Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

Pediatr Surg Int (2013) 29:855–872

DOI 10.1007/s00383-013-3351-3

REVIEW ARTICLE

Classification and diagnostic criteria of variants


of Hirschsprung’s disease
Florian Friedmacher • Prem Puri

Published online: 14 August 2013


Ó Springer-Verlag Berlin Heidelberg 2013

Abstract ‘‘Variants of Hirschsprung’s disease’’ are con- Introduction


ditions that clinically resemble Hirschsprung’s disease
(HD), despite the presence of ganglion cells in rectal suc- The typical patient with Hirschsprung’s disease (HD) is
tion biopsies. The diagnosis and management of these either a newborn presenting with delayed passage of
patients can be challenging. Specific histological, immu- meconium and abdominal distension or a young child with
nohistochemical and electron microscopic investigations severe chronic constipation. 80–90 % of all HD cases
are required to characterize this heterogeneous group of produce clinical symptoms and are diagnosed in the neo-
functional bowel disorders. Variants of HD include intes- natal period. The diagnosis of HD is confirmed by histo-
tinal neuronal dysplasia, intestinal ganglioneuromatosis, logical and histochemical evaluation of rectal suction
isolated hypoganglionosis, immature ganglia, absence of biopsies, which demonstrate the absence of ganglion cells
the argyrophil plexus, internal anal sphincter achalasia and in the submucosa and increased acetylcholinesterase
congenital smooth muscle cell disorders such as mega- (AChE) activity in the lamina propria. However, there are a
cystis microcolon intestinal hypoperistalsis syndrome. This number of patients who clinically resemble HD despite the
review article systematically classifies variants of HD presence of ganglion cells in rectal biopsies. Various terms
based on current diagnostic criteria with an additional such as ‘‘chronic idiopathic intestinal pseudo-obstruction’’,
focus on pathogenesis, epidemiology, clinical presentation, ‘‘intestinal hypoperistalsis syndrome’’ or ‘‘pseudo-Hirsch-
management and outcome. sprung’s disease’’ have been used to describe these
conditions. The diagnosis and management of these
Keywords Hirschsprung’s disease  Intestinal patients can be challenging for both pediatric surgeons and
neuronal dysplasia  Ganglioneuromatosis  gastroenterologists.
Hypoganglionosis  Immature ganglia  Internal anal During the past three decades, our group has focused its
sphincter achalasia  Megacystis microcolon research interest in delineating these conditions based on
intestinal hypoperistalsis syndrome specific histological, immunohistochemical and electron
microscopic studies. In 1997, we suggested that ‘‘Variant
Hirschsprung’s disease’’ [1, 2] may be a more appropriate
description for this heterogeneous group of functional
bowel disorders (Table 1) in patients who continue to have
persistent bowel symptoms despite a ganglionic rectal
F. Friedmacher  P. Puri (&)
biopsy. At present, there are only a few articles in the
National Children’s Research Centre, Our Lady’s Children’s
Hospital, Crumlin, Dublin 12, Ireland literature which have attempted to standardize the termi-
e-mail: prem.puri@ucd.ie nology of HD and allied intestinal disorders [3, 4]. This
review article systematically classifies variants of HD
P. Puri
based on current diagnostic criteria (Fig. 1) with an addi-
Conway Institute of Biomolecular and Biomedical Research,
School of Medicine and Medical Science, University College tional focus on pathogenesis, epidemiology, clinical pre-
Dublin, Dublin, Ireland sentation, management and outcome.

123
856 Pediatr Surg Int (2013) 29:855–872

Intestinal neuronal dysplasia 95 % of all IND cases. Typical histological features of IND
B are hyperganglionosis, giant ganglia, ectopic ganglion
Intestinal neuronal dysplasia (IND) was first described in cells and increased AChE activity in the lamina propria and
1971 by Meyer–Ruge as a hyperplastic malformation of the around submucosal blood vessels [2]. IND occurring in
enteric plexus [5]. Shortly afterwards, Puri et al. [6] association with HD is always IND B.
reported a case of IND in association with HD, who had
rectosigmoid aganglionosis combined with IND of the Pathogenesis of IND
descending and transverse colon. Fadda et al. [7] subse-
quently classified IND into two clinical and histological There still remains a controversy surrounding the existence
distinct subtypes: IND type A (IND A), which occurs in of IND as a distinct histopathological entity [3, 8–10]. It
less than 5 % of all IND cases, is characterized by con- has been suggested by several authors that the observed
genital aplasia or hypoplasia of the sympathetic innerva- changes seen in IND may be either a variant of normal
tion. Patients with IND A typically present in the neonatal bowel development or a secondary acquired phenomenon
period with abdominal distension, bowel obstruction and caused by congenital obstruction or inflammation [11, 12].
episodes of diarrhea with hemorrhagic stools. IND type B An underlying autoimmune mechanism has also been
(IND B) is characterized by hyperplasia of the parasym- proposed [13]. However, multiple familial cases of IND
pathetic submucosal plexus and accounts for more than have been described in the literature, suggesting that there
might also be a genetic component [14, 15]. The strongest
evidence that IND is a real entity arose from experimental
Table 1 Variants of Hirschsprung’s disease animal models. Hox11L1 is a homeobox gene involved in
Intestinal neuronal dysplasia
the proliferation and differentiation of peripheral neural
Intestinal ganglioneuromatosis
crest cells. Two different Hox11L1 knockout mouse mod-
Isolated hypoganglionosis
els have been generated [16, 17]. In both cases, homozy-
gous mutant mice developed megacolon at the age of
Immature ganglia
3–5 weeks. Histological and immunohistochemical evalu-
Absence of the argyrophil plexus
ation revealed hyperplasia of ganglia similar to the phe-
Internal anal sphincter achalasia
notype observed in human IND. Another animal model
Megacystis microcolon intestinal hypoperistalsis syndrome
resulting in a phenotype similar to IND was reported in rats

Fig. 1 Diagnostic algorithm for investigating chronic constipation and functional bowel obstruction in newborn infants and young children

123
Pediatr Surg Int (2013) 29:855–872 857

with a heterozygous mutation of the endothelin B receptor more specific staining techniques were required to assess
(EDNRB) showing features of hyperganglionosis, giant accurately the enteric nervous system [45]. Meier-Ruge et al.
ganglia and hypertrophied nerve fiber strands in the sub- [46–48] have proposed enzyme histochemistry for lactate
mucosal plexus [18]. However, mutational screening of dehydrogenase, succinate dehydrogenase and nitric oxide
Hox11L1 and EDNRB genes in human patients with IND synthase to evaluate and diagnose IND B quantitatively.
demonstrated no mutations in these genes [19–21]. Various other neuronal and glial markers such as nicotin-
amide adenine dinucleotide phosphate diaphorase (NADPH-
Epidemiology of IND d) (Fig. 2c, d), neural cell adhesion molecule (NCAM),
neuron-specific enolase (NSE), cathepsin D, protein gene
Intestinal neuronal dysplasia occurs with an incidence of protein 9.5 (PGP9.5), S-100 protein, peripherin, synapto-
approximately 1 in every 7,500 newborns [22]. However, physin and cuprolinic blue have also been used [49–52].
the frequency of isolated IND varies highly between the Cuprolinic blue staining has been suggested as it stains the
different centers worldwide with reported rates between 0.3 whole population of ganglion cells [52, 53], but only the cell
and 40 % of all rectal suction biopsies [12, 23, 24]. IND bodies and not the axons, which makes it relatively easy to
proximal to a segment of aganglionosis is not uncommon distinguish between the individual cells. Furthermore, a
and has been suggested as a possible cause of persistent defective innervation of the neuromuscular junction within
bowel problems after definitive surgery for HD [25]. Some the affected bowel segment of patients with IND has been
authors have found IND associated with HD in up to 44 % shown [54]. Abnormal submucosal vasculature is a further
of patients, while others have rarely observed this combi- histological finding in isolated IND and IND associated with
nation [26–30]. The high variability of patient age, speci- HD and may be a useful additional diagnostic feature [55]. In
men type and staining methods has resulted in considerable addition, a reduced number of c-Kit positive interstitial cells
confusion in the literature regarding the accurate diagnostic of Cajal (ICCs) has been demonstrated in the myenteric
criteria [31]. plexus and muscle layers [56, 57]. More recently, a marked
reduction in PTEN expression in the submucosal and
Clinical presentation of IND myenteric plexuses of patients with IND has been discov-
ered, which may explain the observed motility dysfunction
The majority of patients with IND present with chronic [58]. Due to the controversy in staining techniques and age-
constipation with or without abdominal distension, thus dependent AChE expression, the most commonly used
resembling HD [32], but with a normal barium contrast diagnostic criteria at present are: (1) more than 20 % of 25
enema. It has been shown that intestinal obstruction is the submucosal ganglia must be giant ganglia containing 9 or
most characteristic clinical symptom of IND in infants and more ganglion cells and (2) the patient must be older than
young children [33]. Furthermore, there is a high incidence 1 year, as before that age, giant ganglia may be misinter-
of additional congenital anomalies, currently ranging from preted due to the fact that immature ganglia often have an
25 to 30 % [34]. The most common ones are anorectal incomplete differentiation in nerve cells [59–61]. Most
malformations, intestinal malrotation, megacystis, con- patients with IND do not display any specific radiological
genital short bowel, hypertrophic pyloric stenosis, necro- features on barium contrast enemas other than rectosigmoid
tizing enterocolitis and Down syndrome [35, 36]. distension. The rectosphincteric reflex has often been shown
to be present, absent or atypical in these patients [34].
Diagnosis of IND
Management of IND
Rectal suction biopsy is the method of choice for the diag-
nosis of IND. It is essential to include a sufficient amount of There is broad compliance with the recommendation that
submucosa in the biopsy specimens. The diagnosis of IND the management of IND B should be conservative in the
was previously based on qualitative criteria, thus resulting in first instance, consisting of laxatives and enemas [62]. The
a high intraobserver variation [37, 38]. Therefore, the debate majority of patients have been shown to respond well to
about the existence of IND as a distinct histopathological this treatment strategy. However, if bowel symptoms per-
entity remains controversial, which is mainly due to the lack sist longer than 6 months of conservative bowel manage-
of consensus in diagnostic criteria [11, 39]. IND was initially ment, surgical treatment options should be considered [63].
diagnosed on the basis of AChE histochemistry of nerve Myectomy of the internal anal sphincter has been per-
fibers in rectal suction biopsies [40, 41] (Fig. 2a, b). How- formed by several authors with satisfactory results [2].
ever, since the AChE activity in the lamina propria mucosae Some investigators also recommend injection of botulinum
has been shown to be an age-dependent phenomenon that toxin into the internal anal sphincter. Resection of the
disappears on maturation of the submucosal plexus [42–44], affected bowel segment and pull-through procedure is

