hand, heart, thy-
from anucleated debris, but as mentioned above also mus, testes and spleen contained the lowest numbers of
236 M.Z. Ratajczak et al.

A a) b)
700
B
a) Sca-1+/Lin-/CD45- cell

AR of Brightfield
1 R1 -
600
R2 CD45
R8
.8

Frequency
R1 500

.6 400

300
.4
200
.2
100

0
Single cells b) Negative cell with positive fragment
0
0 2 00 40 0 600 8 00 1e3 -100 0 0 1 000 1e4 1e 5 1e6

Area of nucleus (7-AAD) CD45 (PE-Cy5)


c) d)
250 90
c) Dead/ degrading cell
200 R3 R7 -
Lin Lin-/CD45-/Sca-1+
Frequency

Frequency
60
150 R4
100
30
50
d) Cellular fragment/ debris
0 0
-100 0 0 1 000 1e4 1 e5 1 e6 0 10 00 1e4 1e 5 1 e6

Lineage (PE) Sca-1 (FITC)

a) b) c) d)

Fig. 14.3 ImageStream system analysis of kidney- derived of CD45 (Panel A.b) and Lin (Panel A.c). Lin /CD45 objects
Sca-1+ /Lin /CD45 cells. Panel A shows analysis of whole from logical gate including regions R2 and R3 are visualized
population of cells isolated from kidney following enzymatic on the histogram presenting their Sca-1 expression (Panel A.d).
digestion of the tissue and the staining for Sca-1, CD45 and Sca-1+ /Lin /CD45 cells are included in region R4. Panel B
hematopoietic lineages markers (Lin). (Panel A.a) shows all shows normal, nucleated Sca-1+ /Lin /CD45 cells (Panel B.a)
acquired objects according to their morphological parameters when compared to the objects imitating them phenotypically.
including the area of the nucleus and the aspect ratio (AR) of Selected images of falsely positive artifacts (Panel B.b), dam-
brightfield. The aspect ratio (AR) is calculated based on bright- aged/degradating cells (Panel B.c) and cellular debris (Panel
field image of each cell as the ratio of cellular minor axis (width) B.d) are shown on the next panels. Each photograph presents
to major axis (height). Round, non-elongated cells possess an brightfield image as well as separate fluorescent images related
aspect ratio close to 1.0, while the elongated cells or clumps to nuclear image (7-AAD; red) and expression of Sca-1 (FITC,
lower than 1.0. Round, single cells containing DNA are included green), Lin (PE; orange) and CD45 (PE-Cy5; yellow). Moreover
in region R1 for further analysis. Objects from Region R1 are magnified combined images of each presented object are shown
subsequently visualized on histograms based on the expression on the lower panel. The scale bar indicates 10 m

these cells/organ (1.350.56, 2.030.37, 2.381.25 and heart (3.780.64 and 4.740.93 m, respectively)
and 3.860.43 103 , respectively) (Table 14.1). (Zuba-Surma et al., 2008a). At the same time Oct-
Thus, by employing flow cytometry- and ISS-based 4+ Sca-1+ Lin CD45 cells detected in other organs
approaches, we identified Oct-4+ cells correspond- were also small (<6 m). Furthermore, relatively high
ing phenotypically to VSEL in various adult organs nuclear to cytoplasmic ratio confirmed primitive char-
(Ratajczak et al., 2008d, a). Furthermore, ISS allowed acter of analyzed populations. BM Bone marrow and
us again to distinguish Oct-4+ Sca-1+ lin CD45 testes were identified as organs containing Oct-4+ Sca-
VSEL from cellular debris and artifacts commonly 1+ Lin CD45 cells with the highest N/C (2.120.33
present in enzymatically digested samples. and 2.110.40, respectively). Cells with the lower N/C
In the next step, we employed collected images of ratio were observed in all other organs. The presence of
these cells to calculate their morphological features small Oct-4+ Sca-1+ lin CD45 cells detected by ISS
such as average size and nuclear to cytoplasmic (N/C) was subsequently confirmed by confocal microscopy
ratio which were computed based on brightfield and on Sca-1+ lin CD45 cells sorted from various organs
nuclear images of cells. We noticed that the small- and we identified small (<5m) Oct-4+ cells with the
est Oct-4+ Sca-1+ lin CD45 cells reside in the BM VSEL phenotype in all organs tested (Ratajczak et al.,
14 Potential Application of VSEL Stem Cells in Neural Regeneration 237

A Heart (1.1%) a highest number of Oct-4 mRNA transcripts in Sca-1+


Thymus (1.7%) Lin CD45 cells isolated from BM.
100% Testes (2.0%)
Spleen (3.3%)
80% Lungs (4.5%)
Liver (5.9%)
60% 14.4 Bone-Marrow-Derived VSEL
Bone marrow (7.1%)
40% Pancreas (7.9%) as Population of Circulating
S. Muscles (12.8%) Pluripotent Stem Cells
20% Kidneys (16.7%)
Brain (37.0%)
0%
The peripheral blood could be envisioned as a high-
Lin/
way by which stem cells are trafficking in the body to
B BF Oct-4 CD45 7-AAD Sca-1 combo keep in balance a pool of stem cells located in different
niches in peripheral tissues (Fig. 14.5). In this con-
Bone marrow 10m
text, BM has been proposed to be a main reservoir that
Brain contains and releases during mobilization these circu-
lating cells (Levesque et al., 2007; Moog, 2006). To
Pancreas
support this it is well known that BM secretes several
Kidneys chemoattractants not only for HSC but also for other
Liver
non-hematopoietic stem cells including VSEL e.g.,
stromal derived factor-1 (SDF-1), hepatocyte growth
factor/scatter factor (HGF/SF), leukemia inhibitory
Fig. 14.4 Oct-4 + VSEL present in adult murine tis-
sues. Panel A shows percentage distribution of Oct-4+ /Sca-
factor (LIF), and vascular endothelial growth fac-
1+ /Lin /CD45 VSEL between analyzed organs which was tor (VEGF) (Ratajczak et al., 2006; Hill et al.,
computed based on the ImageStream system analysis and their 2004; Ponomaryov et al., 2000; Tacchini et al.,
absolute numbers in each tissue. Panel B presents representative 2003). Therefore, stem cells circulate in PB both
images of small Oct-4+ VSEL identified in selected analyzed
organs by ISS. VSEL are stained for Oct-4 expression (FITC,
during fetal development or postnatal while relo-
green). They are negative for Lin markers and CD45 (PE, cating between tissue specific niches may become
orange) and express Sca-1 (PE-Cy5, magenta). Scale bars show chemoattracted from circulating PB to settle down
10 m in BM, where they find a permissive environment.
This also explains why in addition to HSC, several
2008d, a). Furthermore, Oct-4 expression was also other types of non-hematopoietic stem cells have been
confirmed at mRNA level by employing normal RT- described in the BM including mesenchymal stem cells
PCR and real-time PCR performed on sorted by FACS (MSC) (Peister et al., 2004; Prockop, 1997), multi-
populations. Of note, real-time-PCR analysis revealed potent adult progenitor cells (MAPC) (Jiang et al.,

Table 14.1 Content of Absolute number of Oct-4+ VSEL [103 ]


Oct-4+ /Sca-1+ /Lin /CD45
Organ/tissue Per organ Per 1 g of tissue
cells in adult murine organs
and tissues analyzed by ISS. MeanSEM MeanSEM
Cells were isolated from Bone marrow 8.392.00 10.912.61
tissues of adult 48 week old Thymus 2.030.37 24.444.42
mice (C57BL/6) by enzymatic Spleen 3.860.43 47.105.21
digestion. Analysis was Pancreas 9.414.71 27.5313.78
performed three times (N=3) Brain 43.9712.38 116.0132.67
on samples prepared from
Kidneys 19.872.03 46.104.70
organs of three independent
animals. In case of geminate Lungs 5.390.33 34.112.07
organs, the absolute numbers Heart 1.350.56 9.934.13
of cells were calculated per Skeletal m. 15.186.79 4.161.86
one animal Testes 2.381.25 9.5510.04
Liver 6.981.38 4.790.95
238 M.Z. Ratajczak et al.

Circulation of Stem Cells in Blood


Network of Interactions between
Stem Cells and Niches in Peripheral
Tissues

Gradient of Stem Cells


Chemoatractants: with Receptors:

SDF 1
SDF-1 CXCR4
HGF C-met
LIF LIFR
VEGF VEGFRs

Fig. 14.5 BM-derived VSEL circulate in peripheral blood. hiding place for both HSC and circulating CXCR4+ non-HSC
The concept is presented based on the assumption that CXCR4+ (e.g. EPC, MSC and VSEL). These cells are deposited early in
VSEL circulate in the PB. Although the percentage of these cells development and consequently reside in the BM. We hypothe-
circulating in PB is much lower than that of early hematopoietic size that they could serve an important role in tissue/organ repair
stem/progenitor cells, these mobilized non-HSC may have an as a mobile reserve pool of PSC. Besides the SDF-1-CXCR4
important role in tissue repair following injury. The mobilization axis, the trafficking of these cells involves several other moto-
of these cells occurs during tissue damage or injury (e.g., stroke, morphogens such as HGF, VEGF, and leukemia inhibitory factor
heart infarct, toxic liver damage). It is postulated that after (LIF) that create unique network guiding the circulation of stem
mobilization into PB, CXCR4+ non-HSC may subsequently be cells between various tissues/ organs through the blood stream
chemoattracted to the damaged tissues by the gradient of SDF-
1 and other chemokines. In this context, BM tissue becomes a

2002), marrow-isolated adult multilineage inducible and LIF gradient but their abundance and responsive-
(MIAMI) (DIppolito et al., 2004), multipotent adult ness to gradients of SDF-1, HGF, and LIF, decreases
stem cells (Beltrami et al., 2007) and identified by our with age (Kucia et al., 2006a, b; Zuba-Surma et al.,
group VSEL (Kucia et al., 2006a, 2007a). 2009). Subsequent flow cytometric analysis, com-
The circulating BM-derived stem/progenitor cells bined with analysis of neural markers at the mRNA
could play an important role in turn-over of stem cells and protein levels, revealed that these cells reside in
in peripheral tissues. In accordance with this notion we the non-hematopoietic CXCR4+ Sca-1+ lin CD45
recently reported that in mice after an experimental BMNC fraction that is enriched in VSEL (Kucia et al.,
stroke or heart infarct a number of cells circulat- 2006a).
ing in PB (Zuba-Surma et al., 2008b; Kucia et al., In the murine model of stroke, we demonstrated
2006b), enriched for several tissue specific develop- that the expression of SDF-1 around stroke area as
mental markers including neural ones as well as PSC well as serum SDF-1 level increase during the first
markers increases. Accordingly, we noticed that in week after stroke and that SDF-1 was one of the
mice after an experimental stroke these cells not only important chemoattractants for recruiting stem cells to
express neural lineage markers (-III-tubulin, nestin, supernatants from damaged brain tissue (Kucia et al.,
neuronal nuclear antigen [NeuN] and glial fibrillary 2006b). Of note, the importance of SDF-1-CXCR4
acidic protein [GFAP]) and PSC marker Oct-4, but axis in development of brain has been demonstrated
more importantly form neurospheres in vitro (Zuba- in murine embryos with CXCR4 or SDF-1 knockout
Surma et al., 2008b; Kucia et al., 2006b). These which display several defects in their ability to develop
cells are present in significant amounts in BM har- BM, brain, and cerebellum tissues (Vilz et al., 2005;
vested from young mice and respond to SDF-1, HGF, Zhu et al., 2002). Thus, SDF-1 signaling regulates
14 Potential Application of VSEL Stem Cells in Neural Regeneration 239

key events during early cortical development in vitro able to grow neurospheres in co-cultures with bone
and maintains proliferation of cortical progenitors, marrow derived mesenchymal stem cells (Kucia et al.,
possibly through a mechanism involving connexin 43- 2006a).
mediated intercellular coupling (Pritchett et al., 2007). Interestingly, formation of VSEL-DS was associ-
Moreover, SDF-1 signaling upregulates the differenti- ated with a young age in mice and no VSEL-DS
ation of cortical gamma-aminobutyric acid outputting were observed in cells isolated from older mice (> 2
(GABA-ergic) neurons, which is independent of the years) (Kucia et al., 2006a; Zuba-Surma et al., 2009).
sonic signaling pathway and enables the elongation This age-dependent content of VSEL in BM may
and branching of axons of cortical glutamatergic neu- explain why the regeneration process is more efficient
rons (Bhattacharyya et al., 2008; Kasiyanov et al., in younger individuals. There are also differences in
2008). Furthermore, we noticed that in addition to the content of these cells among BMNC between long-
SDF-1 also HGF/SF and LIF are important chemoat- and short-lived mouse strains. The concentration of
tractants for recruiting VSEL to supernatants from these cells is much higher in BM of long-lived (e.g.,
damaged brain tissue (Ratajczak et al., 2006; Kucia C57Bl6) as compared to short-lived (DBA/2J) mice
et al., 2006b). (Kucia et al., 2006a). This suggests that some genes
In sum, we demonstrated that VSEL are mobi- could be responsible for developmental tissue distribu-
lized into the peripheral blood following stroke and tion and expansion of these cells, and that these genes
are subsequently chemoattracted to the damaged neu- could be involved in controlling the regeneration abil-
ral tissue in an SDF-1-CXCR4-, HGF/SF-c-Met-, and ities (e.g., for neural tissues) and thus mammalian life
LIF-LIF-receptor-dependent manner. This supports a span.
concept that chemotactic factors that are upregulated in Since VSEL express several markers of primor-
damaged brain orchestrate release of VSEL from BM dial germ cells (PGC) such as fetal-type alkaline
into PB. phosphatase, Oct-4, SSEA-1, CXCR4, Mvh, Stella,
Fragilis, Nobox and Hdac6, we envision that they
could be closely related to a population of epiblast-
derived PGC, a very first population of stem cells,
14.5 Biological Properties of VSEL that is specified in developing embryo during early
embryogenesis (Kucia et al., 2006a; Ratajczak et al.,
VSEL residing in adult tissues and circulating in 2008c). VSEL are also highly mobile and respond
mobilized peripheral blood could be potentially har- robustly to an SDF-1 gradient, adhere to fibronectin
nessed to regenerate damaged organs. We noticed and fibrinogen, and may interact with BM-derived
that these cells proliferate/differentiate relatively eas- stromal fibroblasts. Confocal microscopy and time-
ily in co-cultures with other adherent cell populations lapse studies revealed that these cells attach rapidly
(e.g., C2C12 myoblasts, mesenchymal stem cells or to, migrate beneath, and undergo emperipolesis in
OP-9 stroma cells) (Kucia et al., 2006a; Ratajczak marrow-derived fibroblasts (Ratajczak et al., 2008c,
et al., 2008c). For example, if plated over a C2C12 2007). Since fibroblasts secrete SDF-1 that as men-
murine sarcoma cell feeder layer, ~510% of puri- tioned above binds to CXCR4 receptor, they may cre-
fied VSEL are able to form spheres that resemble ate a homing environment for small CXCR4+ VSEL.
embryoid bodies (Kucia et al., 2006a). Cells from This robust interaction of VSEL with BM-derived
these VSEL-derived spheres (VSEL-DS) are com- fibroblasts has an important implication, namely that
posed of immature cells with large nuclei containing isolated BM stromal cells may be contaminated by
euchromatin and are CXCR4+ SSEA-1+ Oct-4+ , just these tiny cells from the beginning. This observation
like purified VSEL. Cells in these spheres however may somehow explain the unexpected plasticity of
already enlarge and show some signs of differentia- marrow-derived fibroblastic cells, e.g., mesenchymal
tion. More important, cells from VSEL-DS are capable stem cells (MSC) or MAPC. Recently, we noticed that
to differentiate into all three germ layers (e.g., neu- similar counterpart of murine VSEL is also present
ral tissue, myocardium and pancreas) (Kucia et al., in the human CB and BM. Furthermore, similar cells
2006a). Furthermore, as reported both murine BM- increases in number and circulate in PB in patients
derived and human cord blood (CB)-derived VSEL are after acute myocardial infarction.
240 M.Z. Ratajczak et al.

14.6 Cells that Express VSEL Markers 12


are Mobilized into PB in Patients control PBNC
After Stroke 10 24h
3rd day after stroke
We become interested if VSEL could be mobilized into 7th day
PB in patients after stroke, similarly seen in patients 8

mRNA (fold difference)


P<0.001 vs control
after acute heart infarct. To address this question,
a total of 44 patients afflicted with ischemic stroke
that were admitted within 24 h of the first symptom 6
onset and 22 healthy control subjects were enrolled.
In each case, the stroke had been precisely docu-
mented in clinic by computer tomography (CT) scan. 4
Based on clinical examination and cranial CT findings,
including volumetric analysis, patients were classi-
fied into the following four clinical subgroups: total 2

anterior circulation infarcts (TACI), partial anterior cir-


culation infarcts (PACI), posterior circulation infarcts
0
(POCI), and lacunar infarcts (LACI). Additionally, we Oct4 GFAP
distinguished two subgroups of patients with differ-
ent extensiveness of stroke (e.g., group A small and Fig. 14.6 Changes in the expression of markers for pluripo-
medium: patients diagnosed as LACI and PACI, and tent and neural stem cells in peripheral blood nucleated cells
(PBNC) in stroke patients and control group. PBNC were har-
group B large: patients diagnosed as TACI). The vested from patients that underwent ischemic stroke within 24 h
patients diagnosed as POCI, were included into group of symptom manifestation and at day+3 and day+7 post-stroke.
A or B on the basis of clinical examination and cranial The expression of mRNA for pluripotent markers (Oct-4) and
CT analysis. neural marker (GFAP) in the same number of cells was quanti-
fied by real-time RT-PCR and compared between groups. Data
As expected we observed an increase in mRNA for are meanSD
both pluripotent (Oct-4, Nanog) and neural SC mark-
ers (GFAP, nestin, -III-tubulin, Olig1, Olig2, Sox2
and Musashi-1) in peripheral blood nucleated cells cir-
culating in stroke patients (Fig. 14.6). The increased level of -chemokine SDF-1. As mentioned above the
expression of mRNA for Oct-4 and Nanog in PBNC upregulation of SDF-1 expression in damaged organs
of patients after stroke corresponded to those observed acts in chemoattracting CXCR4+ stem cells (e.g.,
by us previously in an experimental murine model VSEL) necessary for organ regeneration. We found
of stroke. However, maximal increased expression of that the serum concentration of SDF-1 increases in
neural stem cell markers in humans was delayed by patients after stroke. This may be one of the factors
2 days (1 day mice vs. 3 days human). Moreover, that mobilized VSEL from the BM into PB. However,
the kinetic of changes in expression of mRNA for we were not able to measure the concentration of
early stem cells correspond to changes that we recently SDF-1 in the immediate vicinity of the stroke loca-
observed after heart infarct in humans and mice. tion in these patients. However, it is most likely that the
Furthermore, we observed differences in SC mobiliza- serum increase of SDF-1 positively correlates with an
tion in cases of POCI and PACI as compared to other increase in expression of SDF-1 in the damaged brain.
subtypes of stroke. Of note patients with POCI have the Interestingly, we observed a positive correlation
best chance of recovery and PACI is associated with the between the number of circulating CD34+ CD133+ and
highest risk of early recurrence of stroke (i.e., within 3 CXCR4+ CD34+ cells in PB and the serum concen-
months), but it is not associated with high mortality tration of SDF-1. In the available literature, data on
and significant disability. the correlation between these two variables and organ
The changes in the number of CXCR4+ VSEL cir- damage are contradictory. For example, in patients
culating in PB were paralleled by serum changes in following heart infarct, some authors found a direct
14 Potential Application of VSEL Stem Cells in Neural Regeneration 241

correlation (De Falco et al., 2004) while others deter- References


mined the inverse to be true (Abbott et al., 2004).
However, increase in circulating VSEL in peripheral Abbott JD, Huang Y, Liu D, Hickey R, Krause DS, Giordano FJ
blood (~ 3 times) we observed was relatively small, (2004) Stromal cell-derived factor-1alpha plays a critical role
it is important to remember that stem cell mobilization in stem cell recruitment to the heart after myocardial infarc-
and stem cell homing are dynamic processes. It is very tion but is not sufficient to induce homing in the absence of
injury. Circulation 110:33003305.
likely that cells released into circulation may rapidly Basiji DA, Ortyn WE, Liang L, Venkatachalam V, Morrissey P
home to the damaged tissues or enter stem cell niches (2007) Cellular image analysis and imaging by flow cytome-
in other organs, or even return to stem cell niches in try. Clin Lab Med 27:653670.
BM. This means that the number of cells detected in Beltrami AP, Cesselli D, Bergamin N, Marcon P, Rigo S,
Burelli S, Puppato E, DAurizio F, Bottecchia M, Masolini P,
a frozen time laps is only one of the parameters of Mariuzzi L, Finato N, Beltrami CA (2007) Multipotent cells
VSEL trafficking, and does not reflect a total number can be generated in vitro from several adult human organs
of these cells released into PB during time course of (heart, liver and bone marrow). Blood 110:34383446.
acute stroke. This increase in a given moment how- Bhattacharyya BJ, Banisadr G, Jung H, Ren D, Cronshaw DG,
Zou Y, Miller RJ (2008) The chemokine stromal cell-derived
ever was statistically significant and similar to those factor-1 regulates GABAergic inputs to neural progenitors in
we observed for circulating VSEL in murine mod- the postnatal dentate gyrus. J Neurosci 28:67206730.
els of heart infarct as well toxic skeletal muscle and De Falco E, Porcelli D, Torella AR, Straino S, Iachininoto MG,
liver injuries (Kucia et al., 2008b; Zuba-Surma et al., Orlandi A, Truffa S, Biglioli P, Napolitano M, Capogrossi
MC, Pesce M (2004) SDF-1 involvement in endothelial phe-
2008b). Thus, our study demonstrates for the first time notype and ischemia-induced recruitment of bone marrow
that the mobilization of CXCR4+ CD45 lin VSEL and progenitor cells. Blood 104:34723482.
progenitor cells expressing early neural markers Dirnagl U, Iadecola C, Moskowitz MA (1999) Pathobiology
into PB occurs in patients with ischemic stroke of ischaemic stroke: An integrated view. Trends Neurosci
22:391397.
(Paczkowska et al., 2008). Duan X, Kang E, Liu CY, Ming GL, Song H (2008)
Development of neural stem cell in the adult brain. Curr Opin
Neurobiol 18:108115.
DIppolito G, Diabira S, Howard GA, Menei P, Roos BA,
Schiller PC (2004) Marrow-isolated adult multilineage
inducible (miami) cells, a unique population of postnatal
14.7 Conclusions young and old human cells with extensive expansion and
differentiation potential. J Cell Sci 117:29712981.
Encinas JM, Enikolopov G (2008) Identifying and quantitating
In conclusion, our data indicates that VSEL could
neural stem and progenitor cells in the adult brain. Methods
potentially provide a real therapeutic alternative to Cell Biol 85:243272.
the controversial use of human embryonic stem cells Endres M, Dirnagl U (2002) Ischemia and stroke. Adv Exp Med
(ESC) and therapeutic cloning. Augmented evidence Biol 513:455473.
Hess DC, Borlongan CV (2008) Stem cells and neurological
shows that stress related to the stroke triggers the mobi-
diseases. Cell Prolif 41(Suppl 1):94114.
lization of VSEL from BM and perhaps other stem Hill WD, Hess DC, Martin-Studdard A, Carothers JJ, Zheng J,
cell niches into PB. We hypothesize that if these cells Hale D, Maeda M, Fagan SC, Carroll JE, Conway SJ (2004)
observed in mobilized PB in humans will turn out to be SDF-1 (CXCL12) is upregulated in the ischemic penumbra
following stroke: Association with bone marrow cell homing
a human counterparts of VSEL identified in mice they
to injury. J Neuropathol Exp Neurol 63:8496.
could be potentially purified from the PB, expanded Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD,
ex vivo, and employed for regeneration of damaged Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad
neural tissues. Hence, while the ethical debate on the M, Du J, Aldrich S, Lisberg A, Low WC, Largaespada DA,
Verfaillie CM (2002) Pluripotency of mesenchymal stem
application of ESC in therapy continues, the poten-
cells derived from adult marrow. Nature 418:4149.
tial of VSEL is ripe for exploration. Researchers must Kalluri HS, Dempsey RJ (2008) Growth factors, stem cells, and
determine whether these cells could be efficiently stroke. Neurosurg Focus 24:E14.
employed in the clinic or whether they are merely Kasiyanov A, Fujii N, Tamamura H, Xiong H (2008) Modulation
of network-driven, GABA-mediated giant depolarizing
developmental remnants found in the BM that can-
potentials by sdf-1alpha in the developing hippocampus. Dev
not be harnessed effectively for regeneration. The Neurosci 30:285292.
coming years will bring important answers to these Komitova M, Mattsson B, Johansson BB, Eriksson PS (2005)
questions. Enriched environment increases neural stem/progenitor cell
242 M.Z. Ratajczak et al.

proliferation and neurogenesis in the subventricular zone of Ponomaryov T, Peled A, Petit I, Taichman RS, Habler L,
stroke-lesioned adult rats. Stroke 36:12781282. Sandbank J, Arenzana-Seisdedos F, Magerus A, Caruz A,
Kucia M, Dawn B, Hunt G, Guo Y, Wysoczynski M, Majka Fujii N, Nagler A, Lahav M, Szyper-Kravitz M, Zipori D,
M, Ratajczak J, Rezzoug F, Ildstad ST, Bolli R, Ratajczak Lapidot T (2000) Induction of the chemokine stromal-
MZ (2004a) Cells expressing early cardiac markers reside in derived factor-1 following DNA damage improves human
the bone marrow and are mobilized into the peripheral blood stem cell function. J Clin Invest 106:13311339.
after myocardial infarction. Circ Res 95:11911199. Pritchett J, Wright C, Zeef L, Nadarajah B (2007) Stromal
Kucia M, Halasa M, Wysoczynski M, Baskiewicz-Masiuk M, derived factor-1 exerts differential regulation on distinct
Moldenhawer S, Zuba-Surma E, Czajka R, Wojakowski W, cortical cell populations in vitro. BMC Dev Biol 7:31.
Machalinski B, Ratajczak MZ (2007a) Morphological and Prockop DJ (1997) Marrow stromal cells as stem cells for
molecular characterization of novel population of CXCR4(+) nonhematopoietic tissues. Science 276:7174.
SSEA-4(+) Oct-4(+) very small embryonic-like cells puri- Quesenberry PJ, Colvin G, Dooner G, Dooner M, Aliotta JM,
fied from human cord blood preliminary report. Leukemia Johnson K (2007) The stem cell continuum: Cell cycle,
21:297303. injury, and phenotype lability. Ann N Y Acad Sci 1106:
Kucia M, Ratajczak J, Reca R, Janowska-Wieczorek A, 2029.
Ratajczak MZ (2004b) Tissue-specific muscle, neural and Ratajczak MZ, Machalinski B, Wojakowski W, Ratajczak J,
liver stem/progenitor cells reside in the bone marrow, Kucia M (2007) A hypothesis for an embryonic origin of
respond to an sdf-1 gradient and are mobilized into periph- pluripotent Oct-4+ stem cells in adult bone marrow and other
eral blood during stress and tissue injury. Blood Cells Mol tissues. Leukemia 21:860867.
Dis 32:5257. Ratajczak MZ, Zuba-Surma E, Kucia M, Reca R, Wojakowski
Kucia M, Reca R, Campbell FR, Zuba-Surma E, Majka M, W, Ratajczak J (2006) The pleiotropic effects of the SDF-1-
Ratajczak J, Ratajczak MZ (2006a) A population of very CXCR4 axis in organogenesis, regeneration and tumorigen-
small embryonic-like (vsel) CXCR4(+) SSEA-1(+) Oct- esis. Leukemia 20:19151924.
4+ stem cells identified in adult bone marrow. Leukemia Ratajczak MZ, Zuba-Surma EK, Machalinski B, Ratajczak J,
20:857869. Kucia M (2008a) Very small embryonic-like (VSEL) stem
Kucia M, Wysoczynski M, Ratajczak J, Ratajczak MZ (2008a) cells: Purification from adult organs, characterization, and
Identification of very small embryonic like (VSEL) stem biological significance. Stem Cell Rev 4:8999.
cells in bone marrow. Cell Tissue Res 331:125134. Ratajczak MZ, Zuba-Surma EK, Shin DM, Ratajczak J, Kucia
Kucia MJ, Wysoczynski M, Wu W, Zuba-Surma EK, Ratajczak M (2008b) Very small embryonic-like (VSEL) stem cells in
J, Ratajczak MZ (2008b) Evidence that very small embryonic adult organs and their potential role in rejuvenation of tissues
like (VSEL) stem cells are mobilized into peripheral blood. and longevity. Exp Gerontol 43:10091117.
Stem Cells 26:20832092. Ratajczak MZ, Zuba-Surma EK, Wysoczynski M, Ratajczak
Kucia M, Zhang YP, Reca R, Wysoczynski M, Machalinski B, J, Kucia M (2008c) Very small embryonic-like stem cells:
Majka M, Ildstad ST, Ratajczak J, Shields CB, Ratajczak Characterization, developmental origin, and biological sig-
MZ (2006b) Cells enriched in markers of neural tissue- nificance. Exp Hematol 36:742751.
committed stem cells reside in the bone marrow and Ratajczak MZ, Zuba-Surma EK, Wysoczynski M, Wan W,
are mobilized into the peripheral blood following stroke. Ratajczak J, Wojakowski W, Kucia M (2008d) Hunt for
Leukemia 20:1828. pluripotent stem cell regenerative medicine search for
Kucia M, Zuba-Surma EK, Wysoczynski M, Wu W, Ratajczak almighty cell. J Autoimmun 30:151162.
J, Machalinski B, Ratajczak MZ (2007b) Adult marrow- Shyu WC, Lee YJ, Liu DD, Lin SZ, Li H (2006) Homing genes,
derived very small embryonic-like stem cells and tissue cell therapy and stroke. Front Biosci 11:899907.
engineering. Expert Opin Biol Ther 7:14991514. Tacchini L, Matteucci E, De Ponti C, Desiderio MA (2003)
Lvesque JP, Winkler IG, Larsen SR, Rasko JE (2007) Hepatocyte growth factor signaling regulates transactivation
Mobilization of bone marrow-derived progenitors. Handb of genes belonging to the plasminogen activation system via
Exp Pharmacol 180:336. hypoxia inducible factor-1. Exp Cell Res 290:391401.
Moog R (2006) Mobilization and harvesting of peripheral blood Vilz TO, Moepps B, Engele J, Molly S, Littman DR, Schilling
stem cells. Curr Stem Cell Res Ther 1:189201. K (2005) The SDF-1/CXCR4 pathway and the development
Ortyn WE, Hall BE, George TC, Frost K, Basiji DA, Perry of the cerebellar system. Eur J Neurosci 22:18311839.
DJ, Zimmerman CA, Coder D, Morrissey PJ (2006) Walzlein JH, Synowitz M, Engels B, Markovic DS,
Sensitivity measurement and compensation in spectral imag- Gabrusiewicz K, Nikolaev E, Yoshikawa K, Kaminska
ing. Cytometry A 69A:852862. B, Kempermann G, Uckert W, Kaczmarek L, Kettenmann
Paczkowska E, Kucia M, Koziarska D, Halasa M, Safranow K, H, Glass R (2008) The anti-tumorigenic response of neural
Masiuk M, Karbicka A, Nowik M, Nowacki P, Ratajczak precursors depends on subventricular proliferation and age.
MZ, Machalinski B (2008) Clinical evidence that very small Stem Cells 26:29452954.
embryonic-like (VSEL) stem cells are mobilized into periph- Wojakowski W, Tendera M, Zebzda A, Michalowska A, Majka
eral blood in patients after stroke. Stroke 40:12371244. M, Kucia M, Maslankiewicz K, Wyderka R, Krl M, Ochala
Peister A, Mellad JA, Larson BL, Hall BM, Gibson LF, Prockop A, Kozakiewicz K, Ratajczak MZ (2004) Mobilization
DJ (2004) Adult stem cells from bone marrow (MSCS) iso- of D34/CXCR4+ , CD34/CD117+ , c-Met+ stem cells, and
lated from different strains of inbred mice vary in surface mononuclear cells expressing early cardiac, muscle, and
epitopes, rates of proliferation, and differentiation potential. endothelial markers into peripheral blood in patients with
Blood 103:16621668. acute myocardial infarction. Circulation 110:32133220.
14 Potential Application of VSEL Stem Cells in Neural Regeneration 243

