ArticlePDF Available

Interstitial cells of Cajal in the human fetal small bowel as shown by c-Kit immunohistochemistry

Authors:

Abstract and Figures

Interstitial cells of Cajal (ICCs) express the tyrosine kinase receptor c-kit, which is required for their development and spontaneous pacemaker activity in the bowel. From murine models it has been proposed that ICCs do not develop until after birth, but more recent findings indicate that c-kit is expressed early in the embryonic period. The temporal development of ICCs in the human gut remains unknown. To investigate ICCs in the human fetal small bowel using c-kit immunohistochemistry. Small bowel specimens were obtained at post mortem examination of 16 fetuses and nine neonates, eight of whom were premature, born at gestational ages of 13 to 41 weeks, without gastrointestinal disorders. Immunohistochemical analysis was performed on material fixed in formalin and embedded in paraffin. The specimens were exposed to antibodies raised against c-kit (an ICC marker) and neurone specific enolase (a general neuronal marker). The ABC complex method was used to visualise binding of antibodies to the corresponding antigens. c-kit immunoreactive cells were visualised from 13 weeks of gestation. The immunoreactivity was mainly localised in association with the myenteric plexus. From about 17-18 weeks of gestation, the ICCs formed a layer along the myenteric plexus, whereas this layer appeared to be disrupted at 13-16 weeks of gestation. CONCLUSIONS ICCs are c-kit immunoreactive at least from a gestational age of 13 weeks in the human fetal small intestine. From 17-18 weeks of gestation until birth, they form a continuous layer around the myenteric ganglia.
Content may be subject to copyright.
Interstitial cells of Cajal in the human fetal small
bowel as shown by c-kit immunohistochemistry
T Wester, L Eriksson, Y Olsson, L Olsen
Abstract
Background—Interstitial cells of Cajal
(ICCs) express the tyrosine kinase recep-
tor c-kit, which is required for their devel-
opment and spontaneous pacemaker
activity in the bowel. From murine models
it has been proposed that ICCs do not
develop until after birth, but more recent
findings indicate that c-kit is expressed
early in the embryonic period. The tem-
poral development of ICCs in the human
gut remains unknown.
Aim—To investigate ICCs in the human
fetal small bowel using c-kit immunohisto-
chemistry.
Subjects—Small bowel specimens were
obtained at post mortem examination of
16 fetuses and nine neonates, eight of
whom were premature, born at
gestational ages of 13 to 41 weeks, without
gastrointestinal disorders.
Methods—Immunohistochemical analysis
was performed on material fixed in for-
malin and embedded in paraYn. The
specimens were exposed to antibodies
raised against c-kit (an ICC marker) and
neurone specific enolase (a general neuro-
nal marker). The ABC complex method
was used to visualise binding of antibodies
to the corresponding antigens.
Results—c-kit immunoreactive cells were
visualised from 13 weeks of gestation. The
immunoreactivity was mainly localised in
association with the myenteric plexus.
From about 17–18 weeks of gestation, the
ICCs formed a layer along the myenteric
plexus, whereas this layer appeared to be
disrupted at 13–16 weeks of gestation.
Conclusions—ICCs are c-kit immuno-
reactive at least from a gestational age of
13 weeks in the human fetal small intes-
tine. From 17–18 weeks of gestation until
birth, they form a continuous layer
around the myenteric ganglia.
(Gut 1999;44:65–71)
Keywords: interstitial cells of Cajal; c-kit; myenteric
plexus; human; fetal; development
Although it is now more than a century since
the interstitial cells of Cajal (ICCs) first
attracted the interest of scientists,12 many
questions about these cells remain unan-
swered, particularly with respect to their func-
tion. Furthermore, the ontogeny and develop-
ment of ICCs, especially in the human
gastrointestinal tract, require further investiga-
tion, and these questions were addressed in the
present study.
In the human small bowel, the ICCs are
localised at the level of the myenteric plexus
between the longitudinal and circular muscle
layers, in the deep muscular plexus in the
innermost part of the circular muscle layer, and
within the circular muscle layer itself.3–5 They
are considered to be generators of spontaneous
pacemaker activity in the smooth muscle layers
of the gut.6–9 Furthermore, it has been sug-
gested that they may be involved in
neurotransmission.9A cytokine, steel factor,10
also termed stem cell factor (SCF)11 or mast
cell growth factor (MGF),12 has been identified
as the c-kit ligand. Huizinga et al13 showed that
ICCs associated with the myenteric plexus
express c-kit, which is a proto-oncogene
encoding a cell surface receptor tyrosine
kinase.14 Mice with mutations in the white
spotting (W) locus, resulting in kit gene muta-
tions, lack ICCs associated with the myenteric
plexus as well as intestinal pacemaker activity.
In conclusion, ICCs appear to have a key
role in the normal function of the intestine, and
alterations in these cells may well be involved in
various intestinal disorders. For instance, in
Hirschsprung’s disease there are considerably
fewer ICCs in the aganglionic segment than in
the ganglionic bowel.15 16 The absence or
reduction of c-kit immunoreactive cells in
association with the myenteric plexus has also
been observed in infantile hypertrophic pyloric
stenosis.17 Recently, Isozaki et al18 reported two
cases of myopathic chronic intestinal pseudo-
obstruction with decreased numbers of c-kit
immunoreactive cells associated with the my-
enteric plexus.
To our knowledge, the temporal develop-
ment of ICCs in the human gastrointestinal
tract has not been systematically investigated
previously. The purpose of this study was
therefore to map c-kit immunoreactivity as an
ICC marker in the small intestine from human
fetuses at diVerent gestational ages. Neurone
specific enolase (NSE) immunohistochemistry
was used as a marker of the neuronal
component, which has already been well
defined in the human fetal small bowel.19
Information on the ICCs may contribute to our
understanding of the development of small
bowel motility in premature babies in whom
this motility appears immature in the fasting
state,20 21 although it has been reported that
premature babies respond appropriately to
feeding.22 Furthermore, knowledge of ICC
development may help to clarify the aetiology
Abbreviations used in this paper: ICC, interstitial
cells of Cajal; SCF, stem cell factor; MGF, mast cell
growth factor; NSE, neurone specific enolase; PBS
phosphate buVered saline.
