Hirchsprung Correlated Genetic

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Review article

Hirschsprung disease, associated syndromes, and


genetics: a review
Jeanne Amiel, Stanislas Lyonnet
Abstract
Hirschsprung disease (HSCR, aganglionic
megacolon) is the main genetic cause of
functional intestinal obstruction with an
incidence of 1/5000 live births. This devel-
opmental disorder is a neurocristopathy
and is characterised by the absence of the
enteric ganglia along a variable length of
the intestine. In the last decades, the
development of surgical approaches has
dramatically decreased mortality and
morbidity, which has allowed the emer-
gence of familial cases. HSCR appeared to
be a multifactorial malformation with
low, sex dependent penetrance and vari-
able expression according to the length of
the aganglionic segment, suggesting the
involvement of one or more gene(s) with
low penetrance. So far, eight genes have
been found to be involved in HSCR. This
frequent congenital malformation now
stands as a model for genetic disorders
with complex patterns of inheritance.
(J Med Genet 2001;38:729739)
Keywords: Hirschsprung disease; aganglionic megaco-
lon; genetics
Harald Hirschsprung rst described in 1888
two unrelated boys who died from chronic
severe constipation with abdominal distension
resulting in congenital megacolon.
1
The ab-
sence of intramural ganglion cells of the
myenteric and submucosal plexuses (Auer-
bachs and Meissners plexuses, respectively)
downstream from the dilated part of the colon
was considered to be the cause of the disease in
the 1940s.
2
This allowed simple and reliable
diagnostic conrmation from rectal suction
biopsies using histochemical staining for ace-
tylcholinesterase (AchE).
3
In 1948, Swenson
and Bill developed a surgical procedure
4
and
the survival of patients uncovered familial
transmission of HSCR.
5
In 1973, Bolande
6
proposed the term neurocristopathy for syn-
dromes or tumours involving neural crest (NC)
cells. HSCR resulting from an anomaly of the
enteric nervous system (ENS) of NC origin is
therefore regarded as a neurocristopathy.
6 7
HSCR occurs as an isolated trait in 70% of
patients, is associated with a chromosomal
abnormality in 12% of cases, and with
additional congenital anomalies in 18% of
cases.
813
In the latter group of patients, some
monogenic syndromes can be recognised.
Isolated HSCR appears to be a multifactorial
malformation with low, sex dependent pen-
etrance, variable expression according to the
length of the aganglionic segment, and suggest-
ing the involvement of one or more gene(s)
with low penetrance.
5 14
These parameters
must be taken into account for accurate evalu-
ation of the recurrence risk in relatives.
Segregation analyses suggested an oligogenic
mode of inheritance in isolated HSCR.
14
With
a relative risk as high as 200, HSCR is an
excellent model for the approach to common
multifactorial diseases. So far, genetic hetero-
geneity in HSCR has been shown with eight
specic genes involved. The major susceptibil-
ity gene is RET, which is also involved in mul-
tiple endocrine neoplasia type 2 (MEN 2). The
identication of modier genes is currently
under way. The aim of this paper is to review
the clinical data on syndromic HSCR and the
molecular ndings over the last 10 years.
Denition and classication
HSCR is a congenital malformation of the
hindgut characterised by the absence of
parasympathetic intrinsic ganglion cells in the
submucosal and myenteric plexuses.
2
It is
regarded as the consequence of the premature
arrest of the craniocaudal migration of vagal
neural crest cells in the hindgut between the
fth and twelfth week of gestation to form the
enteric nervous system (ENS) and is therefore
regarded as a neurocristopathy.
6 15
While the
internal anal sphincter is the constant inferior
limit, patients can be classied as short
segment HSCR (S-HSCR, 80%of cases) when
the aganglionic segment does not extend
beyond the upper sigmoid, and long segment
HSCR (L-HSCR, 20% of cases) when agan-
glionosis extends proximal to the sigmoid. Four
HSCR variants have been reported: (1) total
colonic aganglionosis (TCA, 3-8% of cases),
16
(2) total intestinal HSCR when the whole
bowel is involved,
16
(3) ultra short segment
HSCR involving the distal rectum below the
pelvic oor and the anus,
17
and (4) suspended
HSCR, a controversial condition, where a por-
tion of the colon is aganglionic above a normal
distal segment.
J Med Genet 2001;38:729739 729
Dpartement de
Gntique, Unit
INSERM U-393,
Hpital
Necker-Enfants
Malades, 149 rue de
Svres, 75743 Paris
Cedex 15, France
J Amiel
S Lyonnet
Correspondence to:
Dr Lyonnet,
[email protected]
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
Clinical features and diagnosis
In most cases, the diagnosis of HSCR is made
in the newborn period
13
owing to intestinal
obstruction with the following features: (1)
failure to pass meconium within the rst 48
hours of life, (2) abdominal distension that is
relieved by rectal stimulation or enemas, (3)
vomiting, and (4) neonatal enterocolitis. Some
patients are diagnosed later in infancy or in
adulthood with severe constipation, chronic
abdominal distension, vomiting, and failure to
thrive.
18
Finally, although a rare presentation,
unexplained perforation of the caecum or
appendix should make the diagnosis be consid-
ered.
On abdominal x ray, a distended small bowel
and proximal colon with an empty rectum are
common ndings. The classical image is a
dilated proximal colon with the aganglionic
cone narrowing towards the distal gut. On
barium enema a small rectum with uncoordi-
nated contractions is seen. The transition zone
represents the site where the narrow agangli-
onic bowel joins the dilated ganglionic bowel.
On a plain x ray taken later, delayed barium
evacuation is observed. Anorectal manometry
shows absence of relaxation of the internal
sphincter in response to rectal distension.
19
The
reliability of this test becomes excellent from
day 12 after birth when the normal rectoenteric
reex is present.
20
Suction rectal biopsy con-
rms the diagnosis in most cases,
21
but a full
thickness rectal biopsy is needed for diagnosis
of HSCR. Furthermore, extramucosal serial
biopsies will be required at laparotomy to
dene the proximal limit of the aganglionic
segment.
DiVerential diagnosis
Other causes of intestinal obstruction should
be considered when abdominal distension and
failure to pass meconium occur in a newborn
infant, namely: (1) meconium ileus resulting
from cystic brosis, (2) intestinal malforma-
tions such as lower ileal and colonic atresia,
isolated or occasionally associated with HSCR,
intestinal malrotation, or duplication, (3) ENS
anomalies grouped together as chronic intesti-
nal pseudo-obstruction syndromes, and (4)
functional intestinal obstruction resulting from
maternal infection, maternal intoxication, or
congenital hypothyroidism.
Treatment and prognosis
The treatment of HSCR is surgical. After care-
ful preoperative management, the principle is
to place the normal bowel at the anus and to
release the tonic contraction of the internal
anal sphincter. Since the initial protocol of
Swenson described in 1948,
4
a series of opera-
tive approaches have been developed, such as
the Soave and Duhamel procedures.
22 23
A one
stage procedure is possible when diagnosis is
made early, before colonic dilatation. Other-
wise, a primary colostomy is required. Fistula
or stenosis of the anastomosis and enterocolitis
are the main short term complications.
24
Long
term complications include chronic constipa-
tion (10-15%) and soiling.
25 26
Laparoscopic
techniques have recently been proposed in
HSCR surgery.
27
Mortality is under 6% since
the 1980s and may be related to short term
complications or caused by the associated mal-
formations.
25
However, the treatment of chil-
dren with TCA is still hazardous.
28 29
Epidemiology
The incidence of HSCR is estimated at 1/5000
live births.
5
However, the incidence varies
signicantly among ethnic groups (1.5, 2.1,
and 2.8 per 10 000 live births in Caucasians,
African-Americans, and Asians, respectively).
13
S-HSCR is far more frequent than L-HSCR
(80% and 20%, respectively).
8 10
There is a sex
bias with a preponderance of aVected males
and a sex ratio of 4/1.
14
Interestingly, the
male:female ratio is signicantly higher for
S-HSCR than for L-HSCR (table 1).
13 14
HSCR occurs as an isolated trait in 70% of
cases. A chromosomal abnormality is associ-
ated with it in 12% of cases, trisomy 21 being
by far the most frequent (>90%). Associated
congenital anomalies are found in 18% of
HSCR patients. The ones occurring at a
frequency above that expected by chance
include gastrointestinal malformation, cleft
palate, polydactyly, cardiac septal defects, and
craniofacial anomalies.
11 12
The higher rate of
associated anomalies in familial cases than in
isolated cases (39% versus 21%) strongly
suggests syndromes with Mendelian inherit-
ance.
12
Assessment of all HSCR patients
should include a careful evaluation for recog-
nisable syndromes by a trained dysmorpholo-
gist.
Chromosomal anomalies
A large number of chromosomal anomalies
have been described in HSCR patients. Free
trisomy 21 (Down syndrome) is by far the most
frequent, involving 2-10% of ascertained
HSCR cases.
5 913
In these cases, both the
unbalanced sex ratio (5.5-10.5 male:female)
and the predominance of S-HSCR are even
greater than in isolated HSCR. Overexpression
of genes on chromosome 21 predisposing to
HSCR has been hypothesised and a suscepti-
bility gene mapping to 21q22 postulated in a
Mennonite kindred.
30
However, these data
were not conrmed in other populations. Hith-
erto, mutations in genes predisposing to
HSCR, namely RET, EDNRB, and GDNF,
respectively, have been found in only three
patients with Down syndrome and HSCR.
31 32
Some chromosomal interstitial deletions
reported in combination with HSCR have been
important for the identication of HSCR
predisposing genes, namely (1) 10q11.2 inter-
stitial deletion observed in a few patients with
Table 1 Epidemiology and recurrence risk gures in HSCR
14
L-HSCR S-HSCR
% probands 19 81
Sex ratio (male:female) 1.75 5.5
Genetic model Dominant Multigenic or recessive
Penetrance (%) (male:female) 52:40 17:4
Recurrence risk to sibs* (%)
Male proband 17/13 5/1
Female proband 33/9 5/3
*Recurrence risk are given for male/female sibs respectively.
730 Amiel, Lyonnet
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
L-HSCR or TCA,
33 34
leading to the mapping
and identication of the rst gene for HSCR
(RET); (2) 13q22.1-32.1 interstitial deletion in
patients with S-HSCR encompassing a second
gene (EDNRB)
3537
; (3) 2q22-23 interstitial
deletion syndrome in patients with HSCR
(table 2),
3840
leading to the identication of the
SIP1 gene (SMAD interacting protein 1).
41
Rarer chromosomal anomalies reported in
combination with HSCR are summarised in
table 2. DiGeorge syndrome, mosaic trisomy 8,
XXY chromosomal constitution, partial dupli-
cation of chromosome 2q, tetrasomy 9p, and
20p deletion each have been observed once
with HSCR.
