Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

Focus Article

Hirschsprung disease:
a developmental disorder
of the enteric nervous system
Sonja J. McKeown,1 Lincon Stamp,1,2 Marlene M. Hao1 and Heather
M. Young1∗

Hirschsprung disease (HSCR), which is also called congenital megacolon or


intestinal aganglionosis, is characterized by an absence of enteric (intrinsic)
neurons from variable lengths of the most distal bowel. Because enteric neurons
are essential for propulsive intestinal motility, infants with HSCR suffer from
severe constipation and have a distended abdomen. Currently the only treatment
is surgical removal of the affected bowel. HSCR has an incidence of around
1:5,000 live births, with a 4:1 male:female gender bias. Most enteric neurons arise
from neural crest cells that emigrate from the caudal hindbrain and then migrate
caudally along the entire gut. The absence of enteric neurons from variable lengths
of the bowel in HSCR results from a failure of neural crest-derived cells to colonize
the affected gut regions. HSCR is therefore regarded as a neurocristopathy.
HSCR is a multigenic disorder and has become a paradigm for understanding
complex factorial disorders. The major HSCR susceptibility gene is RET. The
penetrance of several mutations in HSCR susceptibility genes is sex-dependent.
HSCR can occur as an isolated disorder or as part of syndromes; for example,
Type IV Waardenburg syndrome is characterized by deafness and pigmentation
defects as well as intestinal aganglionosis. Studies using animal models have
shown that HSCR genes regulate multiple processes including survival, prolifer-
ation, differentiation, and migration. Research into HSCR and the development
of enteric neurons is an excellent example of the cross fertilization of ideas that
can occur between human molecular geneticists and researchers using animal
models. © 2012 Wiley Periodicals, Inc.

How to cite this article:


WIREs Dev Biol 2012. doi: 10.1002/wdev.57

INTRODUCTION with congenital megacolon lacked enteric (intrinsic)


neurons.2 Overall, HSCR has an incidence of around
I n 1888, Harald Hirschprung described two unre-
lated boys who died with abdominal disten-
sion, congenital megacolon, and severe chronic
1:5000 live births, but there are differences in inci-
dence between ethnic groups and there is a 4:1
constipation.1 Although a number of studies from male:female sex bias.
the early 1900s had implicated defects in neurons, HSCR can occur as an isolated disorder (70%
the etiology of Hirschsprung disease (HSCR) was not of patients) or as part of a syndrome in which other
identified until the mid-1900s when it was found neural crest derivatives are commonly affected. One
that all postmortem samples of rectum from patients of these syndromes, Type IV Waardenburg syndrome
(WS4), is discussed in the section Type IV Waarden-
∗ Correspondence to: h.young@unimelb.edu.au burg Syndrome (WS4)—An Example of a Syndrome
1
Department of Anatomy & Cell Biology, University of Melbourne, That Includes Aganglionic Megacolon of this arti-
Melbourne 3010, VIC, Australia
2 cle. Between 2 and 15% of HSCR patients have
Murdoch Childrens Research Institute, Parkville 3052, VIC,
Australia the chromosomal abnormality, trisomy 21 (Down

© 2012 Wiley Periodicals, Inc.


Focus Article wires.wiley.com/devbio

in ENS development. This review briefly describes


the ENS and the stages of ENS development relevant
to HSCR. We then discuss the etiology, diagnosis,
genetics, and current and potential treatments for
HSCR, and finally we briefly discuss one of the
syndromic forms of HSCR, WS4.
D

THE ENTERIC NERVOUS SYSTEM


The ENS is an extensive network of neurons and glia
M
within the wall of the bowel.7,8 The ENS is the largest
part of the peripheral nervous system, and there are
at least as many neurons in the gut as there are in
R the spinal cord.9 In the small and large intestines,
enteric neurons are found in two main plexuses; the
myenteric plexus is located between the circular and
longitudinal muscle layers, and the submucosal plexus
C is found within the connective tissue of the submu-
cosa. In humans, some neurons are also found within
the mucosa.10 Neurons in myenteric ganglia are pri-
marily involved in the control of motility whereas
A
most submucosal neurons regulate transport of ions
across the epithelium and blood flow.
FIGURE 1 | Barium enema study, lateral view, of a 6-month-old There are many different types of enteric neu-
infant with HSCR. The descending colon is greatly dilated (‘megacolon’) rons that differ in their targets, inputs, direction of
while the distal colon and rectum are constricted. (Reprinted with projection, neurotransmitters, and electrophysiolog-
permission from Ref 5. Copyright 1999 Radiological Society of North ical characteristics. The neurons form circuits that
America) regulate a number of gut functions, including motility.
In the small and large intestines, enteric neurons are
essential for all coordinated motor patterns including
syndrome).3 HSCR is a multigenic disorder with mixing and peristalsis. The essential role for enteric
variable penetrance (see the section on Etiology below) neurons in peristalsis is exemplified by the bowel
and severity. In over 80% of cases, aganglionosis is obstruction that occurs in the aganglionic region of
restricted to the rectosigmoid colon (short-segment patients with HSCR.
HSCR) (Figure 1), but aganglionosis can also affect
significant lengths of the colon or even extend into the
distal small intestine (long segment HSCR). Extremely ORIGIN AND EARLY DEVELOPMENT
rarely, the entire small and large intestines are agan-
glionic, which is termed total intestinal aganglionosis.
OF THE ENTERIC NERVOUS SYSTEM
A region of bowel with reduced enteric neuron den- Most enteric neurons arise from neural crest cells
sity, the transition zone, is always present oral to the that emigrate from the neural tube adjacent to somites
aganglionic region.4 1–711 (Figure 2). This region of the neural axis is called
The mechanisms controlling the development of ‘vagal’, and encompasses the most caudal hindbrain
the enteric nervous system (ENS) are highly conserved. (somites 1-5) and rostral trunk (somites 6-7). Sacral
Research into HSCR and ENS development has level neural crest cells also contribute some enteric
consequently benefitted greatly from the exchange neurons, but even in the most distal regions of the
of knowledge and ideas between human molecular bowel, the majority of enteric neurons arise from
geneticists and researchers using animal models, vagal neural crest-derived cells.12–14
including mouse, chick, and zebrafish.6 Several HSCR Studies in a variety of species, including human,
susceptibility genes were first identified following have shown that vagal neural crest-derived cells
studies on mice with targeted inactivation of particular enter the foregut and then migrate caudally along
genes that resulted in HSCR-like phenotypes. the gut16,17 (Figure 2). The colonization of the
Likewise, genetic studies of HSCR patients identified gastrointestinal tract by enteric neural crest-derived
genes that were not previously known to be involved cells (ENCCs) is notable both because of the distance

© 2012 Wiley Periodicals, Inc.


WIREs Developmental Biology Hirschsprung disease

H
H

FIGURE 2 | Immunostaining of E9.5 and E10.5 mice with antibodies to the neural crest cell marker, SOX10 (goat anti-SOX10, Santa Cruz). At
E9.5, vagal neural crest-derived cells from the post-otic hindbrain are migrating ventrally toward and into the foregut (arrow ). At E10.5, the most
caudal neural crest-derived cell is in the midgut (open arrow ). Neural crest cells that emigrate from the neural tube adjacent to somites 1–7 follow
the pathway (dotted line ) that is followed later by the vagus nerve (X). The ectoderm was removed from the E10.5 mouse to enable the gut to be
seen. OV, otic vesicle; BA1, branchial arch 1; BA2, branchial arch 2; asterisks, dorsal root ganglia; V, VII, IX, X—cranial nerves V, VII, IX and X.
(Reprinted with permission from Ref 15. Copyright 2012 Elsevier)

ENCCs migrate, and because of the length of time while ENCCs are colonizing the gut,30,31 inhibition
it takes. Because the gut grows substantially while it of cell death of chick vagal neural crest cells as
is being colonized, ENCCs probably migrate further they are migrating from the hindbrain toward the
than any other embryonic cell population.18 For gut results in hyperganglionosis (increased density of
example, in mice, the colon increases in length fivefold enteric neurons),32 and cell death is common when
between when ENCC first enter the proximal colon RET signaling is conditionally abolished while ENCCs
and when they reach the distal end. In humans, it are migrating along the colon in mice.33
takes 3 weeks for ENCCs to migrate from the foregut Many molecules involved in the colonization
to the anal end,17 and in mice it takes 5 days, which of the gut by ENCCs have been identified. These
is 25% of the gestation period.16 include molecules secreted by the gut mesenchyme
ENS development continues after the gut has that act on receptors expressed by ENCCs, as well
been colonized by ENCCs, and even beyond birth, as transcription factors, adhesion molecules and other
at least in laboratory animals.19,20 In this article, molecules expressed by ENCCs themselves. Some of
however, we focus on the mechanisms involved in the the signaling pathways involved in ENS development
early development of the ENS as HSCR is caused by affect multiple processes including proliferation,
a failure of ENCCs to colonize the entire gut. migration, differentiation, and/or survival. Mutations
ENCCs proliferate rapidly while migrating in genes encoding components of many of these
toward and within the gut.21,22 Moreover, a sub- signaling pathways have been associated with HSCR
population of ENCCs starts to undergo neuronal (Table 1). Here we briefly summarize the main
differentiation as they are migrating within the pathways and molecules known to play a role in
gut wall. The colonization of the gut by ENCCs the colonization of the gut by ENCCs.
requires coordinated proliferation, migration, and
neuronal differentiation, as perturbations to cell
number,21–23 migratory behavior,24,25 or rate of neu- GDNF-GFRA1-RET Signaling Pathway
ronal differentiation26–29 can result in aganglionosis in This is a critical pathway for ENS development.
animal models. Although cell death is not prominent Glial cell line-derived neurotrophic factor (GDNF)

© 2012 Wiley Periodicals, Inc.