123
858 Pediatr Surg Int (2013) 29:855–872

Fig. 2 AChE staining of a normal rectal suction biopsy (a). Rectal NADPH-d staining of a normal submucosal plexus (c). Submucosal
suction biopsy from a patient with IND showing hyperganglionosis, plexus of a patient with IND showing giant ganglia (d)
giant ganglia and increased AChE activity in the lamina propria (b).

rarely indicated in infants and children with IND [34]. In [66]. Gillick et al. [67] reported functional outcome in 33
adolescent and adult patients with IND, resection of the patients with IND. After a mean follow-up of 2.4 years,
dilated and redundant colon segment is often the only 64 % of their patients had a good response to conservative
successful therapeutic option [64]. The indication for sur- management with normal bowel habits and did not require
gery should not be determined on the basis of histopa- any surgical intervention. However, 36 % underwent
thological findings alone, instead the decision must be myectomy of the internal anal sphincter after failed con-
based on the individual patient’s clinical symptoms and servative treatment. Seven out of these 12 patients had
distress [65]. normal bowel habits after surgery and two were able to stay
clean with regular enemas. Three patients continued having
Outcome of IND persistent constipation after myectomy and subsequently
underwent resection of redundant and dilated sigmoid
An intensive long-term follow-up involving a team of colon, which resulted in normal bowel habits. Schärli et al.
experienced pediatric surgeons and gastroenterologists is [29] achieved satisfactory results in 90 % of their patients
necessary in patients with IND and chronic constipation within 6 months after internal sphincter myectomy.

123
Pediatr Surg Int (2013) 29:855–872 859

Intestinal ganglioneuromatosis Diagnosis of intestinal GNM

Intestinal ganglioneuromatosis (GNM) is characterized by The diagnosis of intestinal GNM is mainly based on
diffuse proliferation of nerve fibers with significant clinical presentation and histological examination of
hyperplasia of submucosal and myenteric ganglion cells rectal suction or full-thickness biopsies showing massive
causing thickening of the bowel wall [68]. This extremely proliferation of submucosal and myenteric plexuses
rare but severe neoplastic condition leads to chronic bowel comprising thick nerve trunks with scattered mature
obstruction and is frequently associated with multiple neurons, giant ganglia with often 15–40 nerve cells and a
endocrine neoplasia type 2B (MEN 2B), neurofibromatosis high AChE activity [77–80]. Intestinal GNM appears to
1 or Cowden syndrome [69]. be largely confined to the colon and rectum, unlike
neurofibromatosis, which occurs more commonly in the
Pathogenesis small intestine [81]. Although AChE histochemistry has
been suggested to show the typical submucosal and
The pathogenesis of intestinal GNM is related to complex myenteric changes in this condition, highlighting the
hyperplasia of peptidergic, cholinergic and probably thickness of the nerve fibers, it can also be quite easily
adrenergic nerve fibers and neurons [70]. Transmural GNM appreciated in standard H&E-stained paraffin sections
mainly originates from the myenteric plexus, while [82]. In addition, NSE, synaptophysin and S-100 protein
mucosal GNM predominantly affects the submucosal immunostaining has been introduced to evaluate and
plexus and is often associated with von Recklinghausen’s diagnose intestinal GNM [70]. It has also been demon-
disease [70]. Mutation analysis in patients with MEN 2B strated that the submucosal hyperplasia can be extensive,
further confirmed a de novo germline Met918Thr mutation but not as prominent as the one described in the
in exon 16 of the RET proto-oncogene [71], suggesting a myenteric plexus [82, 83]. As intestinal GNM is fre-
clear genetic component to this condition. In addition, a quently associated with MEN 2B, the diagnosis should
recent experimental study in mice revealed that deletion of prompt additional molecular, endocrinological and
the PTEN gene on chromosome 10 disrupts the develop- oncological evaluation [84, 85]. In general, a mutational
ment of the enteric nerve system resulting in a phenotype analysis of the RET proto-oncogene is strongly recom-
similar to human intestinal GNM [72]. mended in all patients with intestinal GNM and MEN
2B as well as their family members [76].
Epidemiology of intestinal GNM
Management of intestinal GNM
The exact incidence of intestinal GNM is unknown, but it
has been reported that the frequently associated MEN 2B Surgical resection of the affected bowel segment is not
syndrome occurs in approximately 1:4,000,000 live births always mandatory in patients with MEN 2B-associated
[71]. Furthermore, it can be estimated that intestinal GNM intestinal GNM [86]. It has been shown that in the majority
is present in 90 % of patients with MEN 2B [73]. of cases, the gastrointestinal symptoms can be managed
with daily laxatives and enemas [87]. However, some
Clinical presentation of intestinal GNM patients ultimately require surgery for severe intestinal
bowel obstruction or stricture formation [88]. Furthermore,
The vast majority of patients with intestinal GNM present all patients with intestinal GNM that carry MEN 2B
with severe chronic constipation and abdominal distension mutations should undergo a prophylactic total thyroidec-
due to intestinal obstruction [73, 74], similar to those with tomy to prevent development of medullary thyroid carci-
HD. Constipation can alternate with episodes of diarrhea noma [73, 80].
[75, 76]. The similarity of the gastrointestinal symptoms
between patients with IND and MEN 2B-associated Outcome of intestinal GNM
intestinal GNM has suggested that these two conditions
could be the result of mutations affecting the same domain Early diagnosis and treatment of patients with MEN 2B-
of the RET proto-oncogene [23]. Gastrointestinal dysmo- associated intestinal GNM are essential for long-term sur-
tility is a common initial presentation of patients with MEN vival. A yearly follow-up with monitoring of basal plasma
2B, but the rarity of this syndrome often delays the diag- calcitonin and CEA levels for possible tumor recurrence is
nosis. Further findings are mucosal neuromas of the lips recommended [80]. In addition, continued surveillance of
and tongue, as well as medullated corneal nerve fibers, the adrenal glands with abdominal ultrasound scanning
distinctive facies with enlarged lips and an asthenic and urine analysis of catecholamine metabolites includ-
‘‘marfanoid’’ body habitus [71]. ing metanephrine, normetanephrine, dopamine and

123
860 Pediatr Surg Int (2013) 29:855–872

vanillylmandelic acid is required as patients with MEN 2B patients with isolated HG compared to patients with HD,
have at least a 50 % risk of developing pheochromocytoma which is predominantly diagnosed during the newborn
[79]. period. Enterocolitis of the newborn has been reported to
be the most serious complication of isolated HG [96].