Syndromes and associated anomalies
Both the recognition of known entities and the
delineation of novel ones including HSCR as a
feature are of importance for disease prognosis,
accurate genetic counselling, and search for
candidate genes. Syndromes associated with
HSCR can be classied as: (1) pleiotropic neu-
rocristopathies, (2) syndromes with HSCR as a
mandatory feature, (3) occasional association
with recognisable syndromes, and (4) miscella-
neous observations (table 3).
NEUROCRISTOPATHIES
The NC is a transient and multipotent embry-
onic structure that gives rise to neuronal,
endocrine and paraendocrine, craniofacial,
conotruncal heart, and pigmentary tissues.
7
Neurocristopathies encompass tumours, mal-
formations, and single or multifocal abnor-
malities of tissues mentioned above in various
combinations. MEN 2, conotruncal heart
defects, congenital central hypoventilation, and
Table 2 Recurrent chromosomal anomalies with HSCR as a feature
Chromosome Key features Number of reports Gene References
Tri 21 Down syndrome, S-HSCR, 5.5 to 10.5 male:female sex ratio 2 to 10% of HSCR cases ? 5, 913
Del 10q11 Mental retardation, L-HSCR 2 cases RET 33, 34
Del 13q22 Mental retardation, growth retardation, dysmorphic features, S-HSCR 7 cases EDNRB 3537
Del 2q22-q23 Postnatal growth retardation and microcephaly, mental retardation, epilepsy, dysmorphic
features, HSCR*
3 cases SIP1 3841
Del 17q21 4 cases ?
Dup 17q21-q23 MCA/MR 4 cases ?
Tri 22pter-q11 Cat eye syndrome ?
*Both S-HSCR and L-HSCR have been observed. Several patients presenting the same pattern of congenital malformations and normal chromosomes have been
reported.
39
Table 3 Syndromes associated with HSCR (reprinted and adapted from Scriver CM et al, eds. The metabolic and molecular bases of inherited diseases
8th ed. Chap 251. New York: McGraw-Hill: 6231-55.)
Syndromes MIM Key features References
Neurocristopathy syndromes
WS4 (Shah-Waardenburg) 277580 Pigmentary anomalies (white forelock, iris hypoplasia, patchy hypopigmentation), deafness 5357
Yemenite deaf-blind-hypopigmentation 601706 Hearing loss, eye anomalies (microcornea, coloboma, nystagmus), pigmentary anomalies 60
BADS 227010 Hearing loss, hypopigmentation of the skin and retina 61
Piebaldism 172800 Patchy hypopigmentation of the skin 62, 63
Haddad 209880 Congenital central hypoventilation 70, 71
MEN2A 171400 Medullary thyroid carcinoma, phaeochromocytoma, hyperplasia of the parathyroid 4350
Riley-Day 223900 Autonomic nervous system anomalies
HSCR mandatory
Goldberg-Shprintzen 235730 Cleft palate, hypotonia, microcephaly, mental retardation, dysmorphic facial features 79
235740 Polydactyly, unilateral renal agenesis, hypertelorism, deafness 82
HSCR with limbs anomalies 235750 Postaxial polydactyly, ventricular septal defect 83
235760 Hypoplasia of distal phalanges and nails, dysmorphic features 84
604211 Preaxial polydactyly, heart defect, laryngeal anomalies 85
306980 Brachydactyly type D 86
BRESHEK Brain abnormalities, Retardation, Ectodermal dysplasia, Skeletal malformation, Hirschsprung
disease, Ear/eye anomalies, Kidney dysplasia
87
Mesomelic dysplasia, Werner type Mesomelia, polydactyly 170
HSCR occasionally associated
Bardet-Biedl and/or 209900 Pigmentary retinopathy, obesity, hypogenitalism, mild mental retardation, postaxial polydactyly 91, 92
KauVman-McKusick 236700 Hydrometrocolpos, postaxial polydactyly, congenital heart defect 89
Smith-Lemli-Opitz 270400 Growth retardation, microcephaly, mental retardation, hypospadias, 23 toes syndactyly,
dysmorphic features
96
Cartilage-hair hypoplasia 250250 Short limb dwarsm, metaphyseal dysplasia, immunodeciency 97
HSCR rarely associated
Fukuyama congenital muscular dystrophy 253800 Muscular dystrophy, polymicrogyria, hydrocephalus, MR, seizures 99, 100
Clayton-Smith 258840 Dysmorphic features, hypoplastic toes and nails, ichthyosis. 101
Kaplan 304100 Agenesis of corpus callosum, adducted thumbs, ptosis, muscle weakness 102
Okamoto 308840 Hydrocephalus, cleft palate, corpus callosum agenesis 103
Miscellaneous associations
Pallister-Hall (CAVE) 140510
Fryns 229850
Aarskog 100050
Jeune asphyxiating thoracic dystrophy 208500
Frontonasal dysplasia 136760
Osteopetrosis
Goldenhar 164210
Lesch-Nyhan 308000
Rubinstein-Taybi 180849
Toriello-Carey 217980
SEMDJL 271640
Hirschsprung disease, associated syndromes, and genetics 731
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
Waardenburg syndrome illustrate each of these
categories and can be associated with HSCR.
Multiple endocrine neoplasia type 2 (MEN 2).
The MEN 2 syndromes include three types of
cancer predisposition with an autosomal domi-
nant mode of inheritance: familial medullary
thyroid carcinoma (FMTC), MEN type 2A,
(MEN 2A) and type 2B (MEN 2B). MEN 2A
is dened by an age related predisposition to
medullary thyroid carcinoma (MTC, 70% by
the age of 70 years), phaeochromocytoma
(50% of cases), and hyperplasia of the parathy-
roid glands (15-35%). In addition to MTCand
phaeochromocytoma, subjects with MEN 2B
present with oral neuromas, marfanoid habi-
tus, and hyperganglionosis of the hindgut.
42
Germline missense mutations of the RET gene
have been identied in MEN 2A, MEN 2B,
and FMTC. Both FMTC and MEN 2A can be
associated with HSCR in some families.
4350
Interestingly, these families present a germline
RET mutation of the MEN 2A or FMTC type
(see below).
4450
This raises the question of
whether all subjects with HSCR, regardless of a
non-contributory family history, should be
screened for RET exon 10 and 11 mutations to
rule out cancer predisposition (3/160 cases in
our series, C609W, C611R, and C620R RET
gene mutations).
Waardenburg syndromes (WS) and related
pigmentary anomalies
WS, an autosomal dominant condition, is by
far the most frequent condition combining
pigmentary anomalies and sensorineural deaf-
ness (1/50 000 live births and 2-5% of all con-
genital deafness), resulting from the absence of
melanocytes of the skin and the stria vascularis
of the cochlea.
51
WS is clinically and genetically
heterogeneous (MIM 193500, MIM 148820,
MIM 193510).
52
The combination of HSCR
with WS denes the WS4 type (Shah-
Waardenburg syndrome, MIM 277580), a
genetically heterogeneous condition. Indeed,
homozygous mutations of the endothelin
pathway
5356
and heterozygous SOX10 muta-
tions have been identied in WS4 patients (see
below).
57
Patients carrying a SOX10 mutation
may also present with CNS involvement
including seizures, ataxia, and demyelinating
peripheral and central neuropathies.
58 59
Pigment related syndromes that may include
HSCR include: (1) Yemenite deaf-blind hypo-
pigmentation syndrome (MIM 601706). A
SOX10 mutation has been reported in one of
these families
60
; (2) Black locks-Albinism-
Deafness Syndrome (BADS, MIM 227010)
with TCA-HSCR reported in one case
61
; (3)
aganglionic megacolon associated with familial
piebaldism (MIM 172800)
62 63
; (4) HSCR and
profound congenital deafness but with no other
WS features has also been reported.
64
Congenital central hypoventilation syndrome
(CCHS, MIM 209880)
Initially termed Ondines curse, CCHS is a
rare, life threatening condition characterised by
abnormal ventilatory response to hypoxia and
hypercapnia owing to failure of autonomic res-
piratory control.
65
CCHS patients often
present symptoms resulting from a broader
dysfunction of the autonomic nervous system
and neural crest cell derived tumours have also
been observed.
6568
CCHS may be a polygenic
disorder with a major locus being involved.
69
Recurrence risk in sibs is estimated as 5% with
few multicase families reported.
65
Haddad syn-
drome (MIM 209880) is dened by the
combination of CCHS with HSCR and repre-
sents 14-20% of CCHS patients.
70 71
In these
cases, L-HSCR (including TCA) is by far the
most frequent, and the sex ratio is equal,
contrary to what is observed in isolated
HSCR.
71
Mutations of the RET and the
endothelin signalling pathways have been iden-
tied in rare CCHS patients: a RET mutation
inherited from a healthy parent in two patients
with Haddad syndrome,
72 73
a GDNF mutation
inherited from a healthy mother in a CCHS
patient,
72
and an EDN3 mutation in a CCHS
patient.
74
Other neurocristopathies
Familial dysautonomia syndrome (FDS, Riley-
Day syndrome, MIM 223900) has been
reported once in association with HSCR.
Although it could have arisen by chance alone,
it is interesting to note that the FDS gene
(IKBKAP) maps to 9q31 where a susceptibility
locus for HSCR has been identied (see
below). Other occasional associations reported
so far include cleft lip with or without cleft pal-
ate, neural crest derived tumours (neuroblas-
toma, ganglioneuroblastoma),
75 76
neural tube
defects (myelomeningocele),
77
and neuro-
bromatosis type 1.
78
The signicance of these
associations is not yet established.
SYNDROMES WITH HSCR AS A MANDATORY
FEATURE
Goldberg-Shprintzen syndrome (MIM 235730)
This rare, probably autosomal recessive, multi-
ple congenital anomalies-mental retardation
syndrome combines HSCR, cleft palate, hypo-
tonia, microcephaly and mental retardation
with or without facial dysmorphic features
(hypertelorism, prominent nose, synophrys,
sparse hair).
79
The observation of both ven-
tricular dilatation and irregular density of white
matter on brain imaging may suggest a neuro-
nal migration defect. Several reports with vari-
able association of microcephaly, iris colo-
boma, cleft palate, and mental retardation may
be variants of this syndrome.
80 81
In our
opinion, patients with a SIP1 gene mutation
have a diVerent condition.
HSCR with limb anomalies
A series of rare syndromes with HSCR and
distal limb anomalies (polydactyly or hypopla-
sia) have been reported. These are: (1) HSCR
with polydactyly, unilateral renal agenesis,
hypertelorism, and congenital deafness (MIM
235740)
82
; (2) HSCR, postaxial polydactyly,
and ventricular septal defects (MIM235750)
83
;
(3) HSCR, hypoplasia of the distal phalanges
and nails, and mild dysmorphic features (MIM
732 Amiel, Lyonnet
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
235760)
84
; (4) HSCR with preaxial polydac-
tyly, heart defect, and laryngeal anomalies
(MIM 604211)
85
; (5) HSCR with brachydac-
tyly type D (MIM 306980)
86
; (6) HSCR with
brachydactyly, macrocephaly, and vertebral
anomalies
87
; (7) BRESHEKsyndrome,
88
and (8)
mesomelic dysplasia, Werner type.