Gut 1999;44:65–71 65
Department of
Paediatric Surgery,
University Children’s
Hospital, S-751 85
Uppsala, Sweden
T Wester
L Olsen
Laboratory of
Neuropathology,
Department of
Genetics and
Pathology, University
Hospital, Uppsala,
Sweden
L Eriksson
Y Olsson
Correspondence to:
Dr T Wester.
Accepted for publication
22 July 1998
and pathophysiology of disorders characterised
by a deficiency of ICCs.
Methods
Specimens from the mid part of the small
bowel were collected for diagnostic purposes at
routine post mortem examination of 16 fetuses
and nine newborns, eight of whom were born
prematurely. The gestational age ranged from
13 to 41 weeks (table 1). The liveborn babies
died between the first and 17th day of life.
Seven of these were born at 21–27 weeks of
gestation. Post mortem examination was per-
formed one to six days after death. Mild mac-
eration was found in three of the fetuses who
died in utero; stillbirths with severe maceration
were not included. Furthermore, fetuses and
infants displaying grossly pathological condi-
tions of the gastrointestinal tract were not
included. The morphology of the bowel wall
was normal in all cases at routine histopatho-
logical examination of haematoxylin and eosin
stained sections, although the mucosa showed
autolytic alterations in several subjects. Speci-
mens were also obtained from the rectum in
five cases at 13, 19, 19, 23, and 31 weeks of
gestation. As control tissue, specimens from
the distal ileum were collected at the time of
ileocaecal resection in three cases. The resec-
tions were performed because of colonic
cancer, villous adenoma, and perforated ap-
pendicitis, and the patients were 63,75, and 78
years old.
The study was approved by the ethics
committee of the Faculty of Medicine of Upp-
sala University.
TISSUE PREPARATION
The specimens were fixed in 10% formalin and
embedded in paraYn. Sections 5 µm thick were
cut, placed on poly-L-lysine coated slides, and
incubated at 37°C overnight. All sections were
deparaYnised in xylene and hydrated.
IMMUNOHISTOCHEMISTRY
Antigen retrieval by microwave oven heating was
performed for both antibodies. The sections
were boiled (750 W) in citric acid buVer (10
mM, pH 6.0) for five (NSE) or ten (c-kit) min-
utes. The sections were allowed to cool to room
temperature in the buVer and then rinsed in
phosphate buVered saline (PBS, pH 7.4). The
endogenous peroxidase activity was blocked in
2% hydrogen peroxide in distilled water for five
minutes and the sections were then rinsed in
PBS. Thereafter they were incubated with
normal swine serum (Dako, Glostrup, Den-
mark; dilution 1:5) for c-kit or normal goat
serum (Dako; dilution 1:5) for NSE in PBS for
20 minutes at room temperature. Incubation
with the primary antibody took place overnight
at 4°C (c-kit) or for one hour at room tempera-
ture (NSE). Table 2 provides a further descrip-
tion of the primary antibodies. After being
rinsed in PBS, the slides were incubated with a
biotinylated secondary antibody, goat anti-
mouse (Dako, product number E0433; dilution
1:200) for NSE or swine anti-rabbit (Dako,
product number E0353; dilution 1:300) for
c-kit, for 30 minutes at room temperature. The
sections were then incubated in Vectastain elite
ABC kit (Vector Laboratories, Burlingame,
California, USA) for 30 minutes and developed
in 3,3’-diaminobenzidine tetrahydrochloride
(Sigma, London, UK) for about six minutes.
Counterstaining with haematoxylin was per-
formed. Finally, the sections were dehydrated,
cleared in xylene and mounted in Pertex
(Histolab, Göteborg, Sweden).
Immunoreactivity was absent in negative
controls in which the primary antibody was
omitted. For c-kit, the immunostaining was
also abolished by a blocking peptide (1.0 µg/ml;
#sc-168p; Santa Cruz Biotechnology, Santa
Cruz, California, USA), incubated with c-kit
antibody (0.01 µg/ml) for two hours at room
temperature, before the specific immunohisto-
chemical procedure, according to instructions
provided by the suppliers.
The slides were analysed by light microscopy.
Table 1 Data on the subjects
Case
Gestational age
(weeks)
Age at death
(days)/death in utero
(iu) Diagnosis
1 13 iu Anencephaly
2 15 iu Anencephaly
3 15 iu Amnion band syndrome
4 16 iu Anencephaly
5 17 iu Fragile X syndrome
6 17 iu Unknown*
7 18 iu Ablatio placentae, chorioamnionitis
8 18 iu Dysplastic kidneys
9 20 iu Down’s syndrome
10 20 iu Unknown
11 21 1 Cerebral haemorrhage
12 21 1 Pulmonary haemorrhage, cerebral
haemorrhage
13 23 15 Aortic thrombosis, renal infarction,
cerebral haemorrhage
14 25 1 RDS, cerebral haemorrhage
15 25 2 Septicaemia, pulmonary haemorrhage
16 25 14 Pneumonia, septicaemia
17 26 17 Septicaemia, cerebral haemorrhage
18 31 iu Unknown
19 31 iu Meckel-Gruber syndrome
20 34 iu Unknown
21 35 iu Unknown
22 36 2 Polycystic kidney disease
23 36 iu Asphyxia in utero
24 41 iu Unknown
25 41 4 Pulmonary hypertension, bilateral
pneumothorax
*Unknown means that the cause of death was not established at autopsy.
RDS, respiratory distress syndrome.
Table 2 Sur vey of the antibodies used
Antibody Clone/product no Characteristics Source Dilution
c-kit # sc-168 Polyclonal, rabbit, anti-human, raised
against peptide corresponding to
amino acids 958-976 within carboxy
terminal domain of human c-kit
Santa Cruz Biotechnology 0.1 µg/ml
NSE BBS/NC/VI-H14 Monoclonal, mouse, anti-human Dako 1:100
NSE, neurone specific enolase.
66 Wester, Eriksson, Olsson, et al
Figure 1 (A) NSE immunohistochemistry showing immunoreactive ganglion cells in the myenteric plexus (arrow) and in
the inner and outer submucous plexuses (arrowheads) of the small bowel in an infant born at 26 weeks of gestation who
died at the age of 17 days. Nerve fibres associated with the ganglia and in the circular muscle layer are also stained.