TABLE 1 HSCR Susceptibility Genes and Mouse Models of HSCR
Zygosity of Frequency of Expression Pattern in Role in ENS
Mutations in Mutation in Isolated Isolated HSCR, Associated ENS Phenotype of Mice Relevant to Development from
Gene HSCR Patients HSCR Syndrome, Phenotype Mouse Mutants ENS Development Animal Studies
Focus Article

RET Non-coding Non-coding mutations: Mostly isolated but Ret +/− mice have a normal RET tyrosine kinase. Survival, proliferation, migration,
HGNC:9967 mutations may be very common. Coding can also be ENS30 Expressed by ENCCs.34 and neuronal differentiation of
homozygous or mutations: ∼50% of associated with a Expression is maintained ENCCs6
heterozygous; familial and 15–20% syndrome1 in neurons but is
coding mutations of sporadic HSCR1 down-regulated in glia.35
are heterozygous1
Homozygous Extremely rare Isolated. Total Ret −/− mice lack neurons
missense36 intestinal distal to the stomach37
aganglionosis36
GDNF Heterozygous38 Very rare Isolated38 Neurons are present along the Secreted molecule.
HGNC:4232 entire gut of Gdnf +/− mice, Expressed by
but at reduced density30 ; mesenchyme of
Gdnf −/− mice lack enteric embryonic gut39 and
neurons distal to the external muscle of
stomach39 mature gut.40
GFRA1 Heterozygous41 Extremely rare (one Isolated41 Gfra1+/− mice have normal GPI-linked protein to which
HGNC:4243 family only, low enteric neuron density;30 GDNF binds. Expressed
penetrance, Gfra1−/− mice lack neurons by ENCCs.43
42
significance distal to the stomach
unknown)41

© 2012 Wiley Periodicals, Inc.


NRTN Heterozygous44 Extremely rare (one Isolated44 Neurons are present along the Neurturin is a secreted Required for the normal projection,
HGNC:8007 family only and entire gut of Nrtn−/− molecule; member of branching and/or survival of
co-occurred with a mice.45 Myenteric neuron GDNF family ligands. axons of enteric excitatory
RET mutation)44 number is normal, but there Expressed by the motor neurons45
are defects in submucosal mesenchyme of the
neuron number and in the embryonic gut46
density of excitatory nerve
fibers in Nrtn−/− mice30
EDNRB Heterozygous ∼5%48 Usually isolated,48 Ednrb s/sl mice have reduced EDNRB is a G EDN3-EDNRB signaling promotes
HGNC:3180 mutations47,48 very occasional neuron density in the colon protein-coupled receptor. proliferation and inhibits
WS449,50 (note: s is a hypomorphic Main receptor for EDN3 neuronal differentiation of
mutation and sl is a null in ENS development. enteric neural progenitors, and
mutation).51 Expressed by ENCCs and regulates ENCC
some gut mesenchymal migration.26,29,53 EDN3-EDNRB
52
cells signaling is also involved in
melanocyte development.54
wires.wiley.com/devbio
TABLE 1 Continued
Zygosity of Frequency of Expression Pattern in Role in ENS
Mutations in Mutation in Isolated HSCR, Associated ENS Phenotype of Mice Relevant to Development from
Gene HSCR Patients Isolated HSCR Syndrome, Phenotype Mouse Mutants ENS Development Animal Studies
Mutations in both WS4; patients are Ednrb −/− mice lack of neurons
alleles49,55 deaf, have piebald in the distal colon54
pigment pattern,
HSCR and
WIREs Developmental Biology

neurological
deficits49,55
EDN3 Heterozygous <5%57 Isolated56 except Edn3 +/− mice have normal EDN3 is the main ligand for EDNRB
HGNC:3178 mutations56 one case of WS458 enteric neuron density59 in ENS development. Expressed
by the mesenchyme of the
embryonic gut with highest
expression in the cecum27,60
Mutations in both WS449,61 Edn3 −/− mice lack of neurons
alleles61 in the distal colon62
ECE1 Heterozygous63 Very rare (one case Cardiac, craniofacial No reports of ENS defects in ECE1 is involved in the proteolytic
HGNC:3146 reported63 ) and autonomic Ece1+/− mice. Ece1−/− processing of big endothelins to
defects, and mice lack neurons in the biologically active peptides.
HSCR63 distal colon, lack epidermal Expression of ECE-1 does not yet
melanocytes, and also appear to have been examined
exhibit craniofacial and in the developing gut.
cardiac defects64

© 2012 Wiley Periodicals, Inc.


NRG1 Heterozygous SNPs Relatively Isolated65 Nrg1−/− mice die at E10.5 Are multiple isoforms of ND
HGNC:7997 in the coding common65 from heart defects.66 neuregulin-1; type 1 is
sequence that expressed in the dorsal neural
increase the risk tube at the time of neural crest
of HSCR conferred cell emigration.67 ErbB2/ErbB3
by RET 65 receptors are expressed by
migrating neural crest cells.67
SOX10 Heterozygous WS468,69 or PCWH49 Sox10 homozygous null Transcription factor expressed by Required for survival and
HGNC:11190 mutations49,68 or mutants lack neurons in the migrating neural crest cells maintenance of neural
deletions69 entire gastrointestinal including ENCCs.70,74 Also crest stem/progenitors
70,71 +/−
tract. Sox10 mice expressed by enteric glia and and the activation of
lack enteric neurons in the glial progenitors, but is Phox2b and Ret.75–77
distal bowel, with the down-regulated upon enteric Also required later in
severity and penetrance of neuron differentiation.74 development for glial
aganglionosis being and melanocyte
strain-dependent.72,73 differentiation.78,79
Hirschsprung disease
TABLE 1 Continued
Focus Article

Zygosity of Frequency of Expression Pattern in Role in ENS


Mutations in Mutation in Isolated HSCR, Associated ENS Phenotype of Mice Relevant to Development from
Gene HSCR Patients Isolated HSCR Syndrome, Phenotype Mouse Mutants ENS Development Animal Studies
ZEB2 (formerly Heterozyous Mowat–Wilson syndrome; Zeb2 −/− mice die at E9.5 Transcription factor that is Required for the formation
known as nonsense or patients have distinctive and the vagal neural expressed by enteric neural of the vagal neural crest,
ZFHX1B, SIP1) frameshift facial features, epilepsy, crest does not form.83 progenitors and then by and also plays later roles
HGNC:14881 mutations80 or cardiac defects, mental enteric glia but is not in ENS development.83,84
truncating retardation, and a range expressed by enteric
mutations81 of other features neurons.84 Also expressed by
including HSCR82 some mesenchymal cells in
the developing gut.84
PHOX2B Heterozygous for a CCHS (Ondine’s curse). Only Homozygous null mutants Transcription factor expressed Required for the activation
HGNC:9143 polyalanine a small proportion of lack neurons throughout by all ENCCs87 and in of Ret.86
repeat expansion CCHS patients also have entire gastrointestinal differentiated neurons and
mutation85 HSCR.85 tract.86 Phox2b +/− mice glia.88
have no reported ENS
defects.
KIAA1279 Homozygous89 Goldberg–Shprintzen ND KIAA1279 encodes kinesin KBP promotes neurite
HGNC:23419 syndrome; patients have binding protein (KBP). formation and neuronal
microcephaly, mental Expression pattern of KBP differentiation in
retardation, and facial not yet reported in mice. zebrafish brain and

© 2012 Wiley Periodicals, Inc.


dysmorphism. Most also spinal axons,90 and in
have HSCR.89 CNS neurons in culture.91
L1CAM L1CAM is on the X-linked hydrocephalus.92 ENCC migration along the L1CAM is a cell adhesion Required for normal
HGNC:6470 X-chromosome Only a small percentage gut is slightly delayed in molecule that is expressed migration of ENCCs94
of patients also have L1cam null mutants, but by most ENCCs94
HSCR.93 the entire gut is
colonized.94 L1cam+/−
mice have no reported
ENS phenotype