Isolated hypoganglionosis Diagnosis of isolated HG

Isolated hypoganglionosis (HG) is a rare entity and has been It is unclear whether isolated HG represents a severe form
classified as a ‘‘hypogenetic type’’ of intestinal innervation of intestinal dysganglionosis or solely an abnormality of
disorders. The clinical presentation of patients with isolated the enteric nervous system that leads to severe constipation
HG is similar to those with classical HD with non-specific [11]. Hence, the diagnosis can be difficult and a consensus
symptoms of severe constipation or bowel obstruction [1, in diagnostic criteria still remains to be found. A full-
89]. It has been demonstrated that congenital and acquired thickness bowel specimen is required for the definitive
HG are two distinct entities with different clinical charac- diagnosis of isolated HG [97–100]. The vast majority of
teristics and histological findings [90]. There is only a reported cases have been evaluated by immunohisto-
limited number of cases in the published literature as iso- chemical staining showing sparse and small myenteric
lated HG is one of the rarest subtypes of intestinal inner- ganglia, absent or low AChE activity in the lamina propria
vation disorders and there remains a controversy regarding as well as hypertrophy of muscularis mucosae and circular
it as a distinct isolated histopathological entity [11]. muscle [101]. Meier-Ruge et al. [102] found significant
histopathological differences between resected bowel
Pathogenesis of isolated HG specimens from patients with isolated HG and normal
bowel tissue using AChE staining. They showed a 40 %
The pathogenesis and genetic basis of isolated HG both reduction in the number of nerve cells, accompanied by a
remain unclear. Several authors have performed mutational doubled distance between ganglia and a three times smaller
analysis of the RET gene, which has been reported to be plexus area in the hypoganglionic bowel segment. These
associated with HD, but neither causative missense muta- observations currently form the basis for the histopathol-
tion nor neutral substitutions were found [91, 92]. Rolle ogical diagnosis of isolated HG. It has also been suggested
et al. [93] have previously shown that some cases of iso- that the size of the myenteric plexus may be an indicator of
lated HG exhibit deficient expression of c-Kit positive clinical severity [103]. Thus, several neuronal markers
ICCs within the myenteric plexus and the smooth muscle have been introduced to facilitate the diagnosis of isolated
layer, which may contribute to the motility dysfunction in HG. To determine the muscular nitrergic innervation and
the hypoganglionic bowel segment [57]. A lack or reduced differentiation of mature from immature ganglia in patients
expression of NCAM-positive nerve fibers within the with isolated HG, NADPH-d staining has been employed
lamina propria, muscularis mucosae as well as circular and to demonstrate a reduced number of positive nerve fibers in
longitudinal muscle layers of patients with isolated HG has the muscularis mucosae with absent or sparse submucosal
also been described [89, 94, 95]. and myenteric ganglion cells [104] (Fig. 3a–d). Further-
more, c-Kit staining has been used to investigate the
Epidemiology of isolated HG expression of ICCs and thus intestinal pacemaker activity,
which is markedly decreased or even absent in patients
Reports of finding isolated HG in rectal biopsies are rare and with isolated HG [56, 93, 105].
vary between 0.3 and 6.4 % [24, 26, 30]. Since 1978, there
have been a total number of 92 cases published in the English Management of isolated HG
literature [96]. Thirty-two percent of them were diagnosed in
the newborn period. However, the median age at diagnosis was The management of isolated HG is similar to that of HD.
4.8 years, which was due to the fact that in some cases the According to the literature, a majority of patients undergo
diagnosis was not made until the patients were adolescents. resection of the affected bowel segment with subsequent
pull-through procedure [96, 106].
Clinical presentation of isolated HG
Outcome of isolated HG
The most common presenting symptoms of isolated HG are
severe chronic constipation with intestinal obstruction and The postoperative outcome after resection of the hypo-
enterocolitis, thus resembling classical symptoms of HD. ganglionic segment usually is good [107]. Typical com-
The median age at diagnosis is considerably higher in plications of isolated HG are enterocolitis, chronic

123
Pediatr Surg Int (2013) 29:855–872 861

Fig. 3 NADPH-d staining in whole mount preparation of a normal myenteric plexus (a, c). Myenteric plexus of a patient with isolated HG
showing markedly reduced number of ganglion cells (b, d)

constipation, overflow encopresis and the need for redo Pathogenesis of IG


pull-through operation due to residual hypoganglionosis
[34]. An overall mortality rate of 8 % has recently been A combination of large (fully mature) and small (imma-
reported with the majority of patients who died being ture) ganglion cells can be found at birth [108]. In the early
newborns suffering from severe enterocolitis [34]. postnatal period, ganglion cells in the submucosal plexus
are generally less developed than the ones in the myenteric
plexus [109]. It has further been demonstrated that this
Immature ganglia immaturity is a physiological, age-dependent phenomenon
and maturation of IG strongly correlates with the age of the
Immature ganglia (IG) are usually found in biopsies from patient [3, 109]. Strong evidence supporting this theory has
premature infants presenting with functional bowel arisen from several animal studies showing postnatal
obstruction. It has been reported that delayed maturation of maturation of the submucosal and myenteric plexuses
ganglion cells in the submucosal and myenteric plexuses is [110–112]. Therefore, the finding of immature ganglion
the most common cause of chronic constipation during the cells may be an indicator of transient functional immaturity
first year of life [85]. of the bowel [108].

123
862 Pediatr Surg Int (2013) 29:855–872

Epidemiology of IG rare cause of constipation and functional bowel obstruction


in infants and children.
There is not much epidemiological data on the incidence of
IG. Ure et al. [30] found four (2.8 %) cases of immature Pathogenesis of absence of the AP
ganglion cells in their cohort of 141 patients with intestinal
neuronal malformations. More recently, Puri et al. [34] In the normal myenteric plexus there are two distinct sub-
reported ten (5.6 %) cases of IG in bowel specimens of 178 types of nerve cells, which can be distinguished by their
patients with variants of HD. affinity for silver stains: (1) argyrophil cells and (2) argen-
taffin cells. Argyrophil cells usually comprise 5–20 % of the
Clinical presentation of IG total number of myenteric neurons [116]. The processes of
these cells along with extrinsic and parasympathetic fibers
Patients with IG usually present with a history of chronic form a complex neuronal network within the myenteric
constipation or functional bowel obstruction resembling plexus, which controls gastrointestinal peristalsis and transit
HD. Further clinical features may include slow-transit time. Argyrophil cells coordinate the activation of argen-
peristalsis and insufficient defecation [113]. taffin cells, which secrete specific neurotransmitters and
ultimately cause contraction or relaxation of muscle fibers
Diagnosis of IG within the bowel wall [116]. A distinct time-lag has been
demonstrated between the appearance of both cell types with
In general, the diagnosis of IG can be made from rectal argyrophil cells appearing earlier than argentaffin cells [117].
suction biopsies. The ganglion cells appear very small It has been shown that there is a caudocranial gradient in the
and have a less significant nucleus with an inconspicuous differentiation of these neuron cells in the human bowel [117,
nucleolus [109, 114] (Fig. 4a, b). However, it is often 118]. Therefore, it is suspected that the disruption of this
not possible with AChE histochemistry to distinguish differentiation process may lead to an absence of the AP.
between these small ganglion cells and the supporting
enteric glial cells. Thus, NADPH-d and NCAM staining Epidemiology of absence of the AP
have been suggested as neuronal markers to show the
small ganglion cells more clearly [1, 101]. Enzyme There is a paucity of published data on the incidence of this
histochemistry for succinate and lactate dehydrogenase is very rare condition. In 1997, Puri et al. [1] reported three
also commonly used to determine IG showing an absent cases with absence of the AP in their series of patients with
or weak positive reaction [3, 46]. In addition, cathepsin functional bowel disorders. Familial cases in the offspring
D has been recommended to evaluate the maturation of of consanguineous parents and recurrence in siblings sug-
immature ganglion cells in more detail [50]. Another gests that the absence of the AP may be inherited in an
helpful biomarker to detect immature enteric ganglion autosomal-recessive manner [116, 119].
cells is Bcl2 [115], which clearly discriminates these
immature small neurons. Clinical presentation of absence of the AP

Management of IG The clinical symptoms of patients with absence of the AP are


highly similar to HD presenting with severe constipation,
The management of patients with IG is conservative with moderate abdominal distension and lack of peristalsis [120].
use of laxatives and enemas [1, 2].
Diagnosis of absence of the AP
Outcome of IG
The absence of argyrophil cells and their neuronal pro-
The majority of patients with IG can successfully be cesses can only be demonstrated using silver impregnation
managed with conservative treatment until their ganglion [121] (Fig. 5a, b) of full-thickness biopsies, while con-
cells are fully mature. It is recommended to repeat the ventional H&E staining, AChE histochemistry and immu-
biopsy 12–18 months after initial investigation [3]. nohistochemistry with several neuronal markers fail to
show this abnormality [1, 101].