170
OCCASIONAL ASSOCIATION IN RECOGNISABLE
SYNDROMES
The occasional association of HSCRwith three
syndromes or groups of syndromes of auto-
somal recessive mode of inheritance may be of
interest for the identication of susceptibility
genes in HSCR.
McKusick-KauVman syndrome (MKKS, MIM
236700) and Bardet-Biedl syndrome (BBS,
MIM 209900)
MKKS is a rare condition characterised by
hydrometrocolpos, postaxial polydactyly, and
congenital heart defect. HSCR is found in 10%
of cases.
89
The MKKS disease causing gene on
20p12, encoding a chaperonin protein, has
recently been identied.
90
BBS is characterised by progressive pigmen-
tary retinopathy, obesity, hypogenitalism, renal
involvement (including cysts, renal cortical
loss, or reduced ability to concentrate urine),
mild mental retardation, and postaxial poly-
dactyly of the hands and feet. BBS is
genetically heterogeneous with at least ve loci
involved, mapped to chromosome arms 2q, 3p,
11q, 15q, and 16q. HSCR has been reported in
several BBS cases.
91 92
Recently, mutations in
the MKKS gene were identied in some BBS
patients conrming clinical overlap at the
molecular level.
93
Smith-Lemli-Opitz syndrome (SLO, MIM
270400)
SLO is characterised by pre- and postnatal
growth retardation and microcephaly, severe
mental retardation, facial dysmorphic features,
hypospadias, and syndactyly between toes 2
and 3. SLO results from cholesterol metabolic
impairment with mutation of the 7-dehydro-
cholesterol reductase gene (DHCR7, chromo-
some 11q12-q13).
94 95
HSCR is observed in a
signicant number of severe SLO patients.
96
Cartilage-hair hypoplasia syndrome (CHH,
MIM 250250)
This skeletal dysplasia, rst described in the
Old Order Amish community, combines meta-
physeal dysplasia with short limb dwarsm,
ne, sparse, and blond hair, transient macro-
cytic anaemia, and immunodeciency. HSCR
is associated with it in approximately 10% of
the cases.
97
The gene (RMRP) has been
mapped to chromosome 9p13.
98
Interestingly,
HSCR has been reported in the Holmgren-
Connor syndrome (MIM 211120), which may
be allelic to CHH.
MISCELLANEOUS OBSERVATIONS
For rare disorders, whether an association with
HSCR observed once is meaningful or oc-
curred by chance alone is not possible to
decide. These conditions are summarised in
table 3 and can be classied as follows: (1) syn-
dromes with muscular dystrophy,
99 100
(2)
syndromes with dermatological ndings,
101
and
(3) syndromes with central nervous system
anomalies.
102 103
Other rare associations include
the nding of HSCR with Fryns syndrome,
Aarskog syndrome, Jeune asphyxiating thoracic
dystrophy, frontonasal dysplasia, osteopetrosis,
Goldenhar syndrome, Lesch-Nyhan syn-
drome, Rubinstein-Taybi syndrome, Toriello-
Carey syndrome, Pallister-Hall syndrome,
spondyloepimetaphyseal dysplasia with joint
laxity (SEMDJL, MIM 271640), persistent
mullerian duct syndromes, and asplenia with
cardiovascular anomaly.
Isolated anomalies
A wide spectrum of additional isolated anoma-
lies have been described among HSCR cases
with an incidence varying from 5% to 30%
according to series.
8 9 11 104107
No constant
pattern is observed and these anomalies
include distal limb, sensorineural, skin, central
nervous system, genital, kidney, and cardiac
malformations. However, cardiac defects,
mostly atrio- or ventriculoseptal defects, are
found with an incidence of 5% of cases of
HSCR, excluding patients with trisomy 21.
Renal dysplasia or agenesis was found in 4.4%
in a series of 160 HSCR cases and may still be
underestimated (personal data). This is of
interest since homozygous knockout mice for
genes involved in the Ret signalling pathway
present with renal agenesis/dysplasia in addi-
tion to megacolon (see below).
108
Genital
anomalies including hypospadias are reported
in up to 2-3% of HSCR patients. Gastro-
intestinal malformations such as Meckel
diverticulum, pyloric stenosis, single umbilical
artery, inguinal hernia, or small bowel atresia
are also found.
109111
Finally, facial dysmorphic
features seem to be extremely frequent when
looked for. These data highlight the import-
ance of a careful assessment by a clinician
trained in dysmorphology for all newborns
diagnosed with HSCR. Skeletal x ray and car-
diac and urogenital echographic survey should
be systematically performed. The observation
of one additional anomaly to HSCR should
prompt chromosomal studies.
Molecular genetics
Segregation studies in non-syndromic HSCR
have shown that the recurrence risk in sibs
varies from 1% to 33% depending on the gen-
der and the length of the aganglionic segment
in the proband and the gender of the sib (table
1).
5 14
Consequently, HSCR has been assumed
to be a sex modied multifactorial disorder, the
eVect of genes playing a major role as
compared to environmental factors (relative
risk of 200).
So far, eight genes are known to be involved
in HSCR in humans, namely the proto-
oncogene RET (RET), glial cell line derived
neurotrophic factor (GDNF), neurturin
(NTN), endothelin B receptor (EDNRB),
endothelin 3 (EDN3), endothelin converting
enzyme 1 (ECE1), SOX10, and SIP1 genes
(table 4).
Hirschsprung disease, associated syndromes, and genetics 733
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
THE RET SIGNALLING PATHWAY
The rst susceptibility locus was mapped to
10q11.2 in multigenerational families segregat-
ing HSCR as an incompletely penetrant auto-
somal dominant trait.
112 113
This region had
been targeted because of the observation of an
interstitial deletion of chromosome 10q11.2 in
patients with TCA and mental retardation.
33
The proto-oncogene RET, identied as disease
causing in MEN 2
114 115
and mapping in
10q11.2, was regarded as a good candidate gene
owing to the concurrence of MEN 2A and
HSCR in some families and the expression in
neural crest derived cells. Consequently, RET
gene mutations were identied in HSCR
patients.
116 117
RET is a 1114 amino acid trans-
membrane receptor with a cadherin-like extra-
cellular domain, a cysteine rich region, and an
intracellular tyrosine kinase domain.
118
Expres-
sion and penetrance of a RET mutation is vari-
able and sex dependent within HSCR families.
In large series, the estimated penetrance is 72%
in males and 51% in females.
119
Over 80 muta-
tions have been identied including large dele-
tions encompassing the RET gene, microdele-
tions and insertions, nonsense, missense and
splicing mutations.
119121
There is no mutational
hot spot at variance with MEN 2A, where
mutations occur in a cluster of six cysteines
(exon 10, residues 609, 611, 618, 620; exon 11,
residues 630,634),
44 114 115
and MEN 2B where
the mutation is almost unique (M918T, exon
16, tyrosine kinase domain).
122124
In vitro, MEN
2 mutations have been shown to be activating
mutations leading to constitutive dimerisation
of the receptor and to transformation,
125
while
haploinsuYciency is the most likely mechanism
for HSCR mutations.
126128
Biochemical studies
showed variable consequences of some HSCR
mutations (misfolding, failure to transport the
protein to the cell surface, abolished biological
activity).
127 128
However, a simple activating ver-
sus inactivating model of gene action is not suf-
cient to explain the concurrence of HSCR and
MEN 2A in patients with a MEN 2A RET gene
mutation.
Despite extensive mutation screening, a RET
mutation is identied in only 50% of familial
and 15-20% of sporadic HSCR cases.
119
However, most families with fewexceptions are
compatible with linkage at the RET locus.
129
Recent studies of known polymorphisms
within the RET gene in a series of sporadic
HSCR patients showed a signicantly diVerent
distribution as compared to controls. Several
polymorphic haplotypes could be associated
with predisposition to HSCR.
130 131
Epigenetic
factors could also be involved.
GDNF, known as a major survival factor for
many types of neurones, was shown to be the
RET ligand by both phenotypic similarities
between Ret -/- and Gdnf -/- knockout mice
132134
and Xenopus embryo bioassays.
135
GDNF is a
TGF-B related 211 residue protein, proteolyti-
cally cleaved to a 134 residue mature protein
that homodimerises. To activate RET, GDNF
needs the presence of a novel glycosylphos-
phatidylinositol (GPI) linked coreceptor
GFRA1.
136 137
Four related GPI linked corecep-
tors, GFRA1-4,
138
and four related soluble
growth factors ligands of RET have been iden-
tied, namely GDNF, NTN,
139
persephin
(PSPN),
140
and artemin.
141
Specic combina-
tions of these proteins are necessary for devel-
opment and maintenance of both central and
peripheral neurones and all can signal through
RET. GDNF mutations have been identied in
only six HSCR patients to date, and can be
regarded as a rare cause of HSCR
(<5%).
31 142 143
Moreover, GDNF mutations
may not be suYcient to lead to HSCR since
four out of six patients have additional
contributory factors, such as RET mutations or
trisomy 21.
31 142
Similarly, a NTN mutation has
been identied in one family, in conjunction
with a RET mutation.
144
Finally, although
Gfra1 homozygous knockout mice are pheno-
typically very similar to Ret and Gdnf -/- mice,
no GFRA1 mutations have been identied in
HSCR patients.
145148
THE ENDOTHELIN SIGNALLING PATHWAY
The endothelin pathway was rst studied for its
vasoconstrictive eVect and putative role in
hypertension. EDNRB and EDNRA are G
protein coupled heptahelical proteins that
transduce signals through the endothelins
(EDN1, 2, 3).
149 150
Table 4 Genes involved in HSCR in humans and known mouse models of megacolon
Gene
Human Mouse
Map location
Mode of
inheritance
Phenotype in
mutants
Frequence of mutation
in heterozygotes Refs Natural mutant Knock-out Refs
RET 10q11.2 AD HSCR 50% familial cases 119 L 108
15% sporadic cases Renal agenesis
GDNF 5p13 AD HSCR 5 cases 31, 142, 143 L 132134
Renal agenesis
NTN 19p13 AD HSCR 1 case 144
SOX10 22q13 AD WS4 57, 59, 165 Dom (AD) L 164
Coat spotting
EDNRB 13q22 AR/AD WS4/HSCR 5% 53, 157160 s
l
(AR) S 154
Coat spotting
EDN3 20q13 AR/AD WS4/HSCR <5% 156 ls (AR) S 155
Coat spotting
ECE1 1p36 AD HSCR 1 case 162 S 161
CF and cardiac
defect
Coat spotting
CF defects
SIP1 2q22 Spo HSCR, MR, facial
dysmorphism
6 cases 38, 39, 41
AD: autosomal dominant, AR: autosomal recessive, Spo: sporadic, s
l
: Piebald lethal, ls: lethal spotting, S: short segment megacolon, L: long segment megacolon, CF:
craniofacial, MR: mental retardation.