Zhang RL, Zhang ZG, Zhang L, Chopp M (2001) Proliferation to a new era in imaging. Folia Histochem Cytobiol 45:
and differentiation of progenitor cells in the cortex and 279290.
the subventricular zone in the adult rat after focal cerebral Zuba-Surma EK, Kucia M, Dawn B, Guo Y, Ratajczak MZ,
ischemia. Neuroscience 105:3341. Bolli R (2008b) Bone marrow-derived pluripotent very
Zhu Y, Yu T, Zhang XC, Nagasawa T, Wu JY, Rao Y (2002) small embryonic-like stem cells (VSELs) are mobilized
Role of the chemokine SDF-1 as the meningeal attrac- after acute myocardial infarction. J Mol Cell Cardiol 44:
tant for embryonic cerebellar neurons. Nat Neurosci 5: 865873.
719720. Zuba-Surma EK, Kucia M, Ratajczak MZ (2008c) Decoding of
Zuba-Surma EK, Kucia M, Abdel-Latif A, Dawn B, Hall dot: The Imagestream System (ISS) as a supportive tool for
B, Singh R, Lillard JW Jr, Ratajczak MZ (2008a) flow cytometric analysis. Cent Eur J Biol 3:110.
Morphological characterization of very small embryonic-like Zuba-Surma EK, Wu W, Ratajczak J, Kucia M, Ratajczak MZ
stem cells (VSELs) by Imagestream System analysis. J Cell (2009) Very small embryonic-like stem cells in adult tissues-
Mol Med 12:292303. potential implications for aging. Mech Ageing Dev 130:
Zuba-Surma EK, Kucia M, Abdel-Latif A, Lillard JW Jr, 5866.
Ratajczak MZ (2007) The Imagestream System: A key step
Chapter 15

Embryonic Stem Cell Transplantation for the Treatment


of Parkinsons Disease

Asuka Morizane and Jun Takahashi

Contents cells and induced pluripotent stem (iPS) cells. Both


human ES and human iPS cells can differentiate into
15.1 Introduction . . . . . . . . . . . . . . . . 246 dopamine (DA) neurons in vitro. These cells have been
15.2 Rationale for Using Transplantation as a transplanted into animal models of Parkinsons disease
Treatment for Parkinsons Disease . . . . . . 246 and behavioral recovery has been observed in some
15.3 In Vitro Differentiation of Embryonic Stem
cases. Prior to clinical application, however, several
Cells . . . . . . . . . . . . . . . . . . . . 247
15.4 Transplantation in a Parkinsons Disease Model 247 problems remain: instability of DA neurons in vivo,
15.5 Safety Issues for Clinical Application . . . . 248 risk of tumor formation by grafted cells, and the possi-
15.6 Another Donor Candidate: Induced Pluripotent ble transfer of animal-derived pathogens. When these
Stem Cell (iPS cell) . . . . . . . . . . . . . 251
problems are solved, cell transplantation will likely
References . . . . . . . . . . . . . . . . . . . . 251
become one of the standard treatments for Parkinsons
disease.
Abstract Parkinsons disease is one of the most Keywords Parkinsons disease Dopamine neuron
common movement disorders. Medical treatment and Embryonic stem cells Transplantation Clinical
deep brain stimulation (DBS) are useful in some application
patients. However, these two therapies do not recon-
stitute patients lost neuronal circuits and the disease Abbreviations
itself progresses. Thus, it is often difficult to control
the disease in late stages. Hundreds of patients have AA ascorbic Acid
been involved in clinical trials where they received cell BDNF brain-derived neurotrophic factor
transplants from aborted fetal/embryonic brain tissue. ALS amyotrophic lateral sclerosis
Although some patients exhibit enormous improve- DA dopamine
ment, this treatment is not standard due to potential dbc AMP dibutyryl cyclic AMP
problems associated with the use of aborted tissue, DBS deep brain stimulation
including a limited supply. In contrast, stem cell EB embryoid bodies
research is advancing rapidly and many people regard ES cell embryonic stem cell
stem cells as the donor source of the next genera- FACS fluorescent activated cell sorting
tion. Many laboratories have tried to generate donor FN fibronectin-coated dish
cells from stem cells, especially embryonic stem (ES) GABA -aminobutyric acid
GDNF glial cell line-derived neurotrophic factor
HLA human leukocyte antigen
iPS cells induced pluripotent stem cells
ITSF insulin, transferrin, selenite and fibronectin
J. Takahashi ()
Department of Biological Repair, Institute for Frontier Medical L laminin coated dish
Sciences, Kyoto University, Kyoto, Japan MACS magnetic activated cell sorting
e-mail: jbtaka@frontier.kyoto-u.ac.jp MEF mouse embryonic fibroblast

H. Ulrich (ed.), Perspectives of Stem Cells, 245


DOI 10.1007/978-90-481-3375-8_15, Springer Science+Business Media B.V. 2010
246 A. Morizane and J. Takahashi

MPTP 1-methyl-4-phenyl-1,2,3,6- to the potential problem associated with using aborted


tetrahydropyridine tissue, including a limited supply. Many researchers
Neu5Gc N-glycolyl-neuraminic acid residues have been trying to use stem cells as another donor
NPC Neural stem/progenitor cell(s) cell source. In this chapter we will review cell trans-
O/L poly-ornithine and laminin-coated dish plantation for PD, focusing on the use of ES and iPS
6-OHDA 6-hydroxy dopamine cells.
PD Parkinsons disease
SDIA stromal cell-derived inducing activity
SHH sonic hedgehog
TH tyrosine hydroxylase
15.2 Rationale for Using Transplantation
as a Treatment for Parkinsons
Disease
15.1 Introduction
The first clinical trial of cell transplantation for PD
Parkinsons disease (PD) is the most common move- was performed in 1987 using human ventral mesen-
ment disorder. About 1% of the population over 60 cephalon cells from aborted embryos (Brundin et al.,
years is affected. Typical symptoms of Parkinsons dis- 1987). To date, more than 400 PD patients around
ease include resting tremor, bradykinesia, and limb the world have received the same type of donor tis-
rigidity (Samii et al., 2004). The main neuropathology sue. In small, open-label trials, some of these patients
in PD is loss of dopamine (DA) neurons in the substan- have shown dramatic improvement after transplan-
tia nigra pars compacta, which leads to decreased DA tation, attracting interest and expectations (Brundin
level in the striatum. Clinical signs of PD are evident et al., 2000; Hagell and Brundin, 2001; Hauser et al.,
when about 80% of striatal DA and 50% of nigral neu- 1999; Mendez et al., 2000). Recently, three groups
rons are lost (Fearnley and Lees, 1991). Patients are reported that transplanted DA neurons can survive
treated with L-3, 4-dihydroxyphenylalanine (L-dopa) over ten years after surgical postmortem examina-
and DA agonists to compensate for the loss of DA tion of patients who had undergone the procedure (Li
neurotransmission in the nigrostriatal system. Most et al., 2008; Mendez et al., 2008; Kordower et al.,
patients that begin with DA agonist therapy will need 2008). However, Lewy body structures, pathological
the addition of L-dopa within 5 years. Patients treated changes often observed in the brain of PD patients,
with L-dopa long-term gradually develop side effects, have been detected in grafted cells in these long-
such as motor fluctuations and L-dopa-induced dysk- term cases (Li et al., 2008; Kordower et al., 2008).
inesia. Although medical therapy is standard, many Furthermore, two double-blind placebo-controlled tri-
patients have difficulty controlling their symptoms in als in the USA failed to observe an improvement of
the later phase of the disease. symptoms at primary endpoints (Freed et al., 2001;
Surgical therapy is another option. Deep brain stim- Olanow et al., 2003), highlighting several problems
ulation is the most common surgical procedure for PD. that require consideration. The first is patient selection.
Some patients exhibit beneficial effects, but selection Disease stage and preoperative response to L-dopa are
of suitable patients for deep brain stimulation is the important issues for selecting good candidates for the
key to successful results. A good response to L-dopa treatment. Younger patients with less severe pathol-
treatment is a good predictor of a positive outcome. ogy have increased chances for successful outcomes
Although deep brain stimulation is a powerful option after transplantation. Not all the patients suffering from
for controlling PD symptoms, the procedure does not PD are suitable for this cell therapy. Other considera-
slow down disease progression, nor is it reparative in tions are problems associated with the procedure itself,
nature. In contrast, cell transplantation has the poten- including tissue preparation, number of the donor cells,
tial to reconstruct neuronal circuits. This procedure graft implantation techniques, and immunosuppres-
was first applied clinically to PD patients in the late sion, among others. Freshly dissected fetal ventral
1980s using fetal/embryonic brain tissues. However, mesencephalic tissue has been used in most of the
thus far, this treatment is not a standard therapy due open-label trials. However, in one of the double-blind
15 ES Cell Transplantation for the Treatment of Parkinsons Disease 247

trials, the tissue was dissociated and expanded for up to Pitx3, Mash1, Lmx1a, and Msx1 (Kim et al., 2002;
4 weeks before transplantation. The number of donor Chung et al., 2005; Kanda et al., 2004; Andersson
embryos used for one side of the brain has ranged et al., 2006). The most efficient protocol for induc-
from 1 to 8. Although it would be better biologically ing DA neurons from murine ES cells thus far is to
to use fresh tissue, it is difficult in practice to prepare use a combination of Nurr1 overexpression, PA6 cells
enough donor tissues at the same time. Collectively, co-culture, and soluble factors including Shh, FGF8,
previous clinical data indicates that cell transplantation and ascorbic acid (Kim et al., 2006). Ninety percent of
is effective if the PD patients and procedures are cor- neurons are dopaminergic under these conditions (see
rectly selected. Due to their pluripotency and ability to Table 15.1). For human ES cells, a similar combina-
self-renew, ES or iPS cells might be better donor cell tion of co-culture with MS5 and soluble factors (Shh
candidates. and FGF8) generates TuJ1-immunopositive neurons,
among which 6479% of the cells express tyrosine
hydroxylase (TH), a marker for DA neurons (Perrier
et al., 2004).
15.3 In Vitro Differentiation
Studies of ventral mesencephalon transplants in ani-
of Embryonic Stem Cells mals (Brown and Dunnett, 1989; Thompson et al.,
2005) and PD patients (Mendez et al., 2005) indi-
Embryonic stem cells can generate neurons, as well as cate that only A9-type DA neurons can improve
other type of cells, including those with mesodermal Parkinsonian symptoms. A9 DA neurons express a
and endodermal phenotypes. Therefore, to generate specific set of markers, including G-protein-coupled
DA neurons from ES cells, we have to begin by induc- inwardly rectifying potassium channel (Girk) 2, Nurr1,
ing a neural cell fate, for which two methods are Pitx3, Engrailed 1/2 (En1/2), Lmx1a, Lmx1b, and
reported. One is a co-culture of ES cells with feeder DA transporter (Anderson et al., 2006). TH-positive
cells, such as mesenchymal stromal cells (Kawasaki cells are also found in the olfactory bulb (A16 neu-
et al., 2000). As for feeders, PA6 cells (Kawasaki rons), but these cells co-express gamma-aminobutyric
et al., 2000), MS5 cells (Barberi et al., 2003), Sertoli acid (GABA) and neurons in the substantia nigra pars
cells (Yue et al., 2006), amniotic membrane (Ueno compacta (A9 neurons) do not.
et al., 2006), human telomerase-overexpressing astro-
cytes from embryonic midbrain (Roy et al., 2006)
and meninges (Hayashi et al., 2008) all have the
15.4 Transplantation in a Parkinsons
activity needed to induce the generation of DA neu-
rons from ES cells. The second approach for neu- Disease Model
ral induction is to form aggregates called embryoid
bodies (EB) and then add soluble factors to differ- There are several animal models for PD. Treating
entiate the EB into proper neuronal cells (Lee et al., rats with 6-hydroxy dopamine (6-OHDA) is one of
2000). Many cytokines and growth factors that are the most common and established models. Usually
found in the developing midbrain of the embryo/fetus the toxin is injected into one side of the nigrostri-
have been tried as additional factors in the media, atal pathway, where it causes specific degradation of
including sonic hedgehog (Shh), FGF8, ascorbic acid, DA neurons in the substantia nigra. This hemi-lesioned
interleukin-1, glial cell line-derived neurotrophic fac- model is advantageous for functional analysis. Because
tor (GDNF), neurturin, TGF-3, dibutyryl cyclic AMP the DA system is imbalanced between the right and left
(dbcAMP), vitamin B12, brain-derived neurotrophic sides of the brain, the model animal rotates to one side
factor (BDNF), neurotrophin 3, and fibroblast growth when tested with drugs such as amphetamine or apo-
factor (FGF) 20, among others (Rolletschek et al., morphine (Marshall and Ungerstedt, 1977). If grafted
2001; Park et al., 2005; Yamazoe et al., 2006, Takagi cells secrete DA and compensate for decreased DA
et al., 2005; Park et al., 2004). levels, the frequency of rotation decreases. Another
Overexpression of transcription factors that partic- PD model is based on 1-methyl-4-phenyl-1,2,3,6-
ipate in the development of DA neurons can also tetrahydropyridine (MPTP). Monkeys and mice are
induce DA neurons. These factors include Nurr1, sensitive to MPTP. The monkey MPTP model is
248 A. Morizane and J. Takahashi

very similar to human PD patients biochemically and One major problem is tumor formation by the grafted
pathologically, as well as symptomatically, displaying cells. This phenomenon has been reported by the use
rigidity, tremor and bradykinesia. of the term tumor (Yang et al., 2007), teratoma
There are numbers of reports describing transplants (Brederlau et al., 2006; Sonntag et al., 2007), over-
of ES cell-derived DA neurons, or their progenitors, growth, or neoplastic tumor mass (Roy et al., 2006;
into the brains of animal PD models. Murine ES Goldman et al., 2007). Due to pluripotency and the
cell-derived progenitors are able to induce behavioral ability to self-renew, the population of cells in a cul-
recovery in recipients (Kim et al., 2002; Nishimura ture dish is heterogeneous. Some cells have advanced
et al., 2003; Barberi et al., 2003; Baier et al., 2004; in maturation, while other cells might still be imma-
Rodriguez-Gomez et al., 2007). Monkey ES cells ture and proliferating. Although most cells are proper
can also differentiate into DA neurons when co- midbrain DA neurons, the population may be contami-
cultured with PA6 feeder cells (Kawasaki et al., nated with undifferentiated or non-neural cells. If cells
2002). Monkeys treated with MPTP recover from with a non-neural cell fate or high proliferation activ-
motor and posture impairments when monkey ES ity exist in the graft, they keep growing and destroy
cell-derived DA neurons are grafted. In positron the surrounding host brain structures. To avoid this
emission tomography scans of these monkeys, 18F- phenomenon, donor cells must be prepared under con-
fluorodopa uptake in the striatum is increased after ditions that precisely control the direction and extent of
transplantation (Takagi et al., 2005). Human ES differentiation. Furthermore, a strategy for purification
cells are also able to differentiate into DA neurons or selection of ideal donor cells from the differentiated
and survive in the striatum of rat PD models (see culture is essential. Cell sorting using fluorescent acti-
Table 15.1). However, some groups have reported vated cell sorting (FACS) or magnetic activated cell
no functional effect (Brederlau et al., 2006; Park sorting (MACS) has been used for positive (Fukuda
et al., 2005), or only small changes (Ben-Hur et al., et al., 2006; Chung et al., 2006; Pruszak et al., 2007) or
2004), after transplantation, indicating that the num- negative selection (Shibata et al., 2006). Using chem-
ber of surviving DA neurons was insufficient. Other icals or drugs to eliminate undesired cells is another
groups have reported that motor function improves approach (Bieberich et al., 2004). If we can purify
after cell transplantation (Sonntag et al., 2007; Yang suitable donor cells and prepare a completely homoge-
et al., 2007; Ko et al., 2007; Cho et al., 2008). It neous population before grafting, DA neuron survival
appears that DA neurons derived from human ES cells after transplantation would increase and the risk of
have a lower survival rate and are less functional in tumor formation would decrease.
rat striatum than those from murine ES cells. Although For clinical use of ES cells, we also need to consider
grafts contain numerous neurons that express a marker the issue of non-human materials. From the derivation
for neuronal nuclei (NeuN; Schulz et al., 2004; Park of human ES cells to their maintenance and differenti-
et al., 2005; Brederlau et al., 2006), the number of TH- ation into DA neurons, we use materials derived from
positive neurons is several-fold lower than expected non-human species, such as mouse embryonic fibrob-
based on grafts from murine ES cells or fetal tissue. lasts and fetal bovine serum. Exposing human ES
The mechanisms underlying the selective vulnerabil- cells to animal derivatives allows them to incorporate
ity or instability of human ES cell-derived DA neurons N-glycolyl-neuramninic acid residues (Neu5Gc) that
are not understood, but they may be related to species are present in most mammals, with the exception of
differences or improper differentiation prior to trans- humans (Martin et al., 2005). Because contamination
plantation. Further studies are needed to solve this of the xenogenic material might cause transfer of ani-
important problem. mal pathogens or an immunological reaction, we need
to establish a protocol that uses only human-derived or
chemically defined materials.
15.5 Safety Issues for Clinical
Genetic instability of human ES cells is another
Application issue that requires consideration. Karyotypic changes
in several human ES cell lines have been reported
Prior to the clinical application of ES cell transplanta- (Cowan et al., 2004; Maitra et al., 2005; Herszfeld
tion, a number of safety issues need to be addressed. et al., 2006; Draper et al., 2004). Changes commonly
Table 15.1 Induction and transplantation of human ES cell-derived DA neurons
Study Cell lines Methods Culture TH/Tub
days III (%) Transplantation
NPC, DA No. of cells Follow- Function Tumor
neuron transplanted Stage up Histology (rotation) formation
Zeng et al. BG01 SDIA (PA6) 12, 18 n.d. n.d. 8- or 5 No Oct4+ n.d. n.d.
(2004) days 22-day- weeks cells, but
old cells SMA+
cells and
TH+
cells
Schulz BG01, EB (HepG2 10, 28 74 1,000 3- or 8 A few n.d. 1 out of 23
et al. BG03 CM/ FGF2 or days 20,000 4-week- weeks TH+
(2004) DMEM/N2) old cells cells
O/L (serum,
BDNF, GDNF)
Ben-Hur HES-1 MEF (serum, 180, 187 19 400,000 6-weeek- 12 389 TH+ Partial PCNA 0.2%
et al. Noggin) days old weeks cells recovery
(2004) sphere cells
(FGF2)
15 ES Cell Transplantation for the Treatment of Parkinsons Disease

L (FGF
withdrawn)
Park et al. HSF-6, SDIA (PA6) 14, 30 41 500,000 Stage 2 A few No PCNA 53%
(2005) SNU- N2, AA, days 2 or 3 weeks TH+ recovery
hES-3, FGF2 cells cells
Miz- FN (BDNF,
hES-1 GDNF, NT-3,
Shh, FGF8)
Brederlau SA002.5 SDIA (PA6) 14, 28 65 100,000 23-day- 13 1050 No +
et al. O/L days old cells weeks TH+ recovery
(2006) cells
Roy et al. H1, H9 EB ITSF 14, 36 67 500,000 Stage 5 10 58,273 Recovery Overgrowth
(2006) medium O/L days cells weeks TH+
(FGF2, SHH, cells
FGF8, human
midbrain
astrocytes)
249
250

Table 15.1 (continued)


Study Cell lines Methods Culture TH/ Tub
days III (%) Transplantation
NPC, DA No. of cells Follow- Function Tumor
neuron transplanted Stage up Histology (rotation) formation
Sonntag et H7, H9 SDIA (MS5- 21, 49 23 100,000 42-day- 12 160 TH+ Recovery +
al. (2007) Wnt1 days (TH/ old cells weeks cells
Noggin) total
SHH, FGF8, cells)
AA, BDNF
O/L (BDNF,
GDNF,
TGF, AA)
Iacovitti et H9, EB ITSF 14, 21 n.d. 2500,000 Stage 4 23 Many n.d. Ki67+ but no
al. (2007) BG01, medium days cells weeks TH+ cells Oct4 or
HUES7, (Noggin) SSEA4
HUES8 FGF2
Ornithine
(dcAMP)
Yang et al. H9 EB (FGF2) 38, 52 43 200,000 5 1273 Recovery Overgrowth
(2007) adherent days months TH+ cells
(FGF8, SHH)
O/L (Wnt3a,
AA, cAMP,
TGF, BDNF,
GDNF)
Ko et al. HSF-6, SDIA (PA6 or >2 45 1,500,000 Progenitors 8 200 Recovery
(2007) H9 MS5, ITS months weeks TH+ cells
medium,SHH,
AA) O/L
(ITS+FGF2)
ITS (AA,BDNF,
dbcAMP)
Cho et al. SNU- EB Matrigel 30, 44 86 500,000 37-day-old 12 10,732 Recovery
(2008) hES- (FGF2) days cells weeks TH+ cells
1,3,16 suspension
(FGF2)
Matrigel (SHH,
FGF8, AA)
In these studies, human ES cell-derived DA neurons were transplanted into 6-OHDA-lesioned striatum of rats. Brain sections were stained immunohistochemically
with TH antibody, and apomorphine or amphetamine was used to test rotational behavior.
A. Morizane and J. Takahashi
15 ES Cell Transplantation for the Treatment of Parkinsons Disease 251

occur in chromosomes 12 and 17 (Baker et al., 2007). cover all types of human leukocyte antigen (HLA)
We do not fully understand the influence of these populations.
chromosomal abnormalities, but they may affect sur- The risk of tumor formation is another problem
vivability and risk of tumor formation. associated with using iPS cells as the donor for
transplantation. Originally, iPS cells were established
using retrovirus and transducing four genes including
c-myc oncogene. Because of this genetic manipu-
lation, iPS cells may have a higher risk of tumor
or cancer generation after grafting than hES cells.
15.6 Another Donor Candidate: Induced
Mouse and human iPS cells can now be estab-
Pluripotent Stem Cell (iPS cell) lished without the Myc gene, thereby reducing this
risk (Nakagawa et al., 2008). Furthermore, genomic
In 2006, Yamanakas group succeeded in generating integration of these genes by retrovirus is not
iPS cells from mouse fibroblasts by transducing four always needed for generating iPS cells. iPS cells
transcriptional factors, Oct3/4, Sox2, c-Myc and Klf4 have been established by tentative expression of
(Takahashi and Yamanaka, 2006). It took only one year these four genes using adenovirus (Stadtfeld et al.,
to establish human iPS cells (Takahashi et al., 2007; 2008) or a plasmid expression system (Okita et al.,
Yu et al., 2007). The cells are pluripotent and pos- 2008).
sess the potency to differentiate into DA neurons in Currently, iPS cells are most useful for drug screen-
vitro (Takahashi et al., 2007). Although iPS cells retro- ing and/or researching genetic disorders. However,
virally integrate extra genes, cells from patients with because the technology in this field is advancing
genetic disorders might be helpful for understanding rapidly, we can expect safer methods for generating
disease mechanisms, screening drugs, and measuring human iPS cells and can apply them for cell therapy
toxicity. Several groups have started to establish iPS in the near future.
cell lines from patients with genetic diseases such
as Gaucher disease type , Parkinsons disease, Type
1 diabetes, Lesch-Nyhan syndrome, and amyotrophic
lateral sclerosis (ALS), among others (Park et al.,
References
2008; Dimos et al., 2008). It may also be possible
to use iPS cells for transplantation, rather than ES
Andersson E, Tryggvason U, Deng Q, Friling S, Alekseenko
cells. A rat PD model recovers following transplant
Z, Robert B, Perlmann T, Ericson J (2006) Identification of
of DA neurons derived from mouse iPS cells (Wernig intrinsic determinants of midbrain dopamine neurons. Cell
et al., 2008). Theoretically, it is possible to gener- 124:393405.
ate iPS cells from the host patient. This autologous Baier PC, Schindehutte J, Thinyane K, Flugge G, Fuchs E,
Mansouri A, Paulus W, Gruss P, Trenkwalder C (2004)
transplantation only causes a minimal immunological Behavioral changes in unilaterally 6-hydroxy-dopamine
response, and reduces the risk of infection from other lesioned rats after transplantation of differentiated mouse
people. Therapeutic cloning of ES cells for the treat- embryonic stem cells without morphological integration.
ment of PD is also a possibility. Nuclear transferred ES Stem Cells 22:396404.
Baker DE, Harrison NJ, Maltby E, Smith K, Moore HD, Shaw
cells have been generated from 24 PD model mice. The PJ, Heath PR, Holden H, Andrews PW (2007) Adaptation to
cells were successfully differentiated into DA neurons culture of human embryonic stem cells and oncogenesis in
and transplanted back into each host mouse individu- vivo. Nat Biotechnol 25:207215.
ally (Tabar et al., 2008). However, due to time and cost Barberi T, Klivenyi P, Calingasan NY, Lee H, Kawamata H,
Loonam K, Perrier AL, Bruses J, Rubio ME, Topf N, Tabar V,
considerations, it is not practical at this point to estab- Harrison NL, Beal MF, Moore MA, Studer L (2003) Neural
lish, and verify the safety of, an individual iPS cell line subtype specification of fertilization and nuclear transfer
for each patient. Due their genetic make-up, iPS cells embryonic stem cells and application in parkinsonian mice.
from a patient may be more sensitive to the disease Nat Biotechnol 21:12001207.
Ben-Hur T, Idelson M, Khaner H, Pera M, Reinhartz E, Itzik A,
environment or may even cause the disease. Thus, it Reubinoff BE (2004) Transplantation of human embryonic
might be better to use iPS cells derived from healthy stem cell-derived neural progenitors improves behavioral
individuals and establish a bank of iPS cell lines to deficit in Parkinsonian rats. Stem Cells 22:12461255.
252 A. Morizane and J. Takahashi