Absence of the argyrophil plexus Management of absence of the AP

The lack of argyrophil cells in the myenteric plexus, which The majority of patients with absence of the AP can be
is also known as absence of the argyrophil plexus (AP), is a managed conservatively with laxatives and enemas, but in

123
Pediatr Surg Int (2013) 29:855–872 863

Fig. 4 H&E (a) and AChE (b) staining in a patient with IG showing immature ganglion cells

Fig. 5 Silver staining showing normal AP (a) and absence of argyrophil cells (b) in a patient with absence of the AP

some cases internal sphincter myectomy or formation of a Internal anal sphincter achalasia
colostomy may be necessary due to persistent constipation
[1, 120]. Internal anal sphincter achalasia (IASA) is a condition with
clinical presentation similar to HD [122–124], but in con-
Outcome of absence of the AP trast to HD, ganglion cells are present in rectal suction
biopsies. Previously, IASA was referred to as ultrashort-
Conservative and surgical treatment of patients with segment HD, which is characterized by an aganglionic
absence of the AP usually results in a satisfactory outcome segment of 1–3 cm above the pectinate line, normal AChE
[1, 120]. activity in the lamina propria and increased AChE activity

123
864 Pediatr Surg Int (2013) 29:855–872

Fig. 6 NADPH-d staining of normal IAS (a). Reduced NADPH-d positive innervations (b) in a patient with IASA

in the muscularis mucosae [125]. Several investigators Epidemiology of IASA


have suggested that IASA is more accurate for this path-
ological entity as many patients with absence of the rec- Since 1973, a total number of 395 cases with IASA have
tosphincteric reflex on anorectal manometry showed been reported in the literature [134]. However, the exact
presence of ganglion cells combined with normal AChE incidence of IASA is unknown.
activity in rectal biopsies [126–128].
Clinical presentation of IASA
Pathogenesis of IASA
The clinical presentation of IASA is in most cases similar
The exact pathogenesis of IASA remains unclear despite to that of HD. Patients with IASA usually present with
the attempts of several investigators to determine the severe constipation with or without soiling. About one-
pathophysiological mechanisms in more detail. It has third of these patients have a history of abdominal disten-
been suggested that age-related changes in the develop- sion and failure of laxative therapy [125, 126].
ing intramuscular innervation of the internal anal
sphincter (IAS) may form the basis for the observed Diagnosis of IASA
motility dysfunction [129]. By analyzing the NADPH-d
activity (Fig. 6a, b), Hirakawa et al. [130] reported The diagnosis of IASA is based on clinical symptoms
absent to marked reduction of nitrergic innervation combined with finding of absence of the rectosphincteric
within the IAS of patients with IASA as the underlying reflex on rectal balloon inflation with marked increased
pathomechanism leading to spasm or increased tone. A rhythmic activity on anorectal manometry (Fig. 7a, b),
defective innervation of the neuromuscular junction of presence of ganglion cells and normal AChE activity in
the IAS with decreased expression of PGP9.5 and syn- rectal suction biopsies as well as reduction of nitrergic
apsin-1 has also been detected [131]. Kobayashi et al. innervation within the IAS.
[94] further demonstrated absent to markedly reduced
NADPH-d and NCAM activity in the IAS of patients Management of IASA
with IASA. In addition, a reduced number of c-Kit
positive ICCs have been found in the IAS of patients Posterior IAS myectomy has been recommended for the
with IASA [132]. The deficiency in nitrergic innervation treatment of IASA [135–137]. More recently, intrasphinc-
and ICCs may explain the impaired anal relaxation in teric injection of botulinum toxin has been introduced as a
patients with IASA [133]. therapeutic alternative [138–143].

123
Pediatr Surg Int (2013) 29:855–872 865

Fig. 7 Anorectal manometry


showing evidence of the
rectosphincteric reflex in a
normal IAS (a). Absence of the
rectosphincteric reflex on rectal
balloon inflation with marked
increased rhythmic activity of
the IAS (b) in a patient with
IASA

Fig. 8 Voiding
cystourethrogram (a) and
barium contrast enema
(b) showing massively enlarged
bladder and microcolon in a
patient with MMIHS

Outcome of IASA indicated that posterior IAS myectomy appears to be a


more effective treatment option resulting in a better func-
The vast majority of patients with IASA have regular tional outcome compared to intrasphincteric botulinum
bowel movements after treatment irrespective of the ther- toxin injection [134]. The rate of transient fecal inconti-
apeutic approach [34, 135]. However, it has recently been nence, non-response and subsequent surgical procedures

123
866 Pediatr Surg Int (2013) 29:855–872

Fig. 9 Electron microscopy of smooth muscle cells in normal bowel (a). Central core degenerations resulting in empty vacuoles (asterisks) and
increased connective tissue (triangles) in smooth muscle cells (b) of a patient with MMIHS

were significantly higher after injection of botulinum toxin, nicotinic acetylcholine receptor (nAChR) in transgenic
whereas long-term improvements were significantly more mice resulted in a phenotype similar to that of human
frequent following IAS myectomy. Interestingly, no dif- MMIHS [153]. Furthermore, it has been shown that a lack
ferences were found in the postoperative use of laxatives or of expression of the a3, b2 and b4 subunits of nAChR in
enemas, postoperative soiling as well as constipation small bowel tissue from patients with MMIHS contributes
between both procedures. to the pathogenesis of this rare condition [154, 155].
CHRNA3 and CHRNB4 genes, which are both coding for
the b4 subunit of nAChR, are additional strong candidates
Megacystis microcolon intestinal hypoperistalsis [156]. Much published data also points to a dysfunction
syndrome within the smooth muscle layer. Puri et al. [157] reported
vacuolar changes in the smooth muscle cells within the
Megacystis microcolon intestinal hypoperistalsis syndrome bowel and bladder wall of patients with MMIHS (Fig. 9a,
(MMIHS) is the most severe form of functional bowel b). Therefore, it has been suggested that the observed
obstruction in the newborn [144]. This rare condition is smooth muscle myopathy may be the underlying cause of
characterized by massive abdominal distension caused by a MMIHS [148, 158]. The expression of several contractile
large-dilated non-obstructed bladder, microcolon with and cytoskeleton proteins such as aSMA, calponin, cal-
malrotation and decreased or absent intestinal peristalsis desmin and desmin has also been found to be absent or
[34] (Fig. 8a, b). decreased in the colonic smooth muscle tissue of patients
with MMIHS [147, 159, 160]. Further support of this
Pathogenesis of MMIHS theory is derived from the findings of abnormal synapto-
physin distribution in the circular muscle layer of bowel
Megacystis microcolon intestinal hypoperistalsis syndrome and bladder specimens, as well as increased connective
was first described in 1976 by Berdon et al. [145], who tissue and atrophic smooth muscle fibers [161]. Moreover,
observed above characteristic features in five newborn a decreased expression of ICCs in the bladder has been
girls. Since then, various hypotheses have been proposed to observed [56, 159, 162]. In addition to the smooth muscle
explain the pathogenesis of MMIHS. Genetic [146], myo- dysfunction, other studies have focused on neuronal dif-
genic [147–149], neurogenic [150, 151] and hormonal ferences in MMIHS [163]. Previously, immaturity and
[151, 152] etiologies have been discussed. However, most malfunction of autonomic nerve endings in the whole
of these theories are derived from case reports due to the gastrointestinal tract have been demonstrated [150]. Axo-
rarity of this condition. Thus, the exact etiology still nal dystrophy and additional defects of the autonomic
remains unclear. At present, several candidate genes for innervation in the intestine of patients with MMIHS have
MMIHS have been identified. Gene knockout of the also been reported [151, 164]. Furthermore, an intramural

123
Pediatr Surg Int (2013) 29:855–872 867

inflammatory process that affects the gastrointestinal and bowel specimens from patients with MMIHS. The
urinary tract has been proposed in the pathogenesis of remaining 23 % were shown to have various neuronal
MMIHS [152]. abnormalities including hyper-/hypoganglionosis and
immature ganglia [34]. In addition, some authors found
Epidemiology of MMIHS significant anomalies in smooth muscle cells from bowel
and bladder specimens, such as vacuolar degeneration as
Since 1976, there have been a total number of 227 patients well as thinning of the longitudinal muscle [149, 157].
with the diagnosis of MMIHS reported in the published
literature [165]. Among them, a male-to-female ratio of Management of MMIHS
1:2.4 has been observed, suggesting a distinct female pre-
dominance. In addition, it has been reported that male The management of patients with MMIHS is frustrating. A
patients with MMIHS seem to have a shorter life expec- number of prokinetic drugs and gastrointestinal hormones
tancy than affected females. This is most likely due to a have been tried without any success [174]. Surgical treat-
more severe form of this condition in males compared to ment usually has to be performed for malrotation, bowel
females [166, 167]. Furthermore, occurrence of familial obstruction and megacystis to achieve decompression of
cases in the offspring of consanguineous parents and bowel and bladder. However, in most cases, these inter-
recurrence in siblings indicates that MMIHS may be ventions do not result in any improvement in enteral food
inherited in an autosomal-recessive manner [168–170]. intake, functional bowel obstruction or bladder function.
Consequently, the majority of patients with MMIHS are
Clinical presentation of MMIHS maintained on total parenteral nutrition (TPN), which leads
to further comorbidities, such as catheter sepsis, dyslipi-
The enlarged bladder with hydronephrosis, microcolon and demia, TPN-associated liver disease and eventually chronic
intestinal malrotation can often be demonstrated on pre- liver failure [165]. In patients with irreversible intestinal
natal ultrasound or MRI scans [146]. The clinical presen- pathology and failure of TPN, intestinal and multivisceral
tation of MMIHS is similar to that of other severe neonatal transplantation has recently been introduced as a valuable
intestinal obstruction, typically presenting with massive therapeutic alternative. At present, 12 multivisceral trans-
abdominal distension, which is a consequence of the large- plantations have been reported in MMIHS patients [34].
dilated non-obstructed bladder with or without upper uri-
nary tract dilatation. The majority of patients with MMIHS Outcome of MMIHS
are unable to void spontaneously and require either vesi-
costomy or catheterization. It has been shown that the In recent years, the survival rate of MMIHS has consid-
detrusor muscle in these patients is strikingly abnormal, erably improved from 12.6 % initially to 55.6 %. Between
which is most likely the cause for the voiding dysfunction 1977 and 2011, an overall survival rate of 19.7 % has been
[171]. Additional common findings are bile-stained vom- reported with the oldest survivor being 24 years old. The
iting, absent or decreased bowel sounds and failure to pass most frequent cause of death in MMIHS patients has been
meconium [165]. shown to be overwhelming sepsis, followed by multiple
organ failure and malnutrition [165]. A 3-year survival rate
Diagnosis of MMIHS of 50 % with all survivors tolerating enteral feeding and
showing adequate gastric emptying has recently been
There should be an emphasis on prenatal diagnosis of reported [175]. These improvements in outcome have
MMIHS to allow for adequate prenatal counseling to mainly been attributed to more specialized care, innova-
enable future parents to make an informed decision tions in parenteral nutrition and introduction of multivis-
regarding the continuation of the pregnancy, given the poor ceral transplantation.
prognosis of this severe condition. The diagnosis of
MMIHS is usually made on the basis of prenatal ultrasound
findings and characteristic clinical presentation in the References
immediate neonatal period. An enlarged bladder and
hydronephrosis can already be found on fetal ultrasound 1. Puri P (1997) Variant Hirschsprung’s disease. J Pediatr Surg
studies [172]. Analysis of enzymatic changes in amniotic 32(2):149–157
fluid in combination with MRI scans can further contribute 2. Puri P, Rolle U (2004) Variant Hirschsprung’s disease. Semin
Pediatr Surg 13(4):293–299
to the prenatal diagnosis of MMIHS [173]. Histological 3. Holschneider AM, Meier-Ruge W, Ure BM (1994) Hirsch-
evaluation of myenteric and submucosal plexuses has sprung’s disease and allied disorders: a review. Eur J Pediatr
revealed normal ganglion cells in 77 % of the investigated Surg 4(5):260–266

