Вы находитесь на странице: 1из 23

Acta Neuropathol

DOI 10.1007/s00401-016-1645-y

REVIEW

Towards a functional pathology ofhereditary neuropathies


JoachimWeis1 KristlG.Claeys1,2,3 AndreasRoos1,4 HamidAzzedine1
IstvanKatona1 J.MichaelSchrder1 JanSenderek5

Received: 12 May 2016 / Revised: 10 November 2016 / Accepted: 13 November 2016


Springer-Verlag Berlin Heidelberg 2016

AbstractA growing number of hereditary neuropathies


have been assigned to causative gene defects in recent
years. The study of human nerve biopsy samples has contributed substantially to the discovery of many of these
neuropathy genes. Genotypephenotype correlations based
on peripheral nerve pathology have provided a comprehensive picture of the consequences of these mutations.
Intriguingly, several gene defects lead to distinguishable
lesion patterns that can be studied in nerve biopsies. These
characteristic features include the loss of certain nerve
fiber populations and a large spectrum of distinct structural
changes of axons, Schwann cells and other components of
peripheral nerves. In several instances the lesion patterns
are directly or indirectly linked to the known functions of
the mutated gene. The present review is designed to provide an overview on these characteristic patterns. It also
considers other aspects important for the manifestation and
pathology of hereditary neuropathies including the role of

* Joachim Weis
jweis@ukaachen.de
* Jan Senderek
Jan.Senderek@med.unimuenchen.de
1

Institute ofNeuropathology, RWTH Aachen University


Medical School, Pauwelsstr. 30, 52074Aachen, Germany

Department ofNeurology, RWTH Aachen University


Medical School, Pauwelsstr. 30, 52074Aachen, Germany

Department ofNeurology, University Hospitals Leuven


andUniversity ofLeuven (KU Leuven), Leuven, Belgium

Leibniz-Institut fr Analytische Wissenschaften-ISAS-e.V.,


OttoHahnStr. 6b, 44227Dortmund, Germany

FriedrichBaurInstitute, Department ofNeurology,


Ludwig-Maximilians-University, Ziemssenstr. 1a,
80336Munich, Germany

inflammation, effects of chemotherapeutic agents and alterations detectable in skin biopsies.


Keywords CharcotMarieTooth disease (CMT)
Hereditary sensory and motor neuropathy (HMSN)
Hereditary motor neuropathy (HMN) Hereditary sensory
and autonomic neuropathy (HSAN) Inflammatory
neuropathy Chemotherapy

Introduction
An effective approach to unravel disease mechanisms is to
link naturally occurring pathogenic mutations with monogenic disorders. Indeed, a mutant gene that gives rise to a
rare monogenic disorder usually also provides an opportunity to understand the genetic, biochemical, signal transduction and developmental network that underlies similar
common polygenic or acquired phenotypes. One striking
example of how the identification of genes for Mendelian disorders influences our views on the pathophysiology of human disease is hereditary peripheral neuropathy.
The Neuromuscular Home Page of the Neuromuscular
Disease Center, Washington University, St. Louis, MO
(neuromuscular.wustl.edu) currently lists more than 200
forms of hereditary neuropathies. Most of these have
already been assigned to distinct disease genes. Many
neuropathy genes are linked to peripheral nerve development, function or maintenance. The gene discoveries
often supported or extended existing or inferred concepts
of disease such as alterations in myelin development and
structure, length-dependent impairment of axonal transport or defects of the axoskeleton. Other disease genes
unraveled previously unrecognized and unexpected neuropathy mechanisms including nuclear envelope structure,

13

mitochondrial dynamics, protein biosynthesis, Ca2+homeostasis, phosphoinositide metabolism, Rho GTPase


signalling and transcriptional regulation (Table1; Fig.1)
[9, 70, 114, 167].
The wide array of neuropathy genes identified so far
illustrates which structural and functional components are
required for building and maintaining the human peripheral nervous system. The study of nerve pathology provides
a unique opportunity to decipher the functions of these
genes and the pathomechanisms related to their mutations.
It establishes genotypephenotype correlations and yields
information about concomitant diseases such as inflammation or microangiopathy that might contribute to the neuropathy phenotype. It also provides a framework for the
interpretation of the consequences of these gene defects in
cell culture and animal models. Moreover, studying biopsies from patients with defined genetic defects gives clues
to the function of the affected genes and their homo- and
paralogs also in the human central nervous system (CNS),
with the additional advantage that nerve and skin biopsies
provide well-preserved biopsy tissue, whereas material
unaffected by autolysis is hardly available for most degenerative human CNS disorders.
The present review focuses on the histological and ultrastructural human nerve biopsy patterns related to hereditary
neuropathy entities and gene defects. These patterns are
discussed in the context of our current understanding of
the functions of the affected genes (and encoded proteins).
In addition, we consider the consequences of mutations
in more than one neuropathy gene in the same individual,
of concomitant inflammation and of confounding factors,
especially chemotherapy, that influence the manifestation
and pathology of hereditary neuropathies.

Clinical aspects andclassifications ofhereditary


neuropathies
Peripheral neuropathy is one of the most common reasons for referrals to neurological clinics [91]. While most
cases are acquired (due to diabetes mellitus, alcoholism or
inflammatory diseases), the prevalence of the largest group
of hereditary neuropathies, CharcotMarieTooth neuropathies (CMT, also called hereditary motor and sensory
neuropathies, HMSN), is about 1:2500 in Northern Europe
[101, 152]. Hereditary neuropathies are classified as CMT
(or HMSN) if motor and sensory systems are both affected,
as hereditary sensory and autonomic neuropathy (HSAN)
if sensory deficits and/or autonomic dysfunctions prevail
and as distal hereditary motor neuropathy (HMN) if the
motor deficit is predominant [38, 57]. Spinal muscular atrophy (SMA) has been clinically and morphologically classified as a pure lower motor neuron disease; however, the

13

Acta Neuropathol

terms distal SMA (DSMA) and HMN are now used largely
interchangeably.
According to electrophysiological criteria, CMT neuropathies are classified into two main subgroups: the demyelinating type CMT1 (or HMSN I) with reduced nerve
conduction velocities (NCV), and the axonal type CMT2
(or HMSN II) with primarily axonal loss characterized by
normal or only slightly reduced NCV, but decreased amplitudes of motor and sensory nerve action potentials [38, 57].
In practical terms, a motor NCV of 38m/s in upper limb
nerves is commonly used as a threshold for differentiating CMT1 from CMT2. As a rule, histopathological studies reveal segmental demyelination and remyelination with
prominent onion bulb formation in CMT1, while in CMT2
there are signs of axonal degeneration with regenerative
sprouting.
An intermediate CMT subgroup with median MNCV
between 25 and 45m/s has been defined [33, 107]. DejerineSottas syndrome (DSS) or HMSN III is an early-onset,
clinically severe, demyelinating motor and sensory neuropathy with MNCV below 12m/s and marked onion bulb
formation and can, thus, be considered as a severe form
of CMT1 [119]. Further subdivisions of CMT neuropathies are based on the inheritance pattern and the molecular genetic defects. Inheritance in CMT can be autosomal
dominant (AD), autosomal recessive (AR) or X-linked.
By convention, autosomal recessive forms of CMT1 are
sometimes referred to as CMT4 and autosomal recessive
forms of CMT2 are also known as AR-CMT2. Autosomal
dominant and autosomal recessive forms of intermediate
CMT have been named CMTDI and CMTRI, respectively.
Finally, X-linked CMT has also been designated as CMTX.
A modification of the current nomenclature of CMTs and
related disorders which takes into account the causative
gene defects, inheritance patterns and primary pathological phenotype (axonal, demyelinating, or intermediate) has
been proposed recently [94].
Mutations in the same gene may lead to dominant or
recessive inheritance patterns, exemplified by the recent
discovery of MME mutations as a cause of both, autosomal dominant and autosomal recessive CMT2 [6, 60]. In
addition, the clinical spectrum of neuromuscular disorders
associated with certain disease genes may be wider than
initially anticipated. For example, mutations in the gene for
glycyl-tRNA synthetase (GARS) may result in CMT2 and
HMN/DSMA [2, 37], and mutations in the TRPV4 gene
encoding an ion channel protein may cause CMT2, congenital non-progressive DSMA or scapuloperoneal SMA
(SPSMA) [5]. In addition, certain disease genes (e.g., MPZ
or GJB1, see below) may be associated with both axonal
and demyelinating neuropathies.
Although CMT neuropathies are considered non-syndromic conditions, additional involvement of other organs

Acta Neuropathol
Table1Neuropathy genes
classified by their known or
inferred functions

1. Myelin proteins
PMP22

CMT1A, CMT1E, HNPP, DSS, CHN

MPZ/P0

CMT1B, DSS, CHN, CMT2I, CMT2J, CMTDID

2. Cytoskeleton, nucleoskeleton and nuclear envelope


NEFL

CMT1F, CMTDI, CMT2E

LMNA
CCT5

AR-CMT2A (CMT2B1)a,b
HSAN with spastic paraplegia

INF2

CMTDIEb

DST
3. Transcriptional regulation

HSAN7b

EGR2

CMT1D, DSS, CMT4E, CHN

SOX10

MORC2

Peripheral demyelinating neuropathy, central


dysmyelination, Waardenburg syndrome, and
Hirschsprung disease (PCWH)b
CMT2Z

IGHMBP2

CMT2S, SMARD1 (HMN6)

DNMT1
IKBKAP

HSAN1Eb
HSAN3 (RileyDay syndrome)

SETX

HMNa

PRDM12

HSAN8

HINT1

CMT2 (neuromyotonia and axonal neuropathy)

MED25
4. Protein biosynthesis

AR-CMT2B (CMT2B2)b

GARS

CMT2D, HMN5A

AARS
YARS

CMT2Nb
CMTDIC

KARS

CMTRIB, HNPPb

MARS
5. Protein modification, folding and degradation

CMT2Ub

HSPB1

CMT2F, HMN2B

HSPB3

HMN2C

HSPB8

CMT2L, HMN2A

LRSAM1

CMT2P

TRIM2

CMT2R

HSJ1

DSMA5, CMT2

GAN

Giant axonal neuropathy 1

6. Intracellular transport
GJB1

CMTX1

SH3TC2/KIAA1985

CMT4C

RAB7

CMT2B, HSAN1

DNM2
DYNC1H1

CMT2M, CMTDIBa
CMTO, SMALED1b

DCTN1

HMN7Ba,b

KIF1A

HSAN2Ca,b

LITAF

CMT1C

NDRG1

CMT4D/HMSNL

KIAA1840
BICD2

CMT2Xa
SMALED2

7. ER membrane shaping
FAM134B

HSAN2B

ATL1

HSAN1Da

13

Acta Neuropathol

Table1continued

ATL3

HSAN1F

TFG

CMT2a
CMT2, HMN5Aa,b

BSCL2
8. Mitochondrial dynamics
MFN2

CMT2A, HMSN5A, HMSN6, CMT2A2B

GDAP1

CMT4A, CMT2K, CMTRIA

9. Mitochondrial energy production


DHTKD1
PDK3

CMT2Qb
CMTX6

HK1

CMT4G/HMSNR

10. Sphingolipid biosynthesis


SPTLC1

HSAN1A

SPTLC2

HSAN1C

11. Phosphoinositide metabolism


MTMR2

CMT4B1

SBF2/MTMR13

CMT4B2

SBF1/MTMR5

CMT4B3
CMT4Ja,b

FIG4
12. Rho GTPase signalling
PLEKHG5

DSMA4, CMTRIC

ARHGEF10

Slow nerve conduction velocities, thin myelin

FGD4

CMT4H

13. Interaction with the extracellular environment


FBLN5
MME

CMT1b
CMT2T

GNB4

CMTDIF

NGF

HSAN5

NTRK1
DHH

CIPA/HSAN4, HSAN5b
46,XY partial gonadal dysgenesis with minifascicular neuropathyb

WNK1

HSAN2Ab

PRX

CMT4F, DSS

14. Ion channels


CMT2C, SPSMA, congenital non-progressive
DSMA (HMN8)b
CIP, HSAN2D, primary erythermalgia, PEPDb

TRPV4
SCN9A
15. Various
PRPS1 (purine metabolism and nucleotide biosynthesis)

CMTX5b

AIFM1 (mitochondrial-mediated apoptosis)

CMTX4b

SMARD1 spinal muscular atrophy with respiratory distress type 1, SMALED1 spinal muscular atrophy with
lower extremity predominance type 1, SMALED2 spinal muscular atrophy with lower extremity predominance 2, CIPA congenital insensitivity to pain with anhidrosis, CIP congenital insensitivity to pain, PEPD
paroxysmal extreme pain disorder
a

Mutations in this gene can also cause other neuromuscular disorders

Mutations in this gene have been related to non-neuromuscular conditions

such as CNS, eyes, skeletal muscle, etc. may be observed


[94]. Apart from this, a large, heterogeneous group of other
hereditary diseases feature peripheral neuropathy as a prominent or prevailing component of the clinical picture. These

13

conditions include familial amyloid neuropathy (FAP), giant


axonal neuropathy (GAN), polyglucosan body disease, and
neuropathies associated with metabolic disorders such as
metachromatic leukodystrophy and Refsum disease.