734 Amiel, Lyonnet
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
A susceptibility locus for HSCR in 13q22
was suggested for three main reasons: (1) a sig-
nicant lod score at 13q22 in a large inbred
Old Order Mennonite community with multi-
ple cases of HSCR,
30 151 152
(2) de novo intersti-
tial deletion of 13q22 in several patients with
HSCR,
36 37 153
and (3) synteny between the
murine locus for piebald-lethal (s
l
), a model of
aganglionosis, and 13q22 in humans. The
critical role of the endothelin pathway in
HSCR was shown by the nding that piebald-
lethal was allelic to the Ednrb knockout mouse
and harboured an Ednrb mutation (table 4).
154
Subsequently, an EDNRB missense mutation
was identied in the Mennonite kindred
(W276C).
53
However, the W276C mutation
was neither necessary (aVected wild type
homozygotes) nor suYcient (unaVected mu-
tant homozygotes) to cause HSCR, and
penetrance was sex dependent (greater in
males than in females).
53
piebald-lethal was con-
sidered a mouse model for WS4 in humans and
some of the aVected Mennonite subjects had
pigmentary anomalies and sensorineural deaf-
ness in addition to HSCR.
30 151
This prompted
a screen of the EDNRB gene in WS4 and
homozygous mutations in a fraction of WS4
families were found.
54
At the same time, an
Edn3 mutation was identied in the lethal spot-
ting (ls) natural mouse model for WS4
155
and
EDN3 homozygous mutations were identied
in WS4 in humans (table 4).
55 56
Both EDNRB and EDN3 were screened in
large series of isolated HSCR patients. While
EDN3 mutations were seldom found,
156
EDNRB mutations were identied in approxi-
mately 5% of the patients.
157160
It is worth
mentioning that the penetrance of EDN3 and
EDNRB heterozygous mutations is incomplete
in those HSCR patients, de novo mutations
have not hitherto been observed, and that
S-HSCR is largely predominant. Interstitial
13q22 deletions encompassing the EDNRB
gene in HSCR patients make haploinsuY-
ciency the most likely mechanism for HSCR
(table 2). Although EDNRB binds all three
endothelins, the similarity of phenotype of the
Ednrb knockout mice to that of the Edn3
knockout mice suggests that EDNRBs major
ligand is EDN3 in neural crest derived cells.
Preproendothelins are proteolytically
cleaved by two related membrane bound met-
alloproteases to give rise to the mature 21 resi-
due endothelin. Ece1 processes only Edn1 and
Edn3. Ece1 knockout mice show craniofacial
defects and cardiac abnormalities in addition
to colonic aganglionosis.
161
A heterozygous
ECE1 mutation has been identied in a patient
combining HSCR and craniofacial and cardiac
defects (R742C).
162
SOX10
The last de novo mouse model for WS4 in
human is dominant megalon (Dom), homo-
zygous Dom mutation being embryonic le-
thal.
163
The Dom gene is Sox10, a member of
the SRY (sex determining factor)-like, high
mobility group (HMG) DNA binding pro-
teins.
164
Subsequently, heterozygous SOX10
mutations have been identied in familial and
isolated patients with WS4 (including de novo
mutation).
57 59 165
At least some mutations
disrupt the DNAbinding domain and may lead
to a loss of function allele, so that again
haploinsuYciency is the most likely mech-
anism for HSCR. Others disrupt the transacti-
vation domain and may result in a dominant
negative eVect. These latest mutations were
identied in patients presenting neurological
impairment in addition to HSCR and pigmen-
tary anomalies.
59
Penetrance appears to be
high, although sibs sharing a mutation and dis-
cordant for HSCR have been described in one
family.
165
Therefore, SOX10 is unlikely to be a
major gene in isolated HSCR.
INTERACTION BETWEEN PATHWAYS
Ret and Ednrb signalling pathways were
considered biochemically independent. How-
ever, G protein coupled receptors and receptor
tyrosine kinases could be engaged in crosstalk.
Moreover, an HSCR patient heterozygous for
weak hypomorphic mutations in both RET and
EDNRB has recently been reported.
166
Each
mutation was inherited from a healthy parent.
Sox10 is involved in cell lineage determina-
tion and is capable of transactivating MITF
synergistically with PAX3.
167
Similarly, Ednrb
transcripts are either absent or drastically
reduced in Dom-/- and +/- mice, respectively.
168
Therefore, the reduced expression of Ednrb in
the dom mouse could arise either from a direct
eVect of Sox10 or from an indirect eVect on a
subset of NC cells of common faith.
Taken all together, several general comments
can be made. RET is the major gene in HSCR
with a heterozygous mutation found in 50% of
familial cases and 15-20% of isolated cases.
RET mutation penetrance is incomplete and
sex dependent. Genotype-phenotype correla-
tion is poor. HSCR is genetically heterogene-
ous and results from mutations in distinct
pathways. Some patients with mutations in more
than one HSCR susceptibility gene are known
(RET + GDNF, RET + NTN, RET + EDNRB).
Multigenic inheritance of Hirschsprung
disease
As mentioned above, RET plays a key role in
non-syndromic HSCR genesis and multiple
genes may be required to modulate clinical
expression. On the other hand, genetic hetero-
geneity, where mutation in one of several genes
is suYcient for phenotypic expression of
HSCR, has been reported (RET, EDNRB,
EDN3, ECE1, SIP1). However, the observation
of non-random association between HSCR,
RET, and chromosome 21q22 in the Menon-
nite population where HSCR imperfectly
segregates with an EDNRB mutation favours
multigenic inheritance resulting from the
cumulative eVects of multiple mutations.
According to the segregation analysis where an
autosomal dominant model in L-HSCR and a
multifactorial model in S-HSCR were more
likely, two approaches have been chosen to test
these hypotheses in L-HSCR and S-HSCR
independently.
Hirschsprung disease, associated syndromes, and genetics 735
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
LINKAGE ANALYSIS IN 12 HSCR FAMILIES WITH
THREE OR MORE AFFECTED SUBJECTS IN TWO OR
MORE GENERATIONS
129
L-HSCR is largely predominant in these fami-
lies. All but one family showed linkage to the
RET locus. Mutational analysis identied a
nonsense or missense mutation at a highly
conserved residue in six families, a splice
mutation in two families, and no coding
sequence variation in three families. Linkage to
a novel locus in 9q31 was identied only in
families with no or hypomorphic RET gene
mutation. Therefore, a severe RET mutation
may lead to phenotypic expression by haploin-
suYciency, while hypomorphic RET mutations
would require the action of other mutations.
A SIB PAIR ANALYSIS IN 49 FAMILIES WITH S-HSCR
PROBANDS
169
This study shows that only three loci on chro-
mosomes 3p21, 10q11, and 19q12 are both
necessary and suYcient to explain the inci-
dence and sib recurrence risk in HSCR. A
multiplicative risk across loci with most af-
fected subjects being heterozygotes at all three
loci seems the best genetic model. Interest-
ingly, marker analysis showed a signicant par-
ent of origin eVect at the RET locus, 78% of
shared RET alleles being maternally derived,
which could explain the sex diVerence in
HSCR expression. Finally, linkage to 9q31 was
conrmed in the sib pairs with no or hypomor-
phic RET mutation.
Genetic counselling
HSCR is a sex modied, multifactorial,
congenital malformation with an overall recur-
rence risk in sibs of the proband of 4% (relative
risk=200). In isolated HSCR, adequate relative
risk gures will be provided by taking into
account the sex and length of the aganglionic
segment in the proband and the gender of the
sib (1-33%). According to the Carter paradox,
the highest recurrence risk is for a male sib of a
female proband with L-HSCR (table 1).
Because of poor genotype-phenotype correla-
tion so far, the benet of mutation screening
for HSCR patients appears low except for sys-
tematic testing of exon 10 and 11 of the RET
gene owing to cancer predisposition of MEN
2A mutations. This, unfortunately, is not yet
routine practice.
Many HSCR cases are associated with other
congenital anomalies. In these cases, the long
term prognosis is highly dependent on the
severity of the associated anomalies. Several
known syndromes have straight Mendelian
inheritance. This emphasises the importance of
careful assessment by a clinician trained in
syndromology of all newborns diagnosed with
HSCR.
We thank the HSCR patients and their families and the French
Hirschsprung Disease Association (AFMA) for their coopera-
tion and active participation over 10 years. We sincerely
acknowledge colleagues from all over the world for providing us
with samples as well as all the students and collaborators of our
research group on Hirschsprung disease. Some data in the
present review have been reprinted and adapted from Scriver
CR, et al, ed. The metabolic and molecular bases of inherited
diseases. 8th ed. Chap 251. New York: McGraw-Hill:6231-55.
1 Hirschsprung H. Stuhltrgheit neugeborener infolge von
dilatation und hypertrophic des colons. Jb Kinderheilkd
1888;27:1.
2 Whitehouse F, Kernohan J. Myenteric plexuses in congeni-
tal megacolon; study of 11 cases. Arch Intern Med 1948;82:
75.
3 Meier-Ruge W, Lutterbeck PM, Herzog B, Morger R,
Moser R, Scharli A. Acetylcholinesterase activity in suction
biopsies of the rectum in the diagnosis of Hirschsprungs
disease. J Pediatr Surg 1972;7:11-17.
4 Swenson O. Early history of the therapy of Hirschsprungs
disease: facts and personal observations over 50 years. J
Pediatr Surg 1996;31:1003-8.
5 Bodian M, Carter C. A family study of Hirschsprung
disease. Ann Hum Genet 1963;26:261.
6 Bolande RP. The neurocristopathies; a unifying concept of
disease arising in neural crest maldevelopment. Hum Pathol
1973;5:409-29.
7 Le Douarin N, Kalcheim C. The neural crest. Cambridge:
Cambridge University Press, 1999.
8 Passarge E. The genetics of Hirschsprungs disease.
Evidence for heterogeneous etiology and a study of
sixty-three families. N Engl J Med 1967;276:138-43.
9 Goldberg EL. An epidemiological study of Hirschsprungs
disease. Int J Epidemiol 1984;13:479-85.
10 Garver KL, Law JC, Garver B. Hirschsprung disease: a
genetic study. Clin Genet 1985;28:503-8.
11 Spouge D, Baird PA. Hirschsprung disease in a large birth
cohort. Teratology 1985;32:171-7.
12 Brooks AS, Breuning MH, Meijers C. Spectrum of pheno-
types associated with Hirschsprung disease: an evaluation
of 239 patients from a single institution. The Third
International Meeting: Hirschsprung disease and related neuro-
cristopathies, Evian, France, 1998.