Original magnification ×66. (B) NSE immunohistochemistry of the small bowel at 13 weeks of gestation displays the
myenteric plexus (arrow), whereas no immunoreactive ganglion cells were present in the submucosa. Original magnification
×40. (C) From 17–18 weeks of gestation, the ICCs form a continuous layer along the myenteric plexus of the small bowel.
This is illustrated in a case at 20 weeks of gestation using c-kit immunohistochemistry.Original magnification ×80. (D)
c-kit immunoreactive cells are elongated in shape and have an ovoid nucleus as in this case at 20 weeks of gestation. The
cell processes do not seem to penetrate into the ganglia. Original magnification ×200. (E) From 13 to 16 weeks of
gestation, the layer of c-kit immunoreactive ICCs associated with the myenteric plexus of the small bowel appears to be
disrupted. In the submucosa, isolated round c-kit immunoreactive cells, interpreted as mast cells, are seen (arrows). In this
case the submucous plexus had not yet developed at 13 weeks of gestation,as shown in (B). Original magnification ×80.
(F) From 17 weeks of gestation, the processes of the ICCs are often seen to penetrate into the circular muscle layer,whereas
isolated ICCs in this layer are only rarely observed. Original magnification ×50. Abbreviations: LM, longitudinal muscle
layer; CM, circular muscle layer; SM, submucosa.
Development of interstitial cells of Cajal 67
Results
NSE IMMUNOHISTOCHEMISTRY
NSE immunohistochemistry clearly showed
the myenteric and submucous plexuses. NSE
immunoreactivity was observed in the nerve
cell bodies and also in nerve fibres in the inter-
muscular space and submucosa (fig 1A). The
myenteric plexus was established in all cases.
On the other hand, the submucous plexus was
absent in the earliest case (13 weeks of
gestation, fig 1B). In several of the early
subjects submucous ganglia were identifiable,
butitwasdiYcult to distinguish an inner and
outer plexus and most of the immunoreactive
cells were observed close to the circular muscle
layer. In the older subjects, separate inner and
outer plexuses were established and clearly
visualised (fig 1A). We did not observe any
NSE immunoreactive cells with morphological
features characteristic of ICCs.
C-KIT IMMUNOHISTOCHEMISTRY
c-kit immunoreactivity was observed in all but
two cases, at gestational ages of 31 and 35
weeks. NSE immunoreactivity was normal in
these cases. There was no obvious reason for
the negative results, but post mortem changes
may have contributed as these fetuses were
mildly macerated. Both fetuses died in utero
and the cause of death could not be established
atpostmortem examination.Thec-kitimmuno-
reactive cells were situated in the intermuscular
space, and most of the c-kit positive cells were
thin and elongated and had an ovoid nucleus,
surrounding the myenteric ganglia and nerve
fascicles (figs 1C and D). In subjects older than
about 17–18 weeks of gestation, a continuous
layer of c-kit immunoreactive cells was usually
observed around the myenteric plexus (fig 1C).
On the other hand, at a gestational age of
13–16 weeks this layer of cells appeared to be
disrupted, and immunoreactive cells with a
round nucleus and short processes were found.
Furthermore, in these cases the immunoreac-
tivity was weak and few immunopositive cells
were seen (fig 1E). In almost all cases, round
immunoreactive cells were observed in the
submucosa without any association with the
submucous ganglia; these were interpreted as
mast cells, which are known to be c-kit immuno-
reactive (fig 1E).23 Although the c-kit positive
cells often extended into the smooth muscle
layers (fig 1F), isolated immunopositive cells
were rarely seen in the muscle layers. No
diVerences between the liveborn neonates and
the fetuses who died in utero could be
distinguished. In the rectum specimens, similar
findings were encountered. However, at 13
weeks of gestation no c-kit immunoreactivity
could be observed around the myenteric plexus
(fig 2A). At 19 weeks of gestation c-kit immu-
noreactive cells were rare in one of the cases,
whereas in the second case, the myenteric gan-
glia were outlined by c-kit immunoreactive
ICCs (fig 2B). The latter pattern was also
observed at 23 and 31 weeks of gestation. In the
adult small intestine, large numbers of ICCs
were found around the myenteric ganglia. The
number of ICCs in the circular muscle layer was
larger than in the fetal cases (fig 2C).
Discussion
The morphology and distribution of ICCs have
previously been diYcult to study, as standard
staining procedures for conventional light
microscopy do not disclose this cell type.
Figure 2 (A) At 13 weeks of gestation no c-kit
immunoreactivity can be seen in association with the
myenteric plexus of the rectum. Original magnification ×
50. (B) A layer of c-kit immunoreactive ICCs is seen
around the myenteric plexus of the rectum at 19 weeks of
gestation. Original magnification ×50. (C) In the adult
small bowel, c-kit immunoreactive ICCs surround the
myenteric ganglia. ICCs are also frequently found in the
circular muscle layer.Or iginal magnification ×25.
68 Wester, Eriksson, Olsson, et al
Consequently, electron microscopy has been
the method of choice for investigations of the
distribution of ICCs, and several ultrastruc-
tural studies have in fact been performed on
human small bowel using this technique.3–5
Ultrastructurally, ICCs associated with the
myenteric plexus are arranged in bundles
surrounding the ganglia and nerve fascicles,
with which they are always in close contact.