Acta Neuropathol
Hereditary neuropathies: Genes and pathomechanisms

Nuclear envelope
(LMNA)

Nucleotid biosynthesis
(PRPS1)

Intracellular transport,
endosomes
(SH3TC2, RAB7)

Rho GTPase signalling


(PLEKHG5, ARHGEF10,
FGD4)

Heat shock proteins


(HSPB1, HSPB8,
HSJ1)
Protein degradation
(LRSAM1, TRIM2,
GAN)

Transcriptional regulation
(EGR2, SOX10, SETX)

Myelin proteins
(PMP22, MPZ, Cx32)

Sphingolipid
biosynthesis
(SPTLC1, 2)
Phosphoinositide
metabolism
(MTMR2, SBF2)

Amino acid processing


(GARS, YARS, AARS,
MARS, KARS)

ER membrane shaping
(FAM134B, ATL1,
ATL3, TFG, BSCL2)

Cytoskeleton
(NEFL, Gigaxonin)

Axonal transport
(DCTN1, DYNC1H1)

Mitochondrial dynamics
(GDAP1, MFN2)
Energy production
(DHTKD1, PDK3, AIFM1
HK1)

Extracellular matrix interaction


(PRX, GNB4)

Fig.1Hereditary neuropathies: genes and pathomechanisms (based on Weis and Senderek, 2014 [180]

Neuropathy genes andpathology patterns


The following chapters provide an overview of distinct
forms of inherited neuropathies and their causative gene
defects that lead to prototypical morphological changes
detectable in nerve and skin biopsies.
Selective lack, loss, or preservation ofcertain nerve
fiber populations insensory nerves
MFN2, MME, KIAA1840 and MORC2 mutations leading
to CMT2 as well as mutations in the BSCL2 gene leading
to distal hereditary motor neuropathy predominantly affect
large myelinated fibers in the sural nerve [6, 28, 60, 100,
149, 166] (Fig.2b). For two cases of CMT2 due to MORC2
mutation, a peculiar patchy, focally accentuated loss of
nerve fibers has been described [149]. Prominent loss of
large and small myelinated nerve fibers along with relative
preservation of unmyelinated axons has been reported in
neuropathies caused by myelin protein zero (MPZ) [163],
Gap junction beta-1 protein/connexin-32 (GJB1) [59] and
Lamin A/C (LMNA) gene mutations [158]. Slowly progressive loss predominantly of large sensory axons with
preservation of the small myelinated and unmyelinated
fibers is a characteristic feature of Friedreichs ataxia due
to mutations in the FXN gene for the iron-binding protein

frataxin [68, 128] (Table2). A similar pattern of preferential degeneration of large myelinated nerve fibers has been
found in patients with posterior column ataxia and retinitis pigmentosa caused by mutation of the feline leukemia
virus subgroup C cellular receptor (FLVCR1) gene, which
is involved in iron homeostasis [122].
Subtotal loss or lack of myelinated fibers in a sural nerve
biopsy with remarkable preservation of unmyelinated axons
has been described by one of us (JMS) in a sporadic case
of sensorymotor neuropathy [138]. Prominent loss of large
and small myelinated fibers restricted to the sensory system combined with a relative preservation of unmyelinated
nerve fibers is seen in HSAN1 due to SPTLC1 mutations
and in HSAN2A caused by WNK1 gene defects [64, 181].
All nerve fiber populations in the sural nerve are equally
reduced in number in the HSANs due to ATL1, ATL3 [54],
RAB7 [65], DNMT1 [10] and FAM134B [78] gene mutations. Incomplete development and/or early degeneration
mainly of small myelinated sensory axons and of unmyelinated nerve fibers is found in HSAN III (familial dysautonomia; Riley-Day syndrome; inhibitor of kappa light
polypeptide gene enhancer in B-cells, kinase complexassociated protein (IKBKAP) gene mutations) and in neuropathies caused by mutations in the genes encoding nerve
growth factor (NGF), its high affinity receptor neurotrophic
tyrosine kinase receptor type 1 (NTRK1) [116, 181] and of

13

Acta Neuropathol

Fig. 2a Myelinated nerve


fibers in the normal sural nerve.
Semithin section, toluidine blue;
scale bar 12m. b Loss of large
myelinated fibers in CMT2 in a
case with a mutation in MFN2,
showing only few remaining
large fibers (arrows). Semithin
section, toluidine blue; scale
bar 12m. c Normal myelinated and unmyelinated (lower
right corner) nerve fibers. EM;
scale bar 2m. d Schwann cell
processes forming empty collagen pockets indicative of the
loss of non-myelinated fibers
in a case of CMT1 due to MPZ
mutation; scale bar 1m

Table2Preferential loss of sural nerve fiber populations in hereditary neuropathies


Affected gene

Large myelinated nerve fibers

Small myelinated nerve fibers

Unmyelinated nerve fibers

MFN2, MME, KIAA1840, MORC2

+++

++

+++

+++

+++

+++

MPZ, GJB1, LMNA


SPTLC1, WNK1
ATL1, ATL3, RAB7, DNMT1, FAM134B
TTR
BSCL2
IKBKAP, NGF, NTRK1, PRDM12

+++

+++
++

++
+

+++

+++

+++

++

+++

+++, severe reduction; ++, moderate reduction; +, minor reduction; , no reduction; ?, no information found

transcriptional regulator PR/SET domain 12 (PRDM12)


[29]. Predominant loss of small myelinated and of unmyelinated nerve fibers is also a distinctive feature of early
stages of hereditary amyloid neuropathy due to transthyretin (TTR) mutations [118].
Alterations ofunmyelinated nerve fibers
As mentioned above, unmyelinated sensory and autonomic
nerve fibers are reduced in number or absent along with
myelinated sensory nerve fibers in several forms of HSAN,
whereas motor axons are largely preserved in this group of
neuropathies. Empty Remak bundles showing complete

13

or partial loss of unmyelinated nerve fibers are often associated with increased numbers of so-called empty collagen pockets and indicate degeneration of unmyelinated
fibers (Fig.2d; Table3). In these structures, processes of
non-myelinating Schwann cells do not wrap axons, but
bundles of collagen fibers instead. Abnormally extended
and branched processes of Schwann cells of unmyelinated
nerve fibers are a prominent and characteristic feature of
CMT4C due to SH3TC2 (KIAA1985) mutations [44, 145]
(Fig. 6a). They have also been described in intermediate
CMT due to INF2 mutations [19]. However, the pathology
of unmyelinated nerve fibers in most hereditary neuropathies is still largely unexplored.

Acta Neuropathol
Fig. 3a Longitudinal section of a normal myelinated
axon showing low density of
organelles. EM; scale bar 2m.
b Focal axonal accumulation
of mitochondria and other
degenerating organelles in a
myelinated nerve fiber in a case
of NEFL neuropathy (from
Elbracht etal. Clin Neuropathol
2014 [39], with permission);
scale bar 2m. c Prominent
axoplasmic reticulum profiles
(arrows) in INF2 neuropathy;
scale bar 2m. d Giant axons
(arrows) in giant axonal neuropathy (GAN mutation); scale
bar 20m

Alterations ofmotor nerve fibers


Selective degeneration of spinal motor neurons and/or their
axons is a hallmark of spinal muscular atrophies (SMAs)
due to mutation of the RNA binding survival of motor neuron protein (SMN) gene and of the golgin and motor-adaptor protein bicaudal D homolog 2 (BICD2) gene [58, 106,
111, 127]. There can be concomitant, but clinically less
apparent or subclinical involvement of other neuronal cell
populations such as sensory neurons [58]. Histopathological nerve fiber alterations in hereditary motor neuropathies
(HMNs) are largely unexplored due to the limited availability of appropriate tissue specimens.
Focal axonal alterations
Focal accumulations of cytoskeletal elements and organelles as well as autophagic vacuoles are found in several
axonal neuropathies including neuropathies due to MFN2
[166, 169] and NEFL (Fig.3b) mutations [39, 40, 186].
Abnormally positioned, elongated and/or distorted mitochondria are especially frequent in patients with MFN2
mutations (see below); actually, intraaxonal accumulations

of degrading organelles, abnormal mitochondria and


cytoskeletal components as well as abnormally large clusters of regenerating axons should alert to the possibility
of CMT2 [14, 136, 137, 163, 167, 169]. Accumulations
of abnormal mitochondria in axons as well as Schwann
cells have also been reported in mitochondriopathies due
to mitochondrial and nuclear gene defects (for review, see
[172]. Large intraaxonal accumulations of mitochondria,
dense bodies, glycogen granules, neurofilaments and membranotubular profiles are a hallmark of neuroaxonal dystrophies [52, 77, 112, 182]. These so-called spheroids stain
positive for neurofilament, ubiquitin, and alpha-synuclein
[77].
Similar accumulations of neurofilaments leading to an
enlargement of axons are also encountered in neuropathies caused by neurofilament light-chain protein (NEFL)
gene mutations (CMT1F, CMT2E, DI-CMT) [39, 40, 186].
They are even more prominent (Fig.3d) in giant axonal
neuropathy due to GAN mutation [4, 71]. Gigaxonin, the
protein encoded by GAN, is an E3 ligase substrate adaptor and thus is believed to be involved in proteasomal
degradation and/or turnover of intermediate filament proteins (reviewed in [71]). PNS and CNS axons of affected

13

Acta Neuropathol

Fig. 4a Large polyglucosan body in a myelinated axon. Semithin


section, toluidine blue; scale bar 10m. b Electron micrograph
revealing the filamentous ultrastructure of the polyglucosan body;
scale bar 1m. c Lack of myelin around larger diameter axons in
a case of congenital amyelination due to a SOX10 mutation [115].
Note that two of the axons (asterisks) show a one to one relationship
with Schwann cell nuclei, indicating that these axons do not belong

to Remak bundles. EM; scale bar 3m. d Increased nodal width in


CMT1 due to an MPZ mutation. EM; scale bar 3m. e Segmental
demyelination in a sural nerve biopsy of a patient with an MPZ mutation. EM; scale bar 2m. f Segmental demyelination followed by
thin remyelination in a patient harbouring a LITAF mutation; arrow
focal myelin folding close to the node of Ranvier. Semithin section,
toluidine blue; scale bar 10m

patients show marked swellings arranged like pearls on


a string; within these swellings, neurofilaments may lack
visible side arms and show dense packing with incomplete
whorl-like arrangement [71]. Organelles and microtubules
are often displaced; autopsy studies revealed a loss of cell
bodies of the affected neuronal populations [71]. Interestingly, altered neurofilament phosphorylation and spacing is
also observed in peripheral nerves of patients with neurofibromatosis type 2 due to mutations of the NF2 gene [144]
and in common CNS conditions associated with axonal
degeneration including Alzheimers and Parkinsons disease [117].
Polyglucosan bodies (Fig.4a, b) are basophilic, strongly
PAS-positive inclusions with a diameter of 10100m.
They are composed of fine branching filaments as well as
granular and amorphous material [82]. In a large series of
sural nerve biopsies, polyglucosan bodies were present in
8.5% of cases [22]. Their frequency was related to age, and
the authors concluded that the presence of a few polyglucosan bodies in sural nerves is a rather common finding

[22]. Intraaxonal polyglucosan bodies are increased in


number in adult polyglucosan body neuropathy and type IV
glycogen storage disease (GSD IV), both due to mutations
of the glycogen branching enzyme 1 encoded by the GBE1
gene. In these disorders, polyglucosan bodies are present
at a high frequency in CNS neurons as well as astrocytes
and may also be found in Schwann cells, muscle fibers and
other cell types [82, 83, 110, 124, 179].

13

Alterations ofnerve fiber regeneration


Regenerating nerve fibers sprout within bands of Bngner
[135]. Such clusters are seen in nerve biopsies in many
cases of hereditary neuropathy and have been observed at
particularly high frequency in certain neuropathies including CMTX1 due to GJB1 mutations [171] as well as
CMT2A due to MFN2 mutations [32]. Absence of regenerative nerve fiber clusters has been described in several
hereditary neuropathies including cases of neuropathy due
to LMNA mutations [34].

Acta Neuropathol

Lack or early loss ofmyelin

Focal Schwann cell changes

CMT1A. Tomacula become apparent both in semithin sections and in teased nerve fiber preparations and are thought
to be caused by slippage and redundant folding of myelin
loops [86]. Less regular outfoldings of the myelin sheath
are referred to as focally folded myelin (FFM) and may
lead to a globular appearance of myelinated nerve fibers
(Fig.5e, f). Such alterations may occur in CMT1A and B,
but they are especially prominent and considered to be a
characteristic feature of several autosomal recessive forms
of CMT due to mutations in the myotubularin-related protein 2 (MTMR2; CMT4B1), SET binding factor 1 and 2
(SBF1 and SBF2/MTMR13; CMT4B3 and CMT4B2), and
periaxin (PRX; CMT4F) genes [7, 55, 66, 104, 146].
Lack or loss of compaction of the layers of the myelin sheath leads to uncompacted or decompacted myelin
(Fig. 5c). These alterations usually affect the major dense
line of outer myelin layers corresponding to the cytoplasmic component. They are encountered in CMT1B [47]
and in several forms of demyelinating autosomal recessive
CMT (CMT4). A similar pattern, affecting the intermediate
line which corresponds to the original extracellular space,
may be caused by deposition of immunoglobulins between
the myelin layers in disorders associated with monoclonal
gammopathy [165].
Abnormal widening and extensions of the adaxonal
Schwann cell cytoplasm and other abnormalities of the
innermost layers of the Schwann cell plasma membranes
are characteristic for the X-chromosomal form of CMT
(CMTX) due to mutation of the GJB1 gene encoding the
connexin-32 protein [140] (Fig.5d).

De andremyelination

Schwann cell onion bulb formations

Segmental demyelination is a frequent finding in CMT of


the demyelinating type, CMT1. It is defined as the loss of
myelin in one or several internodes, the territory of one
myelinating Schwann cell between two nodes of Ranvier,
and is often followed by remyelination. The process usually starts at the node of Ranvier (paranodal demyelination)
and involves activation of monocytes/macrophages as well
as fibroblasts (for review, see [136]). Regenerated myelin
sheaths of remyelinated nerve fibers usually do not reach
the thickness of the preexisting myelin, making them readily identifiable in teased fiber preparations and in longitudinal semithin sections for long periods of time after active
demyelination has taken place (Fig.4e, f).

In many demyelinating neuropathies including CMT1A


and 1B, myelinated and demyelinated nerve fibers are surrounded by one or several layers of abnormal, flattened,
degenerating Schwann cells and their processes (Fig.6).
These so-called Schwann cell onion bulb formations are
thought to result from an initial proliferation of Schwann
cells that try to remyelinate a demyelinated axon. Because
only one of them subsequently captures the internode, the
others recede to the periphery [14]. Several cycles of deand remyelination as well as ample deposition of collagen
and other extracellular matrix components between the
Schwann cell layers can lead to an impressive size of the
onion bulbs [141], which has led to the designation complex onion bulbs in certain cases [88, 89, 175]. During
these cycles, Schwann cell basal laminae often detach and
can form redundant layers. In the extreme form, the onion
bulbs largely or exclusively consist of left over basal lamina
and other extracellular matrix components [14]. As mentioned above, such basal lamina onion bulb formations are

Congenital amyelination or hypomyelinating neuropathy


is characterized by complete or subtotal lack of myelin as
well as other Schwann cell abnormalities. Such patterns are
observed in neurocristopathies caused by mutations in the
gene for the transcription factor SOX10 (Fig.4c) [115] and
in neuropathies due to certain mutations in myelin protein
zero (MPZ), peripheral myelin protein 22 (PMP22) and
early growth response 2 protein (EGR2) genes [18, 167].
Absent myelination may be associated with increased numbers of Schwann cells around the naked axons. Subsequent
degeneration of these Schwann cells leads to the formation
of onion bulbs of Schwann cell processes and their basal
laminae (basal lamina onion bulbs) around these axons
[42, 56].
The congenital cataracts, facial dysmorphism and neuropathy (CCFDN) syndrome due to mutations in the C-terminal domain phosphatase of RNA polymerase II, subunit
A (CTDP1) gene [168] is associated with hypomyelination;
in older CCFDN patients, de- and remyelination as well as
axonal degeneration is observed [160]. At the mild end of
the phenotypic spectrum, individuals with asymptomatic
congenital hypomyelination due to a single Rho guaninenucleotide exchange factor 10 (ARHGEF10) gene mutation
show disproportionately thin myelination, slowed nerve
conduction velocities, moderately reduced axon numbers
and mild denervation of muscles [170].