Bieberich E, Silva J, Wang G, Krishnamurthy K, Condie BG Freed CR, Greene PE, Breeze RE, Tsai WY, DuMouchel W,
(2004) Selective apoptosis of pluripotent mouse and human Kao R, Dillon S, Winfield H, Culver S, Trojanowski JQ,
stem cells by novel ceramide analogues prevents teratoma Eidelberg D, Fahn S (2001) Transplantation of embryonic
formation and enriches for neural precursors in ES cell- dopamine neurons for severe Parkinsons disease. N Engl
derived neural transplants. J Cell Biol 167:723734. J Med 344:710719.
Brederlau A, Correia AS, Anisimov SV, Elmi M, Roybon Fukuda H, Takahashi J, Watanabe K, Hayashi H, Morizane
L, Paul G, Morizane A, Bergquist F, Riebe I, Nannmark A, Koyanagi M, Sasai Y, Hashimoto N (2006)
U, Carta M, Hanse E, Takahashi J, Sasai Y, Funa K, Fluorescence-activated cell sorting-based purification
Brundin P, Eriksson PS, Li JY (2006) Transplantation of of embryonic stem cell-derived neural precursors averts
human embryonic stem cell-derived cells to a rat model tumor formation after transplantation. Stem Cells 24:
of Parkinsons disease: effect of in vitro differentiation 763771.
on graft survival and teratoma formation. Stem Cells 24: Goldman SA, Roy NS, Beal MF, Cleren C (2007) Large stem
14331440. cell grafts could lead to erroneous interpretations of behav-
Brown VJ, Dunnett SB (1989) Comparison of adrenal and foetal ioral results? Nat Med 13:118119.
nigral grafts on drug-induced rotation in rats with 6-OHDA Hagell P, Brundin P. Cell survival and clinical outcome follow-
lesions. Exp Brain Res 78:214218. ing intrastriatal transplantation in Parkinson disease (2001)
Brundin P, Pogarell O, Hagell P, Piccini P, Widner H, Schrag J Neuropathol Exp Neurol 60:741752.
A, Kupsch A, Crabb L, Odin P, Gustavii B, Bjorklund A, Hauser RA, Freeman TB, Snow BJ, Nauert M, Gauger L,
Brooks DJ, Marsden CD, Oertel WH, Quinn NP, Rehncrona Kordower JH, Olanow CW (1999) Long-term evaluation of
S, Lindvall O (2000) Bilateral caudate and putamen grafts bilateral fetal nigral transplantation in Parkinson disease.
of embryonic mesencephalic tissue treated with lazaroids in Arch Neurol 56:179187.
Parkinsons disease. Brain 123: 13801390. Hayashi H, Morizane A, Koyanagi M, Ono Y, Sasai Y,
Brundin P, Strecker RE, Lindvall O, Isacson O, Nilsson Hashimoto N, Takahashi J (2008) Meningeal cells induce
OG, Barbin G, Prochiantz A, Forni C, Nieoullon dopaminergic neurons from embryonic stem cells. Eur J
A, Widner H, et al. (1987) Intracerebral grafting of Neurosci 27: 261268.
dopamine neurons. Experimental basis for clinical trials Herszfeld D, Wolvetang E, Langton-Bunker E, Chung TL,
in patients with Parkinsons disease. Ann N Y Acad Sci Filipczyk AA, Houssami S, Jamshidi P, Koh K, Laslett AL,
495:473496. Michalska A, Nguyen L, Reubinoff BE, Tellis I, Auerbach
Cho MS, Lee YE, Kim JY, Chung S, Cho YH, Kim DS, Kang JM, Ording CJ, Looijenga LH, Pera MF (2006) CD30 is
SM, Lee H, Kim MH, Kim JH, Leem JW, Oh SK, Choi YM, a survival factor and a biomarker for transformed human
Hwang DY, Chang JW, Kim DW (2008) Highly efficient pluripotent stem cells. Nat Biotechnol 24:351357.
and large-scale generation of functional dopamine neurons Iacovitti L, Donaldson AE, Marshall CE, Suon S, Yang MA
from human embryonic stem cells. Proc Natl Acad Sci USA (2007) Protocol for the differentiation of human embryonic
105:33923397. stem cells into dopaminergic neurons using only chemically
Chung S, Hedlund E, Hwang M, Kim DW, Shin BS, Hwang DY, defined human additives: studies in vitro and in vivo. Brain
Jung Kang U, Isacson O, Kim KS (2005) The homeodomain Res 1127:1925.
transcription factor Pitx3 facilitates differentiation of mouse Kanda S, Tamada Y, Yoshidome A, Hayashi I, Nishiyama T
embryonic stem cells into AHD2-expressing dopaminergic (2004) Over-expression of bHLH genes facilitate neural for-
neurons. Mol Cell Neurosci 28:241252. mation of mouse embryonic stem (ES) cells in vitro. Int
Chung S, Shin BS, Hedlund E, Pruszak J, Ferree A, Kang UJ, J Dev Neurosci 22:149156.
Isacson O, Kim KS (2006) Genetic selection of sox1GFP- Kawasaki H, Mizuseki K, Nishikawa S, Kaneko S, Kuwana
expressing neural precursors removes residual tumorigenic Y, Nakanishi S, Nishikawa SI, Sasai Y (2000) Induction of
pluripotent stem cells and attenuates tumor formation after midbrain dopaminergic neurons from ES cells by stromal
transplantation. J Neurochem 97:14671480. cell-derived inducing activity. Neuron 28:3140.
Cowan CA, Klimanskaya I, McMahon J, Atienza J, Witmyer J, Kawasaki H, Suemori H, Mizuseki K, Watanabe K, Urano
Zucker JP, Wang S, Morton CC, McMahon AP, Powers D, F, Ichinose H, Haruta M, Takahashi M, Yoshikawa K,
Melton DA (2004) Derivation of embryonic stem-cell lines Nishikawa S, Nakatsuji N, Sasai Y (2002) Generation of
from human blastocysts. N Engl J Med 350:13531356. dopaminergic neurons and pigmented epithelia from primate
Dimos JT, Rodolfa KT, Niakan KK, Weisenthal LM, Mitsumoto ES cells by stromal cell-derived inducing activity. Proc Natl
H, Chung W, Croft GF, Saphier G, Leibel R, Goland Acad Sci USA 99:15801585.
R, Wichterle H, Henderson CE, Eggan K (2008) Induced Kim JH, Auerbach JM, Rodriguez-Gomez JA, Velasco I, Gavin
pluripotent stem cells generated from patients with ALS D, Lumelsky N, Lee SH, Nguyen J, Sanchez-Pernaute R,
can be differentiated into motor neurons. Science 321:1218 Bankiewicz K, McKay R (2002) Dopamine neurons derived
1221. from embryonic stem cells function in an animal model of
Draper JS, Smith K, Gokhale P, Moore HD, Maltby E, Johnson Parkinsons disease. Nature 418:5056.
J, Meisner L, Zwaka TP, Thomson JA, Andrews PW (2004) Kim DW, Chung S, Hwang M, Ferree A, Tsai HC, Park
Recurrent gain of chromosomes 17q and 12 in cultured JJ, Chung S, Nam TS, Kang UJ, Isacson O, Kim KS
human embryonic stem cells. Nat Biotechnol 22:5354. (2006) Stromal cell-derived inducing activity, Nurr1, and
Fearnley JM, Lees AJ. Ageing and Parkinsons disease: sub- signaling molecules synergistically induce dopaminergic
stantia nigra regional selectivity (1991) Brain 114 (Pt 5): neurons from mouse embryonic stem cells. Stem Cells 24:
22832301. 557567.
15 ES Cell Transplantation for the Treatment of Parkinsons Disease 253

Ko JY, Park CH, Koh HC, Cho YH, Kyhm JH, Kim YS, Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S
Lee I, Lee YS, Lee SH (2007) Human embryonic stem (2008) Generation of mouse induced pluripotent stem cells
cell-derived neural precursors as a continuous, stable, and on- without viral vectors. Science 322:949953.
demand source for human dopamine neurons. J Neurochem Olanow CW, Goetz CG, Kordower JH, Stoessl AJ, Sossi V,
103:14171429. Brin MF, Shannon KM, Nauert GM, Perl DP, Godbold J,
Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW Freeman TB (2003) A double-blind controlled trial of bilat-
(2008) Lewy body-like pathology in long-term embryonic eral fetal nigral transplantation in Parkinsons disease. Ann
nigral transplants in Parkinsons disease. Nat Med 14: Neurol 54:403414.
504506. Park IH, Arora N, Huo H, Maherali N, Ahfeldt T, Shimamura
Lee SH, Lumelsky N, Studer L, Auerbach JM, McKay RD A, Lensch MW, Cowan C, Hochedlinger K, Daley GQ
(2000) Efficient generation of midbrain and hindbrain neu- (2008) Disease-specific induced pluripotent stem cells. Cell
rons from mouse embryonic stem cells. Nat Biotechnol 134:877886.
18:675679. Park S, Lee KS, Lee YJ, Shin HA, Cho HY, Wang KC, Kim
Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, YS, Lee HT, Chung KS, Kim EY, Lim J (2004) Generation
Lashley T, Quinn NP, Rehncrona S, Bjorklund A, Widner of dopaminergic neurons in vitro from human embryonic
H, Revesz T, Lindvall O, Brundin P (2008) Lewy bodies in stem cells treated with neurotrophic factors. Neurosci Lett
grafted neurons in subjects with Parkinsons disease suggest 359:99103.
host-to-graft disease propagation. Nat Med 14:501503. Park CH, Minn YK, Lee JY, Choi DH, Chang MY, Shim JW,
Maitra A, Arking DE, Shivapurkar N, Ikeda M, Stastny V, Ko JY, Koh HC, Kang MJ, Kang JS, Rhie DJ, Lee YS, Son
Kassauei K, Sui G, Cutler DJ, Liu Y, Brimble SN, Noaksson H, Moon SY, Kim KS, Lee SH (2005) In vitro and in vivo
K, Hyllner J, Schulz TC, Zeng X, Freed WJ, Crook J, analyses of human embryonic stem cell-derived dopamine
Abraham S, Colman A, Sartipy P, Matsui S, Carpenter M, neurons. J Neurochem 92:12651276.
Gazdar AF, Rao M, Chakravarti A (2005) Genomic alter- Perrier AL, Tabar V, Barberi T, Rubio ME, Bruses J, Topf
ations in cultured human embryonic stem cells. Nat Genet N, Harrison NL, Studer L (2004) Derivation of midbrain
37:10991103. dopamine neurons from human embryonic stem cells. Proc
Marshall JF, Ungerstedt U (1977) Striatal efferent fibers play Natl Acad Sci USA 101:1254312548.
a role in maintaining rotational behavior in the rat. Science Pruszak J, Sonntag KC, Aung MH, Sanchez-Pernaute R, Isacson
198:6264. O (2007) Markers and methods for cell sorting of human
Martin MJ, Muotri A, Gage F, Varki A (2005) Human embryonic embryonic stem cell-derived neural cell populations. Stem
stem cells express an immunogenic nonhuman sialic acid. Cells 25:22572268.
Nat Med 11:228232. Rodriguez-Gomez JA, Lu JQ, Velasco I, Rivera S, Zoghbi SS,
Mendez I, Dagher A, Hong M, Hebb A, Gaudet P, Law A, Liow JS, Musachio JL, Chin FT, Toyama H, Seidel J, Green
Weerasinghe S, King D, Desrosiers J, Darvesh S, Acorn MV, Thanos PK, Ichise M, Pike VW, Innis RB, McKay
T, Robertson H (2000) Enhancement of survival of stored RD (2007) Persistent dopamine functions of neurons derived
dopaminergic cells and promotion of graft survival by expo- from embryonic stem cells in a rodent model of Parkinsons
sure of human fetal nigral tissue to glial cell line-derived neu- disease. Stem Cells 25:918928.
rotrophic factor in patients with Parkinsons disease. Report Rolletschek A, Chang H, Guan K, Czyz J, Meyer M, Wobus
of two cases and technical considerations. J Neurosurg AM (2001) Differentiation of embryonic stem cell-derived
92:863869. dopaminergic neurons is enhanced by survival-promoting
Mendez I, Sanchez-Pernaute R, Cooper O, Vinuela A, Ferrari D, factors. Mech Dev 105:93104.
Bjorklund L, Dagher A, Isacson O (2005) Cell type analysis Roy NS, Cleren C, Singh SK, Yang L, Beal MF, Goldman
of functional fetal dopamine cell suspension transplants in SA (2006) Functional engraftment of human ES cell-
the striatum and substantia nigra of patients with Parkinsons derived dopaminergic neurons enriched by coculture with
disease. Brain 128:14981510. telomerase-immortalized midbrain astrocytes. Nat Med
Mendez I, Vinuela A, Astradsson A, Mukhida K, Hallett P, 12:12591268.
Robertson H, Tierney T, Holness R, Dagher A, Trojanowski Samii A, Nutt JG, Ransom BR (2004) Parkinsons disease.
JQ, Isacson O (2008) Dopamine neurons implanted into peo- Lancet 363:17831793.
ple with Parkinsons disease survive without pathology for Schulz TC, Noggle SA, Palmarini GM, Weiler DA,
14 years. Nat Med 14:507509. Lyons IG, Pensa KA, Meedeniya AC, Davidson BP,
Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka Lambert NA, Condie BG (2004) Differentiation of
T, Aoi T, Okita K, Mochiduki Y, Takizawa N, Yamanaka human embryonic stem cells to dopaminergic neu-
S (2008) Generation of induced pluripotent stem cells with- rons in serum-free suspension culture. Stem Cells 22:
out Myc from mouse and human fibroblasts. Nat Biotechnol 12181238.
26:101106. Shibata H, Ageyama N, Tanaka Y, Kishi Y, Sasaki K, Nakamura
Nishimura F, Yoshikawa M, Kanda S, Nonaka M, Yokota H, S, Muramatsu S, Hayashi S, Kitano Y, Terao K, Hanazono
Shiroi A, Nakase H, Hirabayashi H, Ouji Y, Birumachi J, Y (2006) Improved safety of hematopoietic transplantation
Ishizaka S, Sakaki T (2003) Potential use of embryonic with monkey embryonic stem cells in the allogeneic setting.
stem cells for the treatment of mouse parkinsonian mod- Stem Cells 24:14501457.
els: improved behavior by transplantation of in vitro Sonntag KC, Pruszak J, Yoshizaki T, van Arensbergen J,
differentiated dopaminergic neurons from embryonic stem Sanchez-Pernaute R, Isacson O (2007) Enhanced yield of
cells. Stem Cells 21:171180. neuroepithelial precursors and midbrain-like dopaminergic
254 A. Morizane and J. Takahashi

neurons from human embryonic stem cells using the BMP Neural conversion of ES cells by an inductive activity on
antagonist noggin. Stem Cells 25:411418. human amniotic membrane matrix. Proc Natl Acad Sci USA
Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K 103:95549559.
(2008) Induced pluripotent stem cells generated without viral Wernig M, Zhao JP, Pruszak J, Hedlund E, Fu D, Soldner
integration. Science 322:945949. F, Broccoli V, Constantine-Paton M, Isacson O, Jaenisch
Tabar V, Tomishima M, Panagiotakos G, Wakayama S, Menon R (2008) Neurons derived from reprogrammed fibroblasts
J, Chan B, Mizutani E, Al-Shamy G, Ohta H, Wakayama T, functionally integrate into the fetal brain and improve symp-
Studer L (2008) Therapeutic cloning in individual parkinso- toms of rats with Parkinsons disease. Proc Natl Acad Sci
nian mice. Nat Med 14:379381. USA 105:58565861.
Takagi Y, Takahashi J, Saiki H, Morizane A, Hayashi T, Kishi Y, Yamazoe H, Kobori M, Murakami Y, Yano K, Satoh M,
Fukuda H, Okamoto Y, Koyanagi M, Ideguchi M, Hayashi Mizuseki K, Sasai Y, Iwata H (2006) One-step induction
H, Imazato T, Kawasaki H, Suemori H, Omachi S, Iida of neurons from mouse embryonic stem cells in serum-free
H, Itoh N, Nakatsuji N, Sasai Y, Hashimoto N (2005) media containing vitamin B12 and heparin. Cell Transplant
Dopaminergic neurons generated from monkey embryonic 15:135145.
stem cells function in a Parkinson primate model. J Clin Yang D, Zhang ZJ, Oldenburg M, Ayala M, Zhang SC (2007)
Invest 115:102109. Human embryonic stem cell-derived dopaminergic neurons
Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, reverse functional deficit in parkinsonian rats. Stem Cells
Tomoda K, Yamanaka S (2007) Induction of pluripotent stem 26:5563.
cells from adult human fibroblasts by defined factors. Cell Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J,
131:861872. Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart
Takahashi K, Yamanaka S (2006) Induction of pluripotent stem R, Slukvin II, Thomson JA (2007) Induced pluripotent stem
cells from mouse embryonic and adult fibroblast cultures by cell lines derived from human somatic cells. Science 318:
defined factors. Cell 126:663676. 19171920.
Thompson L, Barraud P, Andersson E, Kirik D, Bjorklund Yue F, Cui L, Johkura K, Ogiwara N, Sasaki K (2006) Induction
A (2005) Identification of dopaminergic neurons of nigral of midbrain dopaminergic neurons from primate embry-
and ventral tegmental area subtypes in grafts of fetal onic stem cells by coculture with sertoli cells. Stem Cells
ventral mesencephalon based on cell morphology, pro- 24:16951706.
tein expression, and efferent projections. J Neurosci 25: Zeng X, Cai J, Chen J, Luo Y, You ZB, Fotter E, Wang Y, Harvey
64676477. B, Miura T, Backman C, Chen GJ, Rao MS, Freed WJ
Ueno M, Matsumura M, Watanabe K, Nakamura T, Osakada (2004) Dopaminergic differentiation of human embryonic
F, Takahashi M, Kawasaki H, Kinoshita S, Sasai Y (2006) stem cells. Stem Cells 22:925940.
Chapter 16

Functional Multipotency of Neural Stem Cells


and Its Therapeutic Implications

Yang D. Teng, Serdar Kabatas, Jianxue Li, Dustin R. Wakeman, Evan Y. Snyder,
and Richard L. Sidman

Contents macro-environment towards the formation and self-


maintenance of a physiologically functioning adult
16.1 Background . . . . . . . . . . . . . . . . 256 nervous system. We believe that embracing this view
16.2 The Neural Stem Cell . . . . . . . . . . . . 257 of the NSCs multipotency is pivotal for correctly,
16.2.1 Biological Definition . . . . . . . . 257 efficiently, and optimally exploiting stem cell biology
16.2.2 Issues of Cell Identification:
for therapeutic applications, including reconstituting
Cross-Differentiation and Cell Fusion . 257
16.3 Analysis of Neurogenesis and Neural Stem the dysfunctional CNS.
Cell Fate . . . . . . . . . . . . . . . . . . 258
16.3.1 In Vivo . . . . . . . . . . . . . . . 258 Keywords Homeostasis Multipotency Neural stem
16.3.2 In Vitro . . . . . . . . . . . . . . 259 cells Spinal cord injury Transplantation
16.4 Clinically Oriented Investigations . . . . . . 261
16.4.1 Spinal Cord Injury . . . . . . . . . . 261
16.4.2 Neurodegenerative Diseases . . . . . 264 Abbreviations
16.4.3 Stroke . . . . . . . . . . . . . . . 265
16.5 Conclusion . . . . . . . . . . . . . . . . . 266
References . . . . . . . . . . . . . . . . . . . . 266
AD Alzheimers disease
ALS amyotrophic lateral sclerosis
bFGF basic fibroblast growth factor
Abstract In this review, we propose an updated con- BrdU bromodeoxyuridine
cept of the neural stem cell (NSC). New data of our CNTF ciliary neurotrophic factor
own and others suggest that the fields conventional CS-PG chondroitin sulphate proteoglycans
view which has touched principally on the essential DAT dopamine transporter
multipotency of lineage phenotypes (i.e., the ability DDC dopa-decarboxylase
of NSC to differentiate into all neural cells), should EAE experimental allergic encephalomyelitis
be broadened to include the emerging recognition of EdU 5-ethynyl-2-deoxyuridine
the biofunctional multipotency of the NSC to medi- EGF epidermal growth factor
ate systemic homeostasis. Under this new conceptual GAP growth-associated protein
context, one may begin to appreciate and seek the GRP glial restricted precursors
logic and teleology behind the wide range of molec- HD Huntingtons disease
ular tactics the NSC appear to serve at each devel- hNSC human neural stem cells
opmental stage as they integrate into and prepare, HSC hematopoietic stem cells
modify, and guide the surrounding CNS micro- and LIF leukaemia inhibitory factor
LSD lysosomal storage diseases
MP methylprednisolone
MS multiple sclerosis
Y.D. Teng () MPTP 1-methyl-4-phenyl-1,2,3,6-tetra-
Department of Neurosurgery, HMS/CHB/BWH, Boston, MA, hydropyridine
USA
e-mail: yang_teng@hms.harvard.edu NRP neuronal restricted precursors

H. Ulrich (ed.), Perspectives of Stem Cells, 255


DOI 10.1007/978-90-481-3375-8_16, Springer Science+Business Media B.V. 2010
256 Y.D. Teng et al.

NSC Neural stem cell(s) A decade later, Nottebohm and collaborators


PD Parkinsons disease demonstrated telencephalic neuronal replacement in
SN substantia nigra the adult avian brain related to seasonal song learn-
ing (Graziadei and Graziadei, 1979; Goldman and
Nottlebohm, 1983; Nottlebohm, 2004). Around the
same time, it was observed that newborn neurons in the
16.1 Background adult rodent hippocampus appeared to receive synap-
tic inputs (Kaplan and Bell, 1983), and later it was
In 1959, a new method of marking dividing cells in established that they also extended axon projections to
the mammalian brain was developed (Sidman et al., their target area (Stanfield and Trice, 1988). Parallel to
1959). The method was based on the already known these studies, a new cell tracer was being explored that
fact (Hughes et al., 1958) that [3 H]-thymidine injected would accelerate research in the stem/progenitor cell
systemically in mammals would be incorporated selec- field (Gratzner, 1982). The marker, bromodeoxyuri-
tively into DNA replicating during S-phase of the cell dine (BrdU), is a synthetic thymidine analogue that
cycle (Howard and Pelc, 1953), and their positions and also becomes incorporated into a cells DNA during the
numbers ascertained by autoradiography. S-phase of the cell cycle, much like [3 H]-thymidine,
The availability of this technology set an impor- but could be easily and inexpensively detected by
tant process in motion, through which the conven- immunocytochemistry, in contrast to [3 H]-thymidine.
tional doctrine of neurogenesis was reexamined (Ming A new technique based on incorporation of the thymi-
and Song, 2005). The idea that no new neurons are dine analogue 5-ethynyl-2 -deoxyuridine (EdU), with
formed after birth became dogma in neuroscience subsequent detection by click chemistry, is a very
since Santiago Ramon y Cajal, in 1913, hypothe- sensitive, rapid method, independent of fixation or
sized that neurons are generated exclusively during the harsh denaturing treatment of the tissue, and appli-
prenatal phase of development. Although some investi- cable to thick sections or whole mounts (Salic and
gators held different opinions, it was impossible, with Mitchison, 2008; Buck et al., 2008). By staining a
the resources and methods obtainable then, to prove given cell for dual expression of BrdU or EdU and
that postnatally born cells were actually neurons and a cell type-specific marker, the phenotypic fate of a
not glia. newly divided cell can be readily analyzed and quan-
Using the [3 H]-thymidine labeling technique, pat- tified by unbiased stereological methods. This tech-
terns of neuron genesis and migration were demon- nique has become a mainstay in stem cell research.
strated in several developing areas of the mouse brain Only recently has there been concern that BrdU and
(Angevine and Sidman, 1961; Miale and Sidman, other thymidine analogs might be passed from labeled
1961; Sidman, 1961), and in the adult mouse brain grafted stem cells to dividing host cells, allowing for an
(Smart, 1961). Soon after, it was suggested that not element of misidentification unless adequate controls
only astrocytes and microglial cells, but also oligo- are invoked (Burns et al., 2006). Also, labeling might
dendrocytes (Altman, 1962a) and neurons proliferate represent DNA repair in a non-mitotic cell (Bauer and
(Altman, 1962b) in adult mammals. Subsequently, Patterson, 2005), and cell cycle activity with labeling
Angevine (1965) reported that granule cell neurons by thymidine analogs can be a prelude not only to
were still forming in the mouse at least until post- cell division, but also to cell death of neurons (Herrup
natal day 20, the oldest age he examined, in and et al., 2004).
near the dentate gyrus of the hippocampal formation, In 1992, adult and fetal neural stem cells from the
and Altman and colleagues reported evidence of new CNS of rodents were isolated (Reynolds and Weiss,
neurons forming in various regions of the adult rat 1992; Snyder et al., 1992) and, in 1998, human neu-
brain including the dentate gyrus of the hippocampus ral stem cells (Flax et al., 1998; Kukekov et al., 1999).
(Altman and Das, 1965), the neocortex (Altman, 1966) Also in 1998, taking advantage of the fortuitous use of
and the olfactory bulb, the latter after cell proliferation a BrdU-like chemotherapeutic agent in the treatment
in the wall of the anterior horn of the lateral ventri- of a head and neck cancer, the presence of neurogene-
cle and migration via a pathway he named the rostral sis and rostral migratory stream in adult humans was
migratory stream (Altman, 1969). confirmed, further suggesting that concepts regarding
16 Functional Multipotency of Neural Stem Cells 257

stem cell biology being formulated in rodents may be rise to cell types in addition to themselves through
evolutionarily conserved (Eriksson et al., 1998; Curtis asymmetric cell division (Gage, 2000).
et al., 2007). Stem cells are defined according to their repertoires.
Also in the late 1980s and early 1990s, comple- A totipotent stem cell, if implanted in the uterus
mentary methods for identifying and tracking newly- of a living animal can give rise to a full organism
mitotic cells were being developed and employed for and all its organ systems, including CNS and PNS.
tracing the lineage of cells. The most powerful of A pluripotent stem cell, in the simplest definition,
these was the use of replication incompetent retro- is similar to the totipotent cell, except that it can-
viral vectors that could transduce a reporter trans- not give rise to trophoblasts of the placenta. Such
gene to the genome of cells passing through S-phase cells have been convincingly affirmed to exist only
such that this marker would become permanently inte- in the inner cell mass of the blastocyst, although a
grated in a unique chromosomal site and passed to series of recent controversial papers have suggested
all progeny and generations (Price et al., 1987; Sanes that such pluripotent cells may be harbored in the
et al., 1986). Unlike BrdU, which becomes diluted amniotic fluid, placenta, umbilical cord, and bone mar-
50% with each cell division, retroviral-transduced row mesenchyme (Ortiz-Gonzalez et al., 2004). The
genes are not diluted. Combining such marked cells next developmental stage in the progressive restric-
with electrophysiological analysis could provide con- tion of potency is the multipotent somatic stem cell,
vincing evidence that newborn neurons in the adult which is capable of differentiating into all cell types
mammalian CNS are functional and synaptically inte- of a given organ or tissue and into only cells of that
grated (Belluzzi et al., 2003; Carleton et al., 2003, van organ or tissue (Martinez-Serrano et al., 2001). Even
Praag et al., 2002). Yet, though it is now known that though that is the traditional and most widely accepted
neurogenesis persists in some regions of the human definition of the multipotent stem cell, recent find-
adult CNS and that these new neurons are functional ings have forced a reexamination of this biological
and connected, it remains not entirely certain what concept, based on experiments showing possible cross-
biological roles are served by the newborn cells in differentiation, although some such instances have
these few neurogenic hot-spots (Zhao et al., 2008), been found to be artifacts caused by cell fusion, as
and especially in regions such as the adult spinal cord discussed in the next section. Hence, while an active
where stem cell proliferation only occurs transiently area of inquiry at the time of this writing, the distinc-
following trauma or other non-physiological influences tions above remain the lexicon of the field. Importantly,
(Teng et al., 2006). a common belief among stem cell biologists is that
the distinction between totipotence, pluripotence, and
multipotence is not discrete, but rather a continuum
16.2 The Neural Stem Cell in development. Some investigators also define adult
brain-derived stem cells as neural progenitor cells
(Shihabuddin et al., 2004). In addition, it is currently
16.2.1 Biological Definition not clear if any specific nomenclature will be desig-
nated for neural stem/progenitor-like cells that are
Neural stem cells (NSC) are the most primordial and derived from induced pluripotent stem (iPS) cells (e.g.,
uncommitted cells of the nervous system, and are naming them as neural precursor cells; Wernig et al.,
believed to give rise to the vast array of more special- 2008).
ized cells of the CNS and peripheral nervous system
(PNS). To be considered a neural stem cell in
contrast to a progenitor cell (i.e., cells that have 16.2.2 Issues of Cell Identification:
already become lineage-committed to give rise to only
Cross-Differentiation
one category of neural component, e.g., glial cells
versus neurons) that cell must be capable of (i) and Cell Fusion
generating all neural lineages (neurons, astrocytes and
oligodendrocytes) throughout the nervous system, (ii) Somatic stem cells sometime erroneously called
having some capacity for self-renewal and (iii) giving adult stem cells in the lay press have been viewed
258 Y.D. Teng et al.

as exceptionally plastic but nevertheless restricted to as in pathophysiological functions. This process, as


the production of cells from the tissue of origin but not suggested by some investigators, could provide a
cells of non-related tissues. For example, NSC give rise strategy for de-differentiating committed cells that
to the three main types of nervous system cells (neu- might then be reprogrammed for tissue reconstitution
ron, oligodendrocyte and astrocyte), while hematopoi- and reversal or repair of injury (Ogle et al., 2005).
etic stem cells (HSC) produce only blood derived cells, Resolution of the cross-differentiation controversy
etc. However, various reports in the last few years will obviously have a major impact on our current
have challenged this central dogma by demonstrating definitions of stem cells.
that adult stem cells, under certain microenvironmen-
tal (often very non-physiological) conditions, generate
cell types besides those in the tissue of origin, pos-
sibly indicating that they can switch cell fate. HSC, 16.3 Analysis of Neurogenesis
for instance, in addition to forming blood cells, have and Neural Stem Cell Fate
been reported to develop into liver cells (Petersen et al.,
1999) and NSC to give rise not only to nerve cells
but also to early hematopoietic precursors (Bjornson 16.3.1 In Vivo
et al., 1999). These cell behaviors have been termed
cross-differentiation or stem cell plasticity. Such As briefly described, the analysis of neurogenesis
reports have generated excitement as well as skepti- in vivo was largely established by the use of [3 H]-
cism in the field of stem cell biology, as the concept thymidine in 1959 and bromodeoxyuridine (BrdU)
of plasticity defies the developmental biology principle after 1982 as markers of cell division. Those label-
that lineage restriction is imparted during morpho- ing techniques made it possible to track endogenous as
genesis. However, if correct, the ability of somatic well as donor stem/progenitor cells and do quantitative
stem cells to change fate also holds immense ther- analysis of proliferation, differentiation, and survival
apeutic potential as well in circumventing the con- of all cells born after the injection or prelabeling of
cerns by some of having to obtain pluripotent cells such a marker (Kempermann et al., 1997; Miller and
from human embryos (Lakshmipathy and Verfaillie, Nowakowski, 1988; Redmond et al., 2007; Sidman,
2005). 1970).
The controversy comes when more recent stud- Because all marking techniques carry caveats and
ies suggest that such stem cell plasticity might require rigorous controls, a practical principle is to use
not actually exist. Rather, the illusion of cross- at least two independent markers and marking tech-
differentiation/transdifferentiation might actually niques for identifying cells. For example, a strategy
result from the fusion of donor cells with host cells to label newly born cells might include BrdU or EdU
conferring on these host cells the transplanted cells incorporation and/or the tagging of a cell with a genetic
reporter gene and creating the misperception that marker carried in a replication-incompetent, helper-
these host cells are donor-derived. In other words, the virus-free retroviral vector that lacks nuclear import
markers used to visualize the grafted cells in vivo or mechanisms. With the latter technique, viral integra-
in vitro have simply been taken up by the host cells tion occurs only when the nuclear membrane breaks
but no differentiation of donor cells into differentiated down during mitosis. Also the transgene becomes
organ-specific cells had actually occurred. It is also transduced only when the vector adheres to a spe-
possible that cell fusion may cause a change in the cific cell surface receptor. Therefore, they are good,
phenotype and/or the function of cells (through, e.g., non-diffusible, non-dilutable markers of dividing cells
nuclear reprogramming) and may also explain the (Lewis and Emerman, 1994; Ming and Song, 2005).
apparent inherent plasticity of committed cells. While Other methods for distinguishing donor cells from
cell fusion may have created, in many instances, the host cells in transplantation studies might include mis-
misleading appearance of cross-differentiation, the matching the species or sex between the two; for
prevalence of this process itself has caused stem cell example, transplanting human cells into a rodent organ
biologists to speculate whether fusion may actually or implanting a male cell (recognized by its Y chromo-
play a natural role in some normal biological as well some) into a female host (e.g., Li et al., 2006b).
16 Functional Multipotency of Neural Stem Cells 259