The processes of ICCs are long and usually do
not branch. Characteristically, the ICC proc-
esses contain a well developed smooth endo-
plasmic reticulum and a dense meshwork of
intermediate filaments. The perinuclear cyto-
plasm also contains many intermediate fila-
ments, but less abundant smooth endoplasmic
reticulum. The chromatin pattern of the ICC
nucleus is similar to that observed in smooth
muscle cells. However, in contrast with smooth
muscle cells, ICCs do not contain myosin
filaments.4
It has been shown in mouse,24 guinea pig,25
and human bowel15 that the morphology of the
c-kit immunoreactive cells associated with the
myenteric plexus correlates with the ultrastruc-
tural descriptions of ICCs. ICCs are interca-
lated between varicose nerve endings and
smooth muscle cells. Two types of varicosities
have been identified in the nerve endings. The
first may be cholinergic, whereas the second,
which is more commonly encountered in
association with the deep muscular plexus
ICCs than in those associated with the
myenteric plexus, is morphologically similar to
varicosities seen in inhibitory non-adrenergic
non-cholinergic neurones.9These findings in-
dicate that ICCs may play a role in inhibitory
neurotransmission, which was further investi-
gated by Publicover et al,26 who proposed that
these cells may amplify the inhibitory nitric
oxide signalling in canine colon. The demon-
stration of nitric oxide synthase, the endothelial
isoform, in ICCs indicates that this cell type is
able to synthesise nitric oxide.27
Maeda et al23 found that c-kit plays a major
role in the development of the pacemaker sys-
tem in the gut and proposed that ICCs were
involved. Administration of an antagonistic
anti-c-kit antibody to newborn mice resulted in
abnormal intestinal motility.23 Several other
investigators have presented results supporting
the role of ICCs in generating spontaneous
pacemaker activity. For instance, removal of
the submucosa from the circular muscle layer,
disrupting the submucosa-circular muscle in-
terface which is the location of one class of
ICCs, was found to abolish slow waves in the
canine colonic circular muscle.7Methylene
blue perfusion combined with illumination of
intestinal smooth muscle preparations resulted
in selective damage to the ICCs and loss of
slow wave activity.6Furthermore, exposure of
ICCs to rhodamine 123, which is a fluorescent
dye taken up by ICCs and enteric neurones but
not by smooth muscle cells, altered the electri-
cal rhythmicity of canine colonic circular mus-
cle preparations.28
The embryonic origin of ICCs has been
debated. Recently, however, Lecoin et al29
investigated the origin of the c-kit positive cells
in avian bowel. Quail neural crest was grafted
to chicken embryos at embryonic day 2. The
experiment showed that all enteric neurones
and glial cells were of quail origin, whereas all
c-kit immunoreactive cells originated from the
chick. The authors concluded that the c-kit
positive ICCs have a mesenchymal origin.
In an ultrastructural study of murine small
bowel, Faussone-Pellegrini30 reported that
ICCs were not detected in term fetuses, but
appeared during the first two weeks of life. On
the other hand, Torihashi et al24 detected c-kit
immunoreactivity in the murine small intestine
at embryonic day 12 (E12). At this stage the
c-kit positive cells were found at the outer sur-
face of the bowel wall just beneath the serosal
layer. These cells could not be characterised as
either ICCs or smooth muscle cells. The ICC
network associated with the myenteric plexus
developed between days E15 and E18. The
authors suggested that a population of cells
that were c-kit positive at day E12 diVerenti-
ated into smooth muscle cells, losing the c-kit
immunoreactivity, whereas others continued to
express c-kit immunoreactivity and diVerenti-
ated into ICCs associated with the myenteric
plexus. The c-kit immunoreactivity did not
co-localise with the neuronal marker c-ret,
indicating that ICCs are not of neuronal origin.
The ICCs associated with the deep muscular
plexus developed after birth. The development
of ICCs in the human fetal bowel remains
unclear, which was the reason for the present
study. Matsuda et al31 investigated c-kit expres-
sion in normal adult and fetal human tissue
and found c-kit immunoreactivity in enteric
nervous plexuses in adult tissue, but not in fetal
tissue. On the other hand, they observed c-kit
immunoreactive cells in the brain in both
tissues. Vanderwinden et al15 mentioned that
c-kit immunoreactive ICCs appeared in the
human foregut at gestational week 14 and in
the hindgut at 23 weeks of gestation but with-
out further description. Horie et al32 found c-kit
immunoreactivity in the smooth muscle layers
of human fetal oesophagus, small bowel, and
colon at 18 and 20 weeks of gestation.
We observed c-kit immunoreactivity in elon-
gated cells with an ovoid nucleus around the
myenteric ganglia. The typical localisation and
morphology of the c-kit immunoreactive cells
visualised in our study indicate that they are
ICCs. It has previously been shown that ICCs
associated with the myenteric plexus express
the c-kit tyrosine kinase receptor.13 Prosser et
al33 reported that some ICCs associated with
the myenteric plexus were NSE immunoreac-
tive in the rat intestine. However, these findings
were not corroborated in our study, as we did
not observe NSE immunoreactive cells with
morphological features or localisations charac-
teristic of ICCs. Our findings indicate that
ICCs express c-kit at least as early as at 13
weeks of gestation in the human fetal small
bowel, although the morphology and distribu-
tion of the ICCs at this stage suggest that they
are still immature. At a gestational age of 13–16
weeks, few dispersed ICCs were observed in
the intermuscular space and some of the indi-
vidual cells had short processes and a round
Development of interstitial cells of Cajal 69
nucleus. In the rectum specimens,no immuno-
reactivity was observed at this gestational age.
From 17–18 weeks of gestation a continuous
layer of c-kit immunoreactive ICCs was
observed around the myenteric ganglia and
nerve fascicles in the small bowel, and we
detected no further alterations from this
gestational age until full term. From about 19
weeks of gestation, c-kit immunoreactive ICCs
had developed in association with the rectal
myenteric ganglia. We did not observe any c-kit
immunoreactive cells in the region of the deep
muscular plexus in the fetal cases, although
ICCs have been observed at this site in
ultrastructural studies of adult human small
bowel.5One explanation for the absence of
c-kit immunoreactive ICCs in the deep muscu-
lar plexus in the fetal cases may be that ICCs of
this type develop after birth, as in the murine
small bowel.24 However, Isozaki et al18 only
found a few c-kit immunoreactive cells in the
deep muscular plexus in human adults, which
may indicate that it is mainly the ICCs associ-
ated with the myenteric plexus that are c-kit
positive in the human small bowel. We found
numerous c-kit positive ICCs in the circular
muscle layer in the adult cases, but it was diY-
cult to distinguish a separate deep muscular
plexus.