Tomacula andother focal myelin alterations


Concentric and eccentric myelin thickening associated with
axonal constriction in so-called tomaculous fibers (Fig.5a)
is especially frequent in HNPP, but may also occur in

13

Acta Neuropathol

Fig. 5a Concentric myelin thickening (tomaculum) in HNPP. Semithin section, toluidine blue; scale bar 15m. b Concentric accumulation of Schwann cell basal laminae (basal lamina onion bulb; arrows)
at a node of Ranvier in CMT due to MPZ mutation. EM; scale bar
3m. c Incomplete compaction of the innermost myelin layers in
the same case. EM; scale bar 2m. d Accumulation of membranous

material at the axon-Schwann cell interface in CMTX1 due to GJB1


mutation. EM; scale bar 2m. e Cross and f longitudinal sections of
a sural nerve biopsy of a patient with SFB2 mutation showing marked
focal myelin folding. The axon is labelled with an asterisk. Arrow
node of Ranvier. EM; scale bar in e 2m, in f 5m

a frequent feature of congenital hypomyelination neuropathy (CHN), DSS and autosomal recessive CMT (CMT4)
[44] (Fig.5b).
At the other end of the spectrum, the onion bulbs may
consist of numerous vital Schwann cells. Such formations
are a frequent feature of neuropathies in neurofibromatosis,
especially neurofibromatosis type 2 [155]. In nerve fascicles of these patients, Schwann cells may also be grouped
in clusters not showing onion bulb architecture. The
Schwannoid cells within these so-called tumorlets often
show dysmorphic features such as altered nuclear morphology and nucleuscytoplasm ratios [155, 177]. In addition,
nerve biopsies of patients with neurofibromatosis type 2
may reveal atypical onion bulb formations as illustrated in
Fig.6c.

Other Schwann cell alterations inhereditary neuropathies

13

Abnormal cytoplasmic accumulations of beta-actin have


been observed in Schwann cells in nerve biopsies of patients
with CMTDIE due to INF2 mutation [93]. Neurolipidoses
such as sphingolipidoses and gangliosidoses as well as
certain leukodystrophies lead to characteristic inclusions
detectable by electron microscopy in Schwann cells and
other cell populations. These deposits includeamong
otherstuff stone bodies in metachromatic leukodystrophy, lipid inclusions in NiemannPick type A and Tangier
disease, banana bodies within myelinating and non-myelinating Schwann cells in Farber disease and bilaminar or
trilaminar structures composed of linear profiles in adrenoleukodystrophy/adrenomyeloneuropathy [14, 50, 113].

Acta Neuropathol
Fig. 6a Schwann cell onion
bulb (black arrow), abnormal
processes of non-myelinating
Schwann cells (arrowheads)
and Schwann cell basal lamina
proliferation (white arrow) in
CMT4C due to SH3TC2 mutation. EM; scale bar 2m. b
Semithin section of a CMT1A
sural nerve biopsy showing
Schwann cell onion bulbs (black
arrows) around normally and
thinly myelinated axons as well
as obsolete onion bulbs without
axons (white arrows). Toluidine blue; scale bar 10m. c
Atypical onion bulb formations with (black arrow) and
without (white arrow) central
myelinated axon in a case of
neurofibromatosis type 2 and
peripheral neuropathy due to
NF2 mutation. Semithin section,
toluidine blue; scale bar 15m

Secondary axonal degeneration


In advanced stages of most predominantly demyelinating
neuropathies, axonal loss is encountered in nerve biopsies.
It is generally considered a reactive phenomenon resulting
from secondary degeneration of the axons that have been
stripped of their protective Schwann cell covering and
may be accompanied by signs of nerve fiber regeneration.
Axonal degeneration after formation of onion bulbs results
in a denervated or obsolete onion bulb [99]. Moreover,
unmyelinated axons may be present in the Schwann cell
layers of onion bulbs which might originate from sprouting of the parent central axon within the bulb and/or belong
to neighboring unmyelinated nerve fibers that have been
incorporated into the bulb [99]. Pseudo-onion bulbs are
defined as clusters of regenerating myelinated and unmyelinated nerve fibers that are surrounded by Schwann cell
processes, adopting an onion bulb-like shape [99]. They
correspond to later stages of axonal regeneration within a
band of Bngner (Table3).

Alterations offascicular architecture


andof the neural connective tissue
Hypoplasia of perineurial structures associated with amyelination has been observed as a consequence of mutation
of the differentiation gene SOX10 [115]. A surplus of fascicles (minifascicular neuropathy; similar to minifascicle
formation in neuromas [139]), is caused by mutation of the
desert hedgehog (DHH) gene in patients with 46,XY partial
gonadal dysgenesis [102, 120, 130, 159, 162, 176]. Multiple mucosal neuromas are a regular feature of multiple
endocrine neoplasia type 2B (MEN2B) due to mutations
of the RET protooncogene, but have also been described in
a patient with normal RET gene lacking other features of
MEN2B [120]. These patterns have to be discerned from a
surplus of perineurial cells in the context of perineurioma.
This neoplastic lesion of peripheral nerves is characterized
by onion bulb formations composed of cells with perineurial differentiation including epithelial membrane antigen
(EMA) immunoreactivity [130]. Few cases of multiple

13

Acta Neuropathol

Table3Summary of characteristic nerve pathologies in hereditary neuropathies


Characteristic nerve fiber changes in hereditary neuropathies

Prototypic gene defects/disorders

Preferential loss of certain nerve fiber populations


Axonal alterations
Altered neurofilament phosphorylation and spacing
Accumulations of neurofilaments/giant axons

See Table2
Neurofibromatosis type 2
NEFL, GAN

Spheroids

Neuroaxonal dystrophy: PLA2G6, C19ORF12

Polyglucosan bodies

GBE1

Mitochondrial abnormalities, focal axonal accumulations


of mitochondria and of other organelles
Axonal atrophy
Particularly large clusters of regenerating nerve fibers

MFN2, GDAP1

Absence of regenerative nerve fiber clusters

LMNA

Myelin/Schwann cell alterations


Absence of myelin
Widening of nodes of Ranvier
Paranodal demyelination
Segmental de- and remyelination
Uncompacted/widely spaced myelin

CMT2 (all forms)


MFN2

Congenital amyelinating/hypomyelinating neuropathy


(SOX10, MPZ, EGR2)
Prominent in CMT4C (SH3TC2), but also found
in other forms of CMT1
CMT1 (all forms)
CMT1 (all forms)
MPZ

Widening and abnormal segmentation of the adaxonal Schwann cell


cytoplasm
Abnormal paranodal loops

GJB1

Focally folded myelin, (FFM)/globular neuropathy

Prominent in CMT4B1 (MTMR2), CMT4B2 (SBF2) and CMT4B3


(SBF1), also found in other CMT1 forms
HNPP, sometimes in CMT1
CMT1 (all forms), sometimes in HNPP
CMT1 (all forms)

Tomacula
Schwann cell onion bulb formation
Abundant redundant and detached Schwann cell basal laminae
Basal lamina onion bulbs
Complex Schwann cell onion bulbs
Giant onion bulbs, Schwannoid cell clusters
Abnormal deposits in Schwann cells
Alterations of unmyelinated nerve fibers
Empty Remak bundles, collagen pockets
Abnormally extended and branched processes of Schwann cells of
unmyelinated nerve fibers
Alterations of fascicular architecture and of the neural connective tissue
Reduced number of nerve fascicles/abnormal or incomplete
perineurial covering
Increased number of fascicles
Multiple mucosal neuromas
Multiple perineuriomas, perineurial dysplasia
Endoneurial fibrosis, endoneurial deposition of mucoid substances
Amyloid

perineuriomas as well as hypertrophic perineurial dysplasia have been reported [159] one of which had a possible
positive family history [102]. Endoneurial fibrosis and

13

PRX

Prominent in CMT4C (SH3TC2), but also other forms of autosomal


recessive CMT1 (CMT4)
Prominent in CMT4F (PRX)
Neurofibromatosis type 2
Metachromatic leukodystrophy (MLD), adrenoleukodystrophy/adrenomyeloneuropathy (ALD, AMN), Fabry disease, neurolipidoses
Many CMT and HSAN forms
CMT4C (SH3TC2), also found in intermediate CMTDIE (INF2)

SOX10
Minifascicular neuropathy in cases with 46,XY partial gonadal dysgenesis (DHH)
Multiple endocrine neoplasia type 2 (RET) and related disorders
Familial perineuriosis
CMT1 (all forms)
Familial amyloid neuropathy (TTR), PrP systemic amyloidosis syndrome (PRNP)

deposition of mucoid material in the endoneurial extracellular space are prominent in many cases of demyelinating
neuropathy. Together with the formation of Schwann cell

Acta Neuropathol

onion bulbs mentioned above, these extracellular matrix


alterations may lead to a marked thickening of the nerves
(hypertrophic neuropathy) [176].
Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the GLA gene encoding alphagalactosidase. In nerve biopsies, abnormal granular, osmiophilic inclusions are found predominantly in perineurial
and vessel wall cells. These inclusions produce a so-called
Maltese cross birefringence in polarized light microscopy
of unfixed tissue and show a variable ultrastructural morphology [14]. Extracellular granular osmiophilic deposits
are associated with cells of blood vessel walls in peripheral nerves of patients with CADASIL (cerebral autosomal
dominant arteriopathy with subcortical infarcts and leukoencephalopathy) due to mutations of the NOTCH3 gene
encoding a transmembrane receptor protein [143]. Peculiar
intranuclear granular inclusions in epineurial perivascular
cells are apparent in neuroferritinopathy caused by mutations in the FTL gene encoding the ferritin light polypeptide [134].

Paradigmatic neuropathy genes andtheir


functions ata glance
Genes predominantly associated withan axonal
phenotype
Mitofusin 2 (MFN2)
One of the most frequent axonal types of CMT neuropathy
is CMT2A caused by mutations of the MFN2 gene [185].
Mitofusin 2 is located in the outer mitochondrial membrane. Mutations alter the equilibrium between fusion and
fission of the mitochondrial net and are thought to affect
intra-axonal mitochondrial transport and bioenergetics
including the OXPHOS system [24]. Clinical presentation
may vary from rapid progression in early infancy to adult
onset with slow progression, occasionally with optic atrophy [80], pyramidal features, tremor, and parkinsonism
[41]. Histopathologically, the number of myelinated fibers
is reduced depending on the onset and duration of the disease [142] and larger myelinated fibers may be preferentially affected [166] (Fig.2b). Regenerating axons may be
located in large clusters. EM revealed distinct changes of
axonal mitochondria including condensation, swelling, and
interruption of the outer mitochondrial membrane, predominantly at the site of paranodal axonal protrusions where
mitochondria may accumulate [169]. Occasional mitochondria contain homogeneous or filamentous material [169].
Unusually swollen mitochondria were noted in endoneurial fibroblasts [169] and atypically branched cristae were
observed in Schwann cell mitochondria [153].

Neurofilament lightchain polypeptide (NEFL)


Since NEFL mutations directly affect the intermediate filament network of neurons, NEFL mutations are supposed to
lead to primarily axonal neuropathy. This is reflected by the
axonal loss and the above mentioned focal axonal swellings
and organelle accumulations encountered in sural nerve
biopsies of NEFL mutation cases (Fig.3b). Surprisingly,
however, NEFL mutation may also manifest as a predominantly demyelinating type of neuropathy; neuropathies due
to NEFL mutations therefore may be classified as either
CMT1F, CMT2E, or DI-CMT [39, 40, 186].
Genes predominantly associated witha demyelinating
phenotype
Peripheral myelin protein 22 (PMP22) This was the first
neuropathy gene to be identified [84, 121]. Duplications,
deletions and rare point mutations of PMP22 lead to various types of neuropathy with predominant Schwann cell
pathology (CMT1A; CMT1E; Hereditary Neuropathy with
liability to Pressure Palsy, HNPP; DejernieSottas Syndrome, DSS; Congenital Hypomyelination Neuropathy,
CHN, see above). CMT1A due to PMP22 duplications is
by far the most frequent hereditary neuropathy. PMP22
deletions andrarelypoint mutations lead to HNPP.
PMP22 is a cell membrane glycoprotein; proposed functions include Schwann cell maturation, myelination, myelin maintenance and adhesion of myelin lamellae [45]. In
nerve biopsies of CMT1A patients, de- and remyelinated
axons and classical Schwann cell onion bulb formations are
frequent. A small number of cases with PMP22 mutations
including the typical duplication [11] and the p.Arg159Cys
point mutation [49] may show a predominantly axonal
(CMT2-like) phenotype.
MPZ (P0) Myelin protein zerois a major transmembrane peripheral nerve myelin glycoprotein of the immunoglobulin superfamily. Mutations in MPZ can cause a
similar spectrum of demyelinating neuropathies as PMP22
mutations, including CMT1B, DSS and pressure-induced
neuropathy [69, 87, 174]. Myelin protein zero acts as a
homophilic adhesion protein that is thought to serve important functions in myelin development and compaction as
well as myelin maintenance. Accordingly, nerve biopsies of
CMT1B patients may show uncompacted myelin lamella,
especially inner and outer lamella (Fig.5c); abnormal myelin in- and outfoldings might result in a globular neuropathy pattern [164]. As in CMT1A signs of repeated de- and
remyelination including Schwann cell and basal lamina
onion bulb formations as well as axonal loss and endoneurial fibrosis are present.
Intriguingly, certain MPZ mutations are not associated
with a predominantly demyelinating phenotype, but cause