CCHS, central congenital hypoventilation syndrome; ECE-1, endothelin-converting enzyme 1; EDN3, endothelin-3; EDNRB, endothelin receptor B; ENCC, enteric neural crest-derived cells; HGNC, HUGO Gene
Nomenclature Committee; ND, not determined; PCWH, peripheral demyelinating neuropathy, central dysmyelinating leukodystrophy, Waardenburg syndrome and Hirschsprung’s disease; WS4, Waardenburg
syndrome 4.
wires.wiley.com/devbio
WIREs Developmental Biology Hirschsprung disease

belongs to a family of four secreted neurotrophic (a)


factors that bind to GPI-anchored cell surface pro-
teins called GDNF family receptor alphas (GFRAs).95
The GDNF family ligand–GFRA complex activates
the RET tyrosine kinase.96 GDNF preferentially binds A
to GFRA1. GDNF is first expressed by the gut mes-
enchyme shortly before vagal neural crest cells enter S
A
the foregut,97 while RET and GFRA1 are expressed
by ENCCs. Gdnf −/− , Gfra1−/− , and Ret−/− mice
lack neurons in the small and large intestines, and
from most of the stomach37,42,98 (Table 1). In vivo
and in vitro studies have shown that the GDNF- C
GFRA1-RET signaling pathway is essential for the sur- C
M
vival, proliferation, migration, and differentiation of
ENCCs.26,43,99–102 Increasing RET signaling by inac- (b) (c) T (d)A
tivation of negative regulators of RET signaling, or by
increasing GDNF availability, results in an increased
number of enteric neurons.103–105 There are two main
isoforms of RET, which differ in their C-terminal
domains, and are generated by alternate splicing,
RET9 and RET51. Although one study found that
RET9 was critical for colonization of the distal bowel
by ENCCs, but RET51 was not essential,106 another FIGURE 3 | Mouse model of HSCR. (a) Intestine from a 2-week-old
group subsequently found that mice expressing only Edn3 −/− mouse. The mid-colonic region is distended and termed a
RET9 did not exhibit aganglionosis.107,108 This dis- ‘mega-colon’. (b–d) Whole mount preparations of the external muscle
parity is probably due to differences in the transgenes of colonic wall from the regions indicated in (a) showing
or strains used by the two different laboratories. immunostaining with an antibody to the pan-neuronal marker, Hu.
Neurons, which are clustered into ganglia, are present in the distended
region (b), but are totally absent from the distal colon (d). There is a
Neurturin-GFRA2-Ret Signaling Pathway transition zone of reduced neuron density proximal to the aganglionic
Neurturin (NRTN) belongs to the GDNF family lig- region (c). Scale bar = 100 μm (applies to b–d).
ands and binds to GFRA2 to activate RET. Neurons
are present along the entire gastrointestinal tract of Studies in vivo and in vitro have shown that sig-
Nrtn and Gfra2 homozygous null mutant mice; the naling via EDNRB promotes the colonization of
number of myenteric neurons is normal, but the den- the gut by ENCCs by promoting the proliferation
sity of excitatory nerve fibers is reduced.30,109 Thus, and inhibiting the neuronal differentiation of neu-
NRTN-GFRA2-Ret signaling promotes the projection ral progenitors,26,28,29,110 and by directly promoting
or branching of axons from some classes of enteric ENCC migration.25 EDN3-EDNRB signaling is also
neurons. Although studies in mice have not revealed a required for melanocyte development, as mice null
role for NRTN signaling in colonization of the gut by for Ednrb or Edn3 have pigmentation abnormali-
ENCCs, a mutation in NRTN has been found in one ties as well as colonic aganglionosis (WS4 phenotype,
family with HSCR (see the section on Genetics below). see the section on Type IV Waardenburg Syndrome
(WS4)—An Example of a Syndrome That Includes
Aganglionic Megacolon below), whereas Edn3 het-
Endothelin 3-Endothelin Receptor B erozygotes have no abnormalities and Ednrb heterozy-
Signaling Pathway gous and hypomorphic mutants typically only have
Endothelin 3 (EDN3) is a ligand for the G protein- pigmentation abnormalities.28,49 Hence melanocyte
coupled receptor, endothelin receptor B (EDNRB). precursors and ENCCs appear to have different
Edn3 is expressed by the gut mesenchyme, with high- dosage requirements for EDN3/EDNRB signaling,
est levels in the cecum.27,60 Ednrb is expressed by with melanocytes having a lower minimal threshold.
ENCCs and some gut mesenchymal cells.52 Mice lack-
ing EDN3 or EDNRB exhibit colonic aganglionosis62
(Figure 3) resulting from a delay in the entry of vagal Transcription Factors
neural crest-derived cells into the gut as well as reduced A network of transcription factors controls ENS
speed of colonization of the gut by ENCCs.25,27 development.111 Here we very briefly review the

© 2012 Wiley Periodicals, Inc.


Focus Article wires.wiley.com/devbio

roles of SOX10, PHOX2B, and ZEB2 (formerly TABLE 2 Genetic Interactions That Influence the Penetrance and
known as ZFHX1B and SIP1), as mutations in the Severity of Aganglionosis in Mice
genes encoding these molecules are associated with Interacting Genes References
HSCR (Table 1). However, FOXD3, HAND2, ASCL1
Ret and Ednrb 117
(formerly known as MASH1), PAX3, HLX, and the
TFAP2 (formerly known as AP-2) family also play a Ret and Edn3 27
role in ENS development. Sox10 and Ednrb 72
Sox10 is expressed prior to, and during, neural Sox10 and Edn3 28
crest migration.70 Sox10−/− mice die at birth, and Sox10 and Sox8 118
in null mutant embryos, vagal neural crest cells die
Sox10 and L1cam 119
around E9, prior to their entry into the gut.71 SOX10
Sox10 and Zeb2 84
plays an essential role in ENS development most likely
because it is necessary for the expression of Ret and L1cam and Edrnb 120
Phox2b, and because it maintains the multipotency L1cam and Edn3 120
of neural crest cell stem/progenitors.75,76,112,113 The
expression of Sox10 is down-regulated by ENCCs
that differentiate into neurons, but Sox10 expres- other ENCCs and/or other cell types. Mice in which
sion is maintained in enteric glia and is required for ENCCs lack beta1-integrins exhibit aganglionosis
glial fate acquisition.35,78 In the neural crest-derived because beta1-integrins are required to overcome the
melanocyte progenitors that migrate into the ecto- inhibitory effect of high levels of tenascin-C in the
derm, SOX10 regulates the expression of MITF, a key hindgut.24 L1CAM is expressed by most migrating
transcription factor required for melanocyte develop- ENCCs, and perturbation of L1CAM function in vivo
ment, as well as genes required for melanin synthesis or in cultured explants of gut retards the migration of
in melanocytes.79 ENCCs, although the entire length of the gut in L1cam
Phox2b−/− mice do not have any peripheral homozygous null mutants is eventually colonized.94
autonomic neurons, including enteric neurons, and Other molecules shown to be involved in the devel-
die at birth.86 During ENS development, Phox2b is opment of the ENS from animal studies include
first expressed just prior to the entry of vagal neural neurotrophin-3, sonic hedgehog, Indian hedgehog,
crest cells into the gut. Like SOX10, PHOX2B is bone morphogenetic proteins (BMPs) 2 and 4, Notch,
required for Ret expression in ENCCs.86 Although small GTPases, neuregulin, microRNAs, serotonin,
PHOX2B is thought to be involved primarily in the norepinephrine transporter, vitamin A, and the
neuronal specification, Phox2b is expressed by both axon guidance molecules, SEMA3A and netrin.15
enteric neurons and glial cells.88 The activity of the different pathways involved
ZEB2 (Zinc finger E-box-binding homeobox 2) in ENS development must be coordinated, and
is a transcription factor involved in neural specifica- there is substantial evidence for interactions between
tion and in epithelial–mesenchymal transition (EMT) pathways. The first evidence that interactions between
during early neural crest development. Zeb2−/− mice pathways influence ENS development came from
die around E9.5 and exhibit a variety of cardiovas- studies of humans with HSCR.115,116 Subsequent
cular and neural defects, including a failure of the studies in mice have shown that interactions between a
vagal neural crest to form.83 Mice with targeted abla- variety of signaling pathways influence the penetrance
tion of Zeb2 in neural crest cells have craniofacial, and severity of aganglionosis (Table 2). Some of the
heart, pigment, and peripheral nervous system defor- interactions are direct, for example, SOX10 binds
mities as well as aganglionosis of the entire colon directly to Ednrb regulatory regions,112 whereas other
that extends into the small intestine.114 Zeb2 has a interactions may be indirect.
similar expression pattern to Sox10 in the developing
ENS,84 but the molecular mechanism by which ZEB2
HIRSCHSPRUNG DISEASE
regulates enteric neuron development has not yet been
elucidated. Etiology
Studies using animal models have shown that the
absence of enteric neurons from variable lengths of the
Other Molecules and Interactions gastrointestinal tract in HSCR results from a failure
ENCCs express cell surface molecules, includ- of ENCCs to colonize the affected gut regions during
ing beta1-integrins and the cell adhesion molecule development (Figures 3 and 4). Aganglionosis is asso-
L1CAM, which regulate interactions with the ECM, ciated with a delay in the entry of neural crest-derived

© 2012 Wiley Periodicals, Inc.


WIREs Developmental Biology Hirschsprung disease

HSCR when combined with other mutations.1,124 For


example, a common noncoding mutation, a T>C SNP
C
S lying within an intronic RET enhancer, has a 20-fold
C greater contribution to HSCR susceptibility than RET
C
A coding sequence mutations.123 This mutation appears
S to account for some, but not all, of the sex ratio in
W