In the murine small bowel, it has been shown
that the onset of electrical rhythmicity corre-
lates with the development of ICCs. At E16
and E17 spontaneous electrical activity was
absent, whereas slow waves could be recorded
at E19 when the ICCs and the longitudinal
muscle layers had diVerentiated into two
distinct entities.24 In mice, the slow wave
frequency has been shown to correlate with
variations in the intraluminal pressure, which
results in propulsive peristalsis as shown by
radiological methods.34 McLain35 investigated
human fetal gastrointestinal motility by amni-
ography and could not show any emptying of
contrast from the stomach before about 30
weeks of gestation. However, from 30 weeks of
gestation, the rate of propagation of contrast
increased with increasing gestational age.
Bisset et al20 studied small intestinal pressures
in preterm infants from 28 weeks of gestation
until term. Before 31 weeks of gestation a dis-
organised activity pattern with low amplitude
was found. Between 31 and 34 weeks of gesta-
tion, clustered phasic activity appeared, and
from 34 to 37 weeks of gestation prolonged
phasic activity was observed propagating in
aboral direction. At term a well defined fasting
motor activity with migrating motor complexes
was discernible. However, Berseth22 showed
that preterm infants respond appropriately to
feeding administered as infusion, by develop-
ing persistent activity.The responses to feeding
among prematures did not diVer from that of
term infants. The slow wave frequency which
has been associated with ICC function in-
creases with gestational age from 10.5 cycles
per minute at 28 weeks of gestation to 12.5
cycles per minute at term.20
It is diYcult to investigate normal human
fetal development. Ethical aspects of collecting
and using material must be considered. Fur-
thermore, the great variation seen in human
material as opposed to animal models may
make interpretation diYcult. Our material
comprised specimens obtained at autopsy after
induced termination or spontaneous abortion
and specimens from liveborn babies who died
within the first weeks of life. A wide range of
malformations and disorders were encoun-
tered, but we could not distinguish any abnor-
malities in the expression of NSE or c-kit
immunoreactivity which could be related to
these conditions. However, the absence of c-kit
immunoreactivity in two of our cases may well
be explained by post mortem changes or events
occurring before death. It has previously been
pointed out that events before death, such as
hypoxaemia, may influence the staining prop-
erties of enteric ganglia in studies using silver
impregnation36 or NADPH diaphorase
histochemistry.37 The eVects of autolysis are
always a concern when autopsy material is
used, but it has been reported that many anti-
gens are surprisingly resistant to post mortem
changes.38
In conclusion, ICCs associated with the
myenteric plexus in the human fetal small
bowel express the c-kit tyrosine kinase receptor
at least from 13 weeks of gestation. From about
17–18 weeks of gestation the c-kit immuno-
reactive ICCs form a layer of cells surrounding
the myenteric ganglia and nerve fascicles.
However, at 13–16 weeks this cell layer
appeared to be disrupted, which may indicate
that ICCs are still in a phase of development at
that stage.
This work was supported by the HRH Crown Princess Louisa’s
Association for Child Medical Care and the Ollie and Elof Eric-
sson Foundation for Scientific Research. We are also grateful to
Gunilla Tibbling and Frank Bittkowski for their photographic
assistance and to Bengt Sandstedt who contributed specimens.
1 Cajal SR. Sur les ganglions nerveux de l’intestin. C R Seances
Soc Biol Fil 1893;45:217–23.
2 Cajal SR. Histologie du système ner veux de l’homme et des
vertébrés. Paris: Maloine, 1911;2:891–942.
3 Rumessen JJ, Mikkelsen B, Qvortr up,et al. Ultrastr ucture of
interstitial cells of Cajal in circular muscle of human small
intestine. Gastroenterology 1993;104:343–50.
4 Rumessen JJ, Thuneberg L. Interstitial cells of Cajal in
human small intestine. Ultrastructural identification and
organisation between the main smooth muscle layers. Gas-
troenterology 1991;100:1417–31.
5 Rumessen JJ, Mikkelsen HB, Thuneberg. Ultrastructure of
interstitial cells of Cajal associated with deep muscular
plexus of human small intestine. Gastroenterology 1992;102:
56–68.
6 Liu LWC, Thuneberg L, Huizinga JD. Selective lesioning of
interstitial cells of Cajal by methylene blue and light leads
to loss of slow waves. Am J Physiol 1994;266:G485–96.
7 Durdle NG, Kingma YJ, Bowes KL, et al. Origin of slow
waves in the canine colon. Gastroenterology 1983;84:375–82.
8 Liu LWC, Huizinga JD.Electrical coupling of circular mus-
cle to longitudinal muscle and interstitial cells of Cajal in
canine colon. J Physiol (Lond) 1993;470:445–61.
9 Thuneberg L. Interstitial cells of Cajal: intestinal pacemaker
cells? Adv Anat Embryol Cell Biol 1982;71:1–130.
10 Flanagan JG, Leder P.The kit ligand: a cell surface molecule
altered in steel mutant fibroblasts. Cell 1990;63:185–94.
11 Zsebo KM, Wypych J, McNiece IK, et al. Identification,
purification, and biological characterisation of haematopoi-
etic stem cell factor from BuValo rat liver-conditioned
medium. Cell 1990;63:195–201.
12 Williams DE, Eisenmann J, Baird A, et al. Identification of a
ligand for the c-kit proto-oncogene. Cell 1990;63:167–74.
13 Huizinga JD, Thuneberg L, Klüppel M, et al. W/kit gene
required for interstitial cells of Cajal and for intestinal
pacemaker activity. Nature 1995;373:347–9.
14 Yarden Y, Kuang W-J, Yang-Feng T, et al. Human proto-
oncogene c-kit: a new cell surface receptor tyrosine kinase for
an unidentified ligand. EMBO J 1987;6:3341–51.
15 Vanderwinden J-M, Rumessen JJ, Liu H, et al. Interstitial
cells of Cajal in human colon and in Hirschsprung’s
disease. Gastroenterology 1996;111:901–10.
70 Wester, Eriksson, Olsson, et al
16 Yamataka A, Kato Y, Tibboel D, et al. A lack of intestinal
pacemaker (c-kit) in aganglionic bowel of patients with
Hirschsprung’s disease. J Pediatr Surg 1995;30:441–4.
17 Vanderwinden J-M, Liu H, De Laet M-H, et al. Study of the
interstitial cells of Cajal in infantile hypertrophic pyloric
stenosis. Gastroenterology 1996;111:279–88.