13

a mixed axonal and demyelinating (intermediate dominant


CMT, ID-CMT [92]) or even predominantly axonal, often
late-onset neuropathy (CMT2I) [25, 73, 151]. In nerve biopsies of such cases, no large Schwann cell onion bulb formations are found. There is a reduction predominantly of large
myelinated nerve fibers, and clusters of regenerating nerve
fibers are frequent. Some of these clusters are surrounded
by concentrically arranged Schwann cell processes, forming
so-called pseudo-onion bulbs [148]. It is unclear so far
why certain MPZ point mutations cause a primary axonal
phenotype. One possibility is the interference of these mutations with Schwann cellaxon interactions [151].
Connexin 32 (Cx32) The connexin 32 (Cx32) protein is
encoded by the GJB1 gene, which is localized on chromosome Xq13. Hemizygous or heterozygous GJB1 mutations
are the most frequent cause of X-linked CMT (CMTX1).
Few cases of an HNPP-like phenotype have been reported
[17]. Cx32 is a gap junction protein expressed by Schwann
cells and other cell populations. Cx32 appears to be
involved in the trafficking of small molecules through the
layers of the Schwann cell plasma membranes at sites of
non-compacted myelin, that is, at SchmidtLanterman
incisures and at nodes of Ranvier. GJB1 mutations are
thought to lead to loss of Cx32 functions. Morphologically, GJB1 mutations are associated with reduced myelin
thickness and loss of predominantly large myelinated nerve
fibers and clustered small, regenerated myelinated nerve
fibers. In line with the proposed functions of Cx32, characteristic abnormal widenings and extensions of the adaxonal
Schwann cell cytoplasm and other abnormalities of the
innermost layers of Schwann cell plasma membrane have
been described (reviewed in [140]) (Fig.5d). In addition,
abnormal axonal inclusions and other axonal alterations
have been found in myelinated nerve fibers of CMTX1
nerve biopsies [140]. These changes might be mostly secondary, even though for several patients carrying GJB1
mutations a predominantly axonal phenotype has been
described [16, 131, 147, 183].
SH3TC2 Autosomal recessive mutations in the gene
encoding SH3 domain and tetratricopeptide repeat-containing protein 2 (SH3TC2) cause CMT4C. This early-onset
progressive neuropathy is one of the most common forms
of autosomal recessive CMT [184]. Patients present with
distal muscle weakness and wasting, distal sensory deficits
as well as precocious and rapidly progressive scoliosis or
kyphoscoliosis and foot deformities [8, 145]. Characteristic
sural nerve pathology includes hypomyelination with excess
Schwann cell and basal lamina onion bulbs, peculiar supernumerary extensions of the cytoplasm of non-myelinating
Schwann cells and abnormal nodes of Ranvier (Fig.6a) [3,
44, 75, 145]. Several studies indicate that SH3TC2 protein
serves important functions in the endocytic recycling pathway which is important for myelination [3, 123, 156].

13

Acta Neuropathol

Intermediate/mixed axonal anddemyelinating


neuropathies
As mentioned above, a mixed axonal and demyelinating
phenotype is observed in patients harboring certain mutations in genes that intuitively would be assigned to a primary axonal or demyelinating neuropathy. The NEFL and
the MPZ genes are prime examples of this phenomenon.
Both encode for major structural proteins of the axon
and Schwann cell, respectively. NEFL is not expressed in
Schwann cells, and MPZ is not expressed in neurons and
their axons. And yet, mutations in both genes may lead to
an intermediate neuropathy or even demyelinating (NEFL)
and axonal (MPZ) neuropathy (see above). Autosomal
dominant mutations in the INF2 gene encoding inverted
formin-2 protein are known to cause focal segmental glomerulosclerosis (FSGS) or FSGS combined with intermediate CMT (CMTDIE) [19]. CMTDIE patients present with
a typical CMT phenotype consisting of progressive distal
muscle weakness and atrophy, reduced tendon reflexes and
foot deformities. Nerve conduction velocities denote an
intermediate type of neuropathy with moderate slowing
combined with signs of axonal loss. In some of the CMTDIE patients anomalies of the central nervous system and
mental retardation as well as sensory hearing loss are found
[19]. Neuropathological studies of sural nerve biopsies
revealed varying degrees of axonal loss as well as severe
de- and remyelination with formation of multilayered onion
bulbs and supernumerary elongated cytoplasmic expansions of non-myelinating Schwann cells [19, 85, 93, 126].
In addition, occasional focally folded myelin layers were
linked to INF2 nerve pathology [126]. While INF2 mutations are unlikely to contribute to neuropathies without kidney disease [19], they are detectable in 75% of patients suffering from FSGS-CMT [85, 125, 161]. In line with a role
of INF2 in actin dynamics and its known association with
the endoplasmic reticulum (ER), actin aggregation [93]
and a prominent or even proliferated ER [126] was found
in axons (Fig.3c), Schwann cells and perineurial cells of
FSGS-CMT patients.

Prion protein andhereditary neuropathy


Prion protein (PrP) is widely implicated in spongiform
encephalopathies that occur in both animals and humans
with intra- and inter-species transmissibility. Demyelinating neuropathy has been described in rare familial cases of
CreutzfeldtJakob disease (CJD) caused by mutations in
the prion protein (PRNP) gene [1, 105]. In Prnp knockout
mice, a late-onset demyelinating neuropathy was reported
[20, 108]. Neuropathy was triggered by specific depletion
of Prnp expression in neurons, but not in Schwann cells,

Acta Neuropathol

suggesting that prion protein derived from neurons is


required for myelin maintenance in peripheral nerves [20].
Recently, a truncating PRNP mutation was identified in a
large family with a PrP systemic amyloidosis syndrome
[96, 97]. Patients presented with late-onset (4th decade)
autosomal dominant axonal sensory neuropathy, diarrhoea,
nausea, autonomic failure, neurogenic bladder and urinary
tract infections. The neuropathy was length dependent,
and the disease progressed for over 20years until death
[9597]. PrP amyloid was deposited in the CNS including
cerebral blood vessel walls and in peripheral nerves, skeletal muscle, skin and internal organs. In peripheral nerves,
abnormal PrP immunoreactivity was mainly found between
nerve fibers [96, 97].

Skin biopsy findings inhereditary neuropathies


Skin biopsy coupled with immunohistochemistry using
the pan-neuronal marker PGP9.5, is a reliable and minimal invasive method for the assessment of unmyelinated
sensory fibers in the epidermis as well as myelinated fibers
in the dermis. In the last decade, skin biopsies have been
increasingly used in the diagnostics and research of hereditary peripheral neuropathies [154]. The quantification of
the epidermal nerve fibers and other features (e.g., innervation of Meissner corpuscles) can be used in the evaluation
and follow up of sensory axonal degeneration [109]. In particular, a loss of sensory innervation in the skin is present
in HSANs [43, 62]. Besides the changes in intra-epidermal
innervation, alterations in sudomotor, vascular and pilomotor innervation can specifically be observed and quantified
in skin biopsies of patients with autonomic neuropathies
[35]. Axonal swellings in the epidermal small nerve fibers
filled with accumulated vesicles are thought to represent
early signs of axonal degeneration [79, 178]. Myelinated
fibers can be observed in higher density in glabrous skin
samples. Alterations of the nodes of Ranvier of these fibers
(changes in internodal length, nodal asymmetries, segmental demyelination) can be observed in hereditary as well
as in acquired neuropathies [129]. Changes of the level of
certain myelin proteins (PMP22, MPZ, MBP) as a result of
disease causing mutations (PMP22 duplication in CMT1A,
deletion in HNPP) can be quantified with immunoelectron
microscopy to correlate them with the clinical phenotype
[74, 81].

Hereditary neuropathies anddouble trouble


Patients harboring mutations in more than one CMT
gene have been reported, including cases with combined
EGR2/GJB1 [30], PMP22 (duplication)/GJB1 [63] and

PMP22 (duplication)/LITAF [98] mutations. In addition,


genetic studies of the promotor region of SH3TC2, which
is controlled by the transcription factors CREB and SOX10,
unraveled particular SNP genotypes as potential modifiers of disease severity of CMT1A [21]. Furthermore, the
co-occurrence of CMT and muscular dystrophies or other
neuromuscular disorders has been occasionally described:
CMT1A and facioscapulohumeral muscular dystrophy
(FSHD) [23, 133], X-linked CMT and Becker muscular
dystrophy [12], CMT neuropathy and myotonic dystrophy
type 1 [63], and HNPP and adrenomyeloneuropathy [63].
The phenotypes are generally more severe compared to
cases with mutations in a single gene.

Hereditary neuropathies andinflammation


Hereditary neuropathies are usually associated with nerve
biopsy changes that are equally distributed within the
fascicles and over the whole cross-sectional area of an
affected nerve. However, edema, lymphocytic infiltrates
and focally accentuated involvement of fascicles, which
are characteristic of chronic inflammatory demyelinating
polyneuropathy(CIDP), have also been described in some
cases of CMT [46]. Occasionally, sporadic and familial
inflammatory neuropathies were linked to PMP22 or GJB1
mutations [51, 76, 88, 173]. Moreover, immunological
studies in a group of CMT1A patients revealed serological
changes including anti-PMP22 autoantibodies indicative
of a superimposed autoimmune component [48]. Moreover, upregulated MHC class II expression was found to be
a common feature in CMT nerve biopsy samples [157].
Established mouse models for inherited demyelinating neuropathies, such as heterozygous Mpz deficient mice, show
a mild increase of CD8+ lymphocytes and macrophages
in peripheral nerves [150] and an immunodeficient genetic
background ameliorated the phenotype of myelin mutant
mice [132]. Extended cellcell contacts between endoneurial macrophages and fibroblasts were found in CMT mouse
models and human CMT nerve biopsies and were linked to
colony-stimulating factor-1-mediated macrophage activation [53]; further pathomechanisms linked to the immune
system in CMT mouse models have been reviewed recently
[90]. Finally, occasional patients with CMT1A, CMT1B,
CMT1X or CMT4C have been reported to respond to antiinflammatory or immunomodulatory treatment resulting in
amelioration of symptoms [36, 51, 67, 175].

Hereditary neuropathies andchemotherapy


Chemotherapeutic agents and other medication can
severely aggravate or exacerbate a coexisting hereditary

13

neuropathy [15]. For example, acute neurotoxicity of the


antimicrotubule agent vincristine has been observed even at
low doses in patients with CMT1A [26, 31, 61] or CMT
due to EGR2 mutation [103]. Vincristine-induced neuropathy can be the initial presentation of CMT [27] or HNPP
[72]. Recently, predisposition to chemotherapy-induced
peripheral neuropathy related to treatment with another
antimicrotubule agent, paclitaxel, was found to be associated with CMT gene variants, in particular variants in the
PRX and ARHGEF10 genes [13].

Concluding remarks
The number of patients with hereditary neuropathies is
likely to grow as ever-improving molecular genetic testing
strategies will continue to assign cases previously labelled
sporadic or idiopathic to distinct gene defects. This
should also become relevant for so far etiologically often
unexplained peripheral neuropathies in the growing elderly
population. At the same time, next-generation sequencing
techniques are generating vast numbers of candidate gene
mutations that need to be verified or excluded by careful
genotypephenotype correlation including nerve biopsy
studies. We should also expect to witness discovery of
an increasing number of gene alterations that predispose
to the clinical manifestation of a neuropathy when they
occur together with other genetic factors (e.g., hypomorphic mutations in neuropathy genes) or other endogenous
and exogenous influences (e.g., ageing, autoimmunity or
treatment with neurotoxic drugs). Analysis of archival and
newly obtained nerve biopsies will remain a mainstay for
scrutinizing these pathomechanisms.
Acknowledgements The authors are grateful to Mrs. H. Mader, Mrs.
E. Beck and Mrs. C. Krude (Institute of Neuropathology, RWTH
Aachen University Medical School) for technical assistance and to
Mr. M. Meyenberg (Dept. of Cardiology, RWTH Aachen University
Medical School) for artwork. This study was supported by grants from
the German Research Foundation (DFG WE1406/13-1, to JW), the
German Ministry for Education and Research (BMBF 01GM1511D,
to JW, IK and JS), the Fritz-Thyssen-Stiftung (Az. 10.15.1.021MN,
to JS) and the Interdisciplinary Center for Clinical Research (IZKF)
within the Faculty of Medicine of RWTH Aachen University (N5-3,
to JW, and N5-4, to JS). JW, IK and JS are members of the German
CMT-Network which is supported by the German Federal Ministry of
Education and Research (BMBF). JW is also a member of the German Motor Neuron Disease Network (MND-Net) sponsored by the
BMBF and the German Society for Muscle Diseases (DGM).

References
1. Antoine JC, Laplanche JL, Mosnier JF, Beaudry P, Chatelain
J, Michel D (1996) Demyelinating peripheral neuropathy with
CreutzfeldtJakob disease and mutation at codon 200 of the
prion protein gene. Neurology 46:11231127