S
HSCR.1,123 Like mutations in most genes associated
with HSCR, the penetrance of RET mutations,
C even coding sequence mutations, is incomplete. For
S instance, a missense mutation in RET was found in
C four members of one family, of which one had long
A
C segment HSCR, two had short segment HSCR, and
S S one was unaffected.125
There appear to be some phenotypic differences
in mice and humans with reduced RET signaling.
FIGURE 4 | Diagram showing the location of ENCCs along the gut Unlike humans, Ret heterozygous mice do not show
(green) in E10.5 and E12.5 wild-type and Edn3 −/− mice. Edn3 an HSCR-like phenotype30,37 (Table 1). However,
homozygous null mutants are a mouse model of Hirschsprung’s disease. colonic aganglionosis can be observed in mice when
Already at E10.5, ENCCs are not as caudally advanced along the gut in the levels of Ret are reduced to around 40% of
Edn3 null mice compared to wild-type mice.27 There is a similar delayed wild-type levels.126 In addition to intestinal agan-
entry into the gut in Ednrb homozygous null mutants.25 (Reprinted with glionosis, Ret null homozygous mutant mice lack
permission from Ref 122. Copyright 2001 John Wiley and Sons) kidneys and have sympathetic and parasympathetic
neuron defects. In contrast, infants with HSCR only
cells into the foregut, as well as a delayed progression extremely rarely have kidney defects. Studies in mice
of ENCCs along the gut.25,27 The delay is exacerbated have shown that different tissues appear to require
by changes to the gut microenvironment with age that distinct RET-stimulated signaling pathways, which
make it less permissive for ENCC migration.25 Defects might partly explain why intestinal aganglionosis can
in multiple processes can contribute to the delay in occur as an isolated defect in HSCR patients with
the colonization of the gut by ENCC (see the section RET mutations.108
Origin and Early Development of the Enteric Nervous In addition to RET, mutations in a dozen
System above), but ENCC proliferation is critical, as or more genes have been associated with HSCR
a minimal number of ENCCs are required for normal (Table 1). Although mutation in a single gene (RET,
ENS formation.21,23 Although cell death does not play EDNRB, EDN3) can be sufficient for the development
a major role during normal ENS development, it may of HSCR, patients with mutations in two genes
contribute to the etiology of HSCR.121 have been identified, and complex genetic interactions
contribute to the variability in penetrance and severity
(length of the aganglionic region). Most of the genes
Genetics associated with HSCR in humans also result in
The genetics of HSCR are complex and are reviewed aganglionosis in mice following inactivation (Table 1).
in detail by Amiel et al.1 Mutations in genes encoding However, a heterozygous mutation in NRTN that co-
members of a variety of signaling pathways are occurred with a RET mutation was found in one
associated with HSCR (Table 1), but mutations in family with HSCR,44 but mice lacking NRTN do
the coding sequence of these known genes account not have aganglionosis.30 L1CAM is the only known
for only about 50% of familial cases of HSCR and X-linked gene associated with HSCR, but as only
15% of sporadic cases of HSCR.1 RET is the major 3% of patients with mutations in L1CAM also have
susceptibility gene as more than 80% of identified HSCR, L1CAM is thought to act as an X-linked HSCR
mutations associated with HSCR are in RET; these modifier gene.127 Most of the syndromes involving an
include both coding and noncoding mutations.123,124 HSCR phenotype are each associated with mutations
RET coding sequence mutations have been identified in one or a small number of genes (Table 1).
in 20–30% of HSCR cases, and alone, can result in Copy number variants (CNVs), which are struc-
HSCR by haploinsufficiency.1 Noncoding mutations tural variations in DNA that result in an abnormal
are more common and result in reduced amounts of number of copies of sections of DNA, have been
wild-type RET protein due to a reduction in RET associated with susceptibility or resistance to some
transcription; these mutations probably only result in human diseases. Data from a recent study of 67

© 2012 Wiley Periodicals, Inc.


Focus Article wires.wiley.com/devbio

proven or candidate HSCR genes from human and is predominantly excitatory (mediates muscle con-
mice studies suggest that CNVs, particularly in regu- traction), and may even be hypertrophied in some
latory sequences, can play a role in HSCR.128 HSCR patients.4,130 Relaxation of the gut wall during
HSCR has become a paradigm for understanding propulsive motility is normally mediated by inhibitory
complex multigenetic disorders. For example, studies enteric (intrinsic) motor neurons, and so an absence of
of HSCR have shown that small structural variants in intrinsic inhibitory neurons may also contribute to the
a gene can act as powerful genetic modifiers in human aganglionic segment being persistently constricted.130
disease. Furthermore HSCR studies have blurred the Muscle hypertrophy also occurs in the aganglionic
definition of modifier gene, as depending on the segment in mouse and rat models of HSCR,131,132
mutation, the same gene can act as a principal gene or probably as a secondary effect.
a modifier gene. For example, large genomic mutations Perturbation of intestinal motility is not lim-
in RET, alone, can result in HSCR (‘principal gene’) ited to the aganglionic gut segment. HSCR patients
but small structural variants in RET probably only often have motility disturbances following surgical
result in HSCR when combined with other mutations resection. In mouse models of HSCR, spontaneous
(‘modifier gene’).124 Humans appear to be more propagating motility patterns are absent or abnormal
sensitive to decreased levels of RET and EDNRB along the entire colon, including the proximal colon
signaling than mice, which might be due, at least (oral to the transition zone) where enteric ganglia are
in part, to the larger area of bowel that has to be present.51,59 The most likely cause is a reduction in
colonized by ENCCs in humans compared with mice. enteric neuron density, although inflammation and
Although it has been assumed that all cases damage to the interstitial cells of Cajal (ICC), a pop-
of HSCR have a genetic basis, vitamin A deficiency ulation of cells involved in gut motility, in regions
increases the penetrance and severity of aganglionosis proximal to the aganglionic bowel could also con-
in a mouse model of HSCR.129 Thus, environmental tribute to abnormal motility.133
factors could contribute to susceptibility to HSCR.

Current Treatment and Potential Therapies


Diagnosis, Pathology, and Physiology If left untreated, HSCR can be fatal due to enterocoli-
Healthy infants normally pass meconium (the earliest tis, perforation of the bowel, or malnutrition. Current
stools) within the first 24–48 h following birth. Most treatment for HSCR involves the surgical removal of
infants with HSCR, however, fail to pass meconium the defective, aganglionic bowel, and reanastomosis
and suffer from severe constipation. Other symptoms of the distal-most, normal (ganglionated) bowel to
include gradual distension of the abdomen, vomiting, the anus. Depending on the severity of the disease
and fever. Although infants with HSCR normally (length of aganglionosis) and the general health of the
show symptoms within the first weeks after birth, patient, the surgery may be performed in one or two
other children with HSCR may not exhibit symptoms stages; for patients with long-segment aganglionosis or
for several months. major health problems, an initial primary colostomy
HSCR is usually diagnosed by a barium enema is often performed with the resection of the agan-
(Figure 1), anorectal manometry, and a biopsy of the glionic bowel performed later. While these operative
rectum. The pathological evaluation of rectal biopsies procedures are lifesaving, they can be associated with
for HSCR is reviewed by Kapur.4 Most laborato- acute (anastomotic stricture, enterocolitis) and chronic
ries rely on hematoxylin and eosin-stained paraffin (constipation, fecal incontinence) complications.
sections to identify neuronal cell bodies, although his- Cell transplantation therapy has been proposed
tochemical and immunohistochemical techniques are as an alternative to surgical resection of aganglionic
also used by some pathologists to assist the diagnosis. bowel for the treatment of HSCR.10,57,134,135
During surgery to remove the aganglionic region of This would involve the transplantation of neural
bowel, frozen sections are taken to localize the bound- stem/progenitor cells into the aganglionic bowel,
ary between the ganglionic and aganglionic regions which would colonize the defective region, generate
prior to resection.4 neurons, and form the neuronal circuits required for
The megacolon and severe constipation experi- propulsive motility. Neural stem/progenitor cells can
enced by patients with HSCR are caused by a lack be isolated from the bowel of postnatal and adult
of propulsive motility patterns in the distal bowel laboratory animals and humans, including HSCR
due to the lack of intrinsic enteric neurons. The patients.10,136 These cells are capable of both self-
persistent constriction of the aganglionic segment is renewal and of giving rise to multiple lineages in vitro
probably mediated by the extrinsic innervation, which including neurons and glia (Figure 5). Pluripotent stem

© 2012 Wiley Periodicals, Inc.


WIREs Developmental Biology Hirschsprung disease

(a) (b)

FIGURE 5 | Enteric neural crest stem/progenitor cells form neurospheres in vitro. (a) Neural crest stem/progenitors were isolated from the gut of
an E14.5 mouse and cultured in suspension conditions. Neurospheres formed, which were then plated onto fibronectin for 48 h to allow for migration
of neural crest-like cells (identified using an antibody to SOX10, green, Santa Cruz) and neurite outgrowth (identified using the neuronal marker,
TUBB3, formerly known as TuJ1, red, Covance). (b) Higher magnification image of the outgrowth of TUBB3+ neurites and SOX10+ cells that have
migrated away from the neurosphere. There are also a small number of TUBB3+ cell bodies in the outgrowth (arrows ). Scale bars: 200 μm (a);
50 μm (b).

cells, such as embryonic stem (ES) cells and induced occurred spontaneously and is a single base pair
pluripotent stem (iPS) cells, are also a potential source insertion that results in loss of the SOX10 pro-
of cells to generate enteric neurons as ES cells can tein transcriptional activation domain.70 Sox10Dom/+
give rise to neural crest-like cells and to neurons when mice exhibit aganglionosis and pigmentation defects
co-cultured with explants of embryonic gut.137,138 iPS and therefore exhibit a similar phenotype to humans
cells and ‘adult’ stem cells have the added benefit of with WS4. The severity and penetrance of agan-
being patient-derived and thus have reduced chance glionosis in Sox10Dom/+ mice varies with the genetic
of immunological rejection after engraftment. Cell background.72 SOX10 mutations in humans can
therapy to treat HSCR is currently a very active field also result in a more severe phenotype that also
of research.10,57,134,135 includes central and peripheral demyelinating neu-
ropathies, which is called PCWH syndrome (periph-
eral demyelinating neuropathy, central dysmyelinat-
TYPE IV WAARDENBURG SYNDROME ing leukodystrophy, Waardenburg syndrome, and
(WS4)—AN EXAMPLE OF A Hirschsprung disease).69 This neurological phenotype
SYNDROME THAT INCLUDES has been mostly associated with SOX10 mutations
resulting in a dominant negative effect, rather than
AGANGLIONIC MEGACOLON
haploinsufficiency.69
There are four types of Waardenburg syndrome, Of the approximately 20–30% cases of WS4 due
all of which involve sensorineural hearing loss and to mutations in EDNRB or EDN3, about 70% are
pigmentation abnormalities.49 Type IV Waardenburg recessive (homozygous or compound heterozygous)
syndrome (WS4) also involves HSCR and sometimes mutations of EDNRB or EDN3.49 Heterozygous
neurological features. The sensorineural hearing loss mutations in EDNRB or EDN3 are most commonly
and depigmentation are due to abnormal development associated with isolated HSCR, although rare cases of
of neural crest-derived melanocytes in the stria WS4 with heterozygous EDNRB mutations have been
vascularis of the cochlea, skin, hair, or eyes.139 reported.49
Mutations in three genes, SOX10, EDNRB, and Phenotypes associated with SOX10, EDNRB
EDN3, account for approximately 60–80% of cases and EDN3 mutations show a large degree of
of WS4.69 These three genes are required for the variability.49 Moreover, heterozygous disruptions in
development of both the ENS and melanocytes (see SOX10 account for approximately 15% of cases of
the section Origin and Early Development of the Type II Waardenburg syndrome (WS2), which lacks
Enteric Nervous System above). HSCR,69 while heterozygous mutations in EDNRB
Heterozygous mutations in SOX10 account for or EDN3 have also been associated with WS2.49
approximately 50% of WS4 cases, mostly occur- These findings further highlight the genetic complexity
ring de novo.49 The Sox10 ‘Dom’ mutation in mice of HSCR and Waardenburg syndromes, and the