18 Isozaki K, Hirota S, Miyagawa J-I, et al. Deficiency of c-kit+
cells in patients with a myopathic form of chronic
idiopathic intestinal pseudo-obstruction. Am J Gastroenterol
1997;92:332–4.
19 Fekete E, Benedeczky I, Timmermans J-P, et al. Sequential
pattern of nerve-muscle contacts in the small intestine of
developing human fetus. An ultrastructural and immuno-
histochemical study. Histol Histopathol 1996;11:845–50.
20 Bisset WM, Watt JB, Rivers RP, et al. Ontogeny of fasting
small intestinal motor activity in the human infant. Gut
1988;29:483–8.
21 Berseth CL. Gestational evolution of small intestine motility
in preterm and term infants. J Pediatr 1989;115:646–51.
22 Berseth CL. Neonatal small intestinal motility: motor
responses to feeding in term and preterm infants. J Pediatr
1990;117:777–82.
23 Maeda H, Yamagata A, Nishikawa S, et al. Requirement of
c-kit for development of intestinal pacemaker system.
Development 1992;116:369–75.
24 Torihashi S, Ward SM, Sanders KM. Development of c-kit-
positive cells and the onset of electrical rhythmicity in
murine small intestine. Gastroenterology 1997;112:144–55.
25 Burns AJ, Herbert TM, Ward SM, et al. Interstitial cells of
Cajal in the guinea-pig gastrointestinal tract as revealed by
c-kit immunohistochemistry. Cell Tissue Res 1997;290:11–
20.
26 Publicover NG, Hammond EM, Sanders KM. Amplification
of nitric oxide signaling by interstitial cells isolated from
canine colon. Proc Natl Acad Sci USA 1993;90:2087–91.
27 Xue C, Pollock J, Schmidt HHHW, et al.Expression of nitric
oxide synthase by interstitial cells of the canine proximal
colon. J Auton Nerv Syst 1994;49:1–14.
28 Ward SM, Burke EP, Sanders KM. Use of Rhodamine 123
to label and lesion interstitial cells of Cajal in canine
colonic circular muscle. Anat Embryol (Berl) 1990;182:
215–24.
29 Lecoin L, Gabella G, Le Douarin N. Origin of the c-kit
positive interstitial cells in the avian bowel. Development
1996;122:725–33.
30 Faussone-Pellegrini MS. CytodiVerentiation of the intersti-
tial cells of Cajal related to the myenteric plexus of mouse
intestinal muscle coat. A EM study from foetal to adult life.
Anat Embryol (Berl) 1985;171:163–9.
31 Matsuda R, Takahashi T, Nakamura S, et al. Expression of
the c-kit protein in human solid tumours and in
corresponding fetal and adult normal tissues. Am J Pathol
1993;142:339–46.
32 Horie K, Fujita J, Takakura K, et al. The expression of c-kit
protein in human adult and fetal tissues. Hum Reprod 1993;
8:1955–62.
33 Prosser CL, Holzwarth MA, Barr L. Immunohistochemistry
of the interstitial cells of Cajal in the rat intestine. J Auton
Nerv Syst 1989;27:17–25.
34 Der-Silaphet T, Malysz J, Hagel S, et al. Interstitial cells of
Cajal direct normal propulsive contractile activity in the
mouse small intestine. Gastroenterology 1998;114:724–36.
35 McLain CR. Amniography studies of the gastrointestinal
motility of the human fetus. Am J Obstet Gynecol 1963;86:
1079–87.
36 Smith B. Changes seen in the myenteric plexus not due to
primary disease. In: Smith B, ed. The neuropathology of the
alimentary tract. London: Edward Arnold, 1972:17–20.
37 Wester T, O’Briain DS, Puri P. NADPH diaphorase
containing nerve fibers and neurons in the myenteric
plexus are resistant to postmortem changes. Arch Pathol
Lab Med 1998;122:461–6.
38 Knuden LM, Pallesen G. The preservation and loss of vari-
ous nonhaematopoietic antigens in human postmortem tis-
sues as demonstrated by monoclonal antibody immunohis-
tological staining. Histopathology 1986;10:1007–14.
Development of interstitial cells of Cajal 71
... A lthough it is now more than 100 years since Cajal described small fusiform cells with prominent nuclei as forming a network in the gastrointestinal tract, many questions about these cells remain unanswered. 1 In the human bowel, the interstitial cells of Cajal (ICCs) are localized at the level of the myenteric plexus between the longitudinal and circular muscle layers, in the deep muscular plexus in the innermost part of the circular muscle layer, and within the circular muscle layer itself. 2,3 Morphologic studies have suggested 3 major functions for ICCs: (1) as pacemaker cells in the muscles of the gastrointestinal tract, (2) as facilitators of active propagation of electrical events, and (3) as mediators of neurotransmission. 4,5 Recent reports indicate that transmembrane tyrosine-kinase receptor c-kit is essential for the development and function of the ICCs. ...
... 4,5 Recent reports indicate that transmembrane tyrosine-kinase receptor c-kit is essential for the development and function of the ICCs. 3,6 The immunoreactivity of c-Kit is present in various cell types, but in the gut, c-Kit is expressed only in ICCs and mast cells. 4 Interstitial cells of Cajal appear to play a key role in the normal function and development of intestine. ...
... ings. 1,3,4 The 3-dimensional network formed by c-Kit-positive cells was located between the circular and longitudinal muscle layers of the bowel wall, at the innermost part of the circular muscle layer, and within the circular muscle layer. The whole-mount preparation elegantly demonstrated nitrergic neuronal network in the myenteric plexus as a mesh of nerve fibers with ganglia, containing clusters of ganglion cells between the 2 layers of the bowel wall. ...