13

Acta Neuropathol
2. Antonellis A, Ellsworth RE, Sambuughin N, Puls I, Abel A,
Lee-Lin SQ, Jordanova A, Kremensky I, Christodoulou K, Middleton LT, Sivakumar K, Ionasescu V, Funalot B, Vance JM,
Goldfarb LG, Fischbeck KH, Green ED (2003) Glycyl tRNA
synthetase mutations in CharcotMarieTooth disease type 2D
and distal spinal muscular atrophy type V. Am J Hum Genet
72:12931299. doi:10.1086/375039
3. Arnaud E, Zenker J, de Preux Charles AS, Stendel C, Roos A,
Medard JJ, Tricaud N, Kleine H, Luscher B, Weis J, Suter U,
Senderek J, Chrast R (2009) SH3TC2/KIAA1985 protein is
required for proper myelination and the integrity of the node of
Ranvier in the peripheral nervous system. Proc Natl Acad Sci
USA 106:1752817533. doi:10.1073/pnas.0905523106
4. Asbury AK, Gale MK, Cox SC, Baringer JR, Berg BO (1972)
Giant axonal neuropathya unique case with segmental neurofilamentous masses. Acta Neuropathol 20:237247
5. Auer-Grumbach M, Olschewski A, Papic L, Kremer H, McEntagart ME, Uhrig S, Fischer C, Frohlich E, Balint Z, Tang B,
Strohmaier H, Lochmuller H, Schlotter-Weigel B, Senderek J,
Krebs A, Dick KJ, Petty R, Longman C, Anderson NE, Padberg GW, Schelhaas HJ, van Ravenswaaij-Arts CM, Pieber TR,
Crosby AH, Guelly C (2010) Alterations in the ankyrin domain
of TRPV4 cause congenital distal SMA, scapuloperoneal SMA
and HMSN2C. Nat Genet 42:160164. doi:10.1038/ng.508
6. Auer-Grumbach M, Toegel S, Schabhuttl M, Weinmann D,
Chiari C, Bennett DL, Beetz C, Klein D, Andersen PM, Bohme
I, Fink-Puches R, Gonzalez M, Harms MB, Motley W, Reilly
MM, Renner W, Rudnik-Schoneborn S, Schlotter-Weigel B,
Themistocleous AC, Weishaupt JH, Ludolph AC, Wieland T,
Tao F, Abreu L, Windhager R, Zitzelsberger M, Strom TM,
Walther T, Scherer SS, Zuchner S, Martini R, Senderek J (2016)
Rare variants in MME, encoding metalloprotease neprilysin, are linked to late-onset autosomal-dominant axonal polyneuropathies. Am J Hum Genet 99:607623. doi:10.1016/j.
ajhg.2016.07.008
7. Azzedine H, Bolino A, Taieb T, Birouk N, Di Duca M,
Bouhouche A, Benamou S, Mrabet A, Hammadouche T, Chkili
T, Gouider R, Ravazzolo R, Brice A, Laporte J, LeGuern E
(2003) Mutations in MTMR13, a new pseudophosphatase
homologue of MTMR2 and Sbf1, in two families with an autosomal recessive demyelinating form of CharcotMarieTooth
disease associated with early-onset glaucoma. Am J Hum Genet
72:11411153. doi:10.1086/375034
8. Azzedine H, Ravise N, Verny C, Gabreels-Festen A, Lammens M, Grid D, Vallat JM, Durosier G, Senderek J, Nouioua S, Hamadouche T, Bouhouche A, Guilbot A, Stendel C,
Ruberg M, Brice A, Birouk N, Dubourg O, Tazir M, LeGuern
E (2006) Spine deformities in CharcotMarieTooth 4C
caused by SH3TC2 gene mutations. Neurology 67:602606.
doi:10.1212/01.wnl.0000230225.19797.93
9. Azzedine H, Senderek J, Rivolta C, Chrast R (2012) Molecular genetics of CharcotMarieTooth disease: from genes to
genomes. Mol Syndromol 3:204214. doi:10.1159/000343487
10. Baets J, Duan X, Wu Y, Smith G, Seeley WW, Mademan I,
McGrath NM, Beadell NC, Khoury J, Botuyan MV, Mer G,
Worrell GA, Hojo K, DeLeon J, Laura M, Liu YT, Senderek
J, Weis J, Van den Bergh P, Merrill SL, Reilly MM, Houlden
H, Grossman M, Scherer SS, De Jonghe P, Dyck PJ, Klein CJ
(2015) Defects of mutant DNMT1 are linked to a spectrum of
neurological disorders. Brain 138:845861. doi:10.1093/brain/
awv010
11. Bergamin G, Boaretto F, Briani C, Pegoraro E, Cacciavillani M,
Martinuzzi A, Muglia M, Vettori A, Vazza G, Mostacciuolo ML
(2014) Mutation analysis of MFN2, GJB1, MPZ and PMP22 in
Italian patients with axonal CharcotMarieTooth disease. Neuromol Med 16:540550. doi:10.1007/s12017-014-8307-9

Acta Neuropathol
12. Bergmann C, Senderek J, Hermanns B, Jauch A, Janssen B,
Schroder JM, Karch D (2000) Becker muscular dystrophy combined with X-linked CharcotMarieTooth neuropathy. Muscle
Nerve 23:818823
13. Beutler AS, Kulkarni AA, Kanwar R, Klein CJ, Therneau TM,
Qin R, Banck MS, Boora GK, Ruddy KJ, Wu Y, Smalley RL,
Cunningham JM, Le-Lindqwister NA, Beyerlein P, Schroth GP,
Windebank AJ, Zuchner S, Loprinzi CL (2014) Sequencing of
CharcotMarieTooth disease genes in a toxic polyneuropathy.
Ann Neurol 76:727737. doi:10.1002/ana.24265
14. Bilbao JM, Schmidt RM (2015) Biopsy diagnosis of peripheral
neuropathy. Springer, New York
15. Bird TD (19932016) CharcotMarieTooth hereditary neuropathy overview. In: Pagon RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean LJH, Bird TD, Dolan CR, Fong CT,
Smith RJH etal (eds) Gene Reviews. University of Washington,
Seattle, WA
16. Birouk N, LeGuern E, Maisonobe T, Rouger H, Gouider R, Tardieu S, Gugenheim M, Routon MC, Leger JM, Agid Y, Brice A,
Bouche P (1998) X-linked CharcotMarieTooth disease with
connexin 32 mutations: clinical and electrophysiologic study.
Neurology 50:10741082
17. Bissar-Tadmouri N, Parman Y, Boutrand L, Deymeer F, Serdaroglu P, Vandenberghe A, Battaloglu E (2000) Mutational analysis
and genotype/phenotype correlation in Turkish CharcotMarie
Tooth Type 1 and HNPP patients. Clin Genet 58:396402
18. Boerkoel CF, Takashima H, Garcia CA, Olney RK, Johnson
J, Berry K, Russo P, Kennedy S, Teebi AS, Scavina M, Williams LL, Mancias P, Butler IJ, Krajewski K, Shy M, Lupski JR
(2002) CharcotMarieTooth disease and related neuropathies:
mutation distribution and genotypephenotype correlation. Ann
Neurol 51:190201
19. Boyer O, Nevo F, Plaisier E, Funalot B, Gribouval O, Benoit G,
Cong EH, Arrondel C, Tete MJ, Montjean R, Richard L, Karras A, Pouteil-Noble C, Balafrej L, Bonnardeaux A, Canaud
G, Charasse C, Dantal J, Deschenes G, Deteix P, Dubourg O,
Petiot P, Pouthier D, Leguern E, Guiochon-Mantel A, Broutin I,
Gubler MC, Saunier S, Ronco P, Vallat JM, Alonso MA, Antignac C, Mollet G (2011) INF2 mutations in CharcotMarie
Tooth disease with glomerulopathy. N Engl J Med 365:2377
2388. doi:10.1056/NEJMoa1109122
20. Bremer J, Baumann F, Tiberi C, Wessig C, Fischer H, Schwarz
P, Steele AD, Toyka KV, Nave KA, Weis J, Aguzzi A (2010)
Axonal prion protein is required for peripheral myelin maintenance. Nat Neurosci 13:310318
21. Brewer MH, Ma KH, Beecham GW, Gopinath C, Baas F, Choi BO,
Reilly MM, Shy ME, Zuchner S, Svaren J, Antonellis A (2014)
Haplotype-specific modulation of a SOX10/CREB response element at the CharcotMarieTooth disease type 4C locus SH3TC2.
Hum Mol Genet 23:51715187. doi:10.1093/hmg/ddu240
22. Busard HL, Gabreels-Festen AA, van t Hof MA, Renier WO,
Gabreels FJ (1990) Polyglucosan bodies in sural nerve biopsies.
Acta Neuropathol 80:554557
23. Butefisch CM, Lang DF, Gutmann L (1998) The devastating
combination of CharcotMarieTooth disease and facioscapulohumeral muscular dystrophy. Muscle Nerve 21:788791
24. Cartoni R, Martinou JC (2009) Role of mitofusin 2 mutations in
the physiopathology of CharcotMarieTooth disease type 2A.
Exp Neurol 218:268273. doi:10.1016/j.expneurol.2009.05.003
25. Chapon F, Latour P, Diraison P, Schaeffer S, Vandenberghe A
(1999) Axonal phenotype of CharcotMarieTooth disease
associated with a mutation in the myelin protein zero gene. J
Neurol Neurosurg Psychiatry 66:779782
26. Chaudhry V, Chaudhry M, Crawford TO, Simmons-OBrien E,
Griffin JW (2003) Toxic neuropathy in patients with pre-existing neuropathy. Neurology 60:337340

27. Chauvenet AR, Shashi V, Selsky C, Morgan E, Kurtzberg J,


Bell B, Pediatric Oncology Group S (2003) Vincristine-induced
neuropathy as the initial presentation of CharcotMarieTooth
disease in acute lymphoblastic leukemia: a Pediatric Oncology
Group study. J Pediatr Hematol Oncol 25:316320
28. Chen B, Zheng R, Luan X, Zhang W, Wang Z, Yuan Y (2009)
Clincial and pathological study of distal motor neuropathy
with N88S mutation in BSCL2. Neuropathology 29:543547.
doi:10.1111/j.1440-1789.2009.01011.x
29. Chen YC, Auer-Grumbach M, Matsukawa S, Zitzelsberger M,
Themistocleous AC, Strom TM, Samara C, Moore AW, Cho LT,
Young GT, Weiss C, Schabhuttl M, Stucka R, Schmid AB, Parman Y, Graul-Neumann L, Heinritz W, Passarge E, Watson RM,
Hertz JM, Moog U, Baumgartner M, Valente EM, Pereira D,
Restrepo CM, Katona I, Dusl M, Stendel C, Wieland T, Stafford
F, Reimann F, von Au K, Finke C, Willems PJ, Nahorski MS,
Shaikh SS, Carvalho OP, Nicholas AK, Karbani G, McAleer
MA, Cilio MR, McHugh JC, Murphy SM, Irvine AD, Jensen
UB, Windhager R, Weis J, Bergmann C, Rautenstrauss B, Baets
J, De Jonghe P, Reilly MM, Kropatsch R, Kurth I, Chrast R,
Michiue T, Bennett DL, Woods CG, Senderek J (2015) Transcriptional regulator PRDM12 is essential for human pain perception. Nat Genet 47:803808. doi:10.1038/ng.3308
30. Chung KW, Sunwoo IN, Kim SM, Park KD, Kim WK, Kim TS,
Koo H, Cho M, Lee J, Choi BO (2005) Two missense mutations of EGR2 R359W and GJB1 V136A in a CharcotMarie
Tooth disease family. Neurogenetics 6:159163. doi:10.1007/
s10048-005-0217-4
31. Cil T, Altintas A, Tamam Y, Battaloglu E, Isikdogan A (2009)
Low dose vincristine-induced severe polyneuropathy in a
Hodgkin lymphoma patient: a case report (vincristine-induced
severe polyneuropathy). J Pediatr Hematol Oncol 31:787789.
doi:10.1097/MPH.0b013e3181b530ad
32. Claeys KG, Zuchner S, Kennerson M, Berciano J, Garcia A, Verhoeven K, Storey E, Merory JR, Bienfait HM, Lammens M, Nelis
E, Baets J, De Vriendt E, Berneman ZN, De Veuster I, Vance JM,
Nicholson G, Timmerman V, De Jonghe P (2009) Phenotypic
spectrum of dynamin 2 mutations in CharcotMarieTooth neuropathy. Brain 132:17411752. doi:10.1093/brain/awp115
33. Davis CJ, Bradley WG, Madrid R (1978) The peroneal muscular atrophy syndrome: clinical, genetic, electrophysiological
and nerve biopsy studies. I. Clinical, genetic and electrophysiological findings and classification. J Genet Hum 26:311349
34. De Sandre-Giovannoli A, Chaouch M, Kozlov S, Vallat JM,
Tazir M, Kassouri N, Szepetowski P, Hammadouche T, Vandenberghe A, Stewart CL, Grid D, Levy N (2002) Homozygous
defects in LMNA, encoding lamin A/C nuclear-envelope proteins, cause autosomal recessive axonal neuropathy in human
(CharcotMarieTooth disorder type 2) and mouse. Am J Hum
Genet 70:726736. doi:10.1086/339274
35. Donadio V, Incensi A, Giannoccaro MP, Cortelli P, Di Stasi
V, Pizza F, Jaber MA, Baruzzi A, Liguori R (2012) Peripheral autonomic neuropathy: diagnostic contribution of skin
biopsy. J Neuropathol Exp Neurol 71:10001008. doi:10.1097/
NEN.0b013e3182729fdc
36. Donaghy M, Sisodiya SM, Kennett R, McDonald B, Haites N,
Bell C (2000) Steroid responsive polyneuropathy in a family
with a novel myelin protein zero mutation. J Neurol Neurosurg
Psychiatry 69:799805
37. Dubourg O, Azzedine H, Yaou RB, Pouget J, Barois A, Meininger V, Bouteiller D, Ruberg M, Brice A, LeGuern E (2006)
The G526R glycyl-tRNA synthetase gene mutation in distal
hereditary motor neuropathy type V. Neurology 66:17211726.
doi:10.1212/01.wnl.0000218304.02715.04
38. Dyck PJ, Chance P, Lebo R, Carney JA (1993) Hereditary
motor and sensory neuropathies. In: Dyck PJ, Thomas PK,