13 Torfs CP. An epidemiological study of Hirschsprung disease
in a multiracial California population. The Third Inter-
national Meeting: Hirschsprung disease and related neuro-
cristopathies, Evian, France, 1998.
14 Badner JA, Sieber WK, Garver KL, Chakravarti A. A
genetic study of Hirschsprung disease. Am J Hum Genet
1990;46:568-80.
15 Taraviras S, Pachnis V. Development of the mammalian
enteric nervous system. Curr Opin Genet Dev 1999;9:321.
16 Nihoul-Fkt C, Ricour C, Martelli H, Lortat Jacob S, Pel-
lerin D. Total colonic aganglionosis (with or without ileal
involvement): a review of 27 cases. J Pediatr Surg 1986;21:
251.
17 Neilson IR, Yazbeck S. Ultrashort Hirschsprungs disease:
myth or reality. J Pediatr Surg 1990;25:1135-8.
18 Parc R, Berrod JL, Tussiot J, Loygue J. Megacolon in adults.
Apropos of 76 cases. Ann Gastroenterol Hepatol (Paris)
1984;20:133-41.
19 Emir H, Akman M, Sarimurat N, Kilic N, Erdogan E,
Soylet Y. Anorectal manometry during the neonatal period:
its specicity in the diagnosis of Hirschsprungs disease.
Eur J Pediatr Surg 1999;9:101-3.
20 Lopez-Alonso M, Ribas J, Hernandez A, Anguita FA,
Gomez de Terreros I, Martinez-Caro A. EYciency of the
anorectal manometry for the diagnosis of Hirschsprungs
disease in the newborn period. Eur J Pediatr Surg
1995;5:160-3.
21 Kurer MH, Lawson JO, Pambakian H. Suction biopsy in
Hirschsprungs disease. Arch Dis Child 1986;61:83-4.
22 Soave F. Hirschsprungs disease. Technique and results of
Soaves operation. Br J Surg 1966;53:1023-7.
23 Newbern WR. Hirschsprungs disease - the Duhamel modi-
cation. Am J Gastroenterol 1967;47:61-8.
24 Hackam DJ, Filler RM, Pearl RH. Enterocolitis after the
surgical treatment of Hirschsprungs disease: risk factors
and nancial impact. J Pediatr Surg 1998;33:830-3.
25 Yanchar NL, Soucy P. Long-term outcome after Hirsch-
sprungs disease: patients perspectives. J Pediatr Surg
1999;34:1152-60.
26 Moore SW, Albertyn R, Cywes S. Clinical outcome and
long-term quality of life after surgical correction of
Hirschsprungs disease. J Pediatr Surg 1996;31:1496-502.
27 Albanese CT, Jennings RW, Smith B, Bratton B, Harrison
MR. Perineal one-stage pull-through for Hirschsprungs
disease. J Pediatr Surg 1999;34:377-80.
28 Jasonni V, Martucciello G. Total colonic aganglionosis.
Semin Pediatr Surg 1998;7:174-80.
29 Tsuji H, Spitz L, Kiely EM, Drake DP, Pierro A.
Management and long-term follow-up of infants with total
colonic aganglionosis. J Pediatr Surg 1999;34:158-62.
30 PuVenberger EG, KauVman ER, Bolk S, Matise TC, Wash-
ington SS, Angrist M, Weissenbach J, Garver KL, Mascari
M, Ladda R, Slaugenhaupt SA, Chakravarti A. Identity-by-
descent and association mapping of a recessive gene for
Hirschsprung disease on human chromosome 13q22. Hum
Mol Genet 1994;3:1217-25.
31 Salomon R, Attie T, Pelet A, Bidaud C, Eng C, Amiel J, Sar-
nacki S, Goulet O, Ricour C, Nihoul-Fekete C, Munnich
A, Lyonnet S. Germline mutations of the RET ligand
GDNF are not suYcient to cause Hirschsprung disease.
Nat Genet 1996;14:345-7.
32 Sakai T, Wakizaka A, Nirasawa Y, Ito Y. Point nucleotidic
changes in both the RET proto-oncogene and the
endothelin-B receptor gene in a Hirschsprung disease
patient associated with Down syndrome. Tohoku J Exp Med
1999;187:43-7.
33 Martucciello G, Biocchini M, Dodero P, Cirillo M, Puliti A,
Gimelli G. Total colonic aganglionosis associated with
interstitial deletion of the long arm of chromosome 10.
Pediatr Surg Int 1992;7:308.
736 Amiel, Lyonnet
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
34 Fewtrell MS, Tam PK, Thomson AH, Fitchett M, Currie J,
Huson SM, Mulligan LM. Hirschsprungs disease associ-
ated with a deletion of chromosome 10 (q11.2q21.2): a
further link with the neurocristopathies? J Med Genet 1994;
31:325-7.
35 Sparkes RS, Sparkes MC, Kalina RE, Pagon RA, Salk DJ,
Disteche CM. Separation of retinoblastoma and esterase D
loci in a patient with sporadic retinoblastoma and
del(13)(q14.1q22.3). Hum Genet 1984;68:258-9.
36 Lamont MA, Fitchett M, Dennis NR. Interstitial deletion of
distal 13q associated with Hirschsprungs disease. J Med
Genet 1989;26:100-4.
37 Bottani A, Xie YG, Binkert F, Schinzel A. A case of
Hirschsprung disease with a chromosome 13 microdele-
tion, del(13)(q32.3q33.2): potential mapping of one
disease locus. Hum Genet 1991;87:748-50.
38 Lurie IW, Supovitz KR, Rosenblum-Vos LS, Wulfsberg EA.
Phenotypic variability of del(2) (q22-q23): report of a case
with a review of the literature. Genet Couns 1994;5:11-14.
39 Mowat DR, Croaker GD, Cass DT, Kerr BA, Chaitow J,
Ades LC, Chia NL, Wilson MJ. Hirschsprung disease,
microcephaly, mental retardation, and characteristic facial
features: delineation of a new syndrome and identication
of a locus at chromosome 2q22-q23. J Med Genet 1998;35:
617-23.
40 McMilin KD, Reiss JA, Brown MG, Black MH, Buckmaster
DA, Durum CT, Gunter KA, Lawce HJ, Berry TL, Lamb
OA, Olson CL, Weeks FF, Yoshitomi MJ, Jacky PB, Olson
SB, Magenis RE. Clinical outcomes of four patients with
microdeletion in the long arm of chromosome 2. Am J Med
Genet 1998;78:36-43.
41 Wakamatsu N, Yamada Y, Yamada K, On T, Nomura N,
Taniguchi H, Kitoh H, Mutoh N, Yamanaka T, Mushiake
K, Kato K, Sonta S, Nagaya M. Mutations in SIP1, encod-
ing Smad interacting protein-1, cause a form of Hirsch-
sprung disease. Nat Genet 2001;27:369-70.
42 Hansford JR, Mulligan LM. Multiple endocrine neoplasia
type 2 and RET: from neoplasia to neurogenesis. J Med
Genet 2000;37:817-27.
43 Verdy M, Weber AM, Roy CC, Morin CL, Cadotte M, Bro-
chu P. Hirschsprungs disease in a family with multiple
endocrine neoplasia type 2. J Pediatr Gastroenterol Nutr
1982;1:603-7.
44 Mulligan LM, Eng C, Attie T, Lyonnet S, Marsh DJ,
Hyland VJ, Robinson BG, Frilling A, Verellen-Dumoulin
C, Safar A, Venter DJ, Munnich A, Ponder BAJ. Diverse
phenotypes associated with exon 10 mutations of the RET
proto-oncogene. Hum Mol Genet 1994;3:2163-7.
45 Borst MJ, VanCamp JM, Peacock ML, Decker RA.
Mutational analysis of multiple endocrine neoplasia type
2A associated with Hirschsprungs disease. Surgery 1995;
117:386-91.
46 Caron P, Attie T, David D, Amiel J, Brousset F, Roger P,
Munnich A, Lyonnet S. C618R mutation in exon 10 of the
RET proto-oncogene in a kindred with multiple endocrine
neoplasia type 2A and Hirschsprungs disease. J Clin Endo-
crinol Metab 1996;81:2731-3.
47 Peretz H, Luboshitsky R, Baron E, Biton A, Gershoni R,
Usher S, Grynberg E, Yakobson E, GraV E, Lapidot M.
Cys 618 Arg mutation in the RET proto-oncogene associ-
ated with familial medullary thyroid carcinoma and mater-
nally transmitted Hirschsprungs disease suggesting a role
for imprinting. Hum Mutat 1997;10:155-9.
48 Decker RA, Peacock ML, Watson P. Hirschsprung disease
in MEN 2A: increased spectrum of RET exon 10
genotypes and strong genotype-phenotype correlation.
Hum Mol Genet 1998;7:129-34.
49 Romeo G, Ceccherini I, Celli J, Priolo M, Betsos N, Bonardi
G, Seri M, Yin L, Lerone M, Jasonni V, Martucciello G.
Association of multiple endocrine neoplasia type 2 and
Hirschsprung disease. J Intern Med 1998;243:515-20.
50 Borrego S, Eng C, Sanchez B, Saez ME, Navarro E,
Antinolo G. Molecular analysis of the ret and GDNF genes
in a family with multiple endocrine neoplasia type 2A and
Hirschsprung disease. J Clin Endocrinol Metab 1998;83:
3361-4.
51 Waardenburg PJ. A new syndrome combining developmen-
tal anomalies of the eyelids, eyebrows and nose root with
pigmentary defects of the iris and head hair and with con-
genital deafness. Am J Hum Genet 1951;3:195-253.
52 Hageman MJD. Heterogeneity in Waardenburg syndrome.
Am J Hum Genet 1977;29:468-85.
53 PuVenberger EG, Hosoda K, Washington SS, Nakao K,
deWit D, Yanagisawa M, Chakravarti A. A missense muta-
tion of the endothelin-B receptor gene in multigenic
Hirschsprungs disease. Cell 1994;79:1257-66.
54 Attie T, Till M, Pelet A, Amiel J, Edery P, Boutrand L,
Munnich A, Lyonnet S. Mutation of the endothelin-
receptor B gene in Waardenburg-Hirschsprung disease.
Hum Mol Genet 1995;4:2407-9.
55 Hofstra RM, Osinga J, Tan-Sindhunata G, Wu Y, Kamsteeg
EJ, Stulp RP, van Ravenswaaij-Arts C, Majoor-Krakauer D,
Angrist M, Chakravarti A, Meijers C, Buys CH. A
homozygous mutation in the endothelin-3 gene associated
with a combined Waardenburg type 2 and Hirschsprung
phenotype (Shah-Waardenburg syndrome). Nat Genet
1996;12:445-7.