Article
Context.—c-Kit–positive interstitial cells of Cajal (ICC) appear to play a key role in the normal motility function and development of intestine. Nitric oxide is considered to be the most important messenger of inhibitory nonadrenergic, noncholinergic nerves in the enteric nervous system. Objectives.—The aims of this study were to examine the distribution of nitrergic innervation and ICCs in normal human bowel and to demonstrate interconnections between ICCs and nitrergic nerves and smooth muscle fibers using histochemical and immunohistochemical double-staining methods with a whole-mount preparation technique and confocal laser scanning microscopy. Methods.—Full-thickness small and large bowel specimens were obtained at autopsy from 18 children who died of nongastrointestinal diseases. A whole-mount preparation was performed for all specimens, and double staining was carried out with nicotinamide adenine dinucleotide phosphate (reduced form, NADPH)-diaphorase and c-Kit immunohistochemistry. Double immunofluorohistochemistry with neuronal nitric oxide synthase and c-Kit using confocal laser scanning microscopy was also performed in all specimens. Results.—The whole-mount preparation facilitated 3-dimensional visualization of the meshlike network of NADPH-diaphorase–positive nerve fibers in the myenteric plexus surrounded by a reticular network of c-Kit–positive ICCs. The dense c-Kit–positive cellular network located between longitudinal and circular muscle layers and at the innermost part of circular muscle layer intermingled with the myenteric plexus. Short, fine processes of ICCs made connections with the muscle fibers and c-Kit–positive cells. Conclusions.—The development of double–NADPH-diaphorase histochemistry and c-Kit immunohistochemistry staining technique in a whole-mount preparation provides an easy and useful method for investigating the association between c-Kit–positive cellular network and nitrergic neuronal network in the human bowel wall. The characteristic profiles of the c-Kit–positive cellular network and nitrergic neuronal network and their relationship with the smooth muscle fibers provide a morphologic basis for investigating intestinal motility disorders.
... Normal ENS development has been previously described by immunohistochemistry using, among other markers, S-100 (Fu et al., 2004), CD117 (Wester, Eriksson, Olsson, & Olsen, 1999), and NSE (Tam & Lister, 1986). In experimental animal models, it has been observed that cranial, trunk, and vagal NC migration do not depend on neural tube closure (Elms, Siggers, Napper, Greenfield, & Arkell, 2003). ...
... Some authors have reported that, at 16 WG, CD117-positive cells can be observed in the small intestine and rectum, between the circular and longitudinal muscle layers, where the myenteric plexus is located. These cells are scattered, mostly found around the ganglia, and overall CD117 positivity is weak (Wester et al., 1999). Similar findings have been observed in the ileum at 17 WG (Faussone-Pellegrini, Vannucchi, Alaggio, Strojna, & Midrio, 2007). ...
... At 18 WG, ICC can also be observed in the myenteric plexus; these cells are evenly distributed on the myenteric plexus and CD117 immunoreactivity is stronger. This has been observed in the small intestine and in the rectum (Wester, Eriksson, Olsson, & Olsen, 1999). Conversely, other authors have reported finding no CD117 reactivity on intestine samples of 16 WG fetuses (Matsuda et al., 1993). ...
Article
Background: The enteric nervous system (ENS), a component of the peripheral nervous system in the intestinal walls, regulates motility, secretion, absorption, and blood flow. Neural crest (NC) migration, fundamental for ENS development, may be altered by central nervous system development alterations, such as neural tube defects (NTD). Intestinal innervation anomalies have been correlated to NTD. We aim to describe the ENS on a fetus with NTD and fetuses without congenital defects (FWCD). Cases: Two male and four female FWCD, 18-20 weeks-gestation (WG), and a 25 WG female anencephalic fetus. Samples from the pancreatoduodenal groove, jejunum, cecum, rectum, and appendix were analyzed by immunohistochemistry. Nervous plexuses were marked with Neuron-specific enolase and S-100; enteric glial cells with CD56; neuroendocrine cells with chromogranin and synaptophysin, and interstitial cells of Cajal (ICC) with CD117. Results and conclusion: The anencephalic fetus presented a rudimentary brainstem with a cerebellum. Partial frontal, temporal, and occipital bones were found. A large atrial septal defect, an enlarged kidney with a duplex collecting system and a single adrenal gland were found. NSE, S100, and CD56, showed the presence of the myenteric and submucous plexuses of the ENS; scarce interplexus reactivity may indicate inadequate development. Pancreatic and gut neuroendocrine cells, identified with chromogranin and CD56, showed that the enteroendocrine system is present. Findings on FWCD using these markers are consistent with literature descriptions. Vagal NC migration appears to be unaffected despite the presence of anencephaly, although maturation of the ENS may be altered.
... Several studies have assessed the role of immunohistochemical methods both in investigating aspects related to etiopathogenesis and in improving the histopathological diagnosis of IND-B. These studies [39,46,54,[60][61][62][63][64][65][66][67][68][69][70][71] Table 3. ...
... Positive expression in ganglion cells S100 [60,61,66] Positive expression in nerve fibers in the muscular layers and mucosa Synaptophysin [60,62,63,65,66,70] Positive expression in synaptic vesicles in plexuses and nerve fibers SMA (1a4) [61,71] Positive expression in muscle fibers from muscular layers and muscularis mucosa O'Donnell and Puri [69] evaluated the immunohistochemical expression of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) in patients with IND-B. PTEN is a protein involved in cell proliferation, survival, and migration and has a potential modulatory role in neurogenesis and synaptic plasticity. ...
Article
Full-text available
Intestinal neuronal dysplasia type B (IND-B) is a controversial condition among gastrointestinal neuromuscular disorders. Constipation is its most common clinical manifestation in patients. Despite intense scientific research, there are still knowledge gaps regarding the diagnostic criteria for IND-B in the histopathological analysis of rectal biopsies. The guidelines published in the past three decades have directed diagnostic criteria for quantifying the number of ganglion cells in the nervous plexus of the enteric nervous system. However, it is very complex to distinguish numerically what is pathological from what is normal, mainly because of the difficulty in determining a reliable control group composed of healthy children without intestinal symptoms. Thus, a series of immunohistochemical markers have been proposed to assist in the histopathological analysis of the enteric nervous system. Several of these markers facilitate the identification of other structures of the enteric nervous system, in addition to ganglion cells. These structures may be related to the etiopathogenesis of IND-B and represent new possibilities for the histopathological diagnosis of this disease, providing a view beyond the number of ganglion cells. This review critically discusses the aspects related to the disease definitions and diagnostic criteria of this organic cause of constipation.