© 2012 Wiley Periodicals, Inc.


Focus Article wires.wiley.com/devbio

importance of the genetic background for the severity the molecular mechanisms underlying the interactions
of the phenotype. between pathways, and how particular mutations
impact on those interactions, are not well understood.
The 4:1 male:female bias in the incidence of HSCR
CONCLUSION is only partly explained by current knowledge. The
Molecular genetics of HSCR patients and studies using transplantation of neural stem/progenitor cells into
animal models of HSCR have, in parallel, revealed the aganglionic region of HSCR patients, as an alter-
much about the pathogenesis of HSCR. However, native treatment to surgical removal, is an exciting,
there are many outstanding questions. For example, but currently untested possibility.

ACKNOWLEDGMENTS
The authors’ work is supported by NHMRC (Australia) Senior Research Fellowship #1002506, CJ Martin
Fellowship #400433 and Project grant #546473. We thank Annette Bergner for assistance with obtaining the
images shown in Figures 2 and 3, and Lauren Young and John Stephenson for assistance with processing the
embryos shown in Figure 2.

REFERENCES
1. Amiel J, Sproat-Emison E, Garcia-Barcelo M, Lan- human gut mucosa for the treatment of aganglionic gut
tieri F, Burzynski G, Borrego S, Pelet A, Arnold S, disorders. Gastroenterology 2009, 136:2214–2225
Miao X, Griseri P, et al. Hirschsprung disease, associ- 11. Le Douarin NM, Teillet MA. Experimental analysis
ated syndromes and genetics: a review. J Med Genet of the migration and differentiation of neuroblasts of
2008, 45:1–14. the autonomic nervous system and of neurectoder-
2. Whitehouse FR, Kernohan JW. Myenteric plexus in mal mesenchymal derivatives, using a biological cell
congenital megacolon; study of 11 cases. Arch Intern marking technique. Dev Biol 1974, 41:162–184.
Med (Chic) 1948, 82:75–111. 12. Burns AJ, Le Douarin NM. The sacral neural crest
3. Moore SW. Down syndrome and the enteric nervous contributes neurons and glia to the post-umbilical
system. Pediatr Surg Int 2008, 24:873–883. gut: spatiotemporal analysis of the development of
the enteric nervous system. Development (Cambridge,
4. Kapur RP. Practical pathology and genetics of
England) 1998, 125:4335–4347.
Hirschsprung’s disease. Semin Pediatr Surg 2009,
18:212–223. 13. Kapur RP. Colonization of the murine hindgut by
sacral crest-derived neural precursors: experimental
5. Berrocal T, Lamas M, Gutieerrez J, Torres I, Prieto
support for an evolutionarily conserved model. Dev
C, del Hoyo ML. Congenital anomalies of the small
Biol 2000, 227:146–155.
intestine, colon, and rectum. Radiographics 1999,
19:1219–1236. 14. Wang X, Chan AK, Sham MH, Burns AJ, Chan WY.
Analysis of the sacral neural crest cell contribution
6. Burzynski G, Shepherd IT, Enomoto H. Genetic model to the hindgut enteric nervous system in the mouse
system studies of the development of the enteric ner- embryo. Gastroenterology 2011, 141:992–1002.
vous system, gut motility and Hirschsprung’s disease.
15. Young HM, Newgreeen DF, Enomoto H. Develop-
Neurogastroenterol Motil 2009, 21:113–127.
ment of the enteric nervous system. In: Johnson L,
7. Furness JB. The Enteric Nervous System. MA: Black- ed. Physiology of the Gastrointestinal Tract. 5th ed.
well Publishing; 2006. Burlington, MA: Elsevier; 2012.
8. Gershon MD. Developmental determinants of the 16. Kapur RP, Yost C, Palmiter RD. A transgenic model
independence and complexity of the enteric nervous for studying development of the enteric nervous sys-
system. Trends Neurosci 2010, 33:446–456. tem in normal and aganglionic mice. Development
9. Karaosmanoglu T, Aygun B, Wade PR, Gershon MD. (Cambridge, England) 1992, 116:167–175.
Regional differences in the number of neurons in the 17. Wallace AS, Burns AJ. Development of the enteric
myenteric plexus of the guinea pig small intestine and nervous system, smooth muscle and interstitial cells of
colon: an evaluation of markers used to count neurons. Cajal in the human gastrointestinal tract. Cell Tissue
Anat Rec 1996, 244:470–480. Res 2005, 319:367–382.
10. Metzger M, Caldwell C, Barlow AJ, Burns AJ, Tha- 18. Newgreen DF, Southwell B, Hartley L, Allan IJ.
par N. Enteric nervous system stem cells derived from Migration of enteric neural crest cells in relation to

© 2012 Wiley Periodicals, Inc.


WIREs Developmental Biology Hirschsprung disease

growth of the gut in avian embryos. Acta Anat 1996, requirements for endothelin receptor B in the gen-
157:105–115. eration and migration of gut neural crest stem cells.
19. Pham TD, Gershon MD, Rothman TP. Time of ori- Neuron 2003, 40:917–929.
gin of neurons in the murine enteric nervous system: 32. Wallace AS, Barlow AJ, Navaratne L, Delalande JM,
sequence in relation to phenotype. J Comp Neurol Tauszig-Delamasure S, Corset V, Thapar N, Burns AJ.
1991, 314:789–798. Inhibition of cell death results in hyperganglionosis:
20. Roberts RR, Ellis M, Gwynne RM, Bergner AJ, Lewis implications for enteric nervous system development.
MD, Beckett EA, Bornstein JC, Young HM. The first Neurogastroenterol Motil 2009, 21:768–e49.
intestinal motility patterns in fetal mice are not medi- 33. Uesaka T, Jain S, Yonemura S, Uchiyama Y,
ated by neurons or interstitial cells of Cajal. J Physiol Milbrandt J, Enomoto H. Conditional ablation of
2010, 588:1153–1169. GFRalpha1 in postmigratory enteric neurons trig-
21. Barlow AJ, Wallace AS, Thapar N, Burns AJ. Critical gers unconventional neuronal death in the colon and
numbers of neural crest cells are required in the path- causes a Hirschsprung’s disease phenotype. Develop-
ways from the neural tube to the foregut to ensure ment (Cambridge, England) 2007, 134:2171–2181.
complete enteric nervous system formation. Develop- 34. Pachnis V, Mankoo B, Costantini F. Expression
ment (Cambridge, England) 2008, 135:1681–1691. of the c-ret proto-oncogene during mouse embryo-
22. Simpson MJ, Landman KA, Hughes BD, Newgreen genesis. Development (Cambridge, England) 1993,
DF. Cell proliferation drives neural crest cell invasion 119:1005–1017.
of the intestine. Dev Biol 2007, 302:553–568. 35. Young HM, Bergner AJ, Muller T. Acquisition of neu-
23. Yntema CL, Hammond WS. The origin of intrinsic ronal and glial markers by neural crest-derived cells in
ganglia of trunk viscera from vagal neural crest in the the mouse intestine. J Comp Neurol 2003, 456:1–11.
chick embryo. J Comp Neurol 1954, 101:515–541. 36. Inoue K, Shimotake T, Iwai N. Mutational analy-
24. Breau MA, Dahmani A, Broders-Bondon F, Thiery JP, sis of RET/GDNF/NTN genes in children with total
Dufour S. Beta1 integrins are required for the invasion colonic aganglionosis with small bowel involvement.
of the caecum and proximal hindgut by enteric neural Am J Med Genet 2000, 93:278–284.
crest cells. Development (Cambridge, England) 2009, 37. Schuchardt A, D’Agati V, Larsson-Blomberg L,
136:2791–2801. Costantini F, Pachnis V. Defects in the kidney and
25. Druckenbrod NR, Epstein ML. Age-dependent enteric nervous system of mice lacking the tyrosine
changes in the gut environment restrict the invasion kinase receptor Ret. Nature 1994, 367:380–383.
of the hindgut by enteric neural progenitors. Develop- 38. Ivanchuk SM, Myers SM, Eng C, Mulligan LM. De
ment (Cambridge, England) 2009, 136:3195–3203. novo mutation of GDNF, ligand for the RET/GDNFR-
26. Hearn CJ, Murphy M, Newgreen D. GDNF and ET-3 alpha receptor complex, in Hirschsprung disease. Hum
differentially modulate the numbers of avian enteric Mol Genet 1996, 5:2023–2026.
neural crest cells and enteric neurons in vitro. Dev Biol 39. Sanchez MP, Silos-Santiago I, Frisen J, He B, Lira
1998, 197:93–105. SA, Barbacid M. Renal agenesis and the absence of
27. Barlow A, de Graaff E, Pachnis V. Enteric nervous enteric neurons in mice lacking GDNF. Nature 1996,
system progenitors are coordinately controlled by the 382:70–73.
G protein-coupled receptor EDNRB and the receptor 40. Peters RJ, Osinski MA, Hongo JA, Bennett GL,
tyrosine kinase RET. Neuron 2003, 40:905–916. Okragly AJ, Haak-Frendscho M, Epstein ML. GDNF
28. Stanchina L, Baral V, Robert F, Pingault V, Lemort N, is abundant in the adult rat gut. J Auton Nerv Sys
Pachnis V, Goossens M, Bondurand N. Interactions 1998, 70:115–122.
between Sox10, Edn3 and Ednrb during enteric ner- 41. Borrego S, Fernandez RM, Dziema H, Niess A,
vous system and melanocyte development. Dev Biol Lopez-Alonso M, Antinolo G, Eng C. Investigation
2006, 295:232–249. of germline GFRA4 mutations and evaluation of
29. Wu JJ, Chen JX, Rothman TP, Gershon MD. the involvement of GFRA1, GFRA2, GFRA3, and
Inhibition of in vitro enteric neuronal develop- GFRA4 sequence variants in Hirschsprung disease.
ment by endothelin-3: mediation by endothelin B J Med Genet 2003, 40:e18.
receptors. Development (Cambridge, England) 1999, 42. Enomoto H, Araki T, Jackman A, Heuckeroth RO,
126:1161–1173. Snider WD, Johnson EM Jr, Milbrandt J. GFR alpha1-
30. Gianino S, Grider JR, Cresswell J, Enomoto H, deficient mice have deficits in the enteric nervous
Heuckeroth RO. GDNF availability determines enteric system and kidneys. Neuron 1998, 21:317–324.
neuron number by controlling precursor prolifer- 43. Young HM, Hearn CJ, Farlie PG, Canty AJ, Thomas
ation. Development (Cambridge, England) 2003, PQ, Newgreen DF. GDNF is a chemoattractant for
130:2187–2198. enteric neural cells. Dev Biol 2001, 229:503–516.
31. Kruger GM, Mosher JT, Tsai YH, Yeager KJ, Iwashita 44. Doray B, Salomon R, Amiel J, Pelet A, Touraine R,
T, Gariepy CE, Morrison SJ. Temporally distinct Billaud M, Attie T, Bachy B, Munnich A, Lyonnet S.