56 Edery P, Attie T, Amiel J, Pelet A, Eng C, Hofstra RM, Mar-
telli H, Bidaud C, Munnich A, Lyonnet S. Mutation of the
endothelin-3 gene in the Waardenburg-Hirschsprung
disease (Shah-Waardenburg syndrome). Nat Genet 1996;
12:442-4.
57 Pingault V, Bondurand N, Kuhlbrodt K, Goerich DE,
Prehu MO, Puliti A, Herbarth B, Hermans-Borgmeyer I,
Legius E, Matthijs G, Amiel J, Lyonnet S, Ceccherini I,
Romeo G, Smith JC, Read AP, Wegner M, Goossens M.
SOX10 mutations in patients with Waardenburg-
Hirschsprung disease. Nat Genet 1998;18:171-3.
58 Jacobs JM, Wilson J. An unusual demyelinating neuropathy
in a patient with Waardenburgs syndrome. Acta Neuro-
pathol (Berl) 1992;83:670-4.
59 Touraine RL, Attie-Bitach T, Manceau E, Korsch E, Sarda
P, Pingault V, Encha-Razavi F, Pelet A, Auge J, Nivelon-
Chevallier A, Holschneider AM, Munnes M, Doerer W,
Goossens M, Munnich A, Vekemans M, Lyonnet S.
Neurological phenotype in Waardenburg syndrome type 4
correlates with novel SOX10 truncating mutations and
expression in developing brain. Am J Hum Genet 2000;66:
1496-503.
60 Bondurand N, Kuhlbrodt K, Pingault V, Enderich J, Sajus
M, Tommerup N, Warburg M, Hennekam RC, Read AP,
Wegner M, Goossens M. A molecular analysis of the Yem-
enite deaf-blind hypopigmentation syndrome: SOX10 dys-
function causes diVerent neurocristopathies. Hum Mol
Genet 1999;8:1785-9.
61 Gross A, Kunze J, Maier RF, Stoltenburg-Didinger G,
Grimmer I, Obladen M. Autosomal-recessive neural crest
syndrome with albinism, black lock, cell migration disorder
of the neurocytes of the gut, and deafness: ABCD
syndrome. Am J Med Genet 1995;56:322-6.
62 Mahakrishnan A, Srinivasan MS. Piebaldness with Hirsch-
sprungs disease. Arch Dermatol 1980;116:1102.
63 Kaplan P, de Chaderevian JP. Piebaldism-Waardenburg
syndrome: histopathologic evidence for a neural crest syn-
drome. Am J Med Genet 1988;31:679-88.
64 Weinberg AG, Currarino G, Besserman AM. Hirsch-
sprungs disease and congenital deafness. Familial associ-
ation. Hum Genet 1977;38:157-61.
65 Gozal D. Congenital central hypoventilation syndrome: an
update. Pediatr Pulmonol 1998;26:273-82.
66 Chatten J, Voorhess ML. Familial neuroblastoma. Report of
a kindred with multiple disorders, including neuroblasto-
mas in four siblings. N Engl J Med 1967;277:1230-6.
67 Roshkow JE, Haller JO, Berdon WE, Sane SM. Hirsch-
sprungs disease, Ondines curse, and neuroblastoma -
manifestations of neurocristopathy. Pediatr Radiol 1988;19:
45-9.
68 Levard G, Boige N, Vitoux C, Aigrain Y, Boureau M, Nav-
arro J. Neurocristopathy. The association of Hirschsprungs
disease-ganglioneuroma with autonomic nervous system
dysfunction in 2 children. Arch Fr Pediatr 1989;46:595-7.
69 Weese-Mayer DE, Silvestri JM, Marazita ML, Hoo JJ. Con-
genital central hypoventilation syndrome: inheritance and
relation to sudden infant death syndrome. Am J Med Genet
1993;47:360-7.
70 Haddad GG, Mazza NM, Defendini R, Blanc WA, Driscoll
JM, Epstein MA, Epstein RA, Mellins RB. Congenital fail-
ure of automatic control of ventilation, gastrointestinal
motility and heart rate. Medicine 1978;57:517-26.
71 Verloes A, Elmer C, Lacombe D, Heinrichs C, RebuVat E,
Demarquez JL, Moncla A, Adam E. Ondine-Hirschsprung
syndrome (Haddad syndrome). Further delineation in two
cases and review of the literature. Eur J Pediatr 1993;152:75-7.
72 Amiel J, Salomon R, Attie T, Pelet A, Trang H, Mokhtari M,
Gaultier C, Munnich A, Lyonnet S. Mutations of the RET-
GDNF signaling pathway in Ondines curse. Am J Hum
Genet 1996;62:715-17.
73 Sakai T, Wakizaka A, Matsuda H, Nirasawa Y, Itoh Y. Point
mutation in exon 12 of the receptor tyrosine kinase proto-
oncogene RET in Ondine-Hirschsprung syndrome. Pediat-
rics 1998;101:924-6.
74 Bolk S, Angrist M, Xie J, Yanagisawa M, Silvestri JM,
Weese-Mayer DE, Chakravarti A. Endothelin-3 frameshift
mutation in congenital central hypoventilation syndrome.
Nat Genet 1996;13:395-6.
75 Gaisie G, Oh KS, Young LW. Coexistent neuroblastoma and
Hirschsprungs disease -another manifestation of the
neurocristopathy? Pediatr Radiol 1979;8:161-3.
76 Michna BA, McWilliams NB, Krummel TM, Hartenberg
MA, Salzberg AM. Multifocal ganglioneuroblastoma coex-
istent with total colonic aganglionosis. J Pediatr Surg 1988;
23:57-9.
77 Merkler RG, Solish SB, Scherzer AL. Meningomyelocele
and Hirschprung disease: theoretical and clinical signi-
cance. Pediatrics 1985;76:299-300.
78 Clausen N, Andersson P, Tommerup N. Familial occur-
rence of neuroblastoma, von Recklinghausens neuro-
bromatosis, Hirschsprungs agangliosis and jaw-winking
syndrome. Acta Paediatr Scand 1989;78:736-41.
79 Goldberg RB, Shprintzen RJ. Hirschsprung megacolon and cleft
palate in two sibs. J Craniofac Genet Dev Biol 1981;1:185-9.
80 Hurst JA, Markiewicz M, Kumar D, Brett EM. Unknown
syndrome: Hirschsprungs disease, microcephaly, and iris
coloboma: a new syndrome of defective neuronal migra-
tion. J Med Genet 1988;25:494-7.
81 Halal F, Morel J. The syndrome of Hirschsprung disease,
microcephaly, unusual face, and mental retardation. Am J
Med Genet 1990;37:106-8.
82 Santos H, Mateus J, Leal MJ. Hirschsprung disease associ-
ated with polydactyly, unilateral renal agenesis, hyperte-
lorism, and congenital deafness: a new autosomal recessive
syndrome. J Med Genet 1988;25:204-5.
83 Laurence KM, Prosser R, Rocker I, Pearson JF, Richard C.
Hirschsprungs disease associated with congenital heart
malformation, broad big toes, and ulnar polydactyly in sibs:
a case for fetoscopy. J Med Genet 1975;12:334-8.
Hirschsprung disease, associated syndromes, and genetics 737
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
84 Al-Gazali LI, Donnai D, Mueller RF. Hirschsprungs
disease, hypoplastic nails, and minor dysmorphic features:
a distinct autosomal recessive syndrome? J Med Genet
1988;25:758-61.
85 Huang T, Elias ER, Mulliken JB, Kirse DJ, Holmes LB. A
new syndrome: heart defect, laryngeal anomalies, preaxial
polydactyly, and colonic aganglionosis in sibs. Genet Med
1999;1:104.
86 Reynolds JF, Barber JC, Alford BA, Chandler JG, Kelly TE.
Familial Hirschsprungs disease and type D brachydactyly:
a report of four aVected males in two generations. Pediatrics
1983;71:246-9.
87 Toriello HV, Komar K, Lawrence C, Higgins JV, Waterman
DF. Macrocephaly, Hirschsprung disease, brachydactyly,
vertebral defects, and other minor anomalies. Dysmorphol
Clin Genet 1988;1:155-7.
88 Reish O, Gorlin RJ, Hordinsky M, Rest EB, Burke B, Berry
SA. Brain anomalies, retardation of mentality and growth,
ectodermal dysplasia, skeletal malformations, Hirsch-
sprung disease, ear deformity and deafness, eye hypoplasia,
cleft palate, cryptorchidism, and kidney dysplasia/
hypoplasia (BRESEK/BRESHECK): new X-linked syn-
drome? Am J Med Genet 1997;68:386-90.
89 Davenport M, Taitz LS, Dickson JA. The Kaufman-
McKusick syndrome: another association. J Pediatr Surg
1989;24:1192-4.
90 Stone DL, Slavotinek A, BouVard GG, Banerjee-Basu S,
Baxevanis AD, Barr M, Biesecker LG. Mutation of a gene
encoding a putative chaperonin causes McKusick-
Kaufman syndrome. Nat Genet 2000;25:79-82.
91 Radetti G, Frick R, Pasquino B, Mengarda G, Savage MO.
Hypothalamic-pituitary dysfunction and Hirschsprungs
disease in the Bardet-Biedl syndrome. Helv Paediatr Acta
1988;43:249-52.
92 Lorda-Sanchez I, Ayuso C, Ibanez A. Situs inversus and
Hirschsprung disease: two uncommon manifestations in
Bardet-Biedl syndrome. Am J Med Genet 2000;90:80-1.
93 Slavotinek AM, Stone EM, Mykytyn K, Heckenlively JR,
Green JS, Heon E, Musarella MA, Parfrey PS, SheYeld
VC, Biesecker LG. Mutations in MKKS cause Bardet-
Biedl syndrome. Nat Genet 2000;26:15-16.
94 Wassif CA, Maslen C, Kachilele-Linjewile S, Lin D, Linck
LM, Connor WE, Steiner RD, Porter FD. Mutations in the
human sterol delta7-reductase gene at 11q12-13 cause
Smith-Lemli-Opitz syndrome. Am J Hum Genet 1998;63:
55-62.
95 Waterham HR, Wijburg FA, Hennekam RC, Vreken P, Poll-
The BT, Dorland L, Duran M, Jira PE, Smeitink JA,
Wevers RA, Wanders RJ. Smith-Lemli-Opitz syndrome is
caused by mutations in the 7-dehydrocholesterol reductase
gene. Am J Hum Genet 1998;63:329-38.
96 Patterson K, Toomey KE, Chandra RS. Hirschsprung
disease in a 46,XY phenotypic infant girl with Smith-
Lemli-Opitz syndrome. J Pediatr 1983;103:425-7.
97 Makitie O, Kaitila I. Cartilage-hair hypoplasia - clinical
manifestations in 108 Finnish patients. Eur J Pediatr 1993;
152:211-17.
98 Sulisalo T, Sistonen P, Hastbacka J, Wadelius C, Makitie O,
de la Chapelle A, Kaitila I. Cartilage-hair hypoplasia gene
assigned to chromosome 9 by linkage analysis. Nat Genet
1993;3:338-41.