Some research might employ cell type- or develop- has been very important for cell isolation and culture.
mental stage-specific markers to track the development In the late 1980s and early 1990s it became evident
and differentiation of a stem cell. For example, a that most regions of the CNS contain a small popula-
developmental neuron is defined as immature when tion of individual cells that could be isolated and could
it expresses immature markers but lack mature ones. give rise to clonally-related populations consisting of
Therefore, antibodies directed against those imma- multiple neural cell types that heretofore had not been
ture markers would stain only newborn, still imma- regarded as deriving from a common progenitor. These
ture cells that are undergoing neuronal differentiation. regions included olfactory bulb and cerebellum (Ryder
Oligodendrocyte (Olg) development can be used to et al., 1990; Snyder et al., 1992); postnatal (Altman,
illustrate the use of this technique. Even though Olgs 1966) as well as embryonic neocortex (where 18% of
express proteolipid protein (PLP) and DM20 (myelin the cells could generate both neurons and oligodendro-
proteins) during most of their development, the ratio cytes in vitro) (Williams et al., 1991), hippocampus
between these two proteins varies during differenti- (Renfranz et al., 1991; Gage et al., 1995); and sep-
ation (Yu et al., 1994; Timsit et al., 1995). While tum (Temple, 1989). From these studies, however, it
DM20s expression precedes that of PLP and is kept became clear that neural progenitors or stem cells,
high during the first stages of differentiation, PLP is when isolated from the CNS, had a predisposition to
expressed in higher levels only at the latter stages of exit the cell cycle and differentiate unless an inter-
differentiation and, therefore, defines a mature myelin- vention was imposed upon them to remain cycling
forming oligodendrocyte (Meng et al., 2005). Multiple and hence hold commitment in abeyance. For exam-
steps at multiple CNS sites have been defined for this ple, cells cultured from the embryonic rat septum were
cell type (Miller, 2002, 2005). Similarly, other markers incapable of more than one cycle of cell division unless
for other cell types can be used to track the devel- they were co-cultured with fetal striatal tissue (Temple,
opment of a particular cell type. It is important to 1989). When they were prodded to continue prolifer-
reiterate that this method is best used in combination ating, some of the cells appeared to be multipotent.
with some or all of the others described above, because Subsequent studies were launched to identify exoge-
antibodies to developmental markers are not always nous growth factors and/or internal genes that could
ideally specific (Brandt et al., 2003; Kempermann be used to maintain multipotent cells in a proliferative
et al., 2004). state that might allow them to be expanded and used
for developmental or therapeutic studies. The princi-
pal mitogens studied and most commonly used to date
16.3.2 In Vitro are basic fibroblast growth factor (bFGF) (Richards
et al., 1992), epidermal growth factor (EGF) (Reynolds
The isolation and culture of NSC in vitro is of great and Weiss, 1992) and leukemia inhibitory factor (LIF)
value, providing an opportunity to scrutinize individ- (Smith et al., 1988).
ual cells and their differentiation more closely, as well As described above, one of the first difficulties
as to prepare them for research and therapeutic appli- encountered in attempting to culture NSC was that
cations. It also allows one to exclude confounding isolated single cells tended to cease dividing and differ-
variables (e.g., intervention by the immune system) entiate. Only cells surrounded by other cells remained
and to manipulate the microenvironment in system- mitotic, suggesting that cell-to-cell contact was impor-
atic and prescribed ways. Stem cells can be propagated tant for cell proliferation (Temple and Qian, 1995).
as clonal populations in vitro by many effective and Further investigation suggested that a likely candi-
safe means that include both epigenetic and genetic date for mediating this inter-cellular interaction was
strategies. basic fibroblast growth factor (bFGF). Besides being
known for its association with extracellular matrix and
cell membranes, bFGF was surprisingly found to be
16.3.2.1 Epigenetic present throughout CNS development (Baird, 1994;
Kilpatrick et al., 1995). Previous studies had already
The identification of cytokines and growth factors that shown that the addition of exogenous bFGF to cultures
affect survival and proliferation of neural stem cells of cortical neuroectoderm cells appeared to stimulate
260 Y.D. Teng et al.

their proliferation and lead to increased numbers of from tripotent cells, bypassing two of the three normal
neurons (Gensburger et al., 1987), but, it wasnt until bipotent intermediates. It also promotes the expansion
the 1990s that the issue was addressed of which type of specified astrocytic progenitors (Ravin et al., 2008).
of cortical cell was the target of bFGF. It was found Therefore, the method most used now for expand-
that bFGF would stimulate multipotent stem cells from ing and maintaining NSC cultures by epigenetic means
many regions of the CNS (Temple and Qian, 1995; involves addition of bFGF, EGF and LIF to a serum-
Gritti et al., 1996). free basal medium. Many studies (as well as our own
In addition to bFGF, studies in the 1980s had experience) suggest that, (at least for short periods),
already shown that epidermal growth factor (EGF) bFGF alone (at 20 ng/ml) is sufficient. As noted above,
bound to CNS cells and stimulated their prolifera- in addition to stimulating proliferation of NSC, respon-
tion (Simpson et al., 1982; Anchan et al., 1991). EGF siveness to these factors may be one technique for
induced proliferation of progenitor cells that began to selecting NSC from a mixed population of cells at
divide and would come to form a cluster of undifferen- various degrees of maturation, potency, and lineage
tiated cells that expressed nestin, an intermediate fila- commitment.
ment present in neuroepithelial stem-like cells. These Culturing cells with mitogens alone is a double-
cells, when they exited the cell cycle, would differenti- edged sword. Because one does not by intent genet-
ate into neurons and astrocytes (Reynolds et al., 1992). ically manipulate the cells, approval by regulatory
These EGF-expanded cells, when then stimulated by agencies for translation to the clinic may be eas-
bFGF, would yield two progenitor cell sub-types: one ier to obtain. On the downside, however, obtaining a
giving rise to neurons and astrocytes, the other gener- sufficient number of cells while avoiding senescence
ating only neurons (Vescovi et al., 1993). Addition of (particularly of human cells) can be quite challeng-
both bFGF and EGF simultaneously in the same cell ing. Furthermore, one may be selecting for cells with
culture yielded better cell survival than either agent particular avidity for these factors and hence may
alone, and would also better maintain their ability to be more prone to neoplastic transformation. Genetic
differentiate into neurons, oligodendrocytes and astro- modifications that might be imposed by tonic expo-
cytes (Vescovi et al., 1999). The notion emerged that sure to high concentrations of a mitogen are mainly
the most immature and hence most multipotent unknown and less controllable than the some of the
neural stem/progenitor cells bore receptors for both well-studied self-regulated or regulatable genetic aug-
mitogens. Selecting for the presence of both receptors mentations described in the next section (Snyder et al.,
(and hence responsiveness to both mitogens) became 1992; Roy et al., 2004).
the basis of a quick and easy screening technique for
selecting neural stem cell populations from a hetero-
geneous CNS culture (including from human material;
Flax et al., 1998). 16.3.2.2 Genetic
The addition of leukaemia inhibitory factor (LIF)
to cell cultures yielded better long term growth than The genetic strategy for culturing NSC in vitro seeks
bFGF and EGF alone. No difference was noted in early to augment and prolong the expression of stemness
stage cultures, but, after 5060 days in vitro, the cul- genes. Myc represents such a gene; it is becoming rec-
tures without LIF consistently showed slower expan- ognized as important to the proliferative state of stem
sion while those with all three mitogens continued to cells. When its expression is prolonged, cells remain
expand (Carpenter et al., 1999). in the cell cycle and hence their differentiation is held
Interestingly, it has been demonstrated that under in abeyance. When they exit the cell cycle, they may
the presence of bFGF, multipotent NSC (i.e., tripotent still respond to regional epigenetic cues and differ-
cells) normally produce specified neural progenitors entiate appropriately spontaneously and inherently
through a bipotent intermediate cell type. In fact, the downregulating myc translation. In other words, the
tripotent state may be reset at each passage. The cil- exogenous extra copy of myc may be regulated by the
iary neurotrophic factor (CNTF) is believed to instruct cell as it does its cellular myc. Interestingly, embry-
multipotent cells to an astrocytic fate. More recently, it onic stem cells and iPS cells are colloquially said to
was reported that CNTF could direct astrogliogenesis be naturally immortalized, with myc being one the
16 Functional Multipotency of Neural Stem Cells 261

four genes first used to induce pluripotency in postmi- Broadly, there are two fundamental interlocking strate-
totic cells (Takahashi and Yamanaka, 2006). However, gies for using NSC therapeutically. The first seeks
the goal, in most studies, for augmenting certain genes to use the stem cell to provide replacement cells for
in somatic stem cells is to temporarily and control- those that have become dysfunctional or lost (Muotri
lably co-opt that quality safely and effectively. The and Gage, 2006). The second exploits the observation
NSC are usually transfected using a retroviral vector that stem cells (particularly in their non-neuronal state)
encoding the stemness gene. These clonal NSC have constitutively produce neuroprotective, immunoregu-
the advantage of being easily expanded to large num- latory and homeostatic molecules that serve to reduce
bers that typically remain stable and homogenous over host cell loss and inhibit the formation of barriers to
long periods of time from experiment to experiment self-repair (Teng et al., 2002, 2006). We will review
and recipient to recipient without senescing. Such cells both lines of research as they apply to the most fre-
have not only proved to be an ideal model for under- quently studied diseases and where NSC seem to be
standing fundamental aspects of stem cell biology, but, most promising.
because they have been, in many published cases, used
safely and effectively as transplant material for a wide
variety of disease models over the past two decades,
they have established a good reference of what ther- 16.4.1 Spinal Cord Injury
apeutic benefit should be safely achievable by a cell
with stem-like qualities (Parker et al., 2005). In fact, Experimental treatment strategies aimed at the acute
whatever expansion technique is selected for use by stage of the injury process currently include neu-
an investigator should be judged in part by its ability ronal protection and preservation of residual axons
to attain a commonly acceptable degree of safety and and white matter. Several approaches have been
efficacy in models of disease and injury. proposed and applied to this end, most notably anti-
Of course, when contemplating clinical strategies, secondary injury therapy using high dose methyl-
a complete knowledge of the fate of these cells as prednisolone (MP) (Hall and Springer, 2004). Ideally,
for any implanted cell must be vouchsafed. As noted with effective neuroprotection in place, treatments
above, while such cells have proven exceptionally safe, to promote axonal regeneration can be started
regulatory agencies generally feel most comfortable simultaneously or immediately thereafter. In pur-
approving the use of non-genetically modified cells. suing this possibility, experimental use of neu-
Ultimately, however, a well-characterized, uniform, rotrophins or neurotrophic factors to increase neuro-
stable, readily available, inexhaustible supply of clon- protection and axonal growth, and neutralizing anti-
ally related NSC may prove to be most efficacious bodies to Nogo and other oligodendrocyte-related
and practical (Vescovi and Snyder, 1999). Other genes inhibitory molecules have been investigated (Blesch
(e.g., telomerase) have been explored and proven valu- and Tuszynski, 2002; Schwab, 2004). While some
able (Roy et al., 2004). To maximize comfort with produced encouraging outcomes, evidence to the con-
stem cells, their stemness genes may be placed under trary has also been reported. The conflicting findings
regulatable control or put in tandem with suicide genes. do not negate the potential for this therapy but sug-
gest that this strategy, when used in isolation, may
not be sufficient to promote functionally meaning-
ful axonal regrowth in the injured spinal cord. For
example, enzymatic or genetic antagonism to chon-
16.4 Clinically Oriented Investigations droitin sulphate proteoglycans (CS-PG) has offered
some encouraging results. This approach was found,
Although the field of NSC biology is very young, however, to exert neuroprotection (Carter et al., 2008),
strategies by which these cells and their emerging and could be strengthened when it was jointly applied
properties might be harnessed to ameliorate a range of with maneuvers to prompt adult CNS neurons to
neurological disorders have already captured the imag- reenter growth mode (Silver and Miller, 2004).
ination of scientists, clinicians and the public, which Hence, combined therapies are likely to be most
has helped fuel the new field of regenerative medicine. effective.
262 Y.D. Teng et al.

Despite the fact that various treatment strategies to a large degree by intrinsic ability of NSC to secrete
have shown benefit in experimental animal models, neurotrophic factors, and/or immunomodulators, and
there is still no effective therapy for clinical spinal form gap junctions with host cells (as demonstrated by
cord injury (SCI). This difficult situation, in our opin- Teng et al., 2002 and subsequently reported by many
ion, is attributable to the following realities. First, others, including Ourednik et al., 2002; Lu et al., 2003;
there has been no conclusive evidence favoring one Pluchino et al., 2005; Llado et al., 2004; Yan et al.,
process as the predominant pathophysiological mech- 2004; Li et al., 2006, Bjugstad et al., 2005; Teng et al.,
anism which can account for all the spinal dysfunction 2006; Redmond et al., 2007; Jderstad et al., 2010).
seen following SCI. Most of the pathophysiological Thus, preserving the multipotency of these cells as
processes (e.g., secondary molecular events: glutamate opposed to attempting to direct them invariantly down
toxicity, sodium and calcium influx, free radical insult, the differentiation pathway of a single cell type might
cytochrome c release; secondary pathophysiological offer the greatest chance for cell-based therapies of
events: ischemia, anoxia, apoptosis, etc.) apparently the different inter-locked stages of SCI, but in a par-
exist either simultaneously or sequentially in an inter- simonious fashion. Harnessing the potentially broad
locked manner throughout the evolution of the injury therapeutic capacity of the NSC for use in an intelligent
and represent different facets of this complicated dis- and rationale manner requires learning the principles
ease entity (Tator and Fehlings, 1991; Teng et al., that can govern interaction between the pathologic tar-
2004). Most interventions reported to date target solely get and host environmental components, the NSC, and
one facet of the injury process which, in isolation, is other therapeutic reagents.
doomed to have limited benefit. To further complicate We believe that the innate biology of NSC (i.e.,
the situation, a given approach that may be useful when their default production and secretion of various neu-
used alone, may become ineffective or even detrimen- rotrophic factors and other molecules in a differentia-
tal when used in combination with other interventions, tion stage-dependent fashion) enables them to interact
perhaps working at cross purposes. Hence, it is crit- with the surrounding environment, including releasing
ical to understand the intricate interactions between trophic factors in an appropriate, regulated, stimulus-
these options and identify the underlying mechanisms appropriate manner. These factors, in our view, are
of their actions so that they may be orchestrated in a components of the stem cells inherent developmen-
safe, synergetic, and clinically feasible fashion (Reier, tal program which calls upon it to exert homeostatic
2004). forces upon a dynamically growing nervous system
Apropos to this point, we have come to view NSC which, otherwise, could become dysequilibrated. The
as the glue that can bind and integrate multiple result of this inherent program a dividend, so
approaches. Several studies have suggested that NSC, to speak, from developmental biology is to pro-
when transplanted into the injured brain or spinal cord mote, enable, induce, or catalyze the host to attempt
of rodents, migrate preferentially to and become inte- to reconstitute its own tissue, to minimize barriers to
grated within the damaged areas. A subpopulation this process, and to protect endangered cells from cell
of the transplanted NSC is re-directed to differenti- death or other toxic influences. Methods to optimize
ate into cell types that might replace the diseased or this process i.e., to work in concert with normal
degenerated host cells (Snyder et al., 1997; Rosario developmental tendencies, is undoubtedly desirable for
et al., 1997; Yandava et al., 1999; Park et al., 1999, optimizing repair.
2002b). More intriguingly and, ultimately, of proba- An example of harnessing and exploiting such
bly greater importance and utility is the observation inherent stem cell programs will be presented here.
that undifferentiated NSC or NSC that have pursued To direct neural repair more effectively following SCI,
a glial lineage seem to recondition the host CNS we cultured NSC ex vivo upon a biosynthetic scaffold
microenvironment and promote functional recovery by that mimicked the general structure of a healthy spinal
protecting pre-existent but threatened host neurons and cord. It had an inner section, engineered to emulate the
circuitry (Teng et al., 2002). The impact of this action gray matter with an isotropic pore structure of 250
is probably greater than neuronal replacement. The 500 m in diameter to facilitate seeding of the NSC.
precise mechanism by which NSC exert this homeo- The outer section of the scaffold, modeled to emu-
static pressure is unclear, though it is likely attributable late the white matter, had long, axially oriented pores
16 Functional Multipotency of Neural Stem Cells 263

for axonal guidance and radial porosity to allow fluid et al., 2000). Indeed, it has been suggested that the
transport while inhibiting the ingrowth of scar tissue injured cord offers a microenvironment that is not
(Teng et al., 2002). Implantation of the scaffold-NSC favorable to the differentiation of multipotent NSC into
unit into an adult rat hemi-section model of SCI pro- neurons (Cao et al., 2002). Rather, it has been proposed
moted long-term improvement in function (persistent that transplanting neuronal and glial restricted precur-
for 1 year in a group of animals specifically designated sors (NRP/GRP) is more tractable, i.e., pre-committing
for long-term studies) relative to control groups. At 70 the cells to a particular lineage ex vivo rather than
days post injury, animals implanted with scaffold-plus- letting the in vivo environment direct their differenti-
NSC exhibited coordinated, weight-bearing hindlimb ation. The transplantation of NRP/GRP into the post-
stepping. Histological and immunocytochemical anal- contusion spinal cord did improve motor and sensory
ysis suggested that this recovery was not caused by function. Histological analysis showed that a subset of
neural cell replacement; rather, it is attributable pre- the NRP/GRP survived, filled the lesion site, differen-
dominantly to a reduction in host tissue loss from tiated into neurons and glia, and migrated selectively
secondary injury processes as well as diminished glial (Mitsui et al., 2005; Cao et al., 2005). Interestingly,
scarring. This work is the first to demonstrate explic- the volume of spinal cord spared was increased in
itly the chaperone neuroprotective effects of the NRP/GRP recipients, suggesting that their action may
NSC. Tract tracing demonstrated spared corticospinal nevertheless have been attributable in a large part
tract fibers coursing by the injury epicenter and their to local neuroprotection. The actual role that donor-
caudal ends expressing classic growth cone-type vari- derived neurons played in recreating neurocircuitry is
cosities, a finding not observed in untreated groups. presently not determined.
Together with evidence of enhanced local growth- We are coming to learn that replacing lost neural
associated protein (GAP)-43 expression by axons, a tissue with its complex connections is more challeng-
marker associated with regenerative processes (also ing than once assumed, even using a cell that can yield
not seen in controls), these findings suggest donor- immunocytochemically- and electrophysiologically-
derived neuroprotection and promotion of local neural proven neurons in vivo. We do believe that, as we better
regenerative and plastic processes that might have understand the molecular milieu of the injured cord,
also contributed to functional recovery. These results, some degree of reconstruction will be achievable per-
besides suggesting a novel approach to SCI, may more haps not of long projection neurons, but perhaps of
broadly serve as a prototype for the use of NSC interneurons with shorter axons. However, it is fortu-
to anchor multidisciplinary strategies in regenerative itous that the very same exogenous cells that we hope
medicine, including gene therapy, material science, will participate in circuit reconstitution may also con-
growth factor delivery, anti-inflammation and anti- currently be providing trophic and immunoregulatory
scarring strategies, and pharmacological interventions factors that can preserve host circuits. Future research
against secondary injury. must clearly pay attention not only to how the donor
The use of scaffolds and cellular bridges are well- cells are changed by the host but also how the host
suited for lesions in which there is large parenchymal is changed by the donor cells i.e., placing a greater
loss (as was created experimentally with the hemisec- research emphasis on dynamic donor-host interactions.
tion model described above) or where a syrinx might Such an understanding may allow this dynamic to be
otherwise form because of extensive cell death follow- optimized such that a greater number of endogenous
ing contusion. For injuries where the amount of tissue stem cells may be effectively mobilized (Konya et al.,
loss is less dramatic, the implantation of cells alone 2008).
(without a template) may be useful. Murine embryonic Endogenous NSC have been established to reside
stem cells (ESC) were observed to survive and promote in a few well-characterized secondary germinal zones
recovery in the contused spinal cord (McDonald et al., of the adult nervous system, most notably the sub-
1999). Although this recovery was at first attributed granular zone (SGZ) of the dentate gyrus of the hip-
to the few neurons that appeared to emerge, a more pocampus and the subventricular zone (SVZ) along the
detailed study showed that functional impact may, in anterior part of the lateral ventricle in the forebrain.
fact, have been more plausibly due to oligodendrocytes Endogenous NSC may also reside in the ependymal
that myelinated some traumatized host axons (Liu region of the spinal cord. In response to injuries like
264 Y.D. Teng et al.

stroke, adult hippocampal NSC may proliferate and literature, as well. Although traditionally, for dis-
differentiate into new, functioning neurons (Schaffer eases like PD, this has entailed arming cells to pro-
and Gage, 2004). However, NSC in the adult spinal vide substrates for dopamine production, such as
ependymal region do not seem to differentiate into tyrosine hydroxylase (TH) and dopa-decarboxylase
neurons when they reside in their normal spinal cord (DDC) (e.g., Kim et al., 2000), we have come to view
niche. Nevertheless, when these same NSC are trans- molecular therapies more broadly as described
planted into the SGZ, they do yield neurons (Doetsch above for SCI, using the NSCs intrinsic propen-
et al., 1999; Johansson et al., 1999; Shihabuddin sity to supply homeostasis-promoting, neuroprotec-
et al., 2000). Hence, limitations to neurogenesis must tive, and neurotrophic agents that directly benefit
emanate in large part from the microenvironment of host neural functioning. In preliminary studies, as
the adult spinal cord rather than from the cells them- proof-of-concept of this notion using human NSC
selves. Therefore, it seems feasible that either altering (hNSC), we have approached a model of PD that
the milieu or changing the cells to respond differently most faithfully mirrors the actual human entity: non-
to that milieu may permit these endogenous spinal human primates (African green monkeys, a macaque)
NSC to play a more prominent role in neuronal recon- exposed to the complex I mitochondrial toxin 1-
stitution in the adult cord (Danilov et al., 2006). To methyl-4-phenyl-1,2,3,6-tetra-hydropyridine (MPTP).
overcome the normal impediments to adult neurogene- hNSC were implanted into the left and right cau-
sis will require a better understanding of the biological date nucleus and the right substantia nigra (SN) of
roles of spinal NSC, especially their proliferation in eight MPTP-treated, severely Parkinsonian monkeys.
response to injury, inflammation, and neuroactivity By 4 months (and clearly by 7 months) post transplan-
all significantly unexplored (Teng et al., 2006). tation, the majority of hNSC were found bilaterally
along the nigrostriatal pathway and in the SN. In the
presence of NSC, the number and size of host TH-
and dopamine transporter (DAT)-expressing neurons
16.4.2 Neurodegenerative Diseases in SN, typically poisoned by MPTP, returned to
nearly non-MPTP-exposure control levels. Host TH+
Neurodegenerative diseases are a prominent target for neurons in the caudate, whose size-to-number ratio
stem cell therapy in general and NSC in particu- had become dysequilibrated, also returned to their nor-
lar. Although adult-onset Alzheimers disease (AD), mal ratio, as if forced towards equipoise by stem cell
Parkinsons disease (PD), amyotrophic lateral scle- transplant (Bjugstad et al., 2005). The host-nigral stri-
rosis (ALS), and Huntingtons disease (HD) tend to atal pathway, normally attenuated and dysfunctional
receive the most attention, there is a growing recog- in the MPTP-lesioned monkey (as in PD) was largely
nition that some of the lysosomal storage diseases preserved (or restored; Bjugstad et al., 2008). Taken
(LSD) affecting the nervous system in childhood are together, these actions resulted in significant and long-
attractive targets because each disease requires the term diminution in Parkinsonian symptoms. Therefore,
replacement of a single well-characterized, usually dif- a broader view of the role of stem cells even for
fusible, enzyme without a significant need for cell diseases characterized by degeneration of a particu-
replacement if treated early. In the case of the LSD, lar neuronal subtype, as in PD may make these
the enzymes to be replaced are typically produced diseases a more tractable therapeutic target than we
constitutively by the NSC as part of its normal house- initially presumed (Redmond et al., 2007). Exogenous
keeping (e.g., Shihabuddin et al., 2004; Sidman et al., NSC may similarly rescue neurons at genetic risk of
2007). In special cases, as when exogenous NSC gain degenaration (Li et al., 2006b)
access to a normal neuronal precursor pool, they may Diseases characterized by glial dysfunction might
differentiate abundantly into neurons (Rosario et al., be viewed in a similar manner. Although we
1997; Flax et al., 1998). had established long ago that NSC could yield
The notion that using NSC for molecular ther- effectively remyelinating oligodendrocytes through-
apies is likely more tractable even for neurode- out the brains of poorly myelinated mouse mutants
generative diseases that do ostensibly require cell (Yandava et al., 1999), some of the most preva-
replacement has begun to spill over into the adult lent white matter diseases of adulthood such
16 Functional Multipotency of Neural Stem Cells 265

as multiple sclerosis (MS) are characterized by Stem cell therapy for stroke may be divided into two
an environment that is likely inhospitable to both approaches: the first focuses on mobilizing endoge-
host and donor-derived oligodendrocytes. For exam- nous NSC and the second depends on providing exo-
ple in the experimental allergic encephalomyelitis geneous NSC. Obviously, as suggested above, not only
(EAE) model of MS (characterized by chronic CNS will both approaches likely be required for optimal
inflammation, multifocal demyelination, and axonal restitution of function, but the two strategies likely act
loss), syngeneic undifferentiated neural precursor cells synergistically.
injected intravascularly, transited from the vascular Typically, the strategy for exploiting the popu-
space into the inflamed intracerebral microenviron- lation of endogenous NSC has been to attempt to
ment (via constitutively activated integrins and func- stimulate their proliferation and neuronal differentia-
tional chemokine receptors), accumulated and survived tion by administering exogenous growth factors and
within perivascular areas (in the company of reactive other pharmacological agents (e.g., bFGF, transform-
astrocytes, inflamed endothelial cells and encephalito- ing growth factor (TGF)-, erythropoietin). Although,
genic T cells that produced neurogenic and gliogenic some human studies suggest safety and efficacy of this
regulators) and helped to preserve host oligodendro- approach (Ehrenreich et al., 2002), the misadventures
cytes (more so than replacing oligodendrocytes) by of the neurotrophic factor field in the 1980s and 1990s,
exerting an anti-inflammatory effect (Pluchino et al., where the unanticipated pleiotrophic actions of sys-
2003, 2005), thus protecting against chronic neural temically administered growth factors produced unto-
tissue loss. We had previously observed this anti- ward effects, suggests extreme caution before large
inflammatory action of NSC in a cerebral-ischemia scale clinical application.
model (Park et al., 2002a); now it was being reca- With regard to using exogenous NSC for ther-
pitulated in a bona fide inflammatory disease. In the apy against ischemic injury, a number of interesting
EAE model, the NSC appeared to counteract the insights have emerged that draw on the growing field
inflammation by inducing apoptosis of blood-borne of tissue engineering. It was observed that, in con-
CNS-infiltrating encephalitogenic T cells. In addition, ditions where the likelihood of parenchymal loss is
there was a significant reduction in astrogliosis as we greatest, use of a biodegradable synthetic scaffold to
had noted also in traumatic diseases and described support exogenous NSC transiently within the injured
above (Teng et al., 2002; Park et al., 2002a). Taken terrain served to prolong the reciprocal interaction
together, these actions resulted in a marked decrease between the donor and host, fix instructive molecules
in the extent of demyelination and axonal loss, and, emanating from both, abet the inhibition of astroglial
in turn, disease-related disability, both clinically and and inflammatory host reactions, and provide a tem-
neurophysiologically (Pluchino et al., 2003). plate for donor-derived and host-fiber regrowth. Using
hypoxic-ischemic injury as a prototype for insults char-
acterized by extensive tissue loss, Park et al. (2002a)
seeded NSC onto a polymer scaffold that was subse-
16.4.3 Stroke quently implanted into the infarction cavities. Indeed,
not only did this approach serve a dramatic therapeutic
The presumptive goal in using cell-based therapies in function, but it allowed the investigators to document
ischemic (stroke) or hypoxic-ischemic injury would for the first time the multiple reciprocal interactions
be to replace infarcted CNS tissue in an appropriate that spontaneously ensue between NSC and the exten-
organotypic manner. Given the data presented above, sively damaged brain: parenchymal loss was dramati-
however, it seems plausible to expect that cell-based cally reduced, an intricate meshwork of many highly
approaches might also provide trophic and neuro- arborized neurites of both host- and donor-derived
protective support to tissue at risk in the penumbra neurons emerged, and some anatomical connections
surrounding the infarct, inhibit inflammation and scar- appeared to be reconstituted. The NSC-scaffold com-
ring, promote angiogenesis, and help promote the plex altered the trajectory and complexity of host
mobilization, migration, survival, and differentiation cortical neurites. Reciprocally, donor-derived neurons
of endogenous precursor cells (Bramlett and Dietrich, appeared to be capable of directed, target-appropriate
2004; Hass et al., 2005). neurite outgrowth. These biobridges appeared to
266 Y.D. Teng et al.