13


Griffin JW, Low PA, Poduslo JF (eds) Peripheral neuropathy.
Saunders, W.B, pp 10941136
39. Elbracht M, Senderek J, Schara U, Nolte K, Klopstock T, Roos
A, Reimann J, Zerres K, Weis J, Rudnik-Schoneborn S (2014)
Clinical and morphological variability of the E396K mutation
in the neurofilament light chain gene in patients with Charcot
MarieTooth disease type 2E. Clin Neuropathol 33:335343.
doi:10.5414/NP300742
40. Fabrizi GM, Cavallaro T, Angiari C, Bertolasi L, Cabrini I, Ferrarini M, Rizzuto N (2004) Giant axon and neurofilament accumulation in CharcotMarieTooth disease type 2E. Neurology
62:14291431
41. Feely SM, Laura M, Siskind CE, Sottile S, Davis M, Gibbons
VS, Reilly MM, Shy ME (2011) MFN2 mutations cause severe
phenotypes in most patients with CMT2A. Neurology 76:1690
1696. doi:10.1212/WNL.0b013e31821a441e
42. Fidzianska A, Drac H, Rafalowska J (2002) Phenomenon of
Schwann cell apoptosis in a case of congenital hypomyelinating
neuropathy with basal lamina onion bulb formation. Brain Dev
24:727731
43. Fridman V, Oaklander AL, David WS, Johnson EA, Pan J,
Novak P, Brown RH Jr, Eichler FS (2015) Natural history and
biomarkers in hereditary sensory neuropathy type 1. Muscle
Nerve 51:489495. doi:10.1002/mus.24336
44. Gabreels-Festen A, van Beersum S, Eshuis L, LeGuern E,
Gabreels F, van Engelen B, Mariman E (1999) Study on the
gene and phenotypic characterisation of autosomal recessive
demyelinating motor and sensory neuropathy (CharcotMarie
Tooth disease) with a gene locus on chromosome 5q23-q33. J
Neurol Neurosurg Psychiatry 66:569574
45. Gabreels-Festen A, Wetering RV (1999) Human nerve pathology caused by different mutational mechanisms of the PMP22
gene. Ann N Y Acad Sci 883:336343
46. Gabreels-Festen AA, Gabreels FJ, Hoogendijk JE, Bolhuis
PA, Jongen PJ, Vingerhoets HM (1993) Chronic inflammatory
demyelinating polyneuropathy or hereditary motor and sensory
neuropathy? Diagnostic value of morphological criteria. Acta
Neuropathol 86:630635
47. Gabreels-Festen AA, Hoogendijk JE, Meijerink PH, Gabreels
FJ, Bolhuis PA, van Beersum S, Kulkens T, Nelis E, Jennekens FG, de Visser M, van Engelen BG, Van Broeckhoven C,
Mariman EC (1996) Two divergent types of nerve pathology in
patients with different P0 mutations in CharcotMarieTooth
disease. Neurology 47:761765
48. Gabriel CM, Gregson NA, Wood NW, Hughes RA (2002) Immunological study of hereditary motor and sensory neuropathy type
1a (HMSN1a). J Neurol Neurosurg Psychiatry 72:230235
49. Gess B, Jeibmann A, Schirmacher A, Kleffner I, Schilling M,
Young P (2011) Report of a novel mutation in the PMP22 gene
causing an axonal neuropathy. Muscle Nerve 43:605609.
doi:10.1002/mus.21973
50. Gibbels E, Schaefer HE, Runne U, Schroder JM, Haupt WF,
Assmann G (1985) Severe polyneuropathy in Tangier disease
mimicking syringomyelia or leprosy. Clinical, biochemical,
electrophysiological, and morphological evaluation, including
electron microscopy of nerve, muscle, and skin biopsies. J Neurol 232:283294
51. Ginsberg L, Malik O, Kenton AR, Sharp D, Muddle JR,
Davis MB, Winer JB, Orrell RW, King RH (2004) Coexistent
hereditary and inflammatory neuropathy. Brain 127:193202.
doi:10.1093/brain/awh017
52. Gregory A, Kurian MA, Maher ER, Hogarth P, Hayflick SJ
(19932016) PLA2G6-associated neurodegeneration. In: Pagon
RA, Adam MP, Ardinger HH, Wallace SE, Amemiya A, Bean
LJH, Bird TD, Dolan CR, Fong CT, Smith RJH etal (eds) Gene
Reviews. University of Washington, Seattle, WA

13

Acta Neuropathol
53. Groh J, Weis J, Zieger H, Stanley ER, Heuer H, Martini R
(2012) Colony-stimulating factor-1 mediates macrophagerelated neural damage in a model for CharcotMarieTooth disease type 1X. Brain 135:88104. doi:10.1093/brain/awr283
54. Guelly C, Zhu PP, Leonardis L, Papic L, Zidar J, Schabhuttl
M, Strohmaier H, Weis J, Strom TM, Baets J, Willems J, De
Jonghe P, Reilly MM, Frohlich E, Hatz M, Trajanoski S, Pieber
TR, Janecke AR, Blackstone C, Auer-Grumbach M (2011) Targeted high-throughput sequencing identifies mutations in atlastin-1 as a cause of hereditary sensory neuropathy type I. Am J
Hum Genet 88:99105. doi:10.1016/j.ajhg.2010.12.003
55. Guilbot A, Williams A, Ravise N, Verny C, Brice A, Sherman
DL, Brophy PJ, LeGuern E, Delague V, Bareil C, Megarbane
A, Claustres M (2001) A mutation in periaxin is responsible for
CMT4F, an autosomal recessive form of CharcotMarieTooth
disease. Hum Mol Genet 10:415421
56. Guzzetta F, Ferriere G, Lyon G (1982) Congenital hypomyelination polyneuropathy. Pathological findings compared with
polyneuropathies starting later in life. Brain 105:395416
57. Harding AE, Thomas PK (1980) The clinical features of
hereditary motor and sensory neuropathy types I and II. Brain
103:259280
58. Harding BN, Kariya S, Monani UR, Chung WK, Benton M, Yum SW, Tennekoon G, Finkel RS (2015) Spectrum of neuropathophysiology in spinal muscular atrophy
type I. J Neuropathol Exp Neurol 74:1524. doi:10.1097/
NEN.0000000000000144
59. Hattori N, Yamamoto M, Yoshihara T, Koike H, Nakagawa M,
Yoshikawa H, Ohnishi A, Hayasaka K, Onodera O, Baba M,
Yasuda H, Saito T, Nakashima K, Kira J, Kaji R, Oka N, Sobue
G, Study Group for Hereditary Neuropathy in J (2003) Demyelinating and axonal features of CharcotMarieTooth disease
with mutations of myelin-related proteins (PMP22, MPZ and
Cx32): a clinicopathological study of 205 Japanese patients.
Brain 126:134151
60. Higuchi Y, Hashiguchi A, Yuan J, Yoshimura A, Mitsui J, Ishiura H, Tanaka M, Ishihara S, Tanabe H, Nozuma S, Okamoto
Y, Matsuura E, Ohkubo R, Inamizu S, Shiraishi W, Yamasaki
R, Ohyagi Y, Kira JI, Oya Y, Yabe H, Nishikawa N, Tobisawa
S, Matsuda N, Masuda M, Kugimoto C, Fukushima K, Yano
S, Yoshimura J, Doi K, Nakagawa M, Morishita S, Tsuji S,
Takashima H (2016) Mutations in MME cause an autosomalrecessive CharcotMarieTooth disease type 2. Ann Neurol
79:659-672. doi:10.1002/ana.24612
61. Hildebrandt G, Holler E, Woenkhaus M, Quarch G, Reichle A,
Schalke B, Andreesen R (2000) Acute deterioration of Charcot
MarieTooth disease IA (CMT IA) following 2mg of vincristine chemotherapy. Ann Oncol 11:743747
62. Hilz MJ, Axelrod FB, Bickel A, Stemper B, Brys M, Wendelschafer-Crabb G, Kennedy WR (2004) Assessing function
and pathology in familial dysautonomia: assessment of temperature perception, sweating and cutaneous innervation. Brain
127:20902098. doi:10.1093/brain/awh235
63. Hodapp JA, Carter GT, Lipe HP, Michelson SJ, Kraft GH, Bird
TD (2006) Double trouble in hereditary neuropathy: concomitant mutations in the PMP-22 gene and another gene produce
novel phenotypes. Arch Neurol 63:112117. doi:10.1001/
archneur.63.1.112
64. Houlden H, King R, Blake J, Groves M, Love S, Woodward
C, Hammans S, Nicoll J, Lennox G, ODonovan DG, Gabriel
C, Thomas PK, Reilly MM (2006) Clinical, pathological and
genetic characterization of hereditary sensory and autonomic
neuropathy type 1 (HSAN I). Brain 129:411425. doi:10.1093/
brain/awh712
65. Houlden H, King RH, Muddle JR, Warner TT, Reilly MM,
Orrell RW, Ginsberg L (2004) A novel RAB7 mutation

Acta Neuropathol
associated with ulcero-mutilating neuropathy. Ann Neurol
56:586590. doi:10.1002/ana.20281
66. Houlden H, King RH, Wood NW, Thomas PK, Reilly MM
(2001) Mutations in the 5 region of the myotubularin-related
protein 2 (MTMR2) gene in autosomal recessive hereditary
neuropathy with focally folded myelin. Brain 124:907915
67. Houlden H, Laura M, Ginsberg L, Jungbluth H, Robb SA,
Blake J, Robinson S, King RH, Reilly MM (2009) The phenotype of CharcotMarieTooth disease type 4C due to SH3TC2
mutations and possible predisposition to an inflammatory
neuropathy. Neuromuscul Disord 19:264269. doi:10.1016/j.
nmd.2009.01.006
68. Hughes JT, Brownell B, Hewer RL (1968) The peripheral sensory pathway in friedreichs ataxia. An examination by light and
electron microscopy of the posterior nerve roots, posterior root
ganglia, and peripheral sensory nerves in cases of Friedreichs
ataxia. Brain 91:803818
69. Iida M, Koike H, Ando T, Sugiura M, Yamamoto M, Tanaka
F, Sobue G (2012) A novel MPZ mutation in CharcotMarie
Tooth disease type 1B with focally folded myelin and multiple
entrapment neuropathies. Neuromuscul Disord 22:166169.
doi:10.1016/j.nmd.2011.08.005
70. Jerath NU, Shy ME (2015) Hereditary motor and sensory neuropathies: understanding molecular pathogenesis could lead to
future treatment strategies. Biochim Biophys Acta 1852:667
678. doi:10.1016/j.bbadis.2014.07.031
71. Johnson-Kerner BL, Roth L, Greene JP, Wichterle H, Sproule
DM (2014) Giant axonal neuropathy: an updated perspective
on its pathology and pathogenesis. Muscle Nerve 50:467476.
doi:10.1002/mus.24321
72. Kalfakis N, Panas M, Karadima G, Floroskufi P, Kokolakis N,
Vassilopoulos D (2002) Hereditary neuropathy with liability to
pressure palsies emerging during vincristine treatment. Neurology 59:14701471
73. Kamholz J, Shy ME (2004) Late onset CharcotMarieTooth
2 syndrome caused by two novel mutations in the MPZ gene.
Neurology 63:194
74. Katona I, Wu X, Feely SM, Sottile S, Siskind CE, Miller LJ,
Shy ME, Li J (2009) PMP22 expression in dermal nerve
myelin from patients with CMT1A. Brain 132:17341740.
doi:10.1093/brain/awp113
75. Kessali M, Zemmouri R, Guilbot A, Maisonobe T, Brice A,
LeGuern E, Grid D (1997) A clinical, electrophysiologic, neuropathologic, and genetic study of two large Algerian families
with an autosomal recessive demyelinating form of Charcot
MarieTooth disease. Neurology 48:867873
76. Korn-Lubetzki I, Argov Z, Raas-Rothschild A, Wirguin I,
Steiner I (2002) Family with inflammatory demyelinating polyneuropathy and the HNPP 17p12 deletion. Am J Med Genet
113:275278. doi:10.1002/ajmg.10725
77. Kruer MC (2013) The neuropathology of neurodegeneration
with brain iron accumulation. Int Rev Neurobiol 110:165194.
doi:10.1016/B978-0-12-410502-7.00009-0
78. Kurth I, Pamminger T, Hennings JC, Soehendra D, Huebner
AK, Rotthier A, Baets J, Senderek J, Topaloglu H, Farrell SA,
Nurnberg G, Nurnberg P, De Jonghe P, Gal A, Kaether C, Timmerman V, Hubner CA (2009) Mutations in FAM134B, encoding a newly identified Golgi protein, cause severe sensory and
autonomic neuropathy. Nat Genet 41:11791181. doi:10.1038/
ng.464
79. Lauria G, Morbin M, Lombardi R, Borgna M, Mazzoleni G,
Sghirlanzoni A, Pareyson D (2003) Axonal swellings predict
the degeneration of epidermal nerve fibers in painful neuropathies. Neurology 61:631636
80. Leonardi L, Marcotulli C, Storti E, Tessa A, Serrao M, Parisi
V, Santorelli FM, Pierelli F, Casali C (2015) Acute optic

neuropathy associated with a novel MFN2 mutation. J Neurol


262:16781680. doi:10.1007/s00415-015-7756-x
81. Li J, Bai Y, Ghandour K, Qin P, Grandis M, Trostinskaia A,
Ianakova E, Wu X, Schenone A, Vallat JM, Kupsky WJ, Hatfield J, Shy ME (2005) Skin biopsies in myelin-related neuropathies: bringing molecular pathology to the bedside. Brain
128:11681177. doi:10.1093/brain/awh483
82. Lossos A, Kahlon O, Soffer D (2014) Polyglucosan body
disease. In: Vallat JM, Weis J (eds) Peripheral nerve disorders: pathology and genetics. Wiley Blackwell, New York, pp
167171
83. Lossos A, Meiner Z, Barash V, Soffer D, Schlesinger I, Abramsky O, Argov Z, Shpitzen S, Meiner V (1998) Adult polyglucosan body disease in Ashkenazi Jewish patients carrying the
Tyr329Ser mutation in the glycogen-branching enzyme gene.
Ann Neurol 44:867872. doi:10.1002/ana.410440604
84. Lupski JR, de Oca-Luna RM, Slaugenhaupt S, Pentao L,
Guzzetta V, Trask BJ, Saucedo-Cardenas O, Barker DF, Killian
JM, Garcia CA, Chakravarti A, Patel PI (1991) DNA duplication associated with CharcotMarieTooth disease type 1A.
Cell 66:219232
85. Mademan I, Deconinck T, Dinopoulos A, Voit T, Schara U,
Devriendt K, Meijers B, Lerut E, De Jonghe P, Baets J (2013)
De novo INF2 mutations expand the genetic spectrum of hereditary neuropathy with glomerulopathy. Neurology 81:1953
1958. doi:10.1212/01.wnl.0000436615.58705.c9
86. Madrid R, Bradley WG (1975) The pathology of neuropathies
with focal thickening of the myelin sheath (tomaculous neuropathy). J Neurol Sci 25:415448
87. Magot A, Latour P, Mussini JM, Mourtada R, Guiheneuc P,
Pereon Y (2008) A new MPZ mutation associated with a mild
CMT1 phenotype presenting with recurrent nerve compression.
Muscle Nerve 38:10551059. doi:10.1002/mus.21050
88. Malandrini A, Villanova M, Dotti MT, Federico A (1999) Acute
inflammatory neuropathy in CharcotMarieTooth disease.
Neurology 52:859861
89. Marchesi C, Milani M, Morbin M, Cesani M, Lauria G, Scaioli
V, Piccolo G, Fabrizi GM, Cavallaro T, Taroni F, Pareyson D
(2010) Four novel cases of periaxin-related neuropathy and
review of the literature. Neurology 75:18301838. doi:10.1212/
WNL.0b013e3181fd6314
90. Martini R, Willison H (2016) Neuroinflammation in the peripheral nerve: cause, modulator, or bystander in peripheral neuropathies? Glia 64:475486. doi:10.1002/glia.22899
91. Martyn CN, Hughes RA (1997) Epidemiology of peripheral
neuropathy. J Neurol Neurosurg Psychiatry 62:310318
92. Mastaglia FL, Nowak KJ, Stell R, Phillips BA, Edmondston JE,
Dorosz SM, Wilton SD, Hallmayer J, Kakulas BA, Laing NG
(1999) Novel mutation in the myelin protein zero gene in a family with intermediate hereditary motor and sensory neuropathy.
J Neurol Neurosurg Psychiatry 67:174179
93. Mathis S, Funalot B, Boyer O, Lacroix C, Marcorelles P, Magy
L, Richard L, Antignac C, Vallat JM (2014) Neuropathologic
characterization of INF2-related CharcotMarieTooth disease:
evidence for a Schwann cell actinopathy. J Neuropathol Exp
Neurol 73:223233. doi:10.1097/NEN.0000000000000047
94. Mathis S, Goizet C, Tazir M, Magdelaine C, Lia AS, Magy L,
Vallat JM (2015) CharcotMarieTooth diseases: an update and
some new proposals for the classification. J Med Genet 52:681
690. doi:10.1136/jmedgenet-2015-103272
95. Matsuzono K, Ikeda Y, Liu W, Kurata T, Deguchi S, Deguchi
K, Abe K (2013) A novel familial prion disease causing panautonomic-sensory neuropathy and cognitive impairment. Eur J
Neurol 20:e67e69. doi:10.1111/ene.12089
96. Mead S, Gandhi S, Beck J, Caine D, Gajulapalli D, Carswell
C, Hyare H, Joiner S, Ayling H, Lashley T, Linehan JM,