© 2012 Wiley Periodicals, Inc.


Focus Article wires.wiley.com/devbio

Mutation of the RET ligand, neurturin, supports multi- Willems PJ, et al. Endothelin-3 gene mutations in iso-
genic inheritance in Hirschsprung disease. Hum Mol lated and syndromic Hirschsprung disease. Eur J Hum
Genet 1998, 7:1449–1452. Genet 1997, 5:247–251.
45. Heuckeroth RO, Enomoto H, Grider JR, Golden 57. Heanue TA, Pachnis V. Enteric nervous system
JP, Hanke JA, Jackman A, Molliver DC, Bardgett development and Hirschsprung’s disease: advances
ME, Snider WD, Johnson EM Jr, Milbrandt J. Gene in genetic and stem cell studies. Nat Rev 2007,
targeting reveals a critical role for neurturin in the 8:466–479.
development and maintenance of enteric, sensory, and 58. Pingault V, Bondurand N, Lemort N, Sancandi M,
parasympathetic neurons. Neuron 1999, 22:253–263. Ceccherini I, Hugot JP, Jouk PS, Goossens M. A
46. Golden JP, DeMaro JA, Osborne PA, Milbrandt J, heterozygous endothelin 3 mutation in Waardenburg-
Johnson EM Jr. Expression of neurturin, GDNF, and Hirschsprung disease: is there a dosage effect of
GDNF family-receptor mRNA in the developing and EDN3/EDNRB gene mutations on neurocristopathy
mature mouse. Exp Neurol 1999, 158:504–528. phenotypes? J Med Genet 2001, 38:205–209.
47. Puffenberger EG, Hosoda K, Washington SS, Na- 59. Roberts RR, Bornstein JC, Bergner AJ, Young HM.
kao K, deWit D, Yanagisawa M, Chakravart A. Disturbances of colonic motility in mouse mod-
A missense mutation of the endothelin-B receptor els of Hirschsprung’s disease. Am J Physiol 2008,
gene in multigenic Hirschsprung’s disease. Cell 1994, 294:G996–G1008.
79:1257–1266. 60. Leibl MA, Ota T, Woodward MN, Kenny SE, Lloyd
48. Amiel J, Attie T, Jan D, Pelet A, Edery P, Bidaud C, DA, Vaillant CR, Edgar DH. Expression of endothelin
Lacombe D, Tam P, Simeoni J, Flori E, et al. Het- 3 by mesenchymal cells of embryonic mouse caecum.
erozygous endothelin receptor B (EDNRB) mutations Gut 1999, 44:246–252.
in isolated Hirschsprung disease. Hum Mol Genet 61. Edery P, Attie T, Amiel J, Pelet A, Eng C, Hofstra RM,
1996, 5:355–357. Martelli H, Bidaud C, Munnich A, Lyonnet S. Muta-
49. Pingault V, Ente D, Dastot-Le Moal F, Goossens M, tion of the endothelin-3 gene in the Waardenburg-
Marlin S, Bondurand N. Review and update of muta- Hirschsprung disease (Shah-Waardenburg syndrome).
tions causing Waardenburg syndrome. Hum Mutat Nat Genet 1996, 12:442–444.
2010, 31:391–406. 62. Baynash AG, Hosoda K, Giaid A, Richardson JA,
50. Syrris P, Carter ND, Patton MA. Novel nonsense Emoto N, Hammer RE, Yanagisawa M. Interaction
mutation of the endothelin-B receptor gene in a family of endothelin-3 with endothelin-B receptor is essential
with Waardenburg-Hirschsprung disease. Am J Med for development of epidermal melanocytes and enteric
Genet 1999, 87:69–71. neurons. Cell 1994, 79:1277–1285.
63. Hofstra RM, Valdenaire O, Arch E, Osinga J, Kroes
51. Ro S, Hwang SJ, Muto M, Jewett WK, Spencer
H, Loffler BM, Hamosh A, Meijers C, Buys CH. A
NJ. Anatomic modifications in the enteric nervous
loss-of-function mutation in the endothelin-converting
system of piebald mice and physiological conse-
enzyme 1 (ECE-1) associated with Hirschsprung dis-
quences to colonic motor activity. Am J Physiol 2006,
ease, cardiac defects, and autonomic dysfunction. Am
290:G710–G718.
J Hum Genet 1999, 64:304–308.
52. Lee HO, Levorse JM, Shin MK. The endothelin
64. Yanagisawa H, Yanagisawa M, Kapur RP, Richard-
receptor-B is required for the migration of neural
son JA, Williams SC, Clouthier DE, de Wit D,
crest-derived melanocyte and enteric neuron precur-
Emoto N, Hammer RE. Dual genetic pathways of
sors. Dev Biol 2003, 259:162–175.
endothelin-mediated intercellular signaling revealed by
53. Bondurand N, Natarajan D, Barlow A, Thapar N, targeted disruption of endothelin converting enzyme-
Pachnis V. Maintenance of mammalian enteric ner- 1 gene. Development (Cambridge, England) 1998,
vous system progenitors by SOX10 and endothelin 3 125:825–836.
signalling. Development (Cambridge, England) 2006,
65. Garcia-Barcelo MM, Tang CS, Ngan ES, Lui VC,
133:2075–2086.
Chen Y, So MT, Leon TY, Miao XP, Shum CK, Liu
54. Hosoda K, Hammer RE, Richardson JA, Baynash FQ, et al. Genome-wide association study identifies
AG, Cheung JC, Giaid A, Yanagisawa M. Targeted NRG1 as a susceptibility locus for Hirschsprung’s dis-
and natural (piebald-lethal) mutations of endothelin- ease. Proc Natl Acad Sci USA 2009, 106:2694–2699.
B receptor gene produce megacolon associated with 66. Meyer D, Birchmeier C. Multiple essential func-
spotted coat color in mice. Cell 1994, 79:1267–1276. tions of neuregulin in development. Nature 1995,
55. Attie T, Till M, Pelet A, Amiel J, Edery P, Boutrand L, 378:386–390.
Munnich A, Lyonnet S. Mutation of the endothelin- 67. Britsch S, Li L, Kirchhoff S, Theuring F, Brinkmann V,
receptor B gene in Waardenburg-Hirschsprung dis- Birchmeier C, Riethmacher D. The ErbB2 and ErbB3
ease. Hum Mol Genet 1995, 4:2407–2409. receptors and their ligand, neuregulin-1, are essential
56. Bidaud C, Salomon R, Van Camp G, Pelet A, Attie T, for development of the sympathetic nervous system.
Eng C, Bonduelle M, Amiel J, Nihoul-Fekete C, Genes Dev 1998, 12:1825–1836.

© 2012 Wiley Periodicals, Inc.