123
868 Pediatr Surg Int (2013) 29:855–872

4. Schärli AF (1995) Standardization of terminology of dysgan- 26. Montedonico S, Caceres P, Munoz N et al (2011) Histochem-
glionoses. Pediatr Surg Int 10(7):440 ical staining for intestinal dysganglionosis: over 30 years
5. Meier-Ruge W (1971) Casuistic of colon disorder with symptoms experience with more than 1,500 biopsies. Pediatr Surg Int
of Hirschsprung’s disease. Verh Dtsch Ges Pathol 55:506–510 27(5):479–486
6. Puri P, Lake BD, Nixon HH et al (1977) Neuronal colonic 27. Schmittenbecher PP, Sacher P, Cholewa D et al (1999) Hir-
dysplasia: an unusual association of Hirschsprung’s disease. schsprung’s disease and intestinal neuronal dysplasia: a frequent
J Pediatr Surg 12(5):681–685 association with implications for the postoperative course. Pe-
7. Fadda B, Maier WA, Meier-Ruge W et al (1983) Neuronal diatr Surg Int 15(8):553–558
intestinal dysplasia. Critical 10-years’ analysis of clinical and 28. Hanimann B, Inderbitzin D, Briner J et al (1992) Clinical rele-
biopsy diagnosis. Z Kinderchir 38(5):305–311 vance of Hirschsprung-associated neuronal intestinal dysplasia
8. Lake BD (1995) Intestinal neuronal dysplasia. Why does it only (HANID). Eur J Pediatr Surg 2(3):147–149
occur in parts of Europe? Virchows Arch 426(6):537–539 29. Schärli AF (1992) Neuronal intestinal dysplasia. Pediatr Surg Int
9. Lumb PD, Moore L (1998) Back to the drawing board. Intestinal 7(1):2–7
neuronal dysplasia type B: not a histological entity yet. Virch- 30. Ure BM, Holschneider AM, Schulten D et al (1997) Clinical
ows Arch 432(2):99–102 impact of intestinal neuronal malformations: a prospective study
10. Sacher P, Briner J, Hanimann B (1993) Is neuronal intestinal in 141 patients. Pediatr Surg Int 12(5–6):377–382
dysplasia (NID) a primary disease or a secondary phenomenon? 31. Csury L, Peña A (1995) Intestinal neuronal dysplasia: myth or
Eur J Pediatr Surg 3(4):228–230 reality? Literature review. Pediatr Surg Int 10(7):441–446
11. Martucciello G, Pini Prato A, Puri P et al (2005) Controversies 32. Schärli AF, Meier-Ruge W (1981) Localized and disseminated
concerning diagnostic guidelines for anomalies of the enteric forms of neuronal intestinal dysplasia mimicking Hirsch-
nervous system: a report from the fourth International Sympo- sprung’s disease. J Pediatr Surg 16(2):164–170
sium on Hirschsprung’s disease and related neurocristopathies. 33. Montedonico S, Acevedo S, Fadda B (2002) Clinical aspects of
J Pediatr Surg 40(10):1527–1531 intestinal neuronal dysplasia. J Pediatr Surg 37(12):1772–1774
12. Milla PJ, Smith VV (1993) Intestinal neuronal dysplasia. J Pe- 34. Puri P, Gosemann JH (2012) Variants of Hirschsprung disease.
diatr Gastroenterol Nutr 17(4):356–357 Semin Pediatr Surg 21(4):310–318
13. Hirobe S, Doody DP, Ryan DP et al (1992) Ectopic class II 35. Martucciello G, Torre M, Pini Prato A et al (2002) Associated
major histocompatibility antigens in Hirschsprung’s disease and anomalies in intestinal neuronal dysplasia. J Pediatr Surg
neuronal intestinal dysplasia. J Pediatr Surg 27(3):357–362 37(2):219–223
14. Kobayashi H, Mahomed A, Puri P (1996) Intestinal neuronal 36. Pini Prato A, Rossi V, Fiore M et al (2011) Megacystis, meg-
dysplasia in twins. J Pediatr Gastroenterol Nutr 22(4):398–401 acolon, and malrotation: a new syndromic association? Am J
15. Moore SW, Kaschula ROC, Cywes S (1993) Familial and Med Genet A 155A(8):1798–1802
genetic-aspects of neuronal intestinal dysplasia and Hirsch- 37. Koletzko S, Jesch I, Faus-Kebetaler T et al (1999) Rectal biopsy
sprungs disease. Pediatr Surg Int 8(5):406–409 for diagnosis of intestinal neuronal dysplasia in children: a
16. Hatano M, Aoki T, Dezawa M et al (1997) A novel pathogenesis prospective multicentre study on interobserver variation and
of megacolon in Ncx/Hox11L.1 deficient mice. J Clin Invest clinical outcome. Gut 44(6):853–861
100(4):795–801 38. Lumb PD, Moore L (1998) Are giant ganglia a reliable marker
17. Shirasawa S, Yunker AM, Roth KA et al (1997) Enx of intestinal neuronal dysplasia type B (IND B)? Virchows Arch
(Hox11L1)-deficient mice develop myenteric neuronal hyper- 432(2):103–106
plasia and megacolon. Nat Med 3(6):646–650 39. Kapur RP (2003) Neuronal dysplasia: a controversial patho-
18. von Boyen GB, Krammer HJ, Suss A et al (2002) Abnormalities logical correlate of intestinal pseudo-obstruction. Am J Med
of the enteric nervous system in heterozygous endothelin B Genet A 122A(4):287–293
receptor deficient (spotting lethal) rats resembling intestinal 40. Meier-Ruge WA, Brönnimann PB, Gambazzi F et al (1995)
neuronal dysplasia. Gut 51(3):414–419 Histopathological criteria for intestinal neuronal dysplasia of the
19. Gath R, Goessling A, Keller KM et al (2001) Analysis of the submucosal plexus (type B). Virchows Arch 426(6):549–556
RET, GDNF, EDN3, and EDNRB genes in patients with intes- 41. Kobayashi H, Hirakawa H, Puri P (1995) What are the diag-
tinal neuronal dysplasia and Hirschsprung disease. Gut 48(5): nostic criteria for intestinal neuronal dysplasia? Pediatr Surg Int
671–675 10(7):459–464
20. Costa M, Fava M, Seri M et al (2000) Evaluation of the 42. Coerdt W, Michel JS, Rippin G et al (2004) Quantitative mor-
HOX11L1 gene as a candidate for congenital disorders of phometric analysis of the submucous plexus in age-related
intestinal innervation. J Med Genet 37(7):E9 control groups. Virchows Arch 444(3):239–246
21. Fava M, Borghini S, Cinti R et al (2002) HOX11L1: a promoter 43. Smith VV (1993) Intestinal neuronal density in childhood: a
study to evaluate possible expression defects in intestinal baseline for the objective assessment of hypo- and hypergan-
motility disorders. Int J Mol Med 10(1):101–106 glionosis. Pediatr Pathol 13(2):225–237
22. Granero Cendón R, Millan Lopez A, Moya Jimenez MJ et al 44. Cord-Udy CL, Smith VV, Ahmed S et al (1997) An evaluation
(2007) Intestinal neuronal dysplasia: association with digestive of the role of suction rectal biopsy in the diagnosis of intestinal
malformations. Cir Pediatr 20(3):166–168 neuronal dysplasia. J Pediatr Gastroenterol Nutr 24(1):1–6
23. Martucciello G, Caffarena PE, Lerone M et al (1994) Neuronal 45. Meyrat BJ, Laurini RN (2003) Plasticity of the enteric nervous
intestinal dysplasia: clinical experience in Italian patients. Eur J system in patients with intestinal neuronal dysplasia associated
Pediatr Surg 4(5):287–292 with Hirschsprung’s disease: a report of three patients. Pediatr
24. Meier-Ruge W (1992) Epidemiology of congenital innervation Surg Int 19(11):715–720
defects of the distal colon. Virchows Arch A Pathol Anat His- 46. Meier-Ruge WA, Bruder E (2005) Pathology of chronic con-
topathol 420(2):171–177 stipation in pediatric and adult coloproctology. Pathobiology
25. Kobayashi H, Hirakawa H, Surana R et al (1995) Intestinal 72(1–2):1–102
neuronal dysplasia is a possible cause of persistent bowel 47. Meier-Ruge WA, Schmid PC, Stoss F (1995) Intestinal neuronal
symptoms after pull-through operation for Hirschsprung’s dis- dysplasia and its morphometric evidences. Pediatr Surg Int
ease. J Pediatr Surg 30(2):253–257 10(7):447–453