unveil or augment a constitutive reparative response by rons, we have come to recognize that the original view
facilitating a series of reciprocal interactions between was overly narrow and simplistic. The inherent biology
NSC and host, including promoting neuronal differen- of the NSC the richness and complexity of which we
tiation, enhancing the elaboration of neural processes, are only now beginning to appreciate offers many
fostering the re-formation of cortical tissue, promoting other therapeutic tools, including effecting neuropro-
connectivity and prompting revascularization of new tection and immunoregulation, as well as direct cell to
parenchyma by the host. Inflammation and scarring cell communication (i.e., via gap junctions), induction
were also reduced. of and inhibition of obstacles to inborn regenerative
Another interesting observation is that NSC admin- processes, axonal guidance, promotion of angiogene-
istered intravenously in the systemic circulation may sis, exertion of homeostatic pressure, and presumably
migrate into lesioned brain sites, differentiate into neu- other actions yet to be unveiled. These multifaceted
rons and glia and improve functional deficits in rat actions of NSC make them ideally suited for anchor-
models of focal ischemia or cerebral hemorrhage (Chu ing the multi-modal interventions that we are coming
et al., 2003; Jeong et al., 2003; Kim, 2004). This to recognize will be needed to restore function in most
approach to ischemic injury using an intravascular neurological disorders. Complex diseases appear to
route for the delivery of NSC intracranially extends require complex solutions. NSC, as we have described
the observations first made using animal models of in this review, have interfaced and worked syner-
intracranial brain tumors (Aboody et al., 2000) and the gistically with gene and growth factor therapy, anti-
above-described EAE (Pluchino et al., 2003, 2005). apoptotic and neuroprotective strategies, stimulation
Although results in animal models of stroke of neurogenesis, anti-inflammatory and anti-scarring
seem promising, challenges remain before attempting approaches, material science and tissue engineering.
human therapies. For example, obtaining the requisite We, therefore, propose an updated concept of the
number of the proper cells that circumvent immunore- NSC. The fields conventional view which has touched
jection yet interact effectively with host neurocircuitry principally on the essential multipotency of lineage
and limit their impact and distribution solely to the phenotypes (i.e., the ability of NSC to differentiate
CNS are important considerations. Some answers may into all neural cells), should be broadened to include
be found through a better understanding of the molec- the emerging recognition on the biofunctional multi-
ular events that underlie each of the key responses of potency of the NSC to mediate systemic homeostasis.
the injured adult brain to donor NSC and vice versa. Under this new conceptual context, one may begin to
appreciate and seek the logic and teleology behind
the wide range of molecular tactics the NSC appears
to serve at each developmental stage as it integrates
16.5 Conclusion
into and prepares, modifies, and guides the surround-
ing CNS micro- and macro-environment towards the
Even though studies regarding neurogenesis date from formation and self-maintenance of a physiologically
1913, only recently has the cellular and molecular functioning adult nervous system. We believe that
basis of this inborn plasticity begun to be under- embracing this view of the NSCs multipotency is
stood. The stem cell appears to be the repository of pivotal for correctly, efficiently, and optimally exploit-
much of this plasticity. The ability to identify, select, ing stem cell biology for therapeutic applications
isolate, clone, culture, differentiate, genetically manip- including reconstituting the dysfunctional CNS.
ulate, and transplant NSC has clearly advanced our
understanding of this biology. Indeed, given that the
NSC is the first stem cell isolated from a solid organ,
insights derived from studying these cells have and References
will continue to help advance our understanding of
development and repair of other solid organs. Aboody KS, Brown A, Rainov NG, Bower KA, Liu S, Yang W,
Small JE, Herrlinger U, Ourednik V, Black PM, Breakefield
Although research into the use of NSC, dating back
XO, Snyder EY (2000) Neural stem cells display extensive
to the late 1980s, initially focused solely on their tropism for pathology in adult brain: evidence from intracra-
potential for replacing injured or dysfunctional neu- nial tumors. Proc Natl Acad Sci USA 97:1284612851.
16 Functional Multipotency of Neural Stem Cells 267

Altman J (1962a) Autoradiographic study of degenerative and pocampal neurogenesis of mice. Mol Cell Neurosci 24:
regenerative proliferation of neuroglia cells with tritiated 603613.
thymidine. Exp Neurol 5:302318. Buck SB, Bradford J, Gee KR, Agnew BJ, Clarke ST, Salic A
Altman J (1962b) Are neurons formed in the brains of adult (2008) Detection of S-phase cell cycle progression using 5-
mammals? Science 135:11271128. ethynyl-2-deoxyuridine incorporation with click chemistry,
Altman J (1966) Autoradiographic and histological studies of an alternative to using 5-bromo-2-deoxyuridine antibodies.
postnatal neurogenesis. II. A longitudinal investigation of the Biotechniques 44:927929.
kinetics, migration and transformation of cells incorporating Burns TC, Ortiz-Gonzalez XR, Gutierrez-Perez M, Keene CD,
tritiated thymidine in infant rats, with special reference to Sharda R, Demorest ZL, Jiang Y, Nelson-Holte M, Soriano
postnatal neurogenesis in some brain regions. J Comp Neurol M, Nakagawa Y, Luquin MR, Garcia-Verdugo JM, Prsper
126:431474. F, Low WC, Verfaillie CM (2006) Thymidine analogs are
Altman J (1969) Autoradiographic and histological studies of transferred from pre-labeled donor to host cells in the cen-
postnatal neurogenesis. IV. Cell proliferation and migration tral nervous system after transplantation: a word of caution.
in the anterior forebrain, with special reference to persist- Stem Cells 24:11211127.
ing neurogenesis in the olfactory bulb. J Comp Neurol 137: Cao QL, Howard RM, Dennison JB, Whittemore SR (2002)
433457. Differentiation of engrafted neuronal-restricted precursor
Altman J, Das GD (1965) Autoradiographic and histological evi- cells is inhibited in the traumatically injured spinal cord. Exp
dence of postnatal hippocampal neurogenesis in rats. J Comp Neurol 177:349359.
Neurol 124:319335. Cao Q, Xu XM, Devries WH, Enzmann GU, Ping P, Tsoulfas P,
Anchan RM, Reh TA, Angello J, Balliet A, Walker M (1991) Wood PM, Bunge MB, Whittemore SR (2005) Functional
EGF and TGF-alpha stimulate retinal neuroepithelial cell recovery in traumatic spinal cord injury after transplanta-
proliferation in vitro. Neuron 6:923936. tion of multineurotrophin-expressing glial-restricted precur-
Angevine JB Jr (1965) Time of neuron origin in the hippocampal sor cells. J Neurosci 25:69476957.
region. An autoradiographic study in the mouse. Exp Neurol Carleton A, Petreanu LT, Lansford R, Alvarez-Buylla A, Lledo
Suppl 2:170. PM (2003) Becoming a new neuron in the adult olfactory
Angevine JB Jr, Sidman RL (1961) Autoradiographic study of bulb. Nat Neurosci 6:507518.
cell migration during histogenesis of cerebral cortex in the Carpenter MK, Cui X, Hu ZH, Jackson J, Sherman S, Seiger
mouse. Nature 192:766768. A, Wahlberg LA (1999) In vitro expansion of a multipo-
Baird A (1994) Fibroblast growth factors: activities and signifi- tent population of human neural progenitor cells. Exp Neurol
cance of non-neurotrophin neurotrophic growth factors. Curr 158:265278.
Opin Neurobiol 4:7886. Carter LM, Starkey ML, Akrimi SF, Davies M, McMahon SB,
Bauer S, Patterson PH (2005) The cell cycle-apoptosis connec- Bradbury EJ (2008) The yellow fluorescent protein (YFP-H)
tion revisited in the adult brain. J Cell Biol 71:641650. mouse reveals neuroprotection as a novel mechanism under-
Belluzzi O, Benedusi M, Ackman J, LoTurco JJ (2003) lying chondroitinase ABC-mediated repair after spinal cord
Electrophysiological differentiation of new neurons in the injury. J Neurosci 28:1410714120.
olfactory bulb. J Neurosci 23:1041110418. Chu K, Kim M, Jeong SW, Kim SU, Yoon BW (2003) Human
Bjornson CRR, Rietze RL, Reynolds BA, Magli MC, Vescovi neural stem cells can migrate, differentiate, and integrate
AL (1999) Turning brain into blood: a hematopoietic fate after intravenous transplantation in adult rats with transient
adopted by adult neural stem cells in vivo. Science 283: forebrain ischemia. Neurosci Lett 343:129133.
534537. Curtis MA, Kam M, Nannmark U, Anderson MF, Axell MZ,
Bjugstad KB, Redmond DE Jr, Teng YD, Elsworth JD, Roth RH, Wikkelso C, Holts S, van Roon-Mom WM, Bjrk-Eriksson
Blanchard BC, Snyder EY, Sladek JR Jr (2005) Neural stem T, Nordborg C, Frisn J, Dragunow M, Faull RL, Eriksson
cells implanted into MPTP-treated monkeys increase the size PS (2007) Human neuroblasts migrate to the olfactory bulb
of endogenous tyrosine hydroxylase-positive cells found in via a lateral ventricular extension. Science 315:12431249.
the striatum: a return to control measures. Cell Transplant Danilov AI, Covacu R, Moe MC, Langmoen IA, Johansson CB,
14:183192. Olsson T, Brundin L (2006) Neurogenesis in the adult spinal
Bjugstad KB, Teng YD, Redmond DE Jr, Elsworth JD, Roth cord in an experimental model of multiple sclerosis. Eur J
RH, Cornelius SK, Snyder EY, Sladek JR Jr (2008) Human Neurosci 23:394400.
neural stem cells migrate along the nigrostriatal pathway Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez-
in a primate model of Parkinsons disease. Exp Neurol Buylla A (1999) Subventricular zone astrocytes are neural
211:362369. stem cells in the adult mammalian brain. Cell 97:703716.
Blesch A, Tuszynski MH (2002) Spontaneous and neurotrophin- Ehrenreich H, Hasselblatt M, Dembowski C, Cepek L, Lewczuk
induced axonal plasticity after spinal cord injury. Prog Brain P, Stiefel M, Rustenbeck HH, Breiter N, Jacob S, Knerlich
Res 137:415423. F, Bohn M, Poser W, Rther E, Kochen M, Gefeller O,
Bramlett HM, Dietrich WD (2004) Pathophysiology of cere- Gleiter C, Wessel TC, De Ryck M, Itri L, Prange H,
bral ischemia and brain trauma: similarities and differences. Cerami A, Brines M, Sirn AL (2002) Erythropoietin ther-
J Cereb Blood Flow Metab 24:133150. apy for acute stroke is both safe and beneficial. Mol Med 8:
Brandt MD, Jessberger S, Steiner B, Kronenberg G, Reuter 495505.
K, Bick-Sander A, von der Behrens W, Kempermann Eriksson PS, Perfilieva E, Bjork-Eriksson T, Alborn AM,
G (2003) Transient calretinin expression defines early Nordborg C, Peterson DA, Gage FH (1998) Neurogenesis in
postmitotic step of neuronal differentiation in adult hip- the adult human hippocampus. Nat Med 4:13131317.
268 Y.D. Teng et al.

Flax JD, Aurora S, Yang C, Simonin C, Wills AM, Billinghurst Kempermann G, Kuhn HG, Gage FH (1997) More hippocam-
LL, Jendoubi M, Sidman RL, Wolfe JH, Kim SU, Snyder EY pal neurons in adult mice living in an enriched environment.
(1998) Engraftable human neural stem cells respond to devel- Nature 386:493495.
opmental cues, replace reurons and express foreign genes. Kilpatrick TJ, Richards LJ, Bartlett PF (1995) The regulation of
Nat Biotechnol 16:10331039. neural precursor cells within the mammalian brain. Mol Cell
Gage FH (2000) Mammalian neural stem cells. Science Neurosci 6:215.
287:14331438. Kim SU (2004) Human neural stem cells genetically modified
Gage FH, Ray J, Fisher LJ (1995) Isolation, characterization, for brain repair in neurological disorders. Neuropathology
and use of stem cells from the CNS. Ann Rev Neurosci 18: 24:159171.
159192. Kim SU, Nakagawa E, Nagai A, Snyder EY, Ryu JK, Kim J, Lee
Gensburger C, Labourdette G, Sensenbrenner M (1987) Brain MA, Mook IH, Jin BK (2000) Generation of human neural
basic fibroblast growth factor stimulates the proliferation of stem cell lines and brain transplantation in animal models
rat neuronal precursor cells in vitro. FEBS Lett 217:15. of Parkinson disease and multiple sclerosis. Arch Neurol
Goldman SA, Nottebohm F (1983) Neuronal production, migra- 57:1238.
tion, and differentiation in a vocal control nucleus of the adult Konya D, Liao WL, Choi H, Yu D, Woodard MC, Newton KM,
female canary brain. Proc Natl Acad Sci USA 80:23902394. King AM, Pamir NM, Black PM, Frontera WR, Sabharwal S,
Gratzner HG (1982) Monoclonal antibody to 5-bromo- and Teng YD (2008) Functional recovery in T13-L1 hemisected
5-iododeoxyuridine: a new reagent for detection of DNA rats resulting from peripheral nerve rerouting: role of central
replication. Science 218:474475. neuroplasticity. Regen Med 3:309327.
Graziadei GA, Graziadei PP (1979) Neurogenesis and neu- Kukekov VG, Laywell ED, Suslov O, Davies K, Scheffler B,
ron regeneration in the olfactory system of mammals. II. Thomas LB, OBrien TF, Kusakabe M, Steindler DA (1999)
Degeneration and reconstitution of the olfactory sensory Multipotent stem/progenitor cells with similar properties
neurons after axotomy. J Neurocytol 8:197213. arise from two neurogenic regions of adult human brain. Exp
Gritti A, Parati EA, Cova L, Frolichsthal P, Galli R, Wanke E, Neurol 156:333344.
Faravelli L, Morassutti DJ, Roisen F, Nickel DD, Vescovi AL Lakshmipathy U, Verfaillie C (2005) Stem cell plasticity. Blood
(1996) Multipotential stem cells from the adult mouse brain Rev 19:2938.
proliferate and self-renew in response to basic fibroblast Lewis PF, Emerman M (1994) Passage through mitosis is
growth factor. J Neurosci 16:10911100. required for oncoretroviruses but not for the human immun-
Hall ED, Springer JE (2004) Neuroprotection and acute spinal odeficiency virus. J Virol 68:510516.
cord injury: a reappraisal. NeuroRx 1:80100. Li J, Imitola J, Snyder EY, Sidman RL (2006b) Neural stem
Hass S, Weidner N, Winkler J (2005) Adult stem cell therapy in cells rescue nervous Purkinje neurons by restoring molecular
stroke. Curr Opin Neurol 18:5964. homeostasis of tissue plasminogen activator and downstream
Herrup K, Neve R, Ackerman SL, Copani A (2004) Divide targets. J Neurosci 26:78397848.
and die: cell cycle events as triggers of nerve cell death. J Li J, Ma Y, Teng YD, Zheng K, Vartanian TK, Snyder EY,
Neurosci 24:92329239. Sidman RL (2006a) Purkinje neuron degeneration in nervous
Howard A, Pelc SR (1953) Synthesis of desoxyribonucleic acid (nr) mutant mice is mediated by a metabolic pathway involv-
in normal and irradiated cells and its relation to chromosome ing excess tissue plasminogen activator. Proc Natl Acad Sci
breakage. Heredity 6:261273. USA 103:78477852.
Hughes WL, Bond VP, Brecher G, Cronkite ER, Painter RB, Liu S, Qu Y, Stewart TJ, Howard MJ, Chakrabortty S, Holekamp
Quastler H, Sherman FG (1958) Cellular proliferation in TF, McDonald JW (2000) Embryonic stem cells differen-
the mouse as revealed by autoradiography with tritiated tiate into oligodendrocytes and myelinate in culture and
thymidine. Proc Natl Acad Sci USA 44:476483. after spinal cord transplantation. Proc Natl Acad Sci USA
Jderstad J, Jderstad LM, Li J, Chintawar S, Salto C, Pandolfo 97:61266131.
M, Ourednik V, Teng YD, Sidman RL, Arenas E, Snyder Llado J, Haenggeli C, Maragakis NJ, Snyder EY, Rothstein JD
EY, Herlenius E (2010) Communication via gap junctions (2004) Neural stem cells protect against glutamate-induced
underlies early functional & beneficial interactions between excitotoxicity and promote survival of injured motor neurons
grafted neural stem cells & the host. Proc Natl Acad Sci USA through secretion of neurotrophic factors. Mol Cell Neurosci
107 (in press). 27:322331.
Jeong SW, Chu K, Jung KH, Kim SU, Kim M, Roh JK (2003) Lu P, Jones LL, Snyder EY, Tuszynski MH (2003) Neural stem
Human neural stem cell transplantation promotes functional cells constitutively secrete neurotrohic factors and promote
recovery in rats with experimental intracerebral hemorrhage. extensive host axonal growth after spinal cord injury. Exp
Stroke 34:22582263. Neurol 181:115129.
Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U, Martinez-Serrano A, Rubio FJ, Navarro B, Bueno C, Villa
Frisen J (1999) Identification of a neural stem cell in the adult A (2001) Human neural stem and progenitor cells: in
mammalian central nervous system. Cell 96:2534. vitro and in vivo properties, and potential gene therapy
Kaplan MS, Bell DH (1983) Neuronal proliferation in the 9- and cell replacement in the CNS. Curr Gene Ther 1:
month-old rodent-radioautographic study of granule cells in 279299.
the hippocampus. Exp Brain Res 52:15. McDonald JW, Liu XZ, Qu Y, Liu S, Mickey SK, Turetsky D,
Kempermann G, Jessberger S, Steiner B, Kronenberg G (2004) Gottlieb DI, Choi DW (1999) Transplanted embryonic stem
Milestones of neuronal development in the adult hippocam- cells survive, differentiate and promote recovery in injured
pus. Trends Neurosci 27:447452. rat spinal cord. Nat Med 5:1410.
16 Functional Multipotency of Neural Stem Cells 269

Meng F, Zolova O, Kokorina NA, Dobretsova A, Wight PA Pluchino S, Zanotti L, Rossi B, Brambilla E, Ottoboni L,
(2005) Characterization of an intronic enhancer that reg- Salani G, Martinello M, Cattalini A, Bergami A, Furlan
ulates myelin proteolipid protein (Plp) gene expression in R, Comi G, Constantin G, Martino G (2005) Neurosphere-
oligodendrocytes. J Neurosci Res 82:346356. derived multipotent precursors promote neuroprotection by
Miale IL, Sidman RL (1961) An autoradiographic analy- an immunomodulatory mechanism. Nature 436: 266271.
sis of histogenesis in the mouse cerebellum. Exp Neurol Price J, Turner D, Cepko C (1987) Lineage analysis in the ver-
4:277296. tebrate nervous system by retrovirus-mediated gene transfer.
Miller RH (2002) Regulation of oligodendrocyte development in Proc Natl Acad Sci USA 84:156160.
the vertebrate CNS. Progress Neurobiol 67:451467. Ravin R, Hoeppner DJ, Munno DM, Carmel L, Sullivan J,
Miller RH (2005) Dorsally derived oligodendrocytes come of Levitt DL, Miller JL, Athaide C, Panchision DM, McKay
age. Neuron 45:13. RD (2008) Potency and fate specification in CNS stem cell
Miller MW, Nowakowski RS (1988) Use of bromodeoxyuridine- populations in vitro. Cell Stem Cell 3:670680.
immunohistochemistry to examine the proliferation, migra- Redmond DE Jr, Bjugstad KB, Teng YD, Ourednik V, Ourednik
tion and time of origin of cells in the central nervous system. J, Wakeman DR, Parsons XH, Gonzalez R, Blanchard BC,
Brain Res 457:4452. Kim SU, Gu Z, Lipton SA, Markakis EA, Roth RH, Elsworth
Ming GL, Song H (2005) Adult neurogenesis in the mammalian JD, Sladek JR Jr, Sidman RL, Snyder EY (2007) Behavioral
central nervous system. Annu Rev Neurosci 28:223250. improvement in a primate Parkinsons model is associated
Mitsui T, Shumsky JS, Lepore AC, Murray M, Fischer I (2005) with multiple homeostatic effects of human neural stem cells.
Transplantation of neuronal and glial restricted precursors Proc Natl Acad Sci USA 104:1217512180.
into contused spinal cord improves bladder and motor Reier PJ (2004) Cellular transplantation strategies for spinal cord
functions, decreases thermal hypersensitivity, and modifies injury and translational neurobiology. NeuroRx 1:424451.
intraspinal circuitry. J Neurosci 25:96249636. Renfranz PJ, Cunningham MG, McKay RD (1991) Region-
Muotri AR, Gage FH (2006) Generation of neuronal variability specific differentiation of the hippocampal stem cell line
and complexity. Nature 441:10871093. HiB5 upon implantation into the developing mammalian
Nottebohm F (2004) The road we travelled: discovery, choreog- brain. Cell 66:713729.
raphy, and significance of brain replaceable neurons. Ann N Reynolds BA, Tetzlaff W, Weiss S (1992) A multipotent EGF-
Y Acad Sci 1016:628658. responsive striatal embryonic progenitor cell produces neu-
Ogle BM, Cascalho M, Platt JL (2005) Biological implications rons and astrocytes. J Neurosci 12:45654574.
of cell fusion. Nat Rev Mol Cell Biol 6:567575. Reynolds BA, Weiss S (1992) Generation of neurons and astro-
Ortiz-Gonzalez XR, Keene CD, Verfaillie CM, Low WC (2004) cytes from isolated cells of the adult mammalian central
Neural induction of adult bone marrow and umbilical cord nervous system. Science 255:17071710.
stem cells. Curr Neurovasc Res 3:207213. Richards LJ, Kilpatrick TJ, Bartlett PF (1992) De novo genera-
Ourednik J, Ourednik V, Lynch WP, Schachner M, Snyder tion of neuronal cells from the adult mouse brain. Proc Natl
EY (2002) Neural stem cells display an inherent mecha- Acad Sci USA 89:85918595.
nism for rescuing dysfunctional neurons. Nat Biotechnol Rosario CM, Yandava BD, Kosaras B, Zurakowski D, Sidman
20:11031110. RL, Snyder EY (1997) Differentiation of engrafted mul-
Park KI, Liu S, Flax JD, Nissim S, Stieg PE, Snyder EY (1999) tipotent neural progenitors towards replacement of miss-
Transplantation of neural progenitor and stem cells: devel- ing granule neurons in meander tail cerebellum may help
opmental insights may suggest new therapies for spinal cord determine the locus of mutant gene action. Development
and other CNS dysfunction. J Neurotrauma 16: 675687. 124:42134224.
Park KI, Ourednik J, Ourednik V, Taylor RM, Aboody KA, Roy NS, Nakano T, Keyoung HM, Windrem M, Rashbaum
Auguste KI, Lachyankar M, Redmond DE, Snyder EY WK, Alonso ML, Kang J, Peng W, Carpenter MK, Lin J,
(2002b) Global gene and cell replacement strategies via stem Nedergaard M, Goldman SA (2004) Telomerase immortal-
cells. Gene Ther 9:613624. ization of neuronally restricted progenitor cells derived from
Park KI, Teng YD, Snyder EY (2002a) The injured brain the human fetal spinal cord. Nat Biotechnol 22: 297305.
interacts reciprocally with neural stem cells supported by Ryder EF, Snyder EY, Cepko CL (1990) Establishment and char-
scaffolds to reconstitute lost tissue. Nat Biotechnol 20: acterization of multipotent neural cell lines using retrovirus
11111117. vector-mediated oncogene transfer. J Neurobiol 21: 356375.
Parker MA, Anderson JK, Corliss DA, Abraria VE, Sidman Salic A, Mitchison TJ (2008) A chemical method for fast and
RL, Park KI, Teng YD, Cotanche DA, Snyder EY (2005) sensitive detection of DNA synthesis in vivo. Proc Natl Acad
Expression profile of an operationally-defined neural stem Sci USA 105:24152420.
cell clone. Exp Neurol 194:320332. Sanes JR, Rubenstein JL, Nicolas JF (1986) Use of a recom-
Petersen BE, Bowen WC, Patrene KD, Mars WM, Sullivan AK, binant retrovirus to study postimplantation cell lineage in
Murase N, Boggs SS, Greenberger JS, Goff JP (1999) Bone mouse embryos. EMBO J 5:31333142.
marrow as a potential source of hepatic oval cells. Science Schaffer DV, Gage FH (2004) Neurogenesis and neuroadapta-
284:11681170. tion. Neuromolecular Med 5:19.
Pluchino S, Quattrini A, Brambilla E, Gritti A, Salani G, Dina G, Schwab ME (2004) Nogo and axon regeneration. Curr Opin
Galli R, Del Carro U, Amadio S, Bergami A, Furlan R, Comi Neurobiol 14:118124.
G, Vescovi AL, Martino G (2003) Injection of adult neu- Shihabuddin LS, Horner PJ, Ray J, Gage FH (2000) Adult spinal
rospheres induces recovery in a chronic model of multiple cord stem cells generate neurons after transplantation in the
sclerosis. Nature 422:688694. adult dentate gyrus. J Neurosci 20:87278735.
270 Y.D. Teng et al.