WIREs Developmental Biology Hirschsprung disease

68. Pingault V, Bondurand N, Kuhlbrodt K, Goerich DE, Yamanaka T, Mushiake K, et al. Mutations in SIP1,
Prehu MO, Puliti A, Herbarth B, Hermans-Borgmeyer encoding Smad interacting protein-1, cause a form of
I, Legius E, Matthijs G, et al. SOX10 mutations in Hirschsprung disease. Nat Genet 2001, 27:369–370.
patients with Waardenburg-Hirschsprung disease. Nat 81. Zweier C, Albrecht B, Mitulla B, Behrens R, Beese M,
Genet 1998, 18:171–173. Gillessen-Kaesbach G, Rott HD, Rauch A. ‘‘Mowat-
69. Bondurand N, Dastot-Le Moal F, Stanchina L, Col- Wilson’’ syndrome with and without Hirschsprung
lot N, Baral V, Marlin S, Attie-Bitach T, Giurgea I, disease is a distinct, recognizable multiple congeni-
Skopinski L, Reardon W, et al. Deletions at the SOX10 tal anomalies-mental retardation syndrome caused by
gene locus cause Waardenburg syndrome types 2 and mutations in the zinc finger homeo box 1B gene. Am
4. Am J Hum Genet 2007, 81:1169–1185. J Med Genet 2002, 108:177–181.
70. Southard-Smith EM, Kos L, Pavan WJ. Sox10 muta- 82. Garavelli L, Donadio A, Zanacca C, Banchini G,
tion disrupts neural crest development in Dom Della Giustina E, Bertani G, Albertini G, Del Rossi C,
Hirschsprung mouse model. Nat Genet 1998, 18: Zweier C, Rauch A, et al. Hirschsprung disease, men-
60–64. tal retardation, characteristic facial features, and muta-
71. Kapur RP. Early death of neural crest cells is tion in the gene ZFHX1B (SIP1): confirmation of the
responsible for total enteric aganglionosis in Sox10 Mowat-Wilson syndrome. Am J Med Genet A 2003,
(Dom)/Sox10(Dom) mouse embryos. Pediatr Dev 116A:385–388.
Pathol 1999, 2:559–569. 83. Van De Putte T, Maruhashi M, Francis A, Nelles L,
Kondoh H, Huylebroeck D, Higashi Y. Mice lack-
72. Cantrell VA, Owens SE, Chandler RL, Airey DC,
ing Zfhx1b, the gene that codes for Smad-interacting
Bradley KM, Smith JR, Southard-Smith EM. Interac-
protein-1, reveal a role for multiple neural crest cell
tions between Sox10 and EdnrB modulate penetrance
defects in the etiology of Hirschsprung disease-mental
and severity of aganglionosis in the Sox10Dom mouse
retardation syndrome. Am J Hum Genet 2003, 72:2.
model of Hirschsprung disease. Hum Mol Genet 2004,
13:2289–2301. 84. Stanchina L, Van de Putte T, Goossens M, Huyle-
broeck D, Bondurand N. Genetic interaction between
73. Walters LC, Cantrell VA, Weller KP, Mosher JT,
Sox10 and Zfhx1b during enteric nervous system
Southard-Smith EM. Genetic background impacts
development. Dev Biol 2010, 341:416–428.
developmental potential of enteric neural crest-derived
progenitors in the Sox10Dom model of Hirschsprung 85. Amiel J, Laudier B, Attie-Bitach T, Trang H, De Pon-
disease. Hum Mol Genet 2010, 19:4353–4372. tual L, Gener B, Trochet D, Etchevers H, Ray P, Simon-
neau M, et al. Polyalanine expansion and frameshift
74. Young HM, Jones BR, McKeown SJ. The projections
mutations of the paired-like homeobox gene PHOX2B
of early enteric neurons are influenced by the direc-
in congenital central hypoventilation syndrome. Nat
tion of neural crest cell migration. J Neurosci 2002,
Genet 2003, 33:459–461.
22:6005–6018.
86. Pattyn A, Morin X, Cremer H, Goridis C, Brunet JF.
75. Lang D, Chen F, Milewski R, Li J, Lu MM, Epstein
The homeobox gene Phox2b is essential for the devel-
JA. Pax3 is required for enteric ganglia formation and
opment of autonomic neural crest derivatives. Nature
functions with Sox10 to modulate expression of c-ret.
1999, 399:366–370.
J Clin Invest 2000, 106:963–971.
87. Young HM, Hearn CJ, Ciampoli D, Southwell BR,
76. Paratore C, Eichenberger C, Suter U, Sommer L. Sox10
Brunet JF, Newgreen DF. A single rostrocaudal col-
haploinsufficiency affects maintenance of progenitor
onization of the rodent intestine by enteric neuron
cells in a mouse model of Hirschsprung disease. Hum precursors is revealed by the expression of Phox2b,
Mol Genet 2002, 11:3075–3085. Ret, and p75 and by explants grown under the kid-
77. Kim J, Lo L, Dormand E, Anderson DJ. SOX10 ney capsule or in organ culture. Dev Biol 1998,
maintains multipotency and inhibits neuronal differ- 202:67–84.
entiation of neural crest stem cells. Neuron 2003, 88. Corpening JC, Cantrell VA, Deal KK, Southard-Smith
38:17–31. EM. A Histone2BCerulean BAC transgene identifies
78. Paratore C, Goerich DE, Suter U, Wegner M, Som- differential expression of Phox2b in migrating enteric
mer L. Survival and glial fate acquisition of neural neural crest derivatives and enteric glia. Dev Dyn
crest cells are regulated by an interplay between the 2008, 237:1119–1132.
transcription factor Sox10 and extrinsic combinatorial 89. Brooks AS, Bertoli-Avella AM, Burzynski GM, Breed-
signaling. Development (Cambridge, England) 2001, veld GJ, Osinga J, Boven LG, Hurst JA, Mancini GM,
128:3949–3961. Lequin MH, de Coo RF, et al. Homozygous nonsense
79. Harris ML, Baxter LL, Loftus SK, Pavan WJ. Sox mutations in KIAA1279 are associated with malfor-
proteins in melanocyte development and melanoma. mations of the central and enteric nervous systems.
Pigment Cell Melanoma Res 2010, 23:496–513. Am J Hum Genet 2005, 77:120–126.
80. Wakamatsu N, Yamada Y, Yamada K, Ono T, 90. Lyons DA, Naylor SG, Mercurio S, Dominguez C,
Nomura N, Taniguchi H, Kitoh H, Mutoh N, Talbot WS. KBP is essential for axonal structure,

© 2012 Wiley Periodicals, Inc.