123
Pediatr Surg Int (2013) 29:855–872 869

48. Meier-Ruge WA, Longo-Bauer CH (1997) Morphometric clinicopathological and immunohistochemical study of 6 cases.
determination of the methodological criteria for the diagnosis of Hum Pathol 22(3):276–286
intestinal neuronal dysplasia (IND B). Pathol Res Pract 71. Moline J, Eng C (2011) Multiple endocrine neoplasia type 2: an
193(7):465–469 overview. Genet Med 13(9):755–764
49. Krammer HJ, Meier-Ruge W, Sigge W et al (1994) Histopa- 72. Puig I, Champeval D, De Santa Barbara P et al (2009) Deletion
thological features of neuronal intestinal dysplasia of the plexus of Pten in the mouse enteric nervous system induces ganglio-
submucosus in whole mounts revealed by immunohistochemis- neuromatosis and mimics intestinal pseudoobstruction. J Clin
try for PGP 9.5. Eur J Pediatr Surg 4(6):358–361 Invest 119(12):3586–3596
50. Tatekawa Y, Kanehiro H, Kanokogi H et al (2000) The evalu- 73. O’Riordain DS, Obrien T, Crotty TB et al (1995) Multiple
ation of meconium disease by distribution of cathepsin D in endocrine neoplasia type 2B: more than an endocrine disorder.
intestinal ganglion cells. Pediatr Surg Int 16(1–2):53–55 Surgery 118(6):936–942
51. Kobayashi H, Miyano T, Yamataka A et al (1997) Use of syn- 74. Dunzendorfer T, Lee VW, Levine S et al (1996) Refractory
aptophysin polyclonal antibody for the rapid intraoperative constipation and megacolon in MEN 2b. Dig Dis Sci 41(11):
immunohistochemical evaluation of functional bowel disorders. 2233–2237
J Pediatr Surg 32(1):38–40 75. Carney JA, Go VL, Sizemore GW et al (1976) Alimentary-tract
52. Wester T, O’Briain S, Puri P (1998) Morphometric aspects of ganglioneuromatosis: a major component of the syndrome of
the submucous plexus in whole-mount preparations of normal multiple endocrine neoplasia type 2b. N Engl J Med 295(23):
human distal colon. J Pediatr Surg 33(4):619–622 1287–1291
53. Wester T, O’Briain DS, Puri P (1999) Notable postnatal alter- 76. de Krijger RR, Brooks A, van der Harst E et al (1998) Consti-
ations in the myenteric plexus of normal human bowel. Gut pation as the presenting symptom in de novo multiple endocrine
44(5):666–674 neoplasia type 2B. Pediatrics 102(2 Pt 1):405–408
54. Kobayashi H, Hirakawa H, Puri P (1996) Is intestinal neuronal 77. Plenat F, Vignaud JM, Floquet J et al (1984) Intestinal gan-
dysplasia a disorder of the neuromuscular junction? J Pediatr glioneuromatosis: histochemical, histoenzymological and ultra-
Surg 31(4):575–579 structural study of a case. Ann Pathol 4(2):131–136
55. Rolle U, Piotrowska AP, Puri P (2003) Abnormal vasculature in 78. de Schryver-Kecskemeti K, Clouse RE, Goldstein MN et al
intestinal neuronal dysplasia. Pediatr Surg Int 19(5):345–348 (1983) Intestinal ganglioneuromatosis: a manifestation of over-
56. Rolle U, Piaseczna-Piotrowska A, Puri P (2007) Interstitial cells production of nerve growth factor? N Engl J Med 308(11):
of Cajal in the normal gut and in intestinal motility disorders of 635–639
childhood. Pediatr Surg Int 23(12):1139–1152 79. Smith VV, Eng C, Milla PJ (1999) Intestinal ganglioneuroma-
57. Yamataka A, Ohshiro K, Kobayashi H et al (1997) Intestinal tosis and multiple endocrine neoplasia type 2B: implications for
pacemaker C-KIT ? cells and synapses in allied Hirschsprung’s treatment. Gut 45(1):143–146
disorders. J Pediatr Surg 32(7):1069–1074 80. Torre M, Martucciello G, Ceccherini I et al (2002) Diagnostic
58. O’Donnell AM, Puri P (2011) A role for Pten in paediatric and therapeutic approach to multiple endocrine neoplasia type
intestinal dysmotility disorders. Pediatr Surg Int 27(5):491–493 2B in pediatric patients. Pediatr Surg Int 18(5–6):378–383
59. Bruder E, Meier-Ruge WA (2007) Intestinal neuronal dysplasia 81. Shekitka KM, Sobin LH (1994) Ganglioneuromas of the gastro-
type B: how do we understand it today? Pathologe 28(2): intestinal tract: relation to Von Recklinghausen disease and other
137–142 multiple tumor syndromes. Am J Surg Pathol 18(3):250–257
60. Meier-Ruge WA, Ammann K, Bruder E et al (2004) Updated 82. Yin M, King SK, Hutson JM et al (2006) Multiple endocrine
results on intestinal neuronal dysplasia (IND B). Eur J Pediatr neoplasia type 2B diagnosed on suction rectal biopsy in infancy:
Surg 14(6):384–391 a report of 2 cases. Pediatr Dev Pathol 9(1):56–60
61. Meier-Ruge WA, Bruder E, Kapur RP (2006) Intestinal neuronal 83. Sonsino E, Nabarra B, Andrianarison I et al (1979) Histological
dysplasia type B: one giant ganglion is not good enough. Pediatr and ultrastructural study of intestinal ganglioneuromatosis: the
Dev Pathol 9(6):444–452 multiple endocrine neoplasms type IIb syndrome. Ann Anat
62. Ure BM, Holschneider AM, Meier-Ruge W (1994) Neuronal Pathol (Paris) 24(1):29–44
intestinal malformations: a retro- and prospective study on 203 84. Demos TC, Blonder J, Schey WL et al (1983) Multiple endo-
patients. Eur J Pediatr Surg 4(5):279–286 crine neoplasia (MEN) syndrome type IIB: gastrointestinal
63. Puri P, Wester T (1998) Intestinal neuronal dysplasia. Semin manifestations. AJR Am J Roentgenol 140(1):73–78
Pediatr Surg 7(3):181–186 85. Feichter S, Meier-Ruge WA, Bruder E (2009) The histopathol-
64. Stoss F, Meier-Ruge W (1994) Experience with neuronal ogy of gastrointestinal motility disorders in children. Semin
intestinal dysplasia (NID) in adults. Eur J Pediatr Surg 4(5): Pediatr Surg 18(4):206–211
298–302 86. Unruh A, Fitze G, Janig U et al (2007) Medullary thyroid car-
65. Puri P (2003) Intestinal neuronal dysplasia. Semin Pediatr Surg cinoma in a 2-month-old male with multiple endocrine neoplasia
12(4):259–264 2B and symptoms of pseudo-Hirschsprung disease: a case
66. Schmittenbecher PP, Gluck M, Wiebecke B et al (2000) Clinical report. J Pediatr Surg 42(9):1623–1626
long-term follow-up results in intestinal neuronal dysplasia 87. Cohen MS, Phay JE, Albinson C et al (2002) Gastrointestinal
(IND). Eur J Pediatr Surg 10(1):17–22 manifestations of multiple endocrine neoplasia type 2. Ann Surg
67. Gillick J, Tazawa H, Puri P (2001) Intestinal neuronal dysplasia: 235 (5):648–654; discussion 654–645
results of treatment in 33 patients. J Pediatr Surg 36(5):777–779 88. Brauckhoff M, Gimm O, Weiss CL et al (2004) Multiple
68. Charagundla SR, Levine MS, Torigian DA et al (2004) Diffuse endocrine neoplasia 2B syndrome due to codon 918 mutation:
intestinal ganglioneuromatosis mimicking Crohn’s disease. AJR clinical manifestation and course in early and late onset disease.
Am J Roentgenol 182(5):1166–1168 World J Surg 28(12):1305–1311
69. Matthews MA, Adler BH, Arnold MA et al (2013) Diffuse 89. Kobayashi H, Yamataka A, Lane GJ et al (2002) Pathophysiology
intestinal ganglioneuromatosis in a child. J Pediatr Surg 48(5): of hypoganglionosis. J Pediatr Gastroenterol Nutr 34(2):231–235
1129–1133 90. Taguchi T, Masumoto K, Ieiri S et al (2006) New classification
70. D’Amore ESG, Manivel JC, Pettinato G et al (1991) Intestinal of hypoganglionosis: congenital and acquired hypoganglionosis.
ganglioneuromatosis - mucosal and transmural types: a J Pediatr Surg 41(12):2046–2051