Shihabuddin LS, Numan S, Huff MR, Dodge JC, Clarke J, after spinal cord injury. Proc Natl Acad Sci USA 101:
Macauley SL, Yang W, Taksir TV, Parsons G, Passini MA, 30713076.
Gage FH, Stewart GR (2004) Intracerebral transplantation of Teng YD, Lavik EB, Qu X, Park KI, Ourednik J, Zurakowski D,
adult mouse neural progenitor cells into the Niemann-Pick- Langer R, Snyder EY (2002) Functional recovery following
A mouse leads to a marked decrease in lysosomal storage traumatic spinal cord injury mediated by a unique polymer
pathology. J Neurosci 24:1064210651. scaffold seeded with neural stem cells. Proc Natl Acad Sci
Sidman RL (1961) Histogenesis of mouse retina studied with USA 99:30243029.
thymidine-H3 . In: The Structure of the Eye (Smelser GK, Teng YD, Liao W-L, Choi C, Konya D, Sabharwal D,
ed.), pp. 487506. Academic Press, New York. Langer L, Sidman RL, Snyder EY, Frontera WR (2006)
Sidman RL (1970) Autoradiographic methods and principles Physical activity-mediated functional recovery after spinal
for study of the nervous system with thymidine-H3 . In: cord injury: potential roles of neural stem cells. Reg Med
Contemporary Research Methods in Neuroanatomy (Nauta 1:763776.
WJ, Ebbesson SOE, eds.), pp. 252274. Springer-Verlag, Teng YD, Yu D, Song G, Poon C-S (2006) NSC-mediated
New York. multimechanistic repair of respiratory function following
Sidman RL, Li J, Stewart GR, Clarke J, Yang W, Snyder EY, spinal cord injury: neurotrophic factors and gap junction.
Shihabuddin LS (2007) Injection of mouse and human neural Symposium on hot topics in stem cell research. The 36th
stem cells into neonatal Niemann-Pick A model mice. Brain annual meeting of the Society for Neuroscience.
Res 1140:195204. Timsit SG, Martinez S, Alliquant B, Peyron F, Puelles L, Zalc
Sidman RL, Miale IL, Feder N (1959) Cell proliferation in B (1995) Oligodendrocytes originate in a restricted zone of
the primitive ependymal zone; an autoradiographic study the embryonic ventral neural tube defined by DM-20 mRNA
of histogenesis in the nervous system. Exp Neurol 1: expression. J Neurosci 15:10121024.
322333. van Praag H, Schinder AF, Christie BR, Toni N, Palmer TD,
Silver J, Miller JH (2004) Regeneration beyond the glial scar. Gage FH (2002) Functional neurogenesis in the adult hip-
Nat Rev Neurosci 5:146156. pocampus. Nature 415:10301034.
Simpson DL, Morrison R, de Vellis J, Herschman HR (1982) Vescovi AL, Parati EA, Gritti A, Poulin P, Ferrario M, Wanke
Epidermal growth factor binding and mitogenic activity on E, Frlichsthal-Schoeller P, Cova L, Arcellana-Panlilio M,
purified populations of cells from the central nervous system. Colombo A, Galli R (1999) Isolation and cloning of mul-
J Neurosci Res 8:453462. tipotential stem cells from the embryonic human CNS and
Smart I (1961) The subependymal layer of the mouse brain establishment of transplantable human neural stem cell lines
and its cell production as shown by autography after [H3]- by epigenetic stimulation. Exp Neurol 156:7183.
thymidine injection. J Comp Neurol 116:325327. Vescovi AL, Reynolds BA, Fraser DD, Weiss S (1993) bFGF
Smith AG, Heath JK, Donaldson DD, Wong GG, Moreau J, Stahl regulates the proliferative fate of unipotent (neuronal) and
M, Rogers D (1988) Inhibition of pluripotential embryonic bipotent (neuronal/astroglial) EGF-generated CNS progeni-
stem cell differentiation by purified polypeptides. Nature tor cells. Neuron 11:951966.
336:688690. Vescovi AL, Snyder EY (1999) Establishment and properties of
Snyder EY, Deitcher DL, Walsh C, Arnold-Aldea S, Hartweig neural stem cell clones: plasticity in vitro and in vivo. Brain
EA, Cepko CL (1992) Multipotent neural cell lines can Pathol 9:569598.
engraft and participate in development of mouse cerebellum. Wernig M, Zhao JP, Pruszak J, Hedlund E, Fu D, Soldner
Cell 68:3351. F, Broccoli V, Constantine-Paton M, Isacson O, Jaenisch
Snyder EY, Yoon CH, Flax JD, Macklis JD (1997) Multipotent R (2008) Neurons derived from reprogrammed fibroblasts
neural progenitors can differentiate toward replacement functionally integrate into the fetal brain and improve symp-
of neurons undergoing targeted apoptotic degeneration in toms of rats with Parkinsons disease. Proc Natl Acad Sci
adult mouse neocortex. Proc Natl Acad Sci USA 94: USA 105:58565861.
1166311668. Williams BP, Read J, Price J (1991) The generation of neurons
Stanfield BB, Trice JE (1988) Evidence that granule cells gen- and oligodendrocytes from a common precursor cell. Neuron
erated in the dentate gyrus of adult rats extend axonal 7:685693.
projections. Exp Brain Res 72:399406. Yan J, Welsh AM, Bora SH, Snyder EY, Koliatsos VE (2004)
Takahashi K, Yamanaka S (2006) Induction of pluripotent stem Differentiation and tropic/trophic effects of exogenous neural
cells from mouse embryonic and adult fibroblast cultures by precursors in the adult spinal cord. J Comp Neurol 480:101
defined factors. Cell 126:663676. 114.
Tator CH, Fehlings MG (1991) Review of the secondary injury Yandava BD, Billinghurst L, Snyder EY (1999) Global cell
theory of acute spinal cord trauma with emphasis on vascular replacement is feasible via neural stem cell transplantation:
mechanisms. J Neurosurg 75:1526. evidence from the dysmyelinated shiverer mouse brain. Proc
Temple S (1989) Division and differentiation of isolated CNS Natl Acad Sci USA 96:70297034.
blast cells in microculture. Nature 340:471473. Yu W-P, Collarini EJ, Pringle NP, Richardson WD (1994)
Temple S, Qian X (1995) bFGF, neurotrophins, and the control Embryonic expression of myelin genes: evidence for a focal
of cortical neurogenesis. Neuron 15:249252. source of oligodendrocyte precursors in the ventricular zone
Teng YD, Choi H, Onario RC, Zhu S, Desilets FC, Lan of the neural tube. Neuron 23:13531362.
S, Woodard EJ, Snyder EY, Eichler ME, Friedlander RM Zhao C, Deng W, Gage FH (2008) Mechanisms and
(2004) Minocycline inhibits contusion-triggered mitochon- functional implications of adult neurogenesis. Cell 132:
drial cytochrome c release and mitigates functional deficits 645660.
Chapter 17

Dual Roles of Mesenchymal Stem Cells in Spinal Cord Injury:


Cell Replacement Therapy and as a Model System
to Understand Axonal Repair

Cecile King, Shyam Patel, Treena Livingston Arinzeh, and Pranela Rameshwar

Contents expected at sites of spinal cord injury (SCI), simple


implantation of MSC and/or their differentiated cells
17.1 Mesenchymal Stem Cells (MSC) . . . . . . . 272 might be premature unless the basic science precedes
17.2 Biology of Spinal Cord Injury . . . . . . . . 273 translational science. This is particularly true since
17.3 Current Interventions for Spinal Cord Injury 273 an individuals immune response might be unique.
17.4 Cytokines and Soluble Factors . . . . . . . 274
Similar argument could be made for embryonic stem
17.4.1 Tumor Necrosis Factor Alpha (TNF-) . 274
17.4.2 Leukemia Inhibitory Factor (LIF) . . . 275 cells (ESC). The unstable behavior of ESC to form
17.4.3 Interlekin-6 (IL-6) . . . . . . . . . . 275 tumors, as well as rapid generation of mixed cell types
17.4.4 Interleukin-1 (IL-1 ) . . . . . . . . 275 makes MSC desirable stem cells for translational
17.4.5 Transforming Growth Factor 1 (TGF-1) 275
science. This review discusses varied interdisciplinary
17.5 Prospects for Axonal Regeneration in the CNS 275
17.6 Stem Cell Therapy for Spinal Cord Injury . . 276 approaches by which MSC can be applied to SCI
17.7 Transdifferentiation of Mesenchymal Stem Cells repair, including bioengineering approach. The mech-
to Neurons . . . . . . . . . . . . . . . . . 277 anism by which microenvironmental factors such as
17.8 Other Neurodegenerative Disorders . . . . . 277
inflammatory mediators can affect stem cell therapies
17.9 Limitations to Stem Cell Therapeutics . . . . 278
17.10 An Interdisciplinary Approach . . . . . . . 279 is also discussed. In addition to the potential of direct
17.11 Experimental Models for SCI . . . . . . . . 280 application of stem cells or their differentiated cells,
17.12 On the Frontier of Stem Cell Therapy for MSC can be used as models to understand axonal
Neural Dysfunction . . . . . . . . . . . . . 281
repair, which could lead to the development of new
References . . . . . . . . . . . . . . . . . . . . 281
drugs for SCI. The model could test novel factors
to repair neurons. Any identified factor could be
delivered directly or by gene therapy. The co-culture
Abstract Mesenchymal stem cells (MSC) are
method is significant to long-term investigation for
pluripotent cells that differentiate into cells of meso-
rapid screening of compounds, with simultaneous
dermal origin and transdifferentiate into ectodermal
understanding of repair mechanisms of axotomized
and endodermal cell types. MSC can transdifferentiate
neurons. The model could be translated in parallel
with high efficiency to functional neurons, microglia
with other stem cell therapies for SCI repair. We also
and oligodendrocytes. MSC and neurons can respond
discuss the potential of stem cell therapies for other
to environmental cues such as cytokines, which
neurodegenerative diseases.
also affect the development to neural cells. Since
cytokines and other inflammatory mediators are also
Keywords Cytokines Mesenchymal stem cells
Neurons Spinal cord injury Tissue repair

Abbreviations
P. Rameshwar ()
UMDNJ-New Jersey Medical School, Department of Medicine,
Newark, NJ, USA BDNF brain-derived neurotrophic factor
e-mail: rameshwa@umdnj.edu ESC embryonic stem cells

H. Ulrich (ed.), Perspectives of Stem Cells, 271


DOI 10.1007/978-90-481-3375-8_17, Springer Science+Business Media B.V. 2010
272 C. King et al.

HPC hematopoietic progenitor cell amounts of bone marrow aspirates. More specifi-
IFN- interferon- cally, the total numbers cells that can be attained
IL-6 interlekin-6 and expanded from one donor are immense. Another
IL-1 interleukin-1 attractive property of MSC is the versatility in their
LIF leukemia inhibitory factor immune properties. MSC are both immune suppressive
MEM microelectricomechanical as well as immune-enhancing (Castillo et al., 2007;
systems Chan et al., 2006; Potian et al., 2003; Stagg et al.,
MSC mesenchymal stem cells 2006; Ong et al., 2006). This factor is important as the
NEM nanoelectromechanical microenvironment of sites of tissue injury could affect
systems the immune response of MSC through the complex
NGF nerve growth factor
array of factors present. Thus, implantation of MSC,
NSC neural stem cell(s)
either as undifferentiated or differentiated cells might
NT-3 neurotrophin-3
be premature unless basic science research precedes
PNS peripheral nervous system
the translational research. In this chapter, the potential
PEG poly-ethylene glycol
PGA polyglycolic acid use of MSC in spinal cord injury (SCI) cases will be
PLA polylactic acid discussed.
SCI spinal cord injury In general MSC represent a promising source for
STAT3 signal transducer and activator of tran- various CNS impairments. However, the potential use
scription of MSC for SCI is especially exciting due to the mostly
TGF-1 transforming growth factor 1 irreversible and severe effects of such injuries (Hardy
TNF- tumor necrosis factor- et al., 2008). Previous studies have shown, MSC medi-
ated functional recovery in animal models of SCI
however the specific mechanism of improvement is
not understood (Hardy et al., 2008). Most scientists
believe that release of various cytokines and soluble
17.1 Mesenchymal Stem Cells (MSC) factors from MSC catalyze and support the repair of
the areas of injury (Hardy et al., 2008). Another less
MSC are pluripotent cells that can differentiate into supported hypothesis implies the transdifferentiation
various cell types including adipocytes, chondrocytes of MSC into neurons in addition to cell fusion events
and stroma (Fig. 17.1) (Castillo et al., 2007; Bianco may be the cause of the functional recovery (Hardy
et al., 2001). MSC also demonstrate the ability to trans- et al., 2008).
differentiate into cells of endodermal and ectodermal The microenvironment of any damaged tissue gen-
lineages such as hepatocytes and neurons (Ong et al., erates a complex array of factors. Thus, implantation of
2006; Greco and Rameshwar, 2007). MSC are found MSC, either as undifferentiated or differentiated cells
in adult and fetal tissues. In adults, the bone marrow could begin a crosstalk with endogenous cells. In this
is the major location of MSC (Lee et al., 2004) how- chapter, various representative factors and their effects
ever, MSC are also found in umbilical cord blood, on endogenous cells will be discussed. A proposed
although at lower frequencies (Bieback et al., 2004). interdisciplinary approach will bring an understand-
Morphologically, MSC are symmetrical cells display- ing on the interactions between tissue microenviron-
ing a fibroblastoid appearance (Blanc and Pittenger, ment and MSC. Studies using such an approach will
2005). Phenotypically, MSC express CD44, CD29, lead to safe and effective delivery of MSC to SCI
CD105, CD73, and CD166 and lack markers that are patients and will determine whether MSC should be
consistent with hematopoietic cells, in particular CD45 transplanted as undifferentiated or transdifferentiated
(Pittenger et al., 1999; Martinez et al., 2007). MSC also (partly vs. fully), and allow for personalized treat-
express neural-associated markers, such as neural gan- ment by incorporating mathematics and engineering.
glioside, GD2 (Deng et al., 2006; Padovan et al., 2003; Future delivery of MSC or other non-embryonic stem
Caplan, 1994). cell will also require the aid of biomaterials that
The advantage in pursuing research on MSC is could assists in dictating the behavior of stem cells
due to their ease in expansion from relatively small in vivo.
17 Dual Roles of MSC in Spinal Cord Injury 273

Fig. 17.1 Human


mesenchymal stem cells
(hMSC) are pluripotent and
can differentiate into cells of
mesodermal origin and
transdifferentiate into cells
of ectodermal and
enodermal lineages. MSC
show high efficiency in the
generation of functional
neurons, microglia and
oligodendrocytes. These
characteristics make MSC a
promising therapy for spinal
cord injury and other central
nervous system disorders

17.2 Biology of Spinal Cord Injury formation, glutamate release and accumulation, free
radical formation, necrosis and apoptosis of cells
and inflammation (Kwon et al., 2004; Sekhon and
Spinal Cord Injury (SCI) is characterized by com- Fehlings, 2001). The prevention or treatment of sec-
pression or tension applied to the spinal cord which ondary complications is essential if irreversible paral-
results in damage to the nerves, vessels or spinal fluid ysis is to be avoided.
(Fig. 17.2) (Divani et al., 2007). The applied force
causes a loss of function or mobility whether it is tran-
sient or permanent. Minor spinal cord injuries result in
compression of the spinal cord and give rise to mostly 17.3 Current Interventions for Spinal
temporary neurological deficits (Kwon et al., 2004). In Cord Injury
contrast, severe trauma directed to the spinal cord has
a resultant damage or axotomy of neurons and often, For the past three decades, physicians have employed
if not always leads to permanent paralysis (Divani the use of corticosteroids, specifically intravenous
et al., 2007). Cases of severe SCI injury are character- methylprednisolone, to reduce the inflammation asso-
ized by two stages a primary stage and a secondary ciated with SCIs (Ducker and Hamit, 1969; Young,
stage (Divani et al., 2007). The primary phase of 2000; Ducker and Zeidman, 1994). The justification
injury is denoted by the initial infarct, which cause a is that the anti-inflammatory properties of such agents
series of events resulting ultimately in the initiation of would lead not only to the decreased inflammation and
the secondary phase (Kwon et al., 2004; Sekhon and edema, but also to a reduction in neurotoxic agents,
Fehlings, 2001). Secondary stage injuries include: vas- modulation of immune cells, enhanced vascular per-
cular irregularities, abnormal ion fluctuations, edema fusion and inhibition of calcium influx (Divani et al.,
274 C. King et al.

Fig. 17.2 Spinal cord


injury occurs when
significant amounts of force
are applied to the spinal
cord. This force can lead to
two initially indistinguishable
outcomes: axotomy or
compression of neurons or
spinal fluid. Severe SCI injury
is characterized by two
phases the primary injury
phase and the secondary
injury phase

2007; Chi et al., 2008). In addition to corticosteroid 17.4.1 Tumor Necrosis Factor Alpha
administration, decompressive surgery may be per- (TNF-)
formed to reduce edema and ultimately decrease the
risk of further compression of the spinal cord. Other
than these two interventional procedures, physicians TNF- is an inflammatory cytokine has been reported
may perform anatomic realignments to reduce dislo- to play a dual role in SCI. It is produced by various
cation, suggest bed rest to decrease opportunities for endogenous cells in the site of injury including neu-
further damage and ultimately suggest physical ther- rons, glial and vascular endothelial cells (Chi et al.,
apy to assist patients with coping with the disability. In 2008). During the secondary injury phase of SCI TNF-
general, none of these practices restore full mechanis- acts by directing apoptosis events (Chi et al., 2008).
tic functionalities to SCI patients but only act to reduce More specifically, studies utilizing neutralizing anti-
the advancement of secondary injuries. bodies to TNF- caused significant reduction in apop-
totic cells (Yune et al., 2003). Another role of TNF- in
secondary stage SCI is the induction of the expression
of endothelial leukocyte adhesion molecules. These
17.4 Cytokines and Soluble Factors molecules release inflammatory mediators such as
reactive oxygen species (ROS) and neutrophil elas-
Cytokines, soluble factors and other inflammatory tase which activate neutrophils and ultimately lead to
mediators are expected at sites of spinal cord injury. increased edema (Carlos and Harlan, 1994; Hernandez
A representative list of some of these factors with et al., 1987). A recent article detailed the dual role of
corresponding descriptions is presented in the follow- TNF- in SCI injury. These researchers characterize
ing section. the cytokine as having an initial deleterious role in the
17 Dual Roles of MSC in Spinal Cord Injury 275

acute phase and a beneficial role in the chronic phase. the intracellular signaling domain and therefore cannot
Early inhibition of this cytokine decreases apoptotic initiate intracellular signaling. The IL-1 cytokines are
events while later secretion of the cytokine is linked induced by other inflammatory mediators and by tis-
to neuroprotective effects (Chi et al., 2008). sue insults such as infectious agents, hypoxia, and at
sites of tissue injuries (Allan et al., 2005). The effects
of IL-1 could be direct or indirectly mediated by other
17.4.2 Leukemia Inhibitory Factor (LIF) cytokines (Moore, 2002; Laver and Moore, 1989).
Neurons can be affected directly by IL-1 through the
LIF is a cytokine originally discovered to play a role type I receptor (Allan et al., 2005). In addition, IL-1
in the inhibition of myeloid leukemic cells (Gearing could cause indirect effects on neurons by inducing
et al., 1987; Pan et al., 2006). Knockout mice stud- other cytokines in the surrounding tissues, which in
ies show that LIF is imperative to the development turn can bind to neurons (Dinarello, 2005; Laver and
and survival of motor neurons (Li et al., 1995; Pan Moore, 1989; Dinarello, 1996; Bagby, 1989).
et al., 2006). More recently it has been proven by var-
ious studies that this cytokine plays an important role
in SCI. Studies have also shown that after one day of
SCI, both transcription of LIF and number of binding 17.4.5 Transforming Growth Factor 1
sites for exogenous LIF are increased (Kurek et al., (TGF-1)
1998a; Pan et al., 2006). During SCI, LIF is secreted
by endogenous cells, specifically fibroblasts, leading TGF-1 belongs to a superfamily of proteins including
to a marked increase in axonal sprouting and release of the activins, inhibins and bone morphogenic proteins
neurotrophin-3 (Blesch et al., 1999; Pan et al., 2006). (Roberts and Sporn, 1993). TGF- receptors are ubiq-
Also, oliodendrocyte survival is enhanced by exoge- uitously expressed on healthy and malignant cells
nous LIF and has been linked to aiding in functional (Massague et al., 1992; Massague and Weis-Garcia,
recovery (Butzkueven et al., 2002; Kurek et al., 1998b; 1996). TGF-1 interacts with two receptor subtypes:
Pan et al., 2006). I and II (Shi and Massague, 2003). Type I signals
through intracellular pathways involving four mem-
bers of the Smad transcription factors (Massague and
17.4.3 Interlekin-6 (IL-6) Wotton, 2000; Sato et al., 2003; Mehra and Wrana,
2002). The only reported function for type II recep-
IL-6 belongs to the family of interleukin cytokines and tor is their ability to activate the type I TGF- receptor
it has been implicated in spontaneous recovery after (Wrana, 2000). The family of TGF- proteins has been
SCI (Kang and Kang, 2008; Zai et al., 2005). After linked to developmental processes such as embryoge-
spinal cord injury IL-6 levels rapidly increase (Kang nesis and development of neural cells (Roberts and
and Kang, 2008; Hayashi et al., 2000) however the spe- Sporn, 1993; Golestaneh and Mishra, 2005). TGF-
cific function of IL-6 is not fully understood. Studies 1 functions as immune modulators, inhibitors of cell
have shown that the cytokine is involved both in neu- proliferation, differentiation and apoptosis (Sokol and
ronal cell death and differentiation (Bonni et al., 1997; Schiemann, 2004; Downing, 2004). TGF-1 has been
Gadient and Otten, 1997) and also in induction of the determined to exhibit both tumor suppressor and onco-
functional recovery of neurons (Yamauchi et al., 2006). genic properties and can therefore inhibit cell prolif-
eration and promotes malignancy (Kim and Letterio,
2003).
17.4.4 Interleukin-1 (IL-1 )
17.5 Prospects for Axonal Regeneration
IL-1 belongs to the family of IL-1 cytokines (Allan
et al., 2005). The IL-1 family of cytokines is central
in the CNS
to inflammation and host defense. IL-1 and IL-1
appear to exhibit similar effects. Each binds to the The process of axotomy has been shown to alter
type I IL-1 receptor. The type II receptor subtype lacks gene expression in neurons (Tonge et al., 2008).
276 C. King et al.

Specifically, the regeneration-associated genes play 2008). The current upsurge of interest in stem cell
a pivotal role in axonal regeneration (Tonge et al., applications in spinal cord injury has been attributed to
2008). The process is speculated to involve reverse the high likelihood of permanent loss of neurological
axonal transport of particular signaling molecules, function upon trauma (Barnab-Heider and Frisn,
such as signal transducer and activator of tran- 2008). In this section, we explore the notion of the
scription (STAT3) (Tonge et al., 2008). Prior in clinical use of various undifferentiated cell types
vitro conditioning of axotomized nerves with neu- with self-renewal properties in scenarios of CNS
rotrophins causes enhanced axonal growth (Tonge dysfunction.
et al., 2008). Such neurotrophins include brain-derived As of date, approximately 2.5 million people expe-
neurotrophic factor, which increases axonal length by rience disability due to spinal cord injury (SCI) (Lo
over 90%. In addition, environmental factors serve to et al., 2008). The phenomenon of SCI has been
guide axons upon regeneration. For example, the Slit suggested to have two stages: physical trauma and
family functions in repulsing axonal growth (Fujiwara intrinsic cellular destruction. During physical trauma
et al., 2008) while HIP/PAP1 function in the formation stage, various gross insults to the spinal cord such
and regeneration of motor neurons (Tebar et al., 2008). as penetration or contusion can result in loss of
Animal models have also assisted in understanding normal tract function. During the cellular destruction
axonal regeneration. The role of the immune response stage, events including ischemia, glutamate-induced
in CNS regeneration has been recently characterized in excitotoxicity, and apoptosis lead to further damage
a medicinal leech model. The production of antibacte- of the nervous system (Lo et al., 2008). Trauma in
rial peptides by neurons demonstrates that a controlled non-nervous tissue does not pose as great a threat
microbial response facilitates regeneration of nervous as damage to nervous tissue because many tissues
system function. can regenerate themselves and allow for survival.
Axotomy has been shown to serve as one compo- The limited capacity of adult neurons to regenerate,
nent, along with motor neuron dysfunction, to induce however, makes the prospect of mesenchymal stem
neural precursor cells (Azari et al., 2008). Neural pre- cell therapy promising for treatment of patients with
cursor cells are positive for nestin, an intermediate injury. Multiple reasons have been proposed for the
filament protein expressed in stem cells of neurolog- failure of ectodermal tissue to successfully repair
ical origin. The process of axotomy in the presence itself. Firstly, the innate capacity for neurons to
of motor neuron disease mobilizes neural precursors grow is less than other cells of the body (Planchamp
to migrate to the spinal cord (Azari et al., 2008). et al., 2008). Secondly, the microenvironment in
Thus, injury to CNS structures can induce a response which neurons reside may be non-permissive for
from cells with stem-like characteristics, suggesting a development.
valuable role for stem cell therapy in the treatment Neural stem cells (NSC) have been investigated to
of axonal injury and nerve severance (Azari et al., determine their regeneration abilities in spinal cord
2008). transaction. Retinoic acid, the same factor used in
the transdifferentiation of MSC into neurons, can
be used to differentiate NSC into neurons. NSC
and neurotrophin-3-expressing Schwann cells can be
17.6 Stem Cell Therapy for Spinal grafted into a site of spinal cord transaction, result-
ing in improvement of neural function and myeli-
Cord Injury
nation (Zhang et al., 2007). Fetal NSC have been
proposed to repair motor neuron dysfunction since
Estimates indicate that over 120 million Americans they have been shown to contact the neuromuscular
benefit from regenerative medicine (Harris, 2008; Cho junction and develop into a functional cholinergic phe-
et al., 2008; Cui et al., 2008). With respect to neuro- notype (Gao et al., 2007). Synaptic transmission is
logical disease, the use of stem cells may encompass intact upon fetal NSC transplantation, and improve-
disease states such as spinal cord injury, stroke, demen- ment in motor function can be observed (Gao et al.,
tias, neurodegeneration, and cerebral palsy (Harris, 2007).
17 Dual Roles of MSC in Spinal Cord Injury 277

17.7 Transdifferentiation natural microenvironmental cues may allow for the use
of Mesenchymal Stem Cells of MSC in the clinic. Upon transdifferentiation into
neural stem cells via retinoic acid, MSC may harbor
to Neurons
the capacity to secrete neural-specific products. The
advantage of this process is evident in the beneficial
Since neurons do not regenerate naturally with ease effects of various molecules including glial-derived
after injury, studies have investigated the role of stem neurotrophic factor (GDNF). GDNF has been shown
cell therapy in CNS regeneration. Of particular inter- to facilitate survival on neurons of various phenotypes,
est are mesenchymal stem cells (MSC), since they including noradrenergic and dopaminergic (Lo et al.,
entail far fewer ethical concerns than embryonic stem 2008). GDNF also hinders cell death of retinal gan-
cells. MSC have been shown to have transdifferenti- glion cells and cortical neurons, suggesting a role in
ation potential. This suggests that cells that are nor- the treatment of vision loss and various dementias (Lo
mally committed to one fate can be induced to give et al., 2008).
rise to cells of another lineage by in vitro manipu- Thus far, the use of MSC in SCI has not been com-
lation (Greco et al., 2007). Mesenchymal stem cells, pletely successful (Saski et al., 2008). In a study that
which are derived from the mesoderm of the mam- assessed the safety of autologous MSC transplant in
malian gastrula stage, normally develop into tissue patients with the motor neuron disease amyotrophic
of mesodermal origin, including adipocytes, chon- lateral sclerosis, also known as Lou Gehrigs disease,
drocytes, osteoblasts, and bone marrow stromal cells MSC expansion and delivery to the spinal cord of
(Fig. 17.1). Recently, studies have demonstrated a role patients showed minimal systemic toxicity at vari-
for MSC in the development of non-mesodermal tis- ous time points and was well tolerated. The reported
sue, including hepatocytes-like cells of the endoderm complications included sensory deficits in the lower
(Kazemnejad et al., 2008). extremities and pain in the thoracic cavity (Mazzini
MSC can also develop into neurons, which nor- et al., 2006). In another clinical study, neural stem cells
mally derive from the ectodermal lineage of the gas- were produced from bone marrow-derived MSC. Upon
trula (Greco et al., 2007). The value of this con- direct administration of these MSC to the target sites
cept is seen in neurodegenerative conditions such as in patients with chronic SCI, no adverse effects were
Parkinsons disease, amyotrophic lateral sclerosis, and reported (Moviglia et al., 2006). The effects of bone
Alzheimers disease, in which dopaminergic, cholin- marrow-derived hematopoietic progenitor cell (HPC)
ergic, and motor neurons, respectively, are not func- transplant have also been evaluated in patients with
tioning at an optimal level (Lang et al., 2006; Perrier chronic SCI (Deda et al., 2008). In this study, autolo-
and Studer, 2003; Shigetomi and Fukuchi, 1994). gous HPC administration resulted in no major adverse
Parkinsons disease has been investigated intensively effects, and patients experienced improvement in sen-
in the past few years, as stem cell therapy extends sory and motor functions (Deda et al., 2008). The
to regeneration of dopaminergic neurons of the mid- majority of investigators shed a positive perspective on
brain (Chiba et al., 2008). We discuss these prospects the use of MSC in SCI and neurological trauma.
in Section 17.8.
MSC have been shown to function in wound
repair by transdifferentiating into keratinocytes, a phe-
nomenon which functions to replace skin cells after 17.8 Other Neurodegenerative
trauma or injury (Sasaki et al., 2008). They have the Disorders
potential for developing into cells of the endothelium
and pericytes (Sasaki et al., 2008). Regarding the employment of stem cells for neurore-
Mesenchymal stem cell therapy may be approved in generative purposes, stem cell therapy is not lim-
the near future based on the large therapeutic poten- ited to spinal cord injury. Perhaps the most well-
tial of these cells. MSC can be obtained from adult known application of stem cell therapy to the com-
adipose tissue (Sasaki et al., 2008). This route of munity is regeneration of neurons for the treatment of
access suggests few ethical concerns, unlike the obtain- Parkinsons disease. Parkinsons disease affects more
ment of embryonic stem cells. The in vitro use of than 1 out of 100 people over the age of 65. Treatment
278 C. King et al.