99 Mandel H, Brik R, Ludatscher R, Braun J, Berant M.
Congenital muscular dystrophy with neurological
abnormalities: association with Hirschsprung disease. Am J
Med Genet 1993;47:37-40.
100 Kim JJ, Armstrong DD, Fishman MA. Multicore myo-
pathy, microcephaly, aganglionosis, and short stature. J
Child Neurol 1994;9:275-7.
101 Mallory SB, Haynie LS, Williams ML, Hall W. Ichthyosis,
deafness, and Hirschsprungs disease. Pediatr Dermatol
1989;6:24-7.
102 Kaplan P. X linked recessive inheritance of agenesis of the
corpus callosum. J Med Genet 1983;20:122-4.
103 Okamoto N, Wada Y, Goto M. Hydrocephalus and
Hirschsprungs disease in a patient with a mutation of
L1CAM. J Med Genet 1997;34:670-1.
104 Ikeda K, Goto S. Additional anomalies in Hirschsprungs
disease: an analysis based on the nationwide survey in
Japan. Z Kinderchir 1986;41:279-81.
105 Edward E, Ecker J, Christakis N, Folkman J. Hirsch-
sprungs disease: associated abnormalities and demogra-
phy. J Pediatr Surg 1992;27:76.
106 Russell MB, Russell CA, Niebuhr E. An epidemiological
study of Hirschsprungs disease and additional anomalies.
Acta Paediatr 1994;83:68-71.
107 Sarioglu A, Tanyel FC, Buyukpamukcu N, Hicsonmez A.
Hirschsprung-associated congenital anomalies. Eur J Pedi-
atr Surg 1997;7:331-7.
108 Schuchardt A, DAgati V, Larsson-Blomberg L, Costantini
F, Pachnis V. Defects in the kidney and enteric nervous sys-
tem of mice lacking the tyrosine kinase receptor Ret. Nature
1994;367:380-3.
109 Whalen TV Jr, Asch MJ. Report of two patients
with hypertrophic pyloric stenosis and Hirschsprungs dis-
ease. Coincident or common etiology? Am Surg 1985;51:
480-1.
110 Sayed M, al-Alaiyan S. Agenesis of corpus callosum,
hypertrophic pyloric stenosis and Hirschsprung disease:
coincidence or common etiology? Neuropediatrics 1996;27:
204-6.
111 Janik JP, Wayne ER, Janik JS, Price MR. Ileal atresia
with total colonic aganglionosis. J Pediatr Surg 1997;32:
1502-3.
112 Lyonnet S, Bolino A, Pelet A, Abel L, Nihoul-Fekete C,
Briard ML, Mok-Siu V, Kaariainen H, Martucciello G,
Lerone M, Puliti A, Luo Y, Weissenbach J, Devoto M,
Munnich A, Romeo G. A gene for Hirschsprung disease
maps to the proximal long arm of chromosome 10. Nat
Genet 1993;4:346-50.
113 Angrist M, KauVman E, Slaugenhaupt SA, Matise TC,
PuVenberger EG, Washington SS, Lipson A, Cass DT,
Reyna T, Weeks DE, Sieber W, Chakravati A. A gene for
Hirschsprung disease (megacolon) in the pericentromeric
region of human chromosome 10. Nat Genet 1993;4:351-6.
114 Mulligan LM, Kwok JB, Healey CS, Elsdon MJ, Eng C,
Gardner E, Love DR, Mole SE, Moore JK, Papi L. Germ-
line mutations of the RET proto-oncogene in multiple
endocrine neoplasia type 2A. Nature 1993;363:458-60.
115 Donis-Keller H, Dou S, Chi D, Carlson KM, Toshima K,
Lairmore TC, Howe JR, Moley JF, Goodfellow P, Wells
SA. Mutations in the RET proto-oncogene are associated
with MEN 2A and FMTC. Hum Mol Genet 1993;2:851-6.
116 Edery P, Lyonnet S, Mulligan LM, Pelet A, Dow E, Abel
L, Holder S, Nihoul-Fekete C, Ponder BA, Munnich A.
Mutations of the RET proto-oncogene in Hirschsprungs
disease. Nature 1994;367:378-80.
117 Romeo G, Ronchetto P, Luo Y, Barone V, Seri M,
Ceccherini I, Pasini B, Bocciardi R, Lerone M, Kaariainen
H, Martucciello G. Point mutations aVecting the tyrosine
kinase domain of the RET proto-oncogene in Hirschs-
prungs disease. Nature 1994;367:377-8.
118 Ceccherini I, Bocciardi R, Luo Y, Pasini B, Hofstra R,
Takahashi M, Romeo G. Exon structure and anking
intronic sequences of the human RET proto-oncogene.
Biochem Biophys Res Commun 1993;196:1288-95.
119 Attie T, Pelet A, Edery P, Eng C, Mulligan LM, Amiel J,
Boutrand L, Beldjord C, Nihoul-Fekete C, Munnich A,
Ponder BAJ, Lyonnet S. Diversity of RET proto-oncogene
mutations in familial and sporadic Hirschsprung disease.
Hum Mol Genet 1995;4:1381-6.
120 Angrist M, Bolk S, Thiel B, PuVenberger EG, Hofstra RM,
Buys CH, Cass DT, Chakravarti A. Mutation analysis of
the RET receptor tyrosine kinase in Hirschsprung disease.
Hum Mol Genet 1995;4:821-30.
121 Seri M, Yin L, Barone V, Bolino A, Celli I, Bocciardi R,
Pasini B, Ceccherini I, Lerone M, KristoVersson U,
Larsson LT, Casasa JM, Cass DT, Abramowicz MJ,
Vanderwinden JM, Kravcenkiene I, Baric I, Silengo M,
Martucciello G, Romeo G. Frequency of RET mutations
in long- and short-segment Hirschsprung disease. Hum
Mutat 1997;9:243-9.
122 Eng C, Smith DP, Mulligan LM, Nagai MA, Healey CS,
Ponder MA, Gardner E, Scheumann GF, Jackson CE,
TunnacliVe A, Ponder BAJ. Point mutation within the tyro-
sine kinase domain of the RET proto-oncogene in multiple
endocrine neoplasia type 2B and related sporadic tumours
[published erratum appears in Hum Mol Genet 1994;3:
686]. Hum Mol Genet 1994;3:237-41.
123 Hofstra RM, Landsvater RM, Ceccherini I, Stulp R, Stel-
wagen T, Luo Y, Pasini B, Hoppener JW, van Amstel
HKGR. A mutation in the RET proto-oncogene associated
with multiple endocrine neoplasia type 2B and sporadic
medullary thyroid carcinoma. Nature 1994;367:375-6.
124 Carlson KM, Dou S, Chi D, Scavarda N, Toshima K, Jack-
son CE, Wells SAJ, Goodfellow PJ, Donis-Keller H. Single
missense mutation in the tyrosine kinase catalytic domain
of the RET protooncogene is associated with multiple
endocrine neoplasia type 2B. Proc Natl Acad Sci USA
1994;91:1579-83.
125 Santoro M, Carlomagno F, Romano A, Bottaro DP,
Dathan NA, Grieco M, Fusco A, Vecchio G, Matoskova B,
Kraus MH. Activation of RET as a dominant transforming
gene by germline mutations of MEN2A and MEN2B.
Science 1995;267:381-3.
126 Pasini B, Borrello MG, Greco A, Bongarzone I, Luo Y,
Mondellini P, Alberti L, Miranda C, Arighi E, Bocciardi R,
Seri M, Barone V, Radice MT, Romeo G, Pierotti MA.
Loss of function eVect of RET mutations causing Hirsch-
sprung disease. Nat Genet 1995;10:35-40.
127 Carlomagno F, De Vita G, Berlingieri MT, de Franciscis V,
Melillo RM, Colantuoni V, Kraus MH, Di Fiore PP, Fusco
A, Santoro M. Molecular heterogeneity of RET loss of
function in Hirschsprungs disease. EMBO J 1996;15:
2717-25.
128 Pelet A, Geneste O, Edery P, Pasini A, Chappuis S, Atti T,
Munnich A, Lenoir G, Lyonnet S, Billaud M. Various
mechanisms cause RET-mediated signaling defects in Hir-
schsprungs disease. J Clin Invest 1998;101:1415-23.
129 Bolk S, Pelet A, Hofstra RM, Angrist M, Salomon R,
Croaker D, Buys CH, Lyonnet S, Chakravarti A. A human
model for multigenic inheritance: phenotypic expression in
Hirschsprung disease requires both the RET gene and a
new 9q31 locus. Proc Natl Acad Sci USA 2000;97:268-73.
130 Borrego S, Saez ME, Ruiz A, Gimm O, Lopez-Alonso M,
Antinolo G, Eng C. Specic polymorphisms in the RET
proto-oncogene are over-represented in patients with
Hirschsprung disease and may represent loci modifying
phenotypic expression. J Med Genet 1999;36:771-4.
131 Borrego S, Ruiz A, Saez ME, Gimm O, Gao X,
Lopez-Alonso M, Hernandez A, Wright FA, Antinolo G,
Eng C. RET genotypes comprising specic haplotypes of
polymorphic variants predispose to isolated Hirschsprung
disease. J Med Genet 2000;37:572-8.
132 Moore MW, Klein RD, Farinas I, Sauer H, Armanini M,
Phillips H, Reichardt LF, Ryan AM, Carver-Moore K,
Rosenthal A. Renal and neuronal abnormalities in mice
lacking GDNF. Nature 1996;382:76-9.
738 Amiel, Lyonnet
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
133 Sanchez MP, Silos-Santiago I, Frisen J, He B, Lira SA,
Barbacid M. Renal agenesis and the absence of enteric
neurons in mice lacking GDNF. Nature 1996;382:70-3.
134 Pichel JG, Shen L, Sheng HZ, Granholm AC, Drago J,
Grinberg A, Lee EJ, Huang SP, Saarma M, HoVer BJ, Sari-
ola H, Westphal H. Defects in enteric innervation and kid-
ney development in mice lacking GDNF. Nature 1996;382:
73-6.
135 Durbec P, Marcos-Gutierrez CV, Kilkenny C, Grigoriou
M, Wartiowaara K, Suvanto P, Smith D, Ponder B, Costan-
tini F, Saarma M, Sariola H, Pachnis V. GDNF signalling
through the Ret receptor tyrosine kinase. Nature 1996;381:
789-93.
136 Jing S, Wen D, Yu Y, Holst PL, Luo Y, Fang M, Tamir R,
Antonio L, Hu Z, Cupples R, Louis JC, Hu S, Altrock BW,
Fox GM. GDNF-induced activation of the ret protein
tyrosine kinase is mediated by GDNFR-alpha, a novel
receptor for GDNF. Cell 1996;85:1113-24.