123
870 Pediatr Surg Int (2013) 29:855–872

91. Do MY, Myung SJ, Park HJ et al (2011) Novel classification and 111. Sri Paran T, Rolle U, Puri P (2006) Postnatal development of the
pathogenetic analysis of hypoganglionosis and adult-onset Hir- mucosal plexus in the porcine small and large intestine. Pediatr
schsprung’s disease. Dig Dis Sci 56(6):1818–1827 Surg Int 22(12):997–1001
92. Inoue K, Shimotake T, Tomiyama H et al (2001) Mutational 112. Sri Paran T, Rolle U, Puri P (2008) Postnatal changes in enteric
analysis of the RET and GDNF gene in children with hypo- plexus axonal thickness. Pediatr Surg Int 24(12):1365–1367
ganglionosis. Eur J Pediatr Surg 11(2):120–123 113. Bughaighis AG, Emergy JL (1971) Functional obstruction of the
93. Rolle U, Yoneda A, Solari V et al (2002) Abnormalities of intestine due to neurological immaturity. Prog Pediatr Surg
C-Kit-positive cellular network in isolated hypoganglionosis. 3:37–52
J Pediatr Surg 37(5):709–714 114. Venugopal S, Mancer K, Shandling B (1981) The validity of
94. Kobayashi H, Hirakawa H, Puri P (1996) Abnormal internal anal rectal biopsy in relation to morphology and distribution of
sphincter innervation in patients with Hirschsprung’s disease ganglion cells. J Pediatr Surg 16(4):433–437
and allied disorders. J Pediatr Surg 31(6):794–799 115. Park SH, Min H, Chi JG et al (2005) Immunohistochemical
95. Kobayashi H, Li Z, Yamataka A et al (2003) Overexpression of studies of pediatric intestinal pseudo-obstruction: bcl2, a valu-
neural cell adhesion molecule (NCAM) antigens on intestinal able biomarker to detect immature enteric ganglion cells. Am J
smooth muscles in hypoganglionosis: is hypoganglionosis a Surg Pathol 29(8):1017–1024
disorder of the neuromuscular junction? Pediatr Surg Int 116. Tanner MS, Smith B, Lloyd JK (1976) Functional intestinal
19(3):190–193 obstruction due to deficiency of argyrophil neurones in the
96. Dingemann J, Puri P (2010) Isolated hypoganglionosis: sys- myenteric plexus: familial syndrome presenting with short small
tematic review of a rare intestinal innervation defect. Pediatr bowel, malrotation, and pyloric hypertrophy. Arch Dis Child
Surg Int 26(11):1111–1115 51(11):837–841
97. Zhang HY, Feng JX, Huang L et al (2008) Diagnosis and sur- 117. Singh I (1963) The prenatal development of enterochromaffin
gical treatment of isolated hypoganglionosis. World J Pediatr cells in the human gastro-intestinal tract. J Anat 97:377–387
4(4):295–300 118. Lolova IS, Davidoff MS, Itzev DE (1998) Histological and
98. Wedel T, Roblick UJ, Ott V et al (2002) Oligoneuronal hypo- immunocytochemical data on the differentiation of intestinal
ganglionosis in patients with idiopathic slow-transit constipa- endocrine cells in human fetus. Acta Physiol Pharmacol Bulg
tion. Dis Colon Rectum 45(1):54–62 23(3–4):61–71
99. Watanabe Y, Ito F, Ando H et al (1999) Morphological inves- 119. Auricchio A, Brancolini V, Casari G et al (1996) The locus for a
tigation of the enteric nervous system in Hirschsprung’s disease novel syndromic form of neuronal intestinal pseudoobstruction
and hypoganglionosis using whole-mount colon preparation. maps to Xq28. Am J Hum Genet 58(4):743–748
J Pediatr Surg 34(3):445–449 120. Puri P, Lake BD, Nixon HH (1977) Adynamic bowel syndrome:
100. Bruder E, Meier-Ruge WA (2007) Hypoganglionosis as a cause report of a case with disturbance of the cholinergic innervation.
of chronic constipation. Pathologe 28(2):131–136 Gut 18(9):754–759
101. Puri P, Fujimoto T (1988) Diagnosis of allied functional bowel 121. Garvey W, Fathi A, Bigelow F et al (1989) A new method for
disorders using monoclonal antibodies and electronmicroscopy. demonstrating argyrophil cells of the pancreas and intestines.
J Pediatr Surg 23(6):546–554 Stain Technol 64(2):87–91
102. Meier-Ruge WA, Brunner LA, Engert J et al (1999) A correl- 122. Bentley JF (1978) Faecal soiling and anal achalasia. Arch Dis
ative morphometric and clinical investigation of hypoganglio- Child 53(3):185–186
nosis of the colon in children. Eur J Pediatr Surg 9(2):67–74 123. Fadda B, Welskop J, Müntefering H et al (1987) Achalasia of
103. Kubota A, Yamauchi K, Yonekura T et al (2001) Clinicopath- the anal sphincter. Pediatr Surg Int 2(2):81–85
ologic relationship of hypoganglionosis. J Pediatr Surg 36(6): 124. Davidson M, Bauer CH (1958) Studies of distal colonic motility
898–900 in children. IV. Achalasia of the distal rectal segment despite
104. Miyazaki E, Ohshiro K, Puri P (1998) NADPH-diaphorase presence of ganglia in the myenteric plexuses of this area.
histochemical staining of suction rectal biopsies in the diagnosis Pediatrics 21(5):746–761
of Hirschsprung’s disease and allied disorders. Pediatr Surg Int 125. de Caluwé D, Yoneda A, Akl U et al (2001) Internal anal
13(7):464–467 sphincter achalasia: outcome after internal sphincter myectomy.
105. Yamataka A, Fujiwara T, Nishiye H et al (1996) Localization of J Pediatr Surg 36(5):736–738
intestinal pacemaker cells and synapses in the muscle layers of a 126. Doodnath R, Puri P (2009) Internal anal sphincter achalasia.
patient with colonic hypoganglionosis. J Pediatr Surg Semin Pediatr Surg 18(4):246–248
31(4):584–587 127. Lake BD, Puri P, Nixon HH et al (1978) Hirschsprung’s disease:
106. Munakata K, Okabe I, Morita K (1992) Hypoganglionosis. Pe- an appraisal of histochemically demonstrated acetylcholines-
diatr Surg Int 7(1):8–11 terase activity in suction rectal biopsy specimens as an aid to
107. Schärli AF, Sossai R (1998) Hypoganglionosis. Semin Pediatr diagnosis. Arch Pathol Lab Med 102(5):244–247
Surg 7(3):187–191 128. Neilson IR, Yazbeck S (1990) Ultrashort Hirschsprung’s dis-
108. Burki T, Kiho L, Scheimberg I et al (2011) Neonatal functional ease: myth or reality. J Pediatr Surg 25(11):1135–1138
intestinal obstruction and the presence of severely immature 129. Fujimoto T, Puri P, Miyano T (1992) Abnormal peptidergic
ganglion cells on rectal biopsy: 6 year experience. Pediatr Surg innervation in internal sphincter achalasia. Pediatr Surg Int
Int 27(5):487–490 7(1):12–17
109. Smith B (1968) Pre- and postnatal development of the ganglion 130. Hirakawa H, Kobayashi H, O’Briain DS et al (1995) Absence of
cells of the rectum and its surgical implications. J Pediatr Surg NADPH-diaphorase activity in internal anal sphincter (IAS)
3(3):386–391 achalasia. J Pediatr Gastroenterol Nutr 20(1):54–58
110. Montedonico S, Sri Paran T, Pirker M et al (2006) Develop- 131. Oue T, Puri P (1999) Altered intramuscular innervation and
mental changes in submucosal nitrergic neurons in the porcine synapse formation in internal sphincter achalasia. Pediatr Surg
distal colon. J Pediatr Surg 41(5):1029–1035 Int 15(3–4):192–194