of patients with this neurodegenerative condition is stem cells may repair nonfunctional neuronal connec-
arguably the most fruitful application of stem cells tions in chronic ischemia of the brain (Yamashita et al.,
(Trzaska and Rameshwar, 2007; Trazaska et al., 2007; 2008; Tansey et al., 2007). Activation or transplanta-
Sandberg, 2007). In Parkinsons disease, dopamine tion of stem cells in the neural environment may assist
neurons of the substantia nigra of the midbrain lose in increasing cerebral blood flow (Yamashita et al.,
function. As a result, motor and cognitive functions 2008). The activation strategy involves stimulating the
are compromised, and patients display bradykinesia, endogenous neural stem cells of the subventricular
resting tremor, rigidity, shuffling gait, and mask-like zone of the brain, and stroke has been shown to induce
facial expression (Osawa et al., 1997). The cur- proliferation of these cells (Yoo et al., 2008). On the
rent standards of treatment for Parkinsons disease other end of the spectrum, the transplantation strategy
include the dopamine precursor L-dopa, dopamine employs an exogenous source of neural stem cells to
receptor agonists, and cholinergic muscarinic antag- help patients recover from ischemic states (Yamashita
onists (Reiss et al., 2007). Such treatments func- et al., 2008).
tion to restore the proper balance between dopamine
and acetylcholine, but their effects tend to wear off
after some time, leading to uncomfortable symptoms
17.9 Limitations to Stem Cell
such as tardive dyskinesia and psychosis (Olanow
et al., 2006). Given these drawbacks of conventional Therapeutics
Parkinsons disease therapy, stem cells appear to be
critical to the improvement of the quality of patient life. Although numerous advances have been made in
Fortunately, numerous advances have been made stem cell research for neurological conditions such as
in this area. Human ESC can develop into neurons Parkinsons disease, mesencephalic dysfunction, and
with a dopaminergic phenotype, facilitating the recov- cerebrovascular accidents, the field is not without its
ery of patients with midbrain dysfunction. Dopamine- limitations. A brief summary of prospective problems
secreting neurons can be generated from human and with cell therapy for neurological disease is warranted.
murine ESC via treatment with the bone morphogenic Some experts in the field of stem cell research cast
protein antagonist Noggin (Chiba et al., 2008). These doubt on the ability of MSC to develop into true neu-
discoveries, along with others, underscore the potential ral derivatives (Hardy et al., 2008). Such qualms merit
for stem cell treatment in Parkinsons disease. investment of time and resources into investigations on
The boundaries of the therapeutic potential for the therapeutic prospects of MSC therapy and stem cell
stem cells have been extended to include treatment therapy in general for SCI and the other neurological
of lesions of the brain stem. Pair box 7 (Pax7), a insults. In any case, the immunosuppressive properties
transcription factor involved in fetal development and of MSC make their clinical use convincing (Ramasamy
cancer cell proliferation, has been shown to enhance et al., 2008) however, adult stem cells may be limited in
development of mesencephalic structures of the brain their therapeutic potential, as regenerative ability tends
(Thompson et al., 2008). Pax3 expression, on the other to decay with age (Roh et al., 2008).
hand, exerts antagonistic effects, maintaining a stem- Immunological interactions in the context of stem
like phenotype of midbrain structures. Stem cell ther- cell therapy may pose a potential dilemma dur-
apy in the future can make use of the opposing roles ing treatment. Investigations into immunological phe-
of Pax3 and Pax7 in differentiation of the midbrain nomenon have been conducted, and it has been shown
(Thompson et al., 2008). that inflammatory cytokines, such as tumor necro-
Stem cell therapy extends to the treatment of cere- sis factor- (TNF-), interferon- (IFN-), and the
brovascular accidents, commonly known as strokes. acute phase reactant interleukin-6 (IL-6) can upregu-
Cerebrovascular accident is due to CNS dysfunction late MHC genes in neural stem cells and progenitor
resulting from ischemic or hemorrhagic events in the cells. These discoveries encompass numerous models,
brain. Adult stem cells from the bone marrow, skele- including monkeys, rats, and humans (Johansson et al.,
tal muscle, peripheral blood and other sites have been 2008). The implications of immunostimulation of such
proposed to enhance recovery after a cerebrovascu- cytokines may cause complications to surface, as these
lar accident (Roh et al., 2008). Scientists believe that endogenous cytokines increase the probability of graft
17 Dual Roles of MSC in Spinal Cord Injury 279

rejection (Johansson et al., 2008). The environment acids (PGA), polyethylene glycol (PEG), and silicone,
of the CNS includes the aforementioned inflammatory have been sought and fabricated into tubes, fibers,
cytokines and can cause immune-mediated rejection of cross-linked hydrogels and gels containing fibers to
transplanted cells (Johansson et al., 2008). guide axonal growth. Although these materials show
Clever methods must be designed to counteract promise, directing the axons from the nerve guide
this phenomenon while keeping these cytokines func- back to the spinal cord and achieving integration
tions intact, as they may be vital to fetal neurogen- with the host synaptic pathways remains a chal-
esis (Johansson et al., 2008; Thompson and Powrie, lenge. Therefore, recent efforts have been exploring
2004). One prospect on preventing an unfavorable combination strategies using guidance channels with
immunological reaction involves transplantation or co- cells and/or neurotrophic or neuroprotective factors
transplantation of MSC, which are immunosuppressive (Nomura et al., 2006).
in nature (Ramasamy et al., 2008). Various cell types have been explored in combina-
tion with scaffolding materials to regenerate the spinal
cord. Schwann cells are of interest since they secrete
numerous neurotrophic factors, produce an extracel-
17.10 An Interdisciplinary Approach lular matrix and express cell adhesion molecules
(Nomura et al., 2006). They have been used in com-
Bioengineering strategies have focused on providing bination with degradable and nondegradable tubular
a permissive environment for the regeneration of the scaffolds; however have been found to be minimally
spinal cord and achieving integration between the CNS effective in treating the injured spinal cord without the
and PNS (Schmidt and Leach, 2003; Nomura et al., use of additional growth factors and other cell types
2006; Willerth and Sakiyama-Elbert, 2007). A com- (Bunge, 2008). Other supportive cell types that provide
mon approach is the use of biomaterials that act as ensheathment for developing axons, such as olfactory-
guidance channels, and/or as delivery vehicles for ensheathing glial cells (OEFs), have also been explored
cells, neuroregenerative factors and anti-inflammatory and show promise on scaffolds in which the biomate-
drugs. This section will review the current work in this rial acts as a structure to support their cell function.
area. Bioengineering approaches also focus on under- Stem cells, such as neural stem cells (NSC), MSC
standing injury to the spinal cord from a biomechanics and embryonic stem cells (ESC), are currently being
perspective, using various mathematical and experi- investigated in combination with scaffolds because
mental models for analyzing the anatomical loading the biomaterial can aid in localizing and promoting
distribution and resulting cellular responses (Maikos the differentiation and orientation of these cells at
et al., 2008; Laplaca et al., 2007). The functional the defect site based on the physical and chemical
response of nerves to deleterious biomechanical load- characteristics of the scaffold (Engler et al., 2006;
ing has lead to discoveries in determining the range Arinzeh et al., 2005; Xie et al., 2008). By combin-
of loads that can, in turn, promote nerve growth, ing the scaffold with stem/neural progenitor cells, the
i.e. stretch-induced growth (Bueno and Shah, 2008; cells may provide the therapeutic benefit of neuropro-
Smith et al., 2001), and may hold promise in develop- tection (Himes et al., 2006) and/or functionally inte-
ing transplantable nerve constructs to bridge extensive grate into the spared spinal cord circuitry (e.g. form-
damage. ing new oligodendrocytes and/or neurons) (Cummings
Bioengineering research efforts have been primar- et al., 2005; Cizkova et al., 2006) to improve the
ily concentrated on developing biomaterials that act outcome.
as scaffolds to physically guide the regeneration of Scaffolds can also be used for controlled-release
nerves across lesions, i.e. nerve guide or nerve of growth factors/drugs alone or in combination with
guidance channel (Schmidt and Leach, 2003). They cells. By incorporating growth factors/drugs in the
direct the axon growth as well as reduce the infiltration scaffold during fabrication, controlled release of that
of scar tissue. Natural materials, such as collagen, fib- drug occurs via diffusion and is dependent upon
rin, hyaluronic acid, agarose, chitosan and alginates, the physical properties of the material, such as pore
as well as degradable and nondegradable synthetic size, degree of cross-linking, and rate of degradation.
materials, such as polylactic (PLA) and polyglycolic Natural materials, such as collagen, agarose and fibrin,
280 C. King et al.

and synthetic polymers like polyethylene glycol-poly groups have been able to generate functional neu-
lactic acid (PEG-PLA) have been fabricated into guid- rons from MSC, including those from adult human
ance channels for the release of neurotrophin-3 (NT-3), bone marrow. The neurons can be used to estab-
nerve growth factor (NGF) and brain-dervied neu- lish a neuro-muscular system. Skeletal muscle cells
rotrophic factor (BDNF). Results demonstrated axonal can be generated from the same MSC to establish
sprouting and an increased functional recovery in SCI an autologous system. The neurons would be able
models (Willerth and Sakiyama-Elbert, 2007). Other to respond to tissue factors similar to those found
therapeutic drugs, such as chondroitinase ABC, anti- at sites of injuries, such as those discussed above.
bodies against Nogo and cyclic AMP, have shown Except for extrapolation on the roles of cytokines and
promise in reducing the inhibitory environment created other inflammatory mediators, it is unclear how the
in the CNS after injury (Willerth and Sakiyama-Elbert, factors at regions of SCI interact with each other to
2007) and are currently being investigated in controlled affect the damaged tissues. If stem cells, such as MSC
release strategies. Other approaches being explored will be used in any protocol for SCI treatment, it
in drug delivery methods are affinity-based systems would be important to determine if the receptors are
and immobilized drug delivery systems, where the clustered following responses to injuries and deter-
desired drug is non-covalently bound or covalently mine if there are synergism or antagonism among
attached to the scaffold, respectively. Electrically con- cytokines, and how this would affect the skeletal
trolled delivery systems, where the release of the muscles.
target drug is controlled by electrical stimulation, An experimental model can be established in which
have been investigated for use as coatings on neu- neurons are placed in contact with skeletal muscle
ral electrodes. Anti-inflammatory drugs were released from the same source of MSC. The responses of mus-
to reduce the number of reactive astrocytes and had cle, namely end plate potential, could be used as an
no negative effect on the viability of neurons (Wadha indication to test axonal repair if the neurons are sub-
et al., 2006). Release of NT-3 via this method has jected to axotomy by microdissection. This type of
been shown to stimulate neurite extension (Thompson model would recapitulate injured neurons, similar to
et al., 2006). Other systems under development use SCI. By adding cytokines to the system, before and
micro- and nano-electromechanical systems (MEMs after injury, the model would be able to mimic the
and NEMs) for on-demand pulsatile or adjustable con- injured nerve within a milleu of tissue factors. The
tinuous administration of the drug for extended periods model would be able to use to screen compound to
using a wireless integrated system that can regulate identify methods to repair axons.
drug release, receive sensor feedback, and transmit The discussed model could lead to the development
updates (Staples et al., 2006). Combining technologies of a genetic engineering tool in which human MSC
of microfabrication, information science and systems could be used to deliver the identified factors for SCI
biology will lead to more sophisticated drug delivery treatment. Similar methods could be used for oligo-
systems in the future. Approaches above are still in dendrocytes. This in vitro system could be quickly
their early stages of investigation and will require a bet- verified by in vivo methods. In addition, the system
ter understanding of the cell source used, drug release could allow for experimental questions to understand
rates and biomaterial properties to obtain functional nerve regrowth, and synapse formation with skeletal
recovery. muscle.
The method discussed above would need to com-
partmentalize the neurons from skeletal muscle. We
describe a co-culture method that has been modified
17.11 Experimental Models for SCI from a chamber model reported by (Campenot, 1977)
(Fig. 17.3). The system allows for the neurons and
SCI requires attention to major areas: repair the region skeletal muscle cells to be cultured in their respective
of injury; prevent rapid atrophy and cell death of dis- media. The insert is sealed after placement on sterile
tal neuronal degeneration; and regrowth of the axons. grease, as shown. Despite the compartmentalization,
To this end, experimental models should be established the cells can form contact through tracks placed within
with the SCI-associated confounds. Several research the gelatin that coats the surface of the Petri dish.
17 Dual Roles of MSC in Spinal Cord Injury 281

References

Allan SM, Tyrrell PJ, Rothwell NJ (2005) Interleukin-1 and


neuronal injury. Nat Rev Immunol 5:629640.
Arinzeh TL, Tran T, Mcalary J, Daculsi G (2005) A com-
parative study of biphasic calcium phosphate ceramics for
human mesenchymal stem cell-induced bone formation.
Biomaterials 26:36313638.
Azari MF, Profyris C, Zang DW, Petratos S, Cheema SS (2005)
Induction of endogenous neural precursors in mouse mod-
els of spinal cord injury and disease. Eur J Neurol 12:
638648.
Bagby GC (1989) Interleukin-1 and hematopoiesis. Blood Rev
3:152161.
Barnab-Heider F, Frisn J (2008) Stem cells for spinal cord
repair. Cell Stem Cell 3:1624.
Bianco P, Riminucci M, Gronthos S, Robey PG (2001) Bone
marrow stromal stem cells: nature, biology, and potential
applications. Stem Cells 19(3):180192.
Bieback K, Kern S, Klter H, Eichler H (2004) Critical param-
eters for the isolation of mesenchymal stem cells from
Fig. 17.3 Petri dishes were coated with 1 endotoxin-free umbilical cord blood. Stem Cells 22:625634.
gelatin. After this, tracks were made along the dried gelatin Blanc KL, Pittenger MF (2005) Mesenchymal stem cells:
then the Teflon inserts were added. The insert was stabilized progress toward promise. Cytotherapy 7:3645.
with sterile Vaseline. Shown are human MSC-derived neurons. Blesch A, Uy HS, Grill RJ, Cheng JG, Patterson PH, Tuszynski
The neurons can contact skeletal muscle in the inner chamber. MH (1999) Leukemia inhibitory factor augments neu-
Axotomy in the neurons can affect end plate potential. This rotrophin expression and corticospinal axon growth after
simple co-culture model could be a tool for rapid screen of adult CNS injury. J Neurosci 19:35563566.
compound for SCI repair Bonni A, Sun Y, Nadal-Vicens M, Bhatt A, Frank DA,
Rozovsky I, Stahl N, Yancopoulos GD, Greenberg ME
(1997) Regulation of gliogenesis in the central nervous sys-
tem by the JAK/STAT signaling pathway. Science 278:477
483.
Bueno FR, Shah SB (2008) Implications of tensile loading for
17.12 On the Frontier of Stem Cell the tissue engineering of nerves. Tissue Eng Part B Rev
Therapy for Neural Dysfunction 14:219233.
Bunge MB (2008) Novel combination strategies to repair the
injured mammalian spinal cord. J Spinal Cord Med 31:262
At the beginning of this twenty-first century, sci- 269.
entists can benefit from vast scientific discoveries. Butzkueven H, Zhang JG, Soilu-Hanninen M, Hochrein
H, Chionh F, Shipham KA, Emery B, Turnley AM,
Researchers must constantly ask, How can todays
Petratos S, Ernst M, Bartlett PF, Kilpatrick TJ (2002)
advances in science lead to benefits for tomorrows LIF receptor signaling limits immune-mediated demyelina-
patients? The past prospects of stem cells on treat- tion by enhancing oligodendrocyte survival. Nat Med 8:
ment of pathological conditions of the nervous sys- 613619.
Campenot RB (1977) Local control of neurite development
tem have shed light onto future discoveries in the
of nerve growth factor. Proc Natl Acad Sci U S A 74:
field. In any case, scientists must exert some cau- 45164519.
tion concerning the therapeutic potential of stem cells, Caplan AI (1994) The mesengenic process. Clin Plast Surg
considering the aforementioned qualms. With a con- 21:429435.
Carlos TM, Harlan JM (1994) Leukocyte-endothelial adhesion
tinued commitment to the advancement of science
molecules. Blood 84:20682101.
and the translation of basic science discoveries to Castillo M, Liu K, Bonilla LM, Rameshwar P (2007) The
the clinic, stem cell therapy may one day alleviate immune properties of mesenchymal stem cells. Intl J Biomed
much suffering while improving the quality of patient Sci 3:100.
Chan JL, Tang KC, Patel AP, Bonilla LM, Pierobon N,
care.
Ponzio NM, Rameshwar P (2006) Antigen-presenting prop-
erty of mesenchymal stem cells occurs during a narrow
Acknowledgement This work was supported by the FM Kirby window at low levels of interferon-gamma. Blood 107:
Foundation. 48174824.
282 C. King et al.

Chi LY, Yu J, Zhu H, Li XG, Zhu SG, Kindy MS (2008) The dual cloning and expression of cDNA encoding a murine
role of tumor necrosis factor-alpha in the pathophysiology of myeloid leukaemia inhibitory factor (LIF). EMBO J 6:
spinal cord injury. Neurosci Lett 438:174179. 39954002.
Chiba S, Lee YM, Zhou W, Freed CR (2008) Noggin enhances Golestaneh N, Mishra B (2005) TGF-beta, neuronal stem cells
dopamine neuron production from human embryonic stem and glioblastoma. Oncogene 24:57225730.
cells and improves behavioral outcome after transplantation Greco SJ, Rameshwar P (2007) Enhancing effect of IL-1alpha
into Parkinsonian rats. Stem Cells 26:28102820. on neurogenesis from adult human mesenchymal stem
Cho SR, Yang MS, Yim SH, Park JH, Lee JE, Eom YW, Jang IK, cells: implication for inflammatory mediators in regenerative
Kim HE, Park JS, Kim HO, Lee BH, Park CI, Kim YJ (2008) medicine. J Immunol 179:33423350.
Neurally induced umbilical cord blood cells modestly repair Greco SJ, Zhou C, Ye JH, Rameshwar P (2007) An interdis-
injured spinal cords. Neuroreport 19:12591263. ciplinary approach and characterization of neuronal cells
Cizkova D, Rosocha J, Vanicky I, Jergova S, Cizek M (2006) transdifferentiated from human mesenchymal stem cells.
Transplants of human mesenchymal stem cells improve func- Stem Cells Dev 16:811826.
tional recovery after spinal cord injury in the rat. Cell Mol Hardy SA, Maltman DJ, Przyborski SA (2008) Mesenchymal
Neurobiol 26:11671180. stem cells as mediators of neural differentiation. Curr Stem
Cui L, Jiang J, Wei L, Zhou X, Fraser JL, Snider BJ, Yu SP Cell Res Ther 3:4352.
(2008) Transplantation of embryonic stem cells improves Harris DT (2008) Cord blood stem cells: a review of potential
nerve repair and functional recovery after severe sciatic nerve neurological applications. Stem Cell Rev 4:269274.
axotomy in rats. Stem Cells 26:13561365. Hayashi M, Ueyama T, Nemoto K, Tamaki T, Senba E
Cummings BJ, Uchida N, Tamaki SJ, Salazar DL, Hooshmand (2000) Sequential mRNA expression for immediate early
M, Summers R (2005) Human neural stem cells differentiate genes, cytokines, and neurotrophins in spinal cord injury. J
and promote locomoter recovery in spinal cord-injured mice. Neurotrauma 17:203218.
Proc Natl Acad Sci U S A 102:1406914074. Hernandez LA, Grisham MB, Twohig B, Arfors KE, Harlan
Deda H, Inci MC, Kurekci AE, Kayihan K, Ozgun E, JM, Granger DN (1987) Role of neutrophils in ischemia-
Ustunsoy GE, Kocabay S (2008) Treatment of chronic spinal reperfusion-induced microvascular injury. Am J Physiol
cord injured patients with autologous bone marrow-derived 253:H699H703.
hematopoietic stem cell transplantation: 1-year follow-up. Himes BT, Neuhuber B, Coleman C, Kushner R, Swanger
Cytotherapy 10:565574. SA, Kopen GC (2006) Recovery of function following
Deng J, Petersen BE, Steindler DA, Jorgensen ML, Laywell grafting of human bone marrow-derived stromal cells into
ED (2006) Mesenchymal stem cells spontaneously express the injured spinal cord. Neurorehabil Neural Repair 20:
neural proteins in culture and are neurogenic after transplan- 278296.
tation. Stem Cells 24:10541064. Johansson S, Price J, Modo M (2008) Effect of inflammatory
Dinarello CA (1996) Biologic basis for interleukin-1 in disease. cytokines on MHC expression and differentiation of human
Blood 87:20952147. neural stem/progenitor cells. Stem Cells 26:24442454.
Dinarello CA (2005) Blocking IL-1 in systemic inflammation. J Kang MK, Kang SK (2008) Interleukin-6 induces prolifera-
Exp Med 201:13551359. tion in adult spinal cord-derived neural progenitors via the
Divani AA, Hussain MS, Magal E, Heary RF, Qureshi AI JAK2/STAT3 pathway with EGF-induced MAPK phospho-
(2007) The use of stem cells hematopoietic stimulating fac- rylation. Cell Prolif 41:377392.
tors therapy following spinal cord injury. Ann Biomed Eng Kazemnejad S, Allameh A, Soleimani M, Gharehbaghian
35:16471656. A, Mohammadi Y, Amirizadeh N, Jazayery M (2008)
Downing JR (2004) TGF- signaling, tumor suppression, and Biochemical and molecular characterization of hepatocyte-
acute lymphoblastic leukemia. N Engl J Med 351(6):528. like cells derived from human bone marrow mesenchy-
Ducker TB, Hamit HF (1969) Experimental treatments of acute mal stem cells on a novel three-dimensional biocompatible
spinal cord injury. J Neurosurg 30:693697. nanofibrous scaffold. J Gastroenterol Hepatol 24:278287.
Ducker TB, Zeidman SM (1994) Spinal cord injury. Role of Kim SJ, Letterio J (2003) Transforming growth factor-beta sig-
steroid therapy. Spine 19:22812287. naling in normal and malignant hematopoiesis. Leukemia
Engler AJ, Sen S, Sweeney HL, Discher DE (2006) Matrix elas- 17:17311737.
ticity directs stem cell lineage specification. Cell 126:677 Kurek JB, Bennett TM, Bower JJ, Muldoon CM, Austin L
689. (1998a) Leukaemia inhibitor factor (LIF) production in a
Fujiwara T, Kubo T, Koyama Y, Tomita K, Yano K, Tohyama mouse model of spinal trauma. Neurosci Lett 249:14.
M, Hosokawa K (2008) mRNA expression changes of slit Kurek JB, Radford AJ, Crump DE, Bower JJ, Feeney SJ, Austin
proteins following peripheral nerve injury in the rat model. J L, Byrne E (1998b) LIF (AM424), a promising growth factor
Chem Neuroanat 36:170176. for the treatment of ALS. J Neurol Sci 160:S106S113.
Gadient RA, Otten UH (1997) Interleukin-6 (IL-6) a Kwon BK, Tetzlaff W, Grauer JN, Beiner J, Vaccaro AR
molecule with both beneficial and destructive potentials. (2004) Pathophysiology and pharmacologic treatment of
Prog Neurobiol 52:379390. acute spinal cord injury. Spine J 4:451464.
Gao J, Coggeshall RE, Chung JM, Wang J, Wu P (2007) LaPlaca MC, Simon CM, Prado GR, Cullen DK (2007) CNS
Functional motoneurons develop from human neural stem injury biomechanics and experimental models. Prog Brain
cell transplants in adult rats. Neuroreport 18:565569. Res 161:1326.
Gearing DP, Gough NM, King JA, Hilton DJ, Nicola NA, Lange KW, Mecklinger L, Walitza S, Becker G, Gerlach M,
Simpson RJ, Nice EC, Kelso A, Metcalf D (1987) Molecular Naumann M, Tucha O (2006) Brain dopamine and kine-
17 Dual Roles of MSC in Spinal Cord Injury 283

matics of graphomotor functions. Human Movement Sci entiated marrow stromal cells and CD133+ stem-like cells.
25:492509. Cell Transplant 12:839848.
Laver J, Moore MAS (1989) Clinical use of recombinant Pan W, Cain C, Yu Y, Kastin AJ (2006) Receptor-mediated trans-
human hematopoietic growth factors. J Natl Cancer Inst 81: port of LIF across blood-spinal cord barrier is upregulated
13701382. after spinal cord injury. J Neuroimmunol 174:119125.
Lee OK, Kuo TK, Chen WM, Lee KD, Hsieh SL, Chen TH Perrier AL, Studer L (2003) Making and repairing the mam-
(2004) Isolation of multipotent mesenchymal stem cells from malian brainin vitro production of dopaminergic neurons.
umbilical cord blood. Blood 103:16691675. Semin Cell Dev Biol 14:181189.
Li M, Sendtner M, Smith A (1995) Essential function of LIF Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R,
receptor in motor neurons. Nature 378:724727. Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak
Lo WC, Hsu CH, Wu AT, Yang LY, Chen WH, Chiu WT, Lai DR (1999) Multilineage potential of adult human mesenchy-
WF, Wu CH, Gelovani JG, Deng WP (2008) A novel cell- mal stem cells. Science 284:143147.
based therapy for contusion spinal cord injury using GDNF- Planchamp V, Bermel C, Tnges L, Ostendorf T, Kgler S, Reed
delivering NIH3T3 cells with dual reporter genes monitored JC, Kermer P, Bhr M, Lingor P (2008) BAG1 promotes
by molecular imaging. J Nucl Med 49:15121519. axonal outgrowth and regeneration in vivo via Raf-1 and
Maikos JT, Qian Z, Metaxas D, Shreiber DI (2008) Finite ele- reduction of ROCK activity. Brain 131:26062619.
ment analysis of spinal cord injury in the rat. J Neurotrauma Potian JA, Aviv H, Ponzio NM, Harrison JS, Rameshwar
25:795816. P (2003) Veto-like activity of mesenchymal stem cells:
Martinez C, Hofmann TJ, Marino R, Dominici M, Horwitz Functional discrimination between cellular responses
EM (2007) Human bone marrow mesenchymal stromal cells to alloantigens and recall antigens. J Immunol 171:
express the neural ganglioside GD2: a novel surface marker 34263434.
for the identification of MSCs. Blood 109:42454248. Ramasamy R, Tong CK, Seow HF, Vidyadaran S, Dazzi
Massague J, Weis-Garcia F (1996) Serine/threonine kinase F (2008) The immunosuppressive effects of human bone
receptors: mediators of transforming growth factor beta fam- marrow-derived mesenchymal stem cells target T cell pro-
ily signals. Cancer Surv 27:4164. liferation but not its effector function. Cell Immunol 251:
Massague J, Wotton D (2000) Transcriptional control by the 131136.
TGF-beta/Smad signaling system. EMBO J 19:17451754. Reis HJ, Rosa DV, Guimares MM, Souza BR, Barros AG,
Massagu J, Andres J, Attisano L, Cheifetz S, Lpez-Casillas Pimenta FJ, Souza RP, Torres KC, Romano-Silva MA (2007)
F, Ohtsuki M, Wrana JL (1992) TGF-beta receptors. Mol Is DARPP-32 a potential therapeutic target? Expert Opin
Reprod Dev 32(2):99104. Ther Targets 11:16491661.
Mazzini L, Mareschi K, Ferrero I, Vassallo E, Oliveri G, Roberts AB, Sporn MB (1993) Physiological actions and clinical
Boccaletti R, Testa L, Livigni S, Fagioli F (2006) Autologous applications of transforming growth factor-beta (TGF-beta).
mesenchymal stem cells: clinical applications in amy- Growth Factors 8:119.
otrophic lateral sclerosis. Neurol Res 28:523526. Roh JK, Jung KH, Chu K (2008) Adult stem cell transplantation
Mehra A, Wrana JL (2002) TGF-beta and the Smad signal in stroke: its limitations and prospects. Curr Stem Cell Res
transduction pathway. Biochem Cell Biol 80:605622. Ther 3:185196.
Moore MA (2002) Cytokine and chemokine networks influ- Sanberg PR (2007) Neural stem cells for Parkinsons disease: to
encing stem cell proliferation, differentiation, and marrow protect and repair. Proc Natl Acad Sci U S A 104:11869
homing. J Cell Biochem 38:2938. 11870.
Moviglia GA, Fernandez Via R, Brizuela JA, Saslavsky J, Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, Shimizu H
Vrsalovic F, Varela G, Bastos F, Farina P, Etchegaray G, (2008) Mesenchymal stem cells are recruited into wounded
Barbieri M, Martinez G, Picasso F, Schmidt Y, Brizuela P, skin and contribute to wound repair by transdifferentiation
Gaeta CA, Costanzo H, Moviglia Brandolino MT, Merino S, into multiple skin cell type. J Immunol 180:25812587.
Pes ME, Veloso MJ, Rugilo C, Tamer I, Shuster GS (2006) Sato M, Muragaki Y, Saika S, Roberts AB, Ooshima A (2003)
Combined protocol of cell therapy for chronic spinal cord Targeted disruption of TGF-beta1/Smad3 signaling protects
injury. Report on the electrical and functional recovery of two against renal tubulointerstitial fibrosis induced by unilateral
patients. Cytotherapy 8:202209. ureteral obstruction. J Clin Invest 112:14861494.
Nomura H, Tator CH, Shoichet MS (2006) Bioengineered strate- Schmidt CE, Leach JB (2003) Neural tissue engineering: strate-
gies for spinal cord repair. J Neurotrauma 23:496507. gies for repair and regeneration. Annu Rev Biomed Eng
Olanow CW, Obeso JA, Stocchi F (2006) Drug insight: continu- 5:293347.
ous dopaminergic stimulation in the treatment of Parkinsons Sekhon LH, Fehlings MG (2001) Epidemiology, demograph-
disease. Nat Clin Pract Neurol 2:382392. ics, and pathophysiology of acute spinal cord injury. Spine
Ong SY, Dai H, Leong KW (2006) Hepatic differentiation poten- 26:S2S12.
tial of commercially available human mesenchymal stem Shi Y, Massagu J (2003) Mechanisms of TGF-beta signaling
cells. Tissue Eng 12:34773485. from cell membrane to the nucleus. Cell 113:685700.
Osawa H, Yamabe H, Kaizuka M, Tamura N, Tsunoda S, Shirato Shigetomi S, Fukuchi S (1994) Recent aspect of the role of
KI, Okumura K (1997) Systemic lupus erythematosus asso- peripheral dopamine and its receptors in the pathogenesis of
ciated with transverse myelitis and parkinsonian symptoms. hypertension. Fukushima J Med Sci 40:6983.
Lupus 6:613615. Smith DH, Wolf JA, Meaney DF (2001) A new strategy to
Padovan CS, Jahn K, Birnbaum T, Reich P, Sostak P, Strupp M, produce sustained growth of central nervous system axons:
Straube A (2003) Expression of neuronal markers in differ- continuous mechanical tension. Tissue Eng 7:131139.
284 C. King et al.