137 Treanor JJ, Goodman L, de Sauvage F, Stone DM, Poulsen
KT, Beck CD, Gray C, Armanini MP, Pollock RA, Hefti F,
Phillips HS, Goddard A, Moore MW, Buj-Bello A, Davies
AM, Asai N, Takahashi M, Vandlen R, Henderson CE,
Rosenthal A. Characterization of a multicomponent recep-
tor for GDNF. Nature 1996;382:80-3.
138 Rosenthal A. The GDNF protein family: gene ablation
studies reveal what they really do and how. Neuron
1999;22:201-3.
139 Kotzbauer PT, Lampe PA, Heuckeroth RO, Golden JP,
Creedon DJ, Johnson EM, Milbrandt J. Neurturin, a rela-
tive of glial-cell-line-derived neurotrophic factor. Nature
1996;384:467-70.
140 Milbrandt J, de Sauvage FJ, Fahrner TJ, Baloh RH,
Leitner ML, Tansey MG, Lampe PA, Heuckeroth RO,
Kotzbauer PT, Simburger KS, Golden JP, Davies JA,
Vejsada R, Kato AC, Hynes M, Sherman D, Nishimura M,
Wang LC, Vandlen R, MoVat B, Klein RD, Poulsen K,
Gray C, Garces A, Johnson EM, Henderson CE, Phillips
HS, Johnson EM Jr. Persephin, a novel neurotrophic factor
related to GDNF and neurturin. Neuron 1998;20:245-53.
141 Baloh RH, Tansey MG, Lampe PA, Fahrner TJ, Enomoto
H, Simburger KS, Leitner ML, Araki T, Johnson EM, Mil-
brandt J. Artemin, a novel member of the GDNF ligand
family, supports peripheral and central neurons and signals
through the GFRalpha3-RET receptor complex. Neuron
1998;21:1291-302.
142 Angrist M, Bolk S, Halushka M, Lapchak PA, Chakravarti
A. Germline mutations in glial cell line-derived neuro-
trophic factor (GDNF) and RET in a Hirschsprung
disease patient. Nat Genet 1996;14:341-4.
143 Ivanchuk SM, Myers SM, Eng C, Mulligan LM. De novo
mutation of GDNF, ligand for the RET/GDNFR-alpha
receptor complex, in Hirschsprung disease. Hum Mol Genet
1996;5:2023-6.
144 Doray B, Salomon R, Amiel J, Pelet A, Touraine R, Billaud
M, Attie T, Bachy B, Munnich A, Lyonnet S. Mutation of
the RET ligand, neurturin, supports multigenic inheritance
in Hirschsprung disease. Hum Mol Genet 1998;7:1449-52.
145 Cacalano G, Farinas I, Wang LC, Hagler K, Forgie A,
Moore M, Armanini M, Phillips H, Ryan AM, Reichardt
LF, Hynes M, Davies A, Rosenthal A. GFRalpha1 is an
essential receptor component for GDNF in the developing
nervous system and kidney. Neuron 1998;21:53-62.
146 Enomoto H, Araki T, Jackman A, Heuckeroth RO, Snider
WD, Johnson EM, Milbrandt J. GFR alpha1-decient mice
have decits in the enteric nervous system and kidneys.
Neuron 1998;21:317-24.
147 Angrist M, Jing S, Bolk S, Bentley K, Nallasamy S,
Halushka M, Fox GM, Chakravarti A. Human GFRA1:
cloning, mapping, genomic structure, and evaluation as a
candidate gene for Hirschsprung disease susceptibility.
Genomics 1998;48:354-62.
148 Myers SM, Salomon R, Goessling A, Pelet A, Eng C, von
Deimling A, Lyonnet S, Mulligan LM. Investigation of
germline GFR-1 mutations in Hirschsprung disease. J
Med Genet 1999;36:217-20.
149 Inoue A, Yanagisawa M, Kimura S, Kasuya Y, Miyauchi T,
Goto K, Masaki T. The human endothelin family: three
structurally and pharmacologically distinct isopeptides
predicted by three separate genes. Proc Natl Acad Sci USA
1989;86:2863-7.
150 Sakurai T, Yanagisawa M, Masaki T. Molecular characteri-
zation of endothelin receptors. Trends Pharmacol Sci 1992;
13:103-8.
151 Cohen IT, Gadd MA. Hirschsprungs disease in a kindred:
a possible clue to the genetics of the disease. J Pediatr Surg
1982;17:632-4.
152 Van Camp G, Van Thienen MN, Handig I, Van Roy B, Rao
VS, Milunsky A, Read AP, Baldwin CT, Farrer LA, Bondu-
elle M, Standaert L, Meire F, Willems PJ. Chromosome
13q deletion with Waardenburg syndrome: further evi-
dence for a gene involved in neural crest function on 13q. J
Med Genet 1995;32:531-6.
153 Kiss P, Orsztovics M. Association of 13q deletion and Hir-
schsprungs disease. J Med Genet 1989;26:793-4.
154 Hosoda K, Hammer RE, Richardson JA, Baynash AG,
Cheung JC, Giaid A, Yanagisawa M. Targeted and natural
(piebald-lethal) mutations of endothelin-B receptor gene
produce megacolon associated with spotted coat color in
mice. Cell 1994;79:1267-76.
155 Baynash AG, Hosoda K, Giaid A, Richardson JA, Emoto
N, Hammer RE, Yanagisawa M. Interaction of
endothelin-3 with endothelin-B receptor is essential for
development of epidermal melanocytes and enteric neu-
rons. Cell 1994;79:1277-85.
156 Bidaud C, Salomon R, Van Camp G, Pelet A, Attie T, Eng
C, Bonduelle M, Amiel J, Nihoul-Fekete C, Willems PJ,
Munnich A, Lyonnet S. Endothelin-3 gene mutations in
isolated and syndromic Hirschsprung disease. Eur J Hum
Genet 1997;5:247-51.
157 Chakravarti A. Endothelin receptor-mediated signaling in
Hirschsprung disease. Hum Mol Genet 1996;5:303-7.
158 Amiel J, Attie T, Jan D, Pelet A, Edery P, Bidaud C,
Lacombe D, Tam P, Simeoni J, Flori E, Nihoul-Fekete C,
Munnich A, Lyonnet S. Heterozygous endothelin receptor
B (EDNRB) mutations in isolated Hirschsprung disease.
Hum Mol Genet 1996;5:355-7.
159 Kusafuka T, Wang Y, Puri P. Novel mutations of the
endothelin-B receptor gene in isolated patients with
Hirschsprungs disease. Hum Mol Genet 1996;5:347-9.
160 Auricchio A, Casari G, Staiano A, Ballabio A.
Endothelin-B receptor mutations in patients with isolated
Hirschsprung disease from a non-inbred population. Hum
Mol Genet 1996;5:351-4.
161 Yanagisawa H, Yanagisawa M, Kapur RP, Richardson JA,
Williams SC, Clouthier DE, de Wit D, Emoto N, Hammer
RE. Dual genetic pathways of endothelin-mediated inter-
cellular signaling revealed by targeted disruption of
endothelin converting enzyme-1 gene. Development 1998;
125:825-36.
162 Hofstra RM, Valdenaire O, Arch E, Osinga J, Kroes H,
LoZer BM, Hamosh A, Meijers C, Buys CH. A
loss-of-function mutation in the endothelin-converting
enzyme 1 (ECE-1) associated with Hirschsprung disease,
cardiac defects, and autonomic dysfunction. Am J Hum
Genet 1999;64:304-8.
163 Lane PW, Liu HM. Association of megacolon with a new
dominant spotting gene (Dom) in the mouse. J Hered
1984;75:435-9.
164 Herbarth B, Pingault V, Bondurand N, Kuhlbrodt K,
Hermans-Borgmeyer I, Puliti A, Lemort N, Goossens M,
Wegner M. Mutation of the Sry-related Sox10 gene in
dominant megacolon, a mouse model for human Hirsch-
sprung disease. Proc Natl Acad Sci USA 1998;95:5161-5.
165 Southard-Smith EM, Angrist M, Ellison JS, Agarwala R,
Baxevanis AD, Chakravarti A, Pavan WJ. The Sox10
(Dom) mouse: modeling the genetic variation of
Waardenburg-Shah (WS4) syndrome. Genome Res 1999;9:
215-25.
166 Auricchio A, Griseri P, Carpentieri ML, Betsos N, Staiano
A, Tozzi A, Priolo M, Thompson H, Bocciardi R, Romeo
G, Ballabio A, Ceccherini I. Double heterozygosity for a
RET substitution interfering with splicing and an EDNRB
missense mutation in Hirschsprung disease. Am J Hum
Genet 1999;64:1216-21.
167 Potterf SB, Furumura M, Dunn KJ, Arnheiter H, Pavan
WJ. Transcription factor hierarchy in Waardenburg
syndrome: regulation of MITF expression by SOX10 and
PAX3. Hum Genet 2000;107:1-6.
168 Southard-Smith EM, Kos L, Pavan WJ. Sox10 mutation
disrupts neural crest development in Dom Hirschsprung
mouse model. Nat Genet 1998;18:60-4.
169 Bolk S, Angrist M, Fornage M, Olson JM, Hofstra R, Buys
C, Chakravarti A (group I). Salomon R, Pelet A, Amiel J,
Atti-Bitach T, SteVann J, Munnich A, Lyonnet S (group
II). Splitting a multigenic disease: segregation at three loci
explains familial and population risk in Hirschsprung
disease (in preparation).
170 Hall CM. Werners mesomelic dysplasia with ventricular
septal defect and Hirschsprungs disease. Pediatr Radiol
1981;10:2479.
Hirschsprung disease, associated syndromes, and genetics 739
www.jmedgenet.com
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from
doi: 10.1136/jmg.38.11.729
2001 38: 729-739 J Med Genet

Jeanne Amiel and Stanislas Lyonnet

and genetics: a review


Hirschsprung disease, associated syndromes,
http://jmg.bmj.com/content/38/11/729.full.html
Updated information and services can be found at:
These include:
References
http://jmg.bmj.com/content/38/11/729.full.html#related-urls
Article cited in:

http://jmg.bmj.com/content/38/11/729.full.html#ref-list-1
This article cites 159 articles, 42 of which can be accessed free at:
service
Email alerting
box at the top right corner of the online article.
Receive free email alerts when new articles cite this article. Sign up in the
Collections
Topic
(236 articles) Clinical genetics
(566 articles) Epidemiology

Articles on similar topics can be found in the following collections


Notes
http://group.bmj.com/group/rights-licensing/permissions
To request permissions go to:
http://journals.bmj.com/cgi/reprintform
To order reprints go to:
http://group.bmj.com/subscribe/
To subscribe to BMJ go to:
group.bmj.com on March 31, 2014 - Published by jmg.bmj.com Downloaded from

You might also like