Focus Article wires.wiley.com/devbio

outgrowth and maintenance in zebrafish, providing 103. Taketomi T, Yoshiga D, Taniguchi K, Kobayashi T,
insight into the cellular basis of Goldberg-Shprintzen Nonami A, Kato R, Sasaki M, Sasaki A, Ishibashi
syndrome. Development (Cambridge, England) 2008, H, Moriyama M, et al. Loss of mammalian Sprouty2
135:599–608. leads to enteric neuronal hyperplasia and esophageal
91. Alves MM, Burzynski G, Delalande JM, Osinga J, achalasia. Nat Neurosci 2005, 8:855–857.
van der Goot A, Dolga AM, de Graaff E, Brooks AS, 104. Zhou R, Niwa S, Homma N, Takei Y, Hirokawa N.
Metzger M, Eisel UL, et al. KBP interacts with SCG10, KIF26A is an unconventional kinesin and regulates
linking Goldberg-Shprintzen syndrome to microtubule GDNF-Ret signaling in enteric neuronal development.
dynamics and neuronal differentiation. Hum Mol Cell 2009, 139:802–813.
Genet 2010, 19:3642–3651. 105. Wang H, Hughes I, Planer W, Parsadanian A, Grider
92. Okamoto N, Wada Y, Goto M. Hydrocephalus and JR, Vohra BP, Keller-Peck C, Heuckeroth RO. The
Hirschsprung’s disease in a patient with a mutation of timing and location of glial cell line-derived neu-
L1CAM. J Med Genet 1997, 34:670–671. rotrophic factor expression determine enteric ner-
93. Okamoto N, Del Maestro R, Valero R, Monros E, vous system structure and function. J Neurosci 2010,
Poo P, Kanemura Y, Yamasaki M. Hydrocephalus and 30:1523–1538.
Hirschsprung’s disease with a mutation of L1CAM. 106. De Graaff E, Srinivas S, Kilkenny C, D’Agati V,
J Hum Genet 2004, 49:334–337. Mankoo BS, Costantini F, Pachnis V. Differential
94. Anderson RB, Turner KN, Nikonenko AG, Hem- activities of the RET tyrosine kinase receptor isoforms
perly J, Schachner M, Young HM. The cell adhesion during mammalian embryogenesis. Genes Dev 2001,
molecule L1 is required for chain migration of neural 15:2433–2444.
crest cells in the developing mouse gut. Gastroenterol- 107. Jain S, Encinas M, Johnson EM Jr, Milbrandt J. Criti-
ogy 2006, 130:1221–1232. cal and distinct roles for key RET tyrosine docking sites
95. Sariola H, Saarma M. Novel functions and signalling in renal development. Genes Dev 2006, 20:321–333.
pathways for GDNF. J Cell Sci 2003, 116:3855–3862. 108. Jain S, Knoten A, Hoshi M, Wang H, Vohra B,
96. Manie S, Massimo S, Alfredo F, Billaud M. The Heuckeroth RO, Milbrandt J. Organotypic specificity
RET receptor: function in development and dysfunc- of key RET adaptor-docking sites in the pathogene-
tion in congenital malformation. Trends Genet 2001, sis of neurocristopathies and renal malformations in
17:580–589. mice. J Clin Invest 2010, 120:778–790.
97. Natarajan D, Marcos-Gutierrez C, Pachnis V, de 109. Rossi J, Herzig KH, Voikar V, Hiltunen PH,
Graaff E. Requirement of signalling by receptor tyro- Segerstrale M, Airaksinen MS. Alimentary tract
sine kinase RET for the directed migration of enteric innervation deficits and dysfunction in mice lacking
nervous system progenitor cells during mammalian GDNF family receptor alpha2. J Clin Invest 2003,
embryogenesis. Development (Cambridge, England) 112:707–716.
2002, 129:5151–5160. 110. Nagy N, Goldstein AM. Endothelin-3 regulates neu-
98. Moore MW, Klein RD, Farinas I, Sauer H, Armanini ral crest cell proliferation and differentiation in the
M, Phillips H, Reichardt LF, Ryan AM, Carver- hindgut enteric nervous system. Dev Biol 2006,
Moore K, Rosenthal A. Renal and neuronal abnormal- 293:203–217.
ities in mice lacking GDNF. Nature 1996, 382:76–79. 111. Howard MJ. Mechanisms and perspectives on dif-
99. Chalazonitis A, Rothman TP, Chen J, Gershon MD. ferentiation of autonomic neurons. Dev Biol 2005,
Age-dependent differences in the effects of GDNF and 277:271–286.
NT-3 on the development of neurons and glia from 112. Zhu L, Lee HO, Jordan CS, Cantrell VA, Southard-
neural crest-derived precursors immunoselected from Smith EM, Shin MK. Spatiotemporal regulation of
the fetal rat gut: expression of GFRalpha-1 in vitro endothelin receptor-B by SOX10 in neural crest-
and in vivo. Dev Biol 1998, 204:385–406. derived enteric neuron precursors. Nat Genet 2004,
100. Heuckeroth RO, Lampe PA, Johnson EM, Mil- 36:732–737.
brandt J. Neurturin and GDNF promote proliferation 113. Kelsh RN. Sorting out Sox10 functions in neural crest
and survival of enteric neuron and glial progenitors in development. Bioessays 2006, 28:788–798.
vitro. Dev Biol 1998, 200:116–129. 114. Van de Putte T, Francis A, Nelles L, van Grunsven
101. Durbec PL, Larsson-Blomberg LB, Schuchardt A, LA, Huylebroeck D. Neural crest-specific removal of
Costantini F, Pachnis V. Common origin and develop- Zfhx1b in mouse leads to a wide range of neuro-
mental dependence on c-ret of subsets of enteric and cristopathies reminiscent of Mowat-Wilson syndrome.
sympathetic neuroblasts. Development (Cambridge, Hum Mol Genet 2007, 16:1423–1436.
England) 1996, 122:349–358. 115. Auricchio A, Griseri P, Carpentieri ML, Betsos N,
102. Laranjeira C, Pachnis V. Enteric nervous system devel- Staiano A, Tozzi A, Priolo M, Thompson H, Boc-
opment: recent progress and future challenges. Auton ciardi R, Romeo G, et al. Double heterozygosity for
Neurosci 2009, 151:61–69. a RET substitution interfering with splicing and an

© 2012 Wiley Periodicals, Inc.


WIREs Developmental Biology Hirschsprung disease

EDNRB missense mutation in Hirschsprung disease. reflux and a balanced translocation t(3;17)(p12;q11).
Am J Hum Genet 1999, 64:1216–1221. Eur J Hum Genet 2009, 17:483–490.
116. Carrasquillo MM, McCallion AS, Puffenberger EG, 128. Jiang Q, Ho YY, Hao L, Nichols Berrios C,
Kashuk CS, Nouri N, Chakravarti A. Genome-wide Chakravarti A. Copy number variants in candidate
association study and mouse model identify interaction genes are genetic modifiers of hirschsprung disease.
between RET and EDNRB pathways in Hirschsprung PLoS One 2011, 6:e21219.
disease. Nat Genet 2002, 32:237–244. 129. Fu M, Sato Y, Lyons-Warren A, Zhang B, Kane
117. McCallion AS, Stames E, Conlon RA, Chakravarti A. MA, Napoli JL, Heuckeroth RO. Vitamin A facilitates
Phenotype variation in two-locus mouse models enteric nervous system precursor migration by reduc-
of Hirschsprung disease: tissue-specific interaction ing Pten accumulation. Development (Cambridge,
between Ret and Ednrb. Proc Natl Acad Sci USA England) 2010, 137:631–640.
2003, 100:1826–1831.
130. Kubota M, Suita S, Kamimura T, Ito Y, Szurszewski
118. Maka M, Stolt CC, Wegner M. Identification of Sox8 JH. Electrophysiological properties of the aganglionic
as a modifier gene in a mouse model of Hirschsprung segment in Hirschsprung’s disease. Surgery 2002,
disease reveals underlying molecular defect. Dev Biol 131:S288–293.
2005, 277:155–169.
131. Won KJ, Torihashi S, Mitsui-Saito M, Hori M, Sato K,
119. Wallace AS, Schmidt C, Schachner M, Wegner M, Suzuki T, Ozaki H, Karaki H. Increased smooth mus-
Anderson RB. L1cam acts as a modifier gene during cle contractility of intestine in the genetic null of the
enteric nervous system development. Neurobiol Dis endothelin ETB receptor: a rat model for long segment
2010, 40:622–633. Hirschsprung’s disease. Gut 2002, 50:355–360.
120. Wallace AS, Tan MX, Schachner M, Anderson RB. 132. Spencer NJ, Bayguinov P, Hennig GW, Park KJ, Lee
L1cam acts as a modifier gene for members of the HT, Sanders KM, Smith TK. Activation of neural
endothelin signaling pathway during enteric nervous
circuitry and Ca2+ waves in longitudinal and cir-
system development. Neurogastroenterol Motil 2011,
cular muscle during CMMCs and the consequences
23:e510–e522.
of rectal aganglionosis in mice. Am J Physiol 2007,
121. Uesaka T, Enomoto H. Neural precursor death 292:G546–555.
is central to the pathogenesis of intestinal agan-
133. Suzuki T, Won KJ, Horiguchi K, Kinoshita K, Hori M,
glionosis in Ret hypomorphic mice. J Neurosci 2010,
Torihashi S, Momotani E, Itoh K, Hirayama K, Ward
30:5211–5218.
SM, et al. Muscularis inflammation and the loss of
122. Young HM, Newgreen D. Enteric neural crest-derived interstitial cells of Cajal in the endothelin ETB receptor
cells: origin, identification, migration, and differentia- null rat. Am J Physiol 2004, 287:G638–G646.
tion. Anat Rec 2001, 262:1–15.
134. Schafer KH, Micci MA, Pasricha PJ. Neural stem
123. Emison ES, McCallion AS, Kashuk CS, Bush RT, cell transplantation in the enteric nervous system:
Grice E, Lin S, Portnoy ME, Cutler DJ, Green ED, roadmaps and roadblocks. Neurogastroenterol Motil
Chakravarti A. A common sex-dependent mutation in 2009, 21:103–112.
a RET enhancer underlies Hirschsprung disease risk.
Nature 2005, 434:857–863. 135. Hotta R, Natarajan D, Thapar N. Potential of cell
therapy to treat pediatric motility disorders. Semin
124. Emison ES, Garcia-Barcelo M, Grice EA, Lantieri F,
Pediatr Surg 2009, 18:263–273.
Amiel J, Burzynski G, Fernandez RM, Hao L, Kashuk
C, West K, et al. Differential contributions of rare 136. Kruger G, Mosher J, Bixby S, Joseph N, Iwashita T,
and common, coding and noncoding Ret mutations Morrison S. Neural crest stem cells persist in the adult
to multifactorial Hirschsprung disease liability. Am gut but undergo changes in self-renewal, neuronal
J Hum Genet 2010, 87:60–74. subtype potential, and factor responsiveness. Neuron
2002, 35:657–669.
125. Edery P, Pelet A, Mulligan LM, Abel L, Attie T,
Dow E, Bonneau D, David A, Flintoff W, Jan 137. Hotta R, Pepdjonovic L, Anderson RB, Zhang D,
D, et al. Long segment and short segment familial Bergner AJ, Leung J, Pebay A, Young HM, Newgreen
Hirschsprung’s disease: variable clinical expression at DF, Dottori M. Small-molecule induction of neural
the RET locus. J Med Genet 1994, 31:602–606. crest-like cells derived from human neural progenitors.
126. Uesaka T, Nagashimada M, Yonemura S, Enomoto H. Stem Cells (Dayton, Ohio) 2009, 27:2896–2905.
Diminished Ret expression compromises neuronal sur- 138. Kawaguchi J, Nichols J, Gierl MS, Faial T, Smith A.
vival in the colon and causes intestinal aganglionosis Isolation and propagation of enteric neural crest pro-
in mice. J Clin Invest 2008, 118:1890–1898. genitor cells from mouse embryonic stem cells and
127. Griseri P, Vos Y, Giorda R, Gimelli S, Beri S, San- embryos. Development (Cambridge, England) 2010,
tamaria G, Mognato G, Hofstra RM, Gimelli G, 137:693–704.
Ceccherini I. Complex pathogenesis of Hirschsprung’s 139. Read AP, Newton VE. Waardenburg syndrome. J Med
disease in a patient with hydrocephalus, vesico-ureteral Genet 1997, 34:656–665.

© 2012 Wiley Periodicals, Inc.

You might also like