Sokol JP, Schiemann WP (2004) Cystatin C antagonizes trans- Wadhwa R, Lagenaur CF, Cui XT (2006) Electrochemically
forming growth factor beta signaling in normal and cancer controlled release of dexamethasone from conducting poly-
cells. Mol Cancer Res 2:183195. mer polypyrrole coated electrode. J Control Release 110:
Stagg J, Pommey S, Eliopoulos N, Galipeau J (2006) Interferon- 531541.
gama-stimulated marrow stromal cells: a new type of Willerth SM, Sakiyama-Elbert SE (2007) Approaches to neu-
nonhematopoietic antigen-presenting cell. Blood 107:2570 ral tissue engineering using scaffolds for drug delivery. Adv
2577. Drug Dev Rev 59:325338.
Staples M, Daniel K, Cima MJ, Langer R (2006) Application of Wrana JL (2000) Regulation of Smad activity. Cell 100:
micro- and nano-electromechanical devices to drug delivery. 189192.
Pharm Res 23:847863. Xie J, Willerth SM, Li X, Macewan MR, Rader A, Sakiyama-
Tansey MG, McCoy MK, Frank-Cannon TC (2007) Elbert SE, Younan X (2008) The differentiation of embryonic
Neuroinflammatory mechanisms in Parkinsons disease: stem cells seeded on electrospun nanofibers into neural
Potential environmental triggers, pathways, and targets for lineages. Biomaterials 30:354362.
early therapeutic intervention. Expl Neurol 208:125. Yamashita T, Deguchi K, Sehara Y, Lukic-Panin V, Zhang H,
Tebar LA, Granton SM, Parsons-Perez C, Fisher AS, Bayne Kamiya T, Abe K (2008) Therapeutic strategy for ischemic
R, Smith AJ, Turmaine M, Perez-Luz S, Sheasby A, De stroke. Neurochem Res 34:707710.
Felipe C, Ruff C, Raivich G, Hunt SP (2008) Deletion Yamashita T, Deguchi K, Sehara Y, Lukic-Panin V, Zhang H,
of the mouse RegIIIbeta (Reg2) gene disrupts ciliary neu- Kamiya T, Abe K, Yamauchi K, Osuka K, Takayasu M,
rotrophic factor signaling and delays myelination of mouse Usuda N, Nakazawa A, Nakahara N, Yoshida M, Aoshima C,
cranial motor neurons. Proc Natl Acad Sci U S A 105: Hara M, Yoshida J (2006) Activation of JAK/STAT signaling
1140011405. in neurons following spinal cord injury in mice. J Neurochem
Thompson BC, Moulton SE, Ding J, Richardson R, Cameron 96:10601070.
A, OLeary S, Wallace GG, Clark GM (2006) Optimising Yoo SW, Kim SS, Lee SY, Lee HS, Kim HS, Lee YD, Suh-Kim
the incorporation and release of a neurotrophic factor using H (2008) Mesenchymal stem cells promote proliferation of
conducting polypyrrole. J Control Release 116:285294. endogenous neural stem cells and survival of newborn cells
Thompson C, Powrie F (2004) Regulatory T cells. Curr Opin in a rat stroke model. Exp Mol Med 40:387397.
Pharmacol 4:408414. Young W (2000) Molecular and cellular mechanisms of spinal
Thompson JA, Zembrzycki A, Mansouri A, Ziman M (2008) cord injury therapies. In: Neurobiology of Spinal Cord Injury
Pax7 is requisite for maintenance of a subpopulation of supe- (Kalb RG, Strittmatter SM, eds.), p. 241. Humana Press,
rior collicular neurons and shows a diverging expression Totowa.
pattern to Pax3 during superior collicular development. BMC Yune TY, Chang MJ, Kim SJ, Lee YB, Shin SW, Rhim H,
Dev Biol 8:62. Kim YC, Shin ML, Oh YJ, Han CT, Markelonis GJ, Oh TH
Tonge D, Chan K, Zhu N, Panjwani A, Arno M, Lynham S, (2003) Increased production of tumor necrosis factor-alpha
Ward M, Snape A, Pizzey J (2008) Enhancement of axonal induces apoptosis after traumatic spinal cord injury in rats. J
regeneration by in vitro conditioning and its inhibition by Neurotrauma 20:207219.
cyclopentenone prostaglandins. J Cell Sci 121:25652577. Zai LJ, Yoo S, Wrathall JR (2005) Increased growth factor
Trzaska KA, Kuzhikandathil EV, Rameshwar P (2007) expression and cell proliferation after contusive spinal cord
Specification of a dopaminergic phenotype from adult human injury. Brain Res 1052:147155.
mesenchymal stem cells. Stem Cells 25:27972808. Zhang X, Zeng Y, Zhang W, Wang J, Wu J, Li J (2007) Co-
Trzaska KA, Rameshwar P (2007) Current advances in the treat- transplantation of neural stem cells and NT-3-overexpressing
ment of Parkinsons disease with stem cells. Curr Neurovas Schwann cells in transected spinal cord. J Neurotrauma
Res 4:99109. 24:18631877.
Index

Acetylcholine (ACh), 53, 107, 109, 116, 121122, 124, 156, Axonal repair, 271281
162, 203204, 208, 278 Axotomy, 273 276, 280281
Acetylcholine (ACh) receptor, 121, 162
Activated microglia, 21, 176, 182, 183
B
Adenosine, 124, 162, 209
Basic fibroblast growth factor (bFGF), 20, 181, 199201, 211,
Adenosine receptor, 124, 209
259260, 265
Adenylyl cyclase-activating polypeptide (PACAP), 209210
Basic helix-loop-helix (bHLH), 38, 136139
Adipose-derived stem cells (ADSC), 119
Bergmann glia, 148149, 161
Adrenergic neurotransmitters, 207
Bipolar disorder, 104, 108
Adult neural stem cells, 21, 100
Bipotent progenitors, 50
Adult neurogenesis, 1221, 88, 93, 100, 116, 149151, 264
glial-melanocytic (GM), 50, 5355
Adult stem cell, 1415, 116, 120, 151, 238, 257258, 278
glial-myofibroblastic (GF), 50
Alkaline phosphatase, 82, 123, 239
Blastocyst, 61, 79, 119, 257
Altman, 1213, 17, 116, 149, 152, 256, 259
B-myb, 76
Alzheimers disease (AD), 14, 2425, 79, 98, 109110, 264,
Bone marrow cells, 15, 2324
277
Bone marrowderived stem/progenitor cells, 119, 238
amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
Bone marrow stromal cells (BMSC), 118, 239, 277
(AMPA) receptor, 122123, 157, 204205
Bone morphogenetic protein (BMP), 38, 38, 49, 54, 99101,
-aminobutyric acid (GABA)ergic, 25, 9899, 102, 104106,
105106, 150151, 160, 183, 202
108109, 123124, 126, 137, 156, 207, 239
transforming Growth Factor beta (TGF-) superfamily, 3,
-aminobutyric-acid (GABA), 25, 9899, 101, 104106, 116,
68, 22, 99, 102103, 151, 154155, 157158, 160, 162,
120122, 124, 126, 128, 156, 206208, 239, 247
175177, 180, 182183, 200, 247, 250, 275
-aminobutyric-acid (GABA) receptor, 104, 120121, 206207
Brain derived neurotrophic factor (BDNF), 2021, 42, 99, 103,
Amniotic membrane, 247
106108, 179, 181182, 200201, 247, 249250, 276,
Amyloid- peptide, 25, 109, 157
280
Amyotrophic lateral sclerosis (ALS), 98, 105, 107, 163, 251,
Brain lipid-binding protein (BLBP), 148149, 151, 153154
264, 277
Brn3a, 105
Aneuploidy, 7383
Bromodeoxyuridine (BrdU), 1314, 1617, 19, 23, 150, 193,
Anophthalmia-microphthalmia (AM), 202
196, 203, 208, 210212, 256258
Apoptosis, 1718, 38, 78, 80, 125, 177, 179, 180, 212217,
Bub1, 75
265, 273276
Bub3, 75
Astrocyte differentiation, 120, 151154, 156158, 181, 183
BubR1, 75, 79
Astrocytes, 1517, 2122, 24, 4142, 60, 63, 65, 6768, 100,
103, 106, 117119, 124, 136, 138, 146163, 172,
176177, 180183, 232, 247, 249, 256257, 260, 265, C
280 Cajal-Retzius cells, 153
Astrocytic stem cells, 16, 150 Calcium, 104, 118, 124, 128129, 262, 273
Astroglial cells, 146152 free intracellular calcium concentration ([Ca2+]i), 120123,
Astroglia-neuron interactions, 147 125127, 203204, 207, 210, 213
Astrogliogenesis, 183, 260 Calcium binding proteins, 104, 148
ATM kinase (DNA-damage response kinase ATM), 78 calbindin (CB), 99, 104
ATP, 124125, 156, 176, 208209, 211, 215 calretinin (CR), 104
ATR kinase, 199 Calcium-induced calcium release (CICR), 122, 125, 128
Autism, 94, 128 Calcium signaling, 123, 125128
Autophagic cell death, 214

H. Ulrich (ed.), Perspectives of Stem Cells, 285


DOI 10.1007/978-90-481-3375-8,  c Springer Science+Business Media B.V. 2010
286 Index

Calcium waves, 120, 125126 Dedifferentiation, 5455, 151, 153, 183


frequency of calcium waves, 120 Deep brain stimulation (DBS), 246
Callcium oscillations in astrocytes, 159 Dendritic cell markers, 180
Camillo Golgi, 146 Dentate gyrus, 1314, 1621, 100, 104, 116, 123, 149, 153,
Cancer stem cells (CSCs), 80 182, 232, 256, 263
Caspase, 179180, 213215, 217 Developmental neural diseases, 128
Catecholaminergic, 53 Dibutyryl cyclic AMP (dbcAMP), 247, 250
Caudal ganglionic eminence (CGE), 105 Dickkopf, 49
CD34, 233, 240 Dlx1/2, 105
CD36 scavenger receptor, 180 DNA damage signaling, 7779, 214
CD45, 233238, 241, 272 Dogma of neuroscience; dogma in neuroscience, 12, 116,
Cdc2, 38, 76, 201 256
Cdc25A phosphatase, 78, 211 Dopamine (DA), 102103, 124, 203, 207208, 211, 246247,
Cdk4, 75, 197 251, 264, 278
Cdk6, 75, 77, 197 Dopaminergic, 18, 25, 52, 9899, 102104, 207208, 247,
Cell-based therapy, 74, 8283, 98, 102, 262, 265 277278
Cell cycle, 14, 18, 7476, 92, 122, 139140, 148, 150, Downs syndrome (trisomy 21), 79
193212, 215217, 256, 259260
Cell cycle checkpoints, 7476, 199
E
Cell-cycle machinery, 197, 199
E2F-DP, 7577
Cell division, 12, 14, 3839, 61, 7678, 8081, 93, 117, 121,
Ectoderm explants (animal caps), 23, 8
148, 193197, 208, 217, 256259
Embryogenesis, 55, 6167, 74, 100102, 106, 124, 175, 193,
asymmetric division, 14, 80, 117118, 136, 148, 195
239, 275
symmetric division, 14, 117, 135136
Embryoid bodies (EB), 100102, 106, 107
Cellular replacement, 18, 24, 53, 55, 103104, 107, 261,
Embryonal carcinoma (EC) cells, 79, 118120, 125
263264, 271281
Embryonic development, 7, 12, 17, 59, 61, 105, 116, 149, 204,
CENP-E, 75, 80
206
Chemokines, 2022, 180181, 183, 200, 233, 238, 240, 265
Embryonic stem (ES) cells, 15, 24, 53, 5969, 7383, 97110,
Chick embryo, 2, 4, 78
119120, 161, 211, 234, 245251, 260, 263, 277, 279
Chk1/2, 78, 199
Endonuclease (EN), 88, 91, 94
Cholecystokinin (CCK), 104
Engrailed-1, engrailed 1/2, 103, 247
Cholinergic, 53, 9899, 106, 109110, 121122, 125126, 203,
Enkephalin, 209210
276278
Epidermal growth factor (EGF), 20, 39, 101, 103, 119, 125,
Chordin, 2, 48, 49
150151, 153, 159, 200201, 211, 259260
Chromatin remodeling, 93
Epigenetic, 88, 93, 95, 155, 163, 182, 192, 259260
Chromosomal aberrations, 7475
Epigenetic gene silencing, 155
Ciliary epithelium (CE), 194
Epilepsy, 1820, 22, 25, 98, 104, 106, 163
Ciliary neurotrophic factor (CNTF), 120, 150, 154155, 211,
5-ethynyl-2-deoxyuridine (EdU), 256, 258
260
Evolution, 89, 92, 94, 163, 262
Colony stimulating factors (CSF), 175177, 179, 232
Evx1/2, 105
Compensatory neurogenesis, 19
Excitatory neurotransmisson, 122
Controlling elements, 9293
Excitotoxicity, 104105, 276
Cross-differentiation/transdifferentiation, 15, 55, 194, 257258,
Experimental allergic encephalomyelitis (EAE), 265266
272, 276277
Extracellular matrix components (ECM) in neurite outgrowth,
CXCR4, 2122, 233, 238241
42, 64, 157, 159, 160, 162, 164, 176177, 180, 259,
Cyclin
279
cyclin A, 7577
Extrinsic factors, 116118, 121, 128, 194, 205, 214
cyclin D, 75, 77, 197198
cyclin E, 7578, 197198
Cyclin-Cdk complexes, 75, 77, 199 F
Cyclin-dependent kinase inhibitors (CKI), 75, 79, 139, 198 Fetal derived striatal neurons, 106
Cyclin-dependent kinases (CDK), 7577, 139, 197199, 201, Fetal neural stem cells, 25, 256
211, 216217 Fibroblast growth factor (FGF), 4, 20, 38, 42, 100, 119, 150,
Cytokeratins, 37, 3940, 6062 181, 199, 247, 259
Cytokines, 2024, 75, 78, 154, 176, 179181, 194, 199203, FGF20, 247
210, 247, 259, 272, 274275, 278280 FGF4, 99, 108
FGF8, 38, 99, 102103, 105106, 108, 247, 249250
D Follistatin, 2, 4, 7, 49
Dbx1, 105 FOM-1, 82
Dbx2, 105 Fractalkine, 180181
Index 287

G Hormones, 20, 93, 116, 120, 150, 159, 177, 179


GABA, 25, 9899, 101, 104106, 116, 120122, 124, 126, 128, [3 H]-thymidine; tritiated thymidine, 1213, 16, 175176, 196,
156, 206208, 239, 247 204, 209210, 212, 256, 258
GAD67, 99, 106, 109 Human neural stem cells (hNSC), 25, 256, 264
GAP-43 (growth associated protein of 43-kDa molecular mass), Huntingtin, 104
37 Huntingtons disease (HD), 14, 104, 106, 163, 264
Gata2, 99, 108 6-hydroxy dopamine (6-OHDA), 247, 250
Genetic mosaicism, 89
Genome integrity, 7476 I
Genomic shocks hypothesis, 93 ImageStream system (ISS) analysis, 234237
GFAP (glial fibrillary acid protein), 16, 60, 6364, 66, 6869, Immune response, 20, 23, 272, 276
137152, 154158, 183, 238, 240 Immune system, 20, 24, 88, 163, 182, 259
Glaucomatous neurodegeneration, 192 innate immune response, 23
Glia-induced synaptogenesis, 161162 Induced pluripotent stem (iPS) cells, 99, 107, 246247, 251,
Glial (cell line)-derived neurotrophic factor (GDNF), 25, 99, 257, 260
103, 107, 175, 181182, 247, 249250, 277 Inflammation, 2023, 103, 107, 125, 180181, 202, 215,
Glial tumors; gliomas, 6768, 174, 183 263266, 273, 275
Gliogenic differentiation potential, 50 inflammatory cytokines, 21, 180182, 278279
Gliotransmitters, 159, 163 Inflammatory mediators, 274275, 280
Globose basal cells (GBC), 3637, 3940 Inner cell mass (ICM); inner cell mass of the blastocyst, 61,
Glutamate, 21, 102, 104, 116, 120, 122124, 126, 128, 137, 119, 257
148, 156158, 162, 181, 204206, 273, 276 Inositol 1, 4, 5-trisphosphate (IP3), 125, 128, 203204, 209210
Glutamate-aspartate transporter (GLAST), 148150, 154156 Insulin-like growth factor (IGF), 20, 22, 99, 199200, 210
Glutamate receptors, 122124, 204205 Interferon gamma (IFN), 21, 22, 179180
Glutamate transporter-1 (GLT-1), 156158 Interkinetic nuclear migration (INM), 147, 193, 196197, 203,
Glutamic acid decarboxylase (GAD), 104, 126, 206 211212
Glutamine synthase, 148149 Interleukin-1 (IL-1), 21, 180, 182, 275
Glycine, 206207 Intermediate filament (IF), 5969, 148, 158, 260, 276
GM-CSF, 179 Interneurons, 16, 25, 98, 101, 104106, 109, 137, 206, 263
GNMFCO progenitors, 51, 5354 -internexin, 60, 62
G-protein-coupled inwardly rectifying potassium channel Intracellular calcium transients, 118, 127
(Girk), 2, 247 Invasion routes (microglial precursors), 175
Group II introns, 94 Irx3, 105
Growth factors, 156158, 199203 Isl1/2, 105
See also Basic fibroblast growth factor (bFGF), Brain
derived neurotrophic factor (BDNF), Epidermal growth
J
factor (EGF), Fibroblast growth factor (FGF),
Jak/STAT pathway, 154155
Hepatocyte growth factor (HGF), Insulin-like growth
Joseph Altman, 12, 116
factor (IGF), Nerve growth factor (NGF), Transforming
Junk DNA, 88
growth factor beta (TGF-)
Gsh2, 148
K
Kainate receptors, 122123, 204205
H
Karyotypes, 76, 8182, 248
Hanging drop cell suspension, 119
Karyotypic instability, 76
Hayflick limit, 81
Keratin
HB9, 99, 107
keratin 5, 37
Hedgehog (Hh), 100101, 201202
keratin 14, 37
Hematopoietic stem cells (HSC), 15, 174, 234, 238, 258
Kinetochore-associated mitotic checkpoint machinery, 75
Hensens Node, 56
Hepatocyte growth factor (HGF), 183, 237239
Hes1, 137141 L
Hes3, 137138 L1- enhanced green fluorescent protein (EGFP) transgenic
Hes5, 137138 animal, 89
Hippocampal development, hippocampal formation, 17, 121, L-aromatic amino acid decarboxylase (AADC), 103
123, 256 Lbx1, 105
Hippocampal subgranular zone, 116 L-DOPA, 102103, 207208, 246, 264, 278
Hippocampus, 1314, 1721, 23, 25, 6667, 100, 104, 106, Leukemia inhibitory factor (LIF), 154, 183, 211, 237239,
109110, 123, 149151, 153, 178, 182, 232 259260, 275
HNK1, 52 Lewy Body, Lewy Bodies, 108109, 246
Horizontal basal cells, 37 LeX, 151152
288 Index

LFA-1/ICAM-1 (lymphocyte function-associated Mller glial cells; Mller cells, 146, 149, 161, 176, 193194,
antigen-1/lymphocytes/ intercellular adhesion 200, 202203, 205
molecule-1) adhesion system, 175 Multiple sclerosis (MS), 265
Lhx1, 105 Multipotency, 14, 55, 63, 149, 255266
Lhx2, 38 Multipotent neurospheres, 152, 182
Lhx5, 105 Multipotent somatic stem cell, 257
Lhx6, 105, 109 Muscarinic acetylcholine receptor (mAChR), 121122,
Lhx8, 99, 109 203204
Lim1/2, 105 Myc, 49, 201, 251, 260
LINE-1 (Long Interspersed Nucleotide Elements-1, or L1)
retroelements, 8895 N
Lipid mediators of inflammation, 181, 203 Nanog, 7980, 123, 240
Lmx1a, 99, 103, 108, 247 Necrosis, 2021, 67, 180, 213215, 273275, 278
Lmx1b, 99, 105, 108, 247 Necturus, 48
Lysophosphatidic acid (LPA), 160 Nerve growth factor (NGF), 42, 109, 124, 150, 160, 181182,
Lysosomal storage diseases (LSD), 264 261, 280
Nestin, 60, 6269, 102, 119, 124, 148, 150, 152, 154, 155, 183,
M 210, 238, 240, 260, 276
Macroglia, 146, 172 Neural crest cells (NCC), 4855, 6465, 68
Mad1, 75 Neural crest (NC), 4755, 6465, 68
Mad2, 75, 77 Neural crest (NC) stem cells, 5055
Mad3, 75 Neural fate, 24, 6, 8, 163
Mash1 (mammalian achaete-scute homolog 1), 3739, 105, neural default fate, 3
119, 136139, 247 Neural fold (NF), 4849, 60, 62, 182
Math1, 105, 119 Neural inducer, 2, 4, 68
Math3, 137139 Neural induction, 18, 100, 124, 182, 247
Medial ganglionic eminence (MGE), 105, 109, 148 Neural lineages, 15, 101, 238, 257
Meninges, 172175, 183, 247 Neural plate border, 49
Mesectoderm (ectomesenchyme), 48 Neural primordium, 49, 51, 55
Mesenchymal stem cells (MSC), 4851, 5355, 6162, 64, 80, Neural progenitor cells (NPC), 15, 2122, 24, 54, 74, 76,
108, 118119, 121, 128, 173, 237, 239, 247, 271281 98103, 108, 116122, 128, 135141, 147, 151,
Mesoderm, 2, 5, 4849, 6162, 6466, 100101, 118119, 182183, 194, 200, 212213, 257, 259260, 279
174175, 183, 247, 273, 277 Neural stem cells (NSC), 12, 1416, 21, 2426, 52, 55, 63, 67,
Metabotropic glutamate receptors (mGluR), 122124, 128, 157, 88, 91, 98, 100, 102, 117118, 147152, 163, 182, 211,
204 240, 255266, 276279
Metachromatic leukodystrophy, 174, 183 Neural tube, 48, 5051, 53, 101102, 105, 107, 116117,
1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP), 103, 135136, 141, 147148, 153, 155, 193, 196
247248, 264 Neurite outgrowth, 21, 122125, 159, 265
Meteorin, 153 Neuroblastic layer (NBL), 193, 195196, 198, 202204,
Michael Kaplan, 13, 116 206207, 209, 212, 215, 217
Microenvironment, 25, 121, 149, 151, 179, 182, 184, 194195, Neuro D, 119
258259, 262265, 272, 276277 Neurodegenerative diseases, 2425, 4041, 98, 147, 154,
Microglial differentiation, 176177 163164, 264265
Microglial neurotoxic factors, 179181 Neurofilaments (NF), 52, 60, 62, 66, 68, 122, 183, 200
Microglial precursors, 173176 Neurogenesis, 1126, 3542, 52, 54, 64, 76, 80, 88, 93, 100,
Microglia; microglial cells, 172183, 256 102, 105, 116125, 128, 147154, 182183, 193, 195,
development, 174175, 177178, 183 202204, 232, 256261, 264, 279
origin, 172174 Neurogenic astrocytes, 149152
Mitogens, 39, 5455, 64, 75, 106, 157, 175, 178, 183, 201202, Neurogenic regions, 1317, 19, 147, 152
208, 210211, 259260 subgranular zone of hippocampal dentate gyrus, 13, 16, 17,
MNR2, 107 100, 116, 149, 232, 263
Mobile introns, 94 subventricular zone, 1314, 16, 52, 100, 116, 122123, 149,
Morphogens, 3, 38, 54, 79, 100, 116, 120121, 150, 183, 202, 176, 183, 232, 263, 278
208, 238, 258, 275, 278 Neurogenin (Ngn), 3738, 136
Motor neurons, 98, 101, 105108, 275277 Ngn1, 3739, 105, 137
Mps1, 75 Ngn2, 137, 139141
MS5 cells, 99, 247, 250 Neurological disorders, 19, 102, 119, 128, 147, 158, 163164,
Msx1, 4999, 103, 247 261, 266
Msx2, 49 Neuromodulators, 159, 162, 194, 203210
Neuron-restrictive silencer element (NRSE), 92
Index 289

Neuropeptides, 209210 PA6 cells, 247


Neuropeptide Y (NPY), 20, 104, 209, 210 Paracrine, 24, 120, 127128, 202
Neuroregulin (NRG)-ErbB2 signaling, 151, 153 Parkinsons disease (PD), 18, 98, 128, 245251, 264, 277278
Neurosphere, 14, 25, 41, 101, 119, 121, 123, 125, 151152, Parvalbumin (PV), 25, 104105
182, 200, 202, 232, 238239 Pax2, 105
Neurotensin, 178 Pax3, 49, 278
Neurotransmitter, 13, 20, 25, 53, 101, 104, 107, 115128, 152, Pax6, 39, 99, 101, 105, 107, 109, 119, 148, 151
156158, 163, 194, 203211 Pax7, 49, 105, 278
Neurotransmitter choice, 53, 126 Peripherin, 60
Neurotransmitter receptors, 116, 118, 120, 128, 156, 163 Pet1, 99, 108
Neurotrophic factors, 20, 25, 42, 103, 120, 150, 161, 175, Phenotypic fate, 116, 256
178179, 181182, 200, 211, 247, 260262, 265, Phospholipase C-, 128
276277, 279280 Photoreceptor precursors, 212
Neurotrophin (NT), 42, 106, 161, 175, 181182, 200201, 247, Phox2b, 108
261, 275276, 280 Po Del Ro Hortega, 146
Neurturin, 247 Pitx3, 99, 103, 108, 247
Neutralizer, 154, 177, 261, 274 Platelet-Activating Factor (PAF), 181, 202203, 211
Nicotine, 121122 Pluripotent, 4748, 5053, 55, 77, 80, 98, 100, 106107, 119,
Nicotinic acetylcholine receptor (nAChR), 121122, 203 232, 237239, 240, 251, 257258, 272273
Nkx2. 1, 99, 105, 109 Polydactyly, 202
Nkx2. 2, 107 Primordial germ cells (PGC), 239
Nkx6. 1, 99, 107 Programmed cell death, 19, 193, 195, 212213, 215217
N-methyl-D-aspartic acid (NMDA) receptor, 122123, 125, Progressive lineage restriction, 194
128, 157, 204205 Protein kinase A (PKA), 200201, 211
Noggin, 2, 4, 68, 49, 150151, 249250, 278 Psd-93, 88, 92
Notch, 38, 54, 100, 135141, 150, 153, 155, 183, 197, 201202 Ptf1, 105
Nottebohm, 1213, 19, 116, 149, 256 Purinergic neurotransmission, 124
NSCL2, 119 Purinergic receptor, 116, 124125, 157, 209, 211
NSC-scaffold (neural-stem cell scaffold), 265 Purines, 116, 124126, 128, 157, 209, 211
Nurr1, 99, 103, 247
Q
O Quail brain, 176
Oct-4, 82, 123, 233240 Quail embryo, 49, 51
Odorant receptors, 36, 3839
Olfactory bulb, 13, 16, 18, 3638, 4142, 150, 178, 232, 247, R
256, 259 Radial glial cell; radial glia (RG), 17, 65, 67, 101, 117118,
Olfactory ensheating cells, 37, 4142 125, 135136, 138, 146149, 152155, 164, 195
Olfactory epithelium, 16, 3542 Radial glia (RG)-astrocyte transition, 152
Olfactory sensory neurons, 3537 Radial migration, 176
Olig2, 99, 107, 148, 240 Ramified microglial cells, 172, 178179
Oligodendrocyte (Olg) development; oligodendrocyte Ramon y Cajal, 12, 87, 116, 146, 256
differentiation, 182183, 259 Raphe nuclei, 108
OMP (olfactory marker protein), 3740 RCA-1 (Ricinus communed agglutinin-1), 174
Opioid peptides, 209 Regeneration of damaged neural tissue, 159, 232
Organizer, 2, 47, 100, 103 Regenerative medicine, 49, 232, 261, 263, 276
Oscillatory expression, 139141 Regulation of gliogenesis, 182
OST (olfactory sulfotransferase), 37, 40 Resting microglia (ramified microglia), 178179
Oxidative injury, 179 Retina, 14, 63, 6566, 137, 146, 148149, 161, 163, 175176,
178179, 181, 192213, 215217
P Retinal development, 193194, 197198, 201, 204, 206207,
p16, 75, 77, 198 209, 210
p18, 75, 198 Retinal dystrophies, 192
p19 mouse embryonic carcinoma cell line, 119 Retinal progenitor cells (RPC), 137, 191218
p21, 7578, 198, 211 proliferation, 194, 199203
p27 glial fibrillary acidic protein, GFAP, 7577 survival, 212
P2X receptors, 209 Retinoblastoma (tumor suppression) protein (Rb), 75, 197, 201,
P2Y receptors, 124, 209, 210 214
P2Y1 receptors, 125 Retinoic acid (RA), 38, 101, 119, 150, 176, 211, 276277
P2Y2 receptors, 125 Retinopathy, 203
p53, 7881, 214, 216217 Retrotransposition, 8794
290 Index

Retrotransposon, 89, 9394 Telomerase-deficient mice, 81


Intracisternal A-particle (IAP) retrotransposon, 89 Telomeres, 8081
L1 retrotransposon, 8892, 94 Temporal lobe epilepsy (TLE), 20, 104
Reverse transcriptase (RT), 88, 94 Teratocarcinoma, 98, 119
reverse transcriptase (RT) phylogeny, 94 Teratoma, 248
Rostral migratory stream, 16, 151, 256 Thrombospondin (TSP), 162, 176, 182
RRMP2, 76 Thyroid hormones (TH), 177178
Ryanodine, 121122, 125 TN-C, 148
Totipotent, 257
S Transdifferentiation/cross-differentiation, 15, 55, 194, 258259,
S100, 148, 150152, 154, 156, 161162 272, 276277
Sca-1, 233238 Transforming growth factor beta (TGF-), 3, 151, 154
Schizophrenia, 89, 9394, 102, 104, 108, 163 Tripartite synapse, 161
Secondary germinal zones, 263 Trisomic zygotic rescue, 79
Selfish DNA, 89, 94 Trisomy 21 (Downs syndrome), 7980
Serotonergic, 9899, 108109 Trophic factors in CNS, 150
Serotonin (5-hydroxytryptamine, 5-HT), 208 3-tubulin, 123
Smad1, 78, 137 Tumor, 13, 2021, 24, 6769, 7475, 8081, 98100, 119, 180,
Somatostatin (SST), 104, 209 197, 200202, 214, 248251
Sonic hedgehog (Shh), 38, 54, 102, 119, 201, 247 Tyrosine hydroxylase (TH), 102, 126, 207, 247, 264
Sox2, 67, 88, 91, 149150, 152, 240, 251
Sox3, 67 U
Sox9, 4950, 54, 183 Unorganized chromatin (euchromatin), 233
Spinal cord injury (SCI), 42, 98, 261264, 271281 Untranslated region (UTR), 9092
Spinal cord lesions, 98
Spinal muscular atrophy (SMA), 105 V
Spindle assembly checkpoint (SAC), 75 Vasoactive intestinal peptide (VIP), 104, 209
SSEA-1, 82, 233234, 239 Very small embryonic like stem cells (VSELs), 233241
SSEA-4, 233 Vesicular monoamine transporter 2 (VMAT2), 103
Stat3, 141, 183, 202, 211, 276 Vimentin, 6063, 6668, 148150, 158
Stem cell therapy, 24, 107, 264265, 276, 281 Vitamin B12, 247
Stemness genes, 260261 Vitronectin receptor, 180
Stroke, 14, 174, 183, 232, 238241, 265266, 276, 278 VSEL-derived spheres, 239
Stromal derived factor-1 (SDF-1), 22, 237240
Subgranular zone (SGZ), 13, 1617, 100, 116, 149, 232, 263
W
Substantia nigra (SNi), 18, 102, 246247, 264, 278
Wnt, 8, 49, 54, 99100, 102103, 106, 119, 151, 153, 201202,
Subventricular zone (SVZ), 1314, 16, 52, 100, 116, 122123,
250
149, 176, 183, 232, 263, 278
Wnt/catenin signaling, 54, 153
Syncoilin, 60
Synemin, 60, 6268
X
Xenopus laevis, 2, 49
T
Tangential migration, 176
Target Primed Reverse Transcription (TPRT), 91 Z
Telomerase, 8081, 247, 261 Zic1, 49

Вам также может понравиться