13


Al-Doujaily H, Sharps B, Revesz T, Sandberg MK, Reilly
MM, Koltzenburg M, Forbes A, Rudge P, Brandner S, Warren
JD, Wadsworth JD, Wood NW, Holton JL, Collinge J (2013)
A novel prion disease associated with diarrhea and autonomic
neuropathy. N Engl J Med 369:19041914. doi:10.1056/
NEJMoa1214747
97. Mead S, Reilly MM (2015) A new prion disease: relationship with central and peripheral amyloidoses. Nat Rev Neurol
11:9097. doi:10.1038/nrneurol.2014.263
98. Meggouh F, de Visser M, Arts WF, De Coo RI, van Schaik IN,
Baas F (2005) Early onset neuropathy in a compound form
of CharcotMarieTooth disease. Ann Neurol 57:589591.
doi:10.1002/ana.20434
99. Midroni G, Bilbao JM (1995) Biopsy diagnosis of peripheral
neuropathy. Butterworth-Heinemann
100. Montecchiani C, Pedace L, Lo Giudice T, Casella A, Mearini
M, Gaudiello F, Pedroso JL, Terracciano C, Caltagirone C,
Massa R, St George-Hyslop PH, Barsottini OG, Kawarai T,
Orlacchio A (2016) ALS5/SPG11/KIAA1840 mutations cause
autosomal recessive axonal CharcotMarieTooth disease.
Brain 139:7385. doi:10.1093/brain/awv320
101. Mygland A, Monstad P (2001) Chronic polyneuropathies in
Vest-Agder, Norway. Eur J Neurol 8:157165
102. Nakamura N, Oka N, Nakamura S, Akiguchi I, Kashii S (1995) A
case with hereditary motor and sensory neuropathy with multiple
cranial nerves involvement. Rinsho Shinkeigaku 35:516520
103. Nakamura T, Hashiguchi A, Suzuki S, Uozumi K, Tokunaga
S, Takashima H (2012) Vincristine exacerbates asymptomatic
CharcotMarieTooth disease with a novel EGR2 mutation.
Neurogenetics 13:7782. doi:10.1007/s10048-012-0313-1
104. Nakhro K, Park JM, Hong YB, Park JH, Nam SH, Yoon BR,
Yoo JH, Koo H, Jung SC, Kim HL, Kim JY, Choi KG, Choi
BO, Chung KW (2013) SET binding factor 1 (SBF1) mutation causes CharcotMarieTooth disease type 4B3. Neurology
81:165173. doi:10.1212/WNL.0b013e31829a3421
105. Neufeld MY, Josiphov J, Korczyn AD (1992) Demyelinating
peripheral neuropathy in CreutzfeldtJakob disease. Muscle
Nerve 15:12341239. doi:10.1002/mus.880151103
106. Neveling K, Martinez-Carrera LA, Holker I, Heister A, Verrips
A, Hosseini-Barkooie SM, Gilissen C, Vermeer S, Pennings M,
Meijer R, te Riele M, Frijns CJ, Suchowersky O, MacLaren L,
Rudnik-Schoneborn S, Sinke RJ, Zerres K, Lowry RB, Lemmink HH, Garbes L, Veltman JA, Schelhaas HJ, Scheffer H,
Wirth B (2013) Mutations in BICD2, which encodes a golgin
and important motor adaptor, cause congenital autosomal-dominant spinal muscular atrophy. Am J Hum Genet 92:946954.
doi:10.1016/j.ajhg.2013.04.011
107. Nicholson G, Myers S (2006) Intermediate forms of Charcot
MarieTooth neuropathy: a review. Neuromol Med 8:123130
108. Nishida N, Tremblay P, Sugimoto T, Shigematsu K, Shirabe S,
Petromilli C, Erpel SP, Nakaoke R, Atarashi R, Houtani T, Torchia M, Sakaguchi S, DeArmond SJ, Prusiner SB, Katamine S
(1999) A mouse prion protein transgene rescues mice deficient
for the prion protein gene from purkinje cell degeneration and
demyelination. Lab Invest 79:689697
109. Nolano M, Provitera V, Crisci C, Stancanelli A, WendelschaferCrabb G, Kennedy WR, Santoro L (2003) Quantification of
myelinated endings and mechanoreceptors in human digital
skin. Ann Neurol 54:197205. doi:10.1002/ana.10615
110. Nolte KW, Janecke AR, Vorgerd M, Weis J, Schroder JM (2008)
Congenital type IV glycogenosis: the spectrum of pleomorphic polyglucosan bodies in muscle, nerve, and spinal cord
with two novel mutations in the GBE1 gene. Acta Neuropathol
116:491506

111. Oates EC, Reddel S, Rodriguez ML, Gandolfo LC, Bahlo
M, Hawke SH, Lamande SR, Clarke NF, North KN (2012)

13

Acta Neuropathol
Autosomal dominant congenital spinal muscular atrophy: a true
form of spinal muscular atrophy caused by early loss of anterior
horn cells. Brain 135:17141723. doi:10.1093/brain/aws108
112. Ouvrier R, Menezes M (2014) Neuroaxonal dystrophy. In: Vallat JM, Weis J (eds) Peripheral nerve disorders: pathology and
genetics. Wiley Blackwell, New York, pp 146148
113. Ouvrier R, Menezes M (2014) Peripheral nerve involvement in
neurolipidoses. In: Vallat JM, Weis J (eds) Peripheral nerve disorders. Pathology and genetics. Wiley, New York, pp 149157
114. Pareyson D, Saveri P, Sagnelli A, Piscosquito G (2015) Mitochondrial dynamics and inherited peripheral nerve diseases.
Neurosci Lett 596:6677. doi:10.1016/j.neulet.2015.04.001
115. Parthey K, Kornhuber M, Kunze C, Wand D, Nolte KW, Nikolin S, Weis J, Schroder JM (2012) SOX10 mutation with peripheral amyelination and developmental disturbance of axons.
Muscle Nerve 45:284290. doi:10.1002/mus.22262
116. Pearson J, Dancis J, Axelrod F, Grover N (1975) The sural
nerve in familial dysautonomia. J Neuropathol Exp Neurol
34:413424
117. Perrot R, Berges R, Bocquet A, Eyer J (2008) Review of the
multiple aspects of neurofilament functions, and their possible
contribution to neurodegeneration. Mol Neurobiol 38:2765.
doi:10.1007/s12035-008-8033-0
118. Plante-Bordeneuve V, Ferreira A, Lalu T, Zaros C, Lacroix C,
Adams D, Said G (2007) Diagnostic pitfalls in sporadic transthyretin familial amyloid polyneuropathy (TTR-FAP). Neurology 69:693698. doi:10.1212/01.wnl.0000267338.45673.f4
119. Plante-Bordeneuve V, Said G (2002) DejerineSottas disease
and hereditary demyelinating polyneuropathy of infancy. Muscle Nerve 26:608621. doi:10.1002/mus.10197
120. Pujol RM, Matias-Guiu X, Miralles J, Colomer A, de Moragas
JM (1997) Multiple idiopathic mucosal neuromas: a minor form
of multiple endocrine neoplasia type 2B or a new entity? J Am
Acad Dermatol 37:349352

121. Raeymaekers P, Timmerman V, Nelis E, De Jonghe P,
Hoogendijk JE, Baas F, Barker DF, Martin JJ, De Visser M,
Bolhuis PA etal (1991) Duplication in chromosome 17p11.2
in CharcotMarieTooth neuropathy type 1a (CMT 1a). The
HMSN Collaborative Research Group. Neuromuscul Disord
1:9397

122. Rajadhyaksha AM, Elemento O, Puffenberger EG, Schierberl KC, Xiang JZ, Putorti ML, Berciano J, Poulin C, Brais
B, Michaelides M, Weleber RG, Higgins JJ (2010) Mutations in FLVCR1 cause posterior column ataxia and retinitis
pigmentosa. Am J Hum Genet 87:643654. doi:10.1016/j.
ajhg.2010.10.013

123. Roberts RC, Peden AA, Buss F, Bright NA, Latouche M,
Reilly MM, Kendrick-Jones J, Luzio JP (2010) Mistargeting of
SH3TC2 away from the recycling endosome causes Charcot
MarieTooth disease type 4C. Hum Mol Genet 19:10091018.
doi:10.1093/hmg/ddp565
124. Robitaille Y, Carpenter S, Karpati G, DiMauro SD (1980) A
distinct form of adult polyglucosan body disease with massive
involvement of central and peripheral neuronal processes and
astrocytes: a report of four cases and a review of the occurrence
of polyglucosan bodies in other conditions such as Laforas disease and normal ageing. Brain 103:315336
125. Rodriguez PQ, Lohkamp B, Celsi G, Mache CJ, Auer-Grumbach M, Wernerson A, Hamajima N, Tryggvason K, Patrakka J
(2013) Novel INF2 mutation p. L77P in a family with glomerulopathy and CharcotMarieTooth neuropathy. Pediatric nephrology 28:339343. doi:10.1007/s00467-012-2299-1
126. Roos A, Weis J, Korinthenberg R, Fehrenbach H, Hausler M,
Zuchner S, Mache C, Hubmann H, Auer-Grumbach M, Senderek J (2015) Inverted formin 2-related CharcotMarieTooth
disease: extension of the mutational spectrum and pathological

Acta Neuropathol
findings in Schwann cells and axons. J Peripher Nerv Syst
20:5259. doi:10.1111/jns.12106
127. Rudnik-Schoneborn S, Deden F, Eggermann K, Eggermann T,
Wieczorek D, Sellhaus B, Yamoah A, Goswami A, Claeys KG,
Weis J, Zerres K (2016) Autosomal dominant spinal muscular
atrophy with lower extremity predominance: a recognizable
phenotype of BICD2 mutations. Muscle Nerve 54:496500.
doi:10.1002/mus.25114
128. Said G, Marion MH, Selva J, Jamet C (1986) Hypotrophic
and dying-back nerve fibers in Friedreichs ataxia. Neurology
36:12921299
129. Saporta MA, Katona I, Lewis RA, Masse S, Shy ME, Li J
(2009) Shortened internodal length of dermal myelinated
nerve fibres in CharcotMarieTooth disease type 1A. Brain
132:32633273. doi:10.1093/brain/awp274
130. Scheithauer BW, Woodruff JM, Antonescu CR (2007) Perineurioma. In: Louis DN, Ohgaki H, Wiestler OD, Cavenee WK
(eds) WHO classification of tumours of the central nervous system. International Agency for Research on Cancer, pp 158159
131. Scherer SS, Fischbeck KH (1999) Is CMTX an axonopathy?
Neurology 52:432433
132. Schmid CD, Stienekemeier M, Oehen S, Bootz F, Zielasek J,
Gold R, Toyka KV, Schachner M, Martini R (2000) Immune
deficiency in mouse models for inherited peripheral neuropathies leads to improved myelin maintenance. J Neurosci
20:729735
133. Schreiber O, Schneiderat P, Kress W, Rautenstrauss B, Senderek
J, Schoser B, Walter MC (2013) Facioscapulohumeral muscular
dystrophy and CharcotMarieTooth neuropathy 1Aevidence
for double trouble overlapping syndromes. BMC Med Genet
14:92. doi:10.1186/1471-2350-14-92

134. Schroder JM (2005) Ferritinopathy: diagnosis by muscle or nerve biopsy, with a note on other nuclear inclusion
body diseases. Acta Neuropathol 109:109114. doi:10.1007/
s00401-004-0949-5
135. Schroder JM (1968) The hyperneurotization of the bands of
Bngner in experimental isoniazid neuropathy: phase and
electron microscopic study. Virchows Arch Abt B Zellpath
1:131156
136. Schroder JM (2006) Neuropathology of CharcotMarieTooth
and related disorders. Neuromol Med 8:2342
137. Schroder JM (2001) Pathology of peripheral nerves: an atlas of
structural and molecular pathological changes. Springer, New
York
138. Schroder JM, Heide G, Ramaekers V, Mortier W (1993) Subtotal aplasia of myelinated nerve fibers in the sural nerve. Neuropediatrics 24:286291. doi:10.1055/s-2008-1071558
139. Schroder JM, Seiffert KE (1970) The fine structure of the neuromatous neurotization of nerve transplants. Virchows Arch B
Cell Pathol 5:219235
140. Schroder JM, Senderek J (2014) X-linked neuropathy of the
CharcotMarieTooth type. In: Vallat JM, Weis J (eds) Peripheral nerve disorders. Pathology and genetics. Wiley, New York,
pp 111118
141. Schroder JM, Weis J (2014) Basic pathology of the peripheral
nervous system. In: Vallat JM, Weis J (eds) Peripheral nerve
disorders. Pathology and genetics. Wiley, New York, pp 3858
142. Schrder JM, Weis J, Senderek J (2014) Autosomal dominant
neuropathy of the axonal CharcotMarieTooth type 2. In: Vallat JM, Weis J (eds) Peripheral nerve disorders. Pathology and
genetics. Wiley, New York, pp 7284
143. Schroder JM, Zuchner S, Dichgans M, Nagy Z, Molnar MJ
(2005) Peripheral nerve and skeletal muscle involvement in
CADASIL. Acta Neuropathol (Berl) 110:587599
144. Schulz A, Baader SL, Niwa-Kawakita M, Jung MJ, Bauer R,
Garcia C, Zoch A, Schacke S, Hagel C, Mautner VF, Hanemann