123
Pediatr Surg Int (2013) 29:855–872 871

132. Piotrowska AP, Solari V, Puri P (2003) Distribution of inter- 151. Taguchi T, Ikeda K, Shono T et al (1989) Autonomic innerva-
stitial cells of Cajal in the internal anal sphincter of patients with tion of the intestine from a baby with megacystis microcolon
internal anal sphincter achalasia and Hirschsprung disease. Arch intestinal hypoperistalsis syndrome: immunohistochemical
Pathol Lab Med 127(9):1192–1195 study. J Pediatr Surg 24(12):1264–1266
133. de Lorijn F, de Jonge WJ, Wedel T et al (2005) Interstitial cells 152. Srikanth MS, Ford EG, Isaacs H Jr et al (1993) Megacystis
of Cajal are involved in the afferent limb of the rectoanal microcolon intestinal hypoperistalsis syndrome: late sequelae
inhibitory reflex. Gut 54(8):1107–1113 and possible pathogenesis. J Pediatr Surg 28(7):957–959
134. Friedmacher F, Puri P (2012) Comparison of posterior internal 153. Xu W, Gelber S, Orr-Urtreger A et al (1999) Megacystis,
anal sphincter myectomy and intrasphincteric botulinum toxin mydriasis, and ion channel defect in mice lacking the alpha3
injection for treatment of internal anal sphincter achalasia: a neuronal nicotinic acetylcholine receptor. Proc Natl Acad Sci
meta-analysis. Pediatr Surg Int 28(8):765–771 USA 96(10):5746–5751
135. Doodnath R, Puri P (2009) Long-term outcome of internal 154. Richardson CE, Morgan JM, Jasani B et al (2001) Megacystis-
sphincter myectomy in patients with internal anal sphincter microcolon-intestinal hypoperistalsis syndrome and the absence
achalasia. Pediatr Surg Int 25(10):869–871 of the alpha3 nicotinic acetylcholine receptor subunit. Gastro-
136. Hecker WC, Holschneider A, Fendel H et al (1973) Chronic enterology 121(2):350–357
constipation in children due to achalasia of the internal anal 155. Xu W, Orr-Urtreger A, Nigro F et al (1999) Multiorgan auto-
sphincter. Dtsch Med Wochenschr 98(49):2334–2340 nomic dysfunction in mice lacking the beta2 and the beta4
137. Heikkinen M, Lindahl H, Rintala RJ (2005) Long-term outcome subunits of neuronal nicotinic acetylcholine receptors. J Neuro-
after internal sphincter myectomy for internal sphincter acha- sci 19(21):9298–9305
lasia. Pediatr Surg Int 21(2):84–87 156. Lev-Lehman E, Bercovich D, Xu W et al (2001) Characteriza-
138. Ciamarra P, Nurko S, Barksdale E et al (2003) Internal anal tion of the human beta4 nAChR gene and polymorphisms in
sphincter achalasia in children: clinical characteristics and CHRNA3 and CHRNB4. J Hum Genet 46(7):362–366
treatment with Clostridium botulinum toxin. J Pediatr Gastro- 157. Puri P, Lake BD, Gorman F et al (1983) Megacystis-micro-
enterol Nutr 37(3):315–319 colon-intestinal hypoperistalsis syndrome: a visceral myopathy.
139. Irani K, Rodriguez L, Doody DP et al (2008) Botulinum toxin for J Pediatr Surg 18(1):64–69
the treatment of chronic constipation in children with internal 158. Schuffler MD, Pagon RA, Schwartz R et al (1988) Visceral
anal sphincter dysfunction. Pediatr Surg Int 24(7):779–783 myopathy of the gastrointestinal and genitourinary tracts in
140. Koivusalo AI, Pakarinen MP, Rintala RJ (2009) Botox injection infants. Gastroenterology 94(4):892–898
treatment for anal outlet obstruction in patients with internal 159. Piotrowska AP, Rolle U, Chertin B et al (2003) Alterations in
anal sphincter achalasia and Hirschsprung’s disease. Pediatr smooth muscle contractile and cytoskeleton proteins and inter-
Surg Int 25(10):873–876 stitial cells of Cajal in megacystis microcolon intestinal hypo-
141. Langer JC, Birnbaum E (1997) Preliminary experience with peristalsis syndrome. J Pediatr Surg 38(5):749–755
intrasphincteric botulinum toxin for persistent constipation after 160. Szigeti R, Chumpitazi BP, Finegold MJ et al (2010) Absent
pull-through for Hirschsprung’s disease. J Pediatr Surg smooth muscle actin immunoreactivity of the small bowel
32(7):1059–1061 muscularis propria circular layer in association with chromo-
142. Messineo A, Codrich D, Monai M et al (2001) The treatment of some 15q11 deletion in megacystis-microcolon-intestinal
internal anal sphincter achalasia with botulinum toxin. Pediatr hypoperistalsis syndrome. Pediatr Dev Pathol 13(4):322–325
Surg Int 17(7):521–523 161. Masetti M, Rodriguez MM, Thompson JF et al (1999) Multi-
143. Minkes RK, Langer JC (2000) A prospective study of botulinum visceral transplantation for megacystis microcolon intestinal
toxin for internal anal sphincter hypertonicity in children with hypoperistalsis syndrome. Transplantation 68(2):228–232
Hirschsprung’s disease. J Pediatr Surg 35(12):1733–1736 162. Piaseczna Piotrowska A, Rolle U, Solari V et al (2004) Inter-
144. Boman F, Sfeir R, Bonnevalle M et al (2006) Complexity of stitial cells of Cajal in the human normal urinary bladder and in
pathological interpretation in megacystis-microcolon-intestinal the bladder of patients with megacystis-microcolon intestinal
hypoperistalsis syndrome. Ann Pathol 26(2):115–121 hypoperistalsis syndrome. BJU Int 94(1):143–146
145. Berdon WE, Baker DH, Blanc WA et al (1976) Megacystis- 163. Kirtane J, Talwalker V, Dastur DK (1984) Megacystis, micro-
microcolon-intestinal hypoperistalsis syndrome - a new cause of colon, intestinal hypoperistalsis syndrome: possible pathogene-
intestinal obstruction in the newborn: report of radiologic find- sis. J Pediatr Surg 19(2):206–208
ings in five newborn girls. AJR Am J Roentgenol 126(5): 164. al-Rayess M, Ambler MW (1992) Axonal dystrophy presenting
957–964 as the megacystis-microcolon-intestinal hypoperistalsis syn-
146. Chamyan G, Debich-Spicer D, Opitz JM et al (2001) Mega- drome. Pediatr Pathol 12(5):743–750
cystis-microcolon-intestinal hypoperistalsis syndrome and 165. Gosemann JH, Puri P (2011) Megacystis microcolon intestinal
aganglionosis in trisomy 18. Am J Med Genet 102(3):293–296 hypoperistalsis syndrome: systematic review of outcome. Pedi-
147. Ciftci AO, Cook RC, van Velzen D (1996) Megacystis micro- atr Surg Int 27(10):1041–1046
colon intestinal hypoperistalsis syndrome: evidence of a primary 166. Köhler M, Pease PW, Upadhyay V (2004) Megacystis-micro-
myocellular defect of contractile fiber synthesis. J Pediatr Surg colon-intestinal hypoperistalsis syndrome (MMIHS) in siblings:
31(12):1706–1711 case report and review of the literature. Eur J Pediatr Surg
148. Puri P, Tsuji M (1992) Megacystis-microcolon-intestinal hypo- 14(5):362–367
peristalsis syndrome (neonatal hollow visceral myopathy). Pe- 167. Young ID, McKeever PA, Brown LA et al (1989) Prenatal
diatr Surg Int 7(1):18–22 diagnosis of the megacystis-microcolon-intestinal hypoperistal-
149. Rolle U, O’Briain S, Pearl RH et al (2002) Megacystis-micro- sis syndrome. J Med Genet 26(6):403–406
colon-intestinal hypoperistalsis syndrome: evidence of intestinal 168. Mc Laughlin D, Puri P (2013) Familial megacystis microcolon
myopathy. Pediatr Surg Int 18(1):2–5 intestinal hypoperistalsis syndrome: a systematic review. Pediatr
150. Gillis DA, Grantmyre EB (1985) Megacystis-microcolon- Surg Int (in press)
intestinal hypoperistalsis syndrome: survival of a male infant. 169. McNamara HM, Onwude JL, Thornton JG (1994) Megacystis-
J Pediatr Surg 20(3):279–281 microcolon-intestinal hypoperistalsis syndrome: a case report

123
872 Pediatr Surg Int (2013) 29:855–872

supporting autosomal recessive inheritance. Prenat Diagn 14(2): 173. Garel C, Dreux S, Philippe-Chomette P et al (2006) Contribution
153–154 of fetal magnetic resonance imaging and amniotic fluid digestive
170. Penman DG, Lilford RJ (1989) The megacystis-microcolon- enzyme assays to the evaluation of gastrointestinal tract abnor-
intestinal hypoperistalsis syndrome: a fatal autosomal recessive malities. Ultrasound Obstet Gynecol 28(3):282–291
condition. J Med Genet 26(1):66–67 174. Puri P, Shinkai M (2005) Megacystis microcolon intestinal
171. Rolle U, Puri P (2006) Structural basis of voiding dysfunction in hypoperistalsis syndrome. Semin Pediatr Surg 14(1):58–63
megacystis microcolon intestinal hypoperistalsis syndrome. 175. Loinaz C, Rodriguez MM, Kato T et al (2005) Intestinal and
J Pediatr Urol 2(4):277–284 multivisceral transplantation in children with severe gastroin-
172. Granata C, Puri P (1997) Megacystis-microcolon-intestinal testinal dysmotility. J Pediatr Surg 40(10):1598–1604
hypoperistalsis syndrome. J Pediatr Gastroenterol Nutr 25(1):
12–19

123

You might also like