CO, Dun XP, Parkinson DB, Weis J, Schroder JM, Gutmann


DH, Giovannini M, Morrison H (2013) Merlin isoform 2 in
neurofibromatosis type 2-associated polyneuropathy. Nat Neurosci 16:426433. doi:10.1038/nn.3348
145. Senderek J, Bergmann C, Stendel C, Kirfel J, Verpoorten N, De
Jonghe P, Timmerman V, Chrast R, Verheijen MH, Lemke G,
Battaloglu E, Parman Y, Erdem S, Tan E, Topaloglu H, Hahn
A, Muller-Felber W, Rizzuto N, Fabrizi GM, Stuhrmann M,
Rudnik-Schoneborn S, Zuchner S, Michael Schroder J, Buchheim E, Straub V, Klepper J, Huehne K, Rautenstrauss B,
Buttner R, Nelis E, Zerres K (2003) Mutations in a gene encoding a novel SH3/TPR domain protein cause autosomal recessive
CharcotMarieTooth type 4C neuropathy. Am J Hum Genet
73:11061119
146. Senderek J, Bergmann C, Weber S, Ketelsen UP, Schorle H,
Rudnik-Schoneborn S, Buttner R, Buchheim E, Zerres K (2003)
Mutation of the SBF2 gene, encoding a novel member of the
myotubularin family, in CharcotMarieTooth neuropathy type
4B2/11p15. Hum Mol Genet 12:349356
147. Senderek J, Hermanns B, Bergmann C, Boroojerdi B, Bajbouj
M, Hungs M, Ramaekers VT, Quasthoff S, Karch D, Schroder
JM (1999) X-linked dominant CharcotMarieTooth neuropathy: clinical, electrophysiological, and morphological phenotype in four families with different connexin32 mutations(1). J
Neurol Sci 167:90101
148. Senderek J, Hermanns B, Lehmann U, Bergmann C, Marx G,
Kabus C, Timmerman V, Stoltenburg-Didinger G, Schroder JM
(2000) CharcotMarieTooth neuropathy type 2 and P0 point
mutations: two novel amino acid substitutions (Asp61Gly; Tyr119Cys) and a possible hotspot on Thr124Met. Brain Pathol
10:235248
149. Sevilla T, Lupo V, Martinez-Rubio D, Sancho P, Sivera R, Chumillas MJ, Garcia-Romero M, Pascual-Pascual SI, Muelas N,
Dopazo J, Vilchez JJ, Palau F, Espinos C (2016) Mutations in
the MORC2 gene cause axonal CharcotMarieTooth disease.
Brain 139:6272. doi:10.1093/brain/awv311
150. Shy ME, Arroyo E, Sladky J, Menichella D, Jiang H, Xu W,
Kamholz J, Scherer SS (1997) Heterozygous P0 knockout mice
develop a peripheral neuropathy that resembles chronic inflammatory demyelinating polyneuropathy (CIDP). J Neuropathol
Exp Neurol 56:811821
151. Shy ME, Jani A, Krajewski K, Grandis M, Lewis RA, Li J,
Shy RR, Balsamo J, Lilien J, Garbern JY, Kamholz J (2004)
Phenotypic clustering in MPZ mutations. Brain 127:371384.
doi:10.1093/brain/awh048
152. Skre H (1974) Genetic and clinical aspects of CharcotMarie
Tooths disease. Clin Genet 6:98118
153. Sole G, Ferrer X, Vital C, Martin-Negrier ML, Vital A, Latour
P (2009) Ultrastructural mitochondrial modifications characteristic of mitofusin 2 mutations (CMT2A). J Peripher Nerv Syst
14:206207. doi:10.1111/j.1529-8027.2009.00234.x
154. Sommer C, Lauria G (2007) Skin biopsy in the management of
peripheral neuropathy. Lancet Neurol 6:632642. doi:10.1016/
S1474-4422(07)70172-2
155. Sperfeld AD, Hein C, Schroder JM, Ludolph AC, Hanemann
CO (2002) Occurrence and characterization of peripheral nerve
involvement in neurofibromatosis type 2. Brain 125:9961004
156. Stendel C, Roos A, Kleine H, Arnaud E, Ozcelik M, Sidiropoulos PN, Zenker J, Schupfer F, Lehmann U, Sobota RM, Litchfield DW, Luscher B, Chrast R, Suter U, Senderek J (2010)
SH3TC2, a protein mutant in CharcotMarieTooth neuropathy, links peripheral nerve myelination to endosomal recycling.
Brain 133:24622474. doi:10.1093/brain/awq168

157. Stoll G, Gabreels-Festen AA, Jander S, Muller HW, Hanemann CO (1998) Major histocompatibility complex class II
expression and macrophage responses in genetically proven

13


CharcotMarieTooth type 1 and hereditary neuropathy with
liability to pressure palsies. Muscle Nerve 21:14191427
158. Tazir M, Azzedine H, Assami S, Sindou P, Nouioua S, Zemmouri R, Hamadouche T, Chaouch M, Feingold J, Vallat JM,
Leguern E, Grid D (2004) Phenotypic variability in autosomal recessive axonal CharcotMarieTooth disease due
to the R298C mutation in lamin A/C. Brain 127:154163.
doi:10.1093/brain/awh021

159. Thomas PK, King RH, Workman JM, Schroder JM (2000)
Hypertrophic perineurial dysplasia in multifocal and generalized peripheral neuropathies. Neuropathol Appl Neurobiol
26:536543
160. Tournev I, King RH, Workman J, Nourallah M, Muddle JR,
Kalaydjieva L, Romanski K, Thomas PK (1999) Peripheral
nerve abnormalities in the congenital cataracts facial dysmorphism neuropathy (CCFDN) syndrome. Acta Neuropathol
98:165170

161. Toyota K, Ogino D, Hayashi M, Taki M, Saito K, Abe A,
Hashimoto T, Umetsu K, Tsukaguchi H, Hayasaka K (2013)
INF2 mutations in CharcotMarieTooth disease complicated
with focal segmental glomerulosclerosis. J Peripher Nerv Syst
18:9798. doi:10.1111/jns5.12014
162. Umehara F, Tate G, Itoh K, Yamaguchi N, Douchi T, Mitsuya
T, Osame M (2000) A novel mutation of desert hedgehog in a
patient with 46, XY partial gonadal dysgenesis accompanied
by minifascicular neuropathy. Am J Hum Genet 67:13021305.
doi:10.1016/S0002-9297(07)62958-9
163. Vallat JM (2003) Dominantly inherited peripheral neuropathies.
J Neuropathol Exp Neurol 62:699714
164. Vallat JM, Gil R, Leboutet MJ, Hugon J, Moulies D (1987)
Congenital hypo- and hypermyelination neuropathy. Two cases.
Acta Neuropathol 74:197201
165. Vallat JM, Magy L, Richard L, Piaser M, Sindou P, Calvo J,
Ghorab K, Cros D (2008) Intranervous immunoglobulin deposits: an underestimated mechanism of neuropathy. Muscle Nerve
38:904911. doi:10.1002/mus.21057
166. Vallat JM, Ouvrier RA, Pollard JD, Magdelaine C, Zhu D,
Nicholson GA, Grew S, Ryan MM, Funalot B (2008) Histopathological findings in hereditary motor and sensory neuropathy of axonal type with onset in early childhood associated with
mitofusin 2 mutations. J Neuropathol Exp Neurol 67:1097
1102. doi:10.1097/NEN.0b013e31818b6cbc
167. Vallat JM, Weis J (2014) Peripheral nerve disorders. Pathology
and genetics. In: Keohane K, Gray F (eds) International society
of neuropathology: pathology and genetics series, Wiley Blackwell, New York
168. Varon R, Gooding R, Steglich C, Marns L, Tang H, Angelicheva
D, Yong KK, Ambrugger P, Reinhold A, Morar B, Baas F, Kwa
M, Tournev I, Guerguelcheva V, Kremensky I, Lochmuller H,
Mullner-Eidenbock A, Merlini L, Neumann L, Burger J, Walter M, Swoboda K, Thomas PK, von Moers A, Risch N, Kalaydjieva L (2003) Partial deficiency of the C-terminal-domain
phosphatase of RNA polymerase II is associated with congenital cataracts facial dysmorphism neuropathy syndrome. Nat
Genet 35:185189. doi:10.1038/ng1243
169. Verhoeven K, Claeys KG, Zuchner S, Schroder JM, Weis J,
Ceuterick C, Jordanova A, Nelis E, De Vriendt E, Van Hul M,
Seeman P, Mazanec R, Saifi GM, Szigeti K, Mancias P, Butler
IJ, Kochanski A, Ryniewicz B, De Bleecker J, Van den Bergh
P, Verellen C, Van Coster R, Goemans N, Auer-Grumbach M,
Robberecht W, Milic Rasic V, Nevo Y, Tournev I, Guergueltcheva V, Roelens F, Vieregge P, Vinci P, Moreno MT, Christen
HJ, Shy ME, Lupski JR, Vance JM, De Jonghe P, Timmerman
V (2006) MFN2 mutation distribution and genotype/phenotype
correlation in CharcotMarieTooth type 2. Brain 129:2093
2102. doi:10.1093/brain/awl126

13

Acta Neuropathol
170. Verhoeven K, De Jonghe P, Van de Putte T, Nelis E, Zwijsen A,
Verpoorten N, De Vriendt E, Jacobs A, Van Gerwen V, Francis
A, Ceuterick C, Huylebroeck D, Timmerman V (2003) Slowed
conduction and thin myelination of peripheral nerves associated
with mutant rho guanine-nucleotide exchange factor 10. Am J
Hum Genet 73:926932. doi:10.1086/378159
171. Vital A, Ferrer X, Lagueny A, Vandenberghe A, Latour P, Goizet
C, Canron MH, Louiset P, Petry KG, Vital C (2001) Histopathological features of X-linked CharcotMarieTooth disease in 8
patients from 6 families with different connexin32 mutations. J
Peripher Nerv Syst 6:7984
172. Vital A, Vital C (2012) Mitochondria and peripheral neuropathies. J Neuropathol Exp Neurol 71:10361046. doi:10.1097/
NEN.0b013e3182764d47
173. Vital A, Vital C, Lagueny A, Ferrer X, Ribiere-Bachelier C,
Latour P, Petry KG (2003) Inflammatory demyelination in a
patient with CMT1A. Muscle Nerve 28:373376. doi:10.1002/
mus.10404
174. Warner LE, Hilz MJ, Appel SH, Killian JM, Kolodry EH, Karpati G, Carpenter S, Watters GV, Wheeler C, Witt D, Bodell A,
Nelis E, Van Broeckhoven C, Lupski JR (1996) Clinical phenotypes of different MPZ (P0) mutations may include CharcotMarieTooth type 1B, DejerineSottas, and congenital
hypomyelination. Neuron 17:451460
175. Watanabe M, Yamamoto N, Ohkoshi N, Nagata H, Kohno Y,
Hayashi A, Tamaoka A, Shoji S (2002) Corticosteroid- responsive asymmetric neuropathy with a myelin protein zero gene
mutation. Neurology 59:767769
176. Webster HD, Schroder JM, Asbury AK, Adams RD (1967) The
role of Schwann cells in the formation of onion bulbs found
in chronic neuropathies. J Neuropathol Exp Neurol 26:276299

177. Weis J (2014) Polyneuropathies associated with neurofibromatosis. In: Vallat JM, Weis J (eds) Peripheral nerve disorders. Pathology and genetics. Wiley Blackwell, New York, pp
172174

178. Weis J, Katona I, Muller-Newen G, Sommer C, Necula G,
Hendrich C, Ludolph AC, Sperfeld AD (2011) Small-fiber
neuropathy in patients with ALS. Neurology 76:20242029.
doi:10.1212/WNL.0b013e31821e553a
179. Weis J, Schroder JM (1988) Adult polyglucosan body myopathy with subclinical peripheral neuropathy: case report and
review of diseases associated with polyglucosan body accumulation. Clin Neuropathol 7:271279
180. Weis J, Senderek J (2014) Introduction to the hereditary neuropathies. In: Vallat JM, Weis J (eds) Peripheral nerve disorders.
Pathology and genetics. Wiley Blackwell, New York, pp 5961
181. Wilmshurst JM, Ouvrier R (2011) Hereditary peripheral neuropathies of childhood: an overview for clinicians. Neuromuscul Disord 21:763775. doi:10.1016/j.nmd.2011.05.013

182. Wu Y, Jiang Y, Gao Z, Wang J, Yuan Y, Xiong H, Chang
X, Bao X, Zhang Y, Xiao J, Wu X (2009) Clinical study and
PLA2G6 mutation screening analysis in Chinese patients with
infantile neuroaxonal dystrophy. Eur J Neurol 16:240245.
doi:10.1111/j.1468-1331.2008.02397.x
183. Young P, Grote K, Kuhlenbaumer G, Debus O, Kurlemann H,
Halfter H, Funke H, Ringelstein EB, Stogbauer F (2001) Mutation analysis in CharcotMarieTooth disease type 1: point
mutations in the MPZ gene and the GJB1 gene cause comparable phenotypic heterogeneity. J Neurol 248:410415

184. Zimon M, Battaloglu E, Parman Y, Erdem S, Baets J, De
Vriendt E, Atkinson D, Almeida-Souza L, Deconinck T, Ozes
B, Goossens D, Cirak S, Van Damme P, Shboul M, Voit T,
Van Maldergem L, Dan B, El-Khateeb MS, Guergueltcheva V,
Lopez-Laso E, Goemans N, Masri A, Zuchner S, Timmerman V,
Topaloglu H, De Jonghe P, Jordanova A (2015) Unraveling the
genetic landscape of autosomal recessive CharcotMarieTooth

Acta Neuropathol
neuropathies using a homozygosity mapping approach. Neurogenetics 16:3342. doi:10.1007/s10048-014-0422-0
185. Zuchner S, Mersiyanova IV, Muglia M, Bissar-Tadmouri N,
Rochelle J, Dadali EL, Zappia M, Nelis E, Patitucci A, Senderek J, Parman Y, Evgrafov O, Jonghe PD, Takahashi Y, Tsuji
S, Pericak-Vance MA, Quattrone A, Battaloglu E, Polyakov AV,
Timmerman V, Schroder JM, Vance JM (2004) Mutations in the
mitochondrial GTPase mitofusin 2 cause CharcotMarieTooth

neuropathy type 2A. Nat Genet 36:449451. doi:10.1038/


ng1341
186. Zuchner S, Vorgerd M, Sindern E, Schroder JM (2004) The
novel neurofilament light (NEFL) mutation Glu397Lys is associated with a clinically and morphologically heterogeneous
type of CharcotMarieTooth neuropathy. Neuromuscul Disord
14:147157

13

Вам также может понравиться