... 24 Recent studies investigated the fetal and postnatal differentiation and development of ICCs in the human gastrointestinal tract. 25,26 c-Kit-positive ICCs were present in the stomach from 9.5 weeks' gestation and in the small and large bowel from 12 to 13 weeks' gestation. The distribution of the ICCs varies with gestational age and region in the gastrointestinal tract, and maturation of ICCs continues postnatally. ...
... The distribution of the ICCs varies with gestational age and region in the gastrointestinal tract, and maturation of ICCs continues postnatally. 25,26 With increasing age, dense networks of ICCs are found around the myenteric plexus, and numerous ICC mus s are expressed within the muscle layers and particularly at the innermost part of the circular muscle. Furthermore, the myenteric ICCs showed an increased cell size and increased number of individual cytoplasmic processes during early development. ...
Article
Full-text available
Context.—Constipation or recurrent intestinal dysmotility problems are common after definitive surgical treatment in Hirschsprung disease (HD). c-Kit–positive interstitial cells of Cajal (ICCs) play a key role in the motility function and development of the gastrointestinal tract. Interstitial cells of Cajal that carry the tyrosine kinase receptor (c-Kit) develop as either myenteric ICCs or muscular ICCs under the influence of the kit ligand, which can be provided by neuronal and nonneuronal cells, for example, smooth muscle cells. Objective.—To investigate the distribution of myenteric and muscular ICCs in different parts of the colon in HD. Methods.—Resected bowel specimens from 8 patients with rectosigmoid HD were investigated using combined staining with c-Kit enzyme and fluorescence immunohistochemistry and acetylcholinesterase and nicotinamide adenine dinucleotide phosphate (NADPH) histochemistry in whole-mount preparations and conventional frozen sections. Results.—In the normal bowel, ICCs formed a dense network surrounding the myenteric plexus and at the innermost part of the circular muscle. Myenteric ICCs were absent or sparse in the aganglionic bowel and sparse in the transitional zone. The expression of myenteric ICCs in the ganglionic bowel in HD was reduced compared to that in the normal bowel, and they formed only sparse networks. Muscular ICCs were found in the aganglionic bowel, transitional zone, and normoganglionic bowel of HD in a reduced density compared to the normal bowel. Conclusion.—This study demonstrates altered distribution of ICCs in the entire resected bowel of HD patients. This finding suggests that persistent dysmotility problems after pull-through operation in HD may be due to altered distribution and impaired function of ICCs.
... Neurons in the SMP are first detected at 11-14 PCW. 32,33 The migrating cells in the MP eventually condense into discrete neuronal stripes, which arise in the SI around 14 PCW and in the colon around 21 PCW. 6 Of note, the human ENS has an added level of complexity with myenteric neurons that occupy an additional distinct plane from the neuronal stripes. ...
Article
Full-text available
The enteric nervous system (ENS) controls gastrointestinal (GI) motility, and defects in ENS development underlie pediatric GI motility disorders. In disorders such as Hirschsprung’s disease (HSCR), pediatric intestinal pseudo-obstruction (PIPO), and intestinal neuronal dysplasia type B (INDB), ENS structure is altered with noted decreased neuronal density in HSCR and reports of increased neuronal density in PIPO and INDB. The developmental origin of these structural deficits is not fully understood. Here, we review the current understanding of ENS development and pediatric GI motility disorders incorporating new data on ENS structure. In particular, emerging evidence demonstrates that enteric neurons are patterned into circumferential stripes along the longitudinal axis of the intestine during mouse and human development. This novel understanding of ENS structure proposes new questions about the pathophysiology of pediatric GI motility disorders. If the ENS is organized into stripes, could the observed changes in enteric neuron density in HSCR, PIPO, and INDB represent differences in the distribution of enteric neuronal stripes? We review mechanisms of striped patterning from other biological systems and propose how defects in striped ENS patterning could explain structural deficits observed in pediatric GI motility disorders.
... Visualization of the human fetal ENS using cross sections, which show the ENS within the intestinal walls, have identified structural changes with the emergence of individual ganglia evident at 14PCW 6,11,12 . Structural progression has also been observed in non-neuronal cells of the human ENS with interstitial cells of Cajal, pacemaker cells of the intestine, forming a more mesh-like appearance later in gestation 13 . ...
Article
Full-text available
Immature gastrointestinal motility impedes preterm infant survival. The enteric nervous system controls gastrointestinal motility, yet it is unknown when the human enteric nervous system matures enough to carry out vital functions. Here we demonstrate that the second trimester human fetal enteric nervous system takes on a striped organization akin to the embryonic mouse. Further, we perform ex vivo functional assays of human fetal tissue and find that human fetal gastrointestinal motility matures in a similar progression to embryonic mouse gastrointestinal motility. Together, this provides critical knowledge, which facilitates comparisons with common animal models to advance translational disease investigations and testing of pharmacological agents to enhance gastrointestinal motility in prematurity.
... At later stages, differentiated ICCs can be identified as c-Kit-positive and aSMA-negative, whereas smooth muscles cells are αSMA-positive and c-Kit-negative (Klüppel et al., 1998;Radenkovic et al., 2010). This also highlights a usefulness of anti-c-Kit antibody, and indeed in mammals, commercially available antibodies have widely been used to study ICCs in embryos and adults (Baker et al., 2021;Maeda et al., 1992;Malysz et al., 2017;Mei et al., 2009;Roberts et al., 2010;Torihashi et al., 1999;Torihashi et al., 1995;Torihashi et al., 1997;Ward et al., 1994;Wester et al., 1999). ...
Preprint
The gut peristaltic movement, a wave-like propagation of a local contraction, is important for the transportation and digestion of ingested materials. Among three types of cells, the enteric nervous system (ENS), smooth muscle cells, and interstitial cells of Cajal (ICCs), the ICCs have been thought to act as a pacemaker, and therefore it is important to decipher the cellular functions of ICCs for the understanding of gut peristalsis. c-Kit, a tyrosine kinase receptor, has widely been used as a marker for ICCs. Most studies with ICCs have been conducted in mammals using commercially available anti-c-Kit antibody. Recently, the chicken embryonic gut has emerged as a powerful model to study the gut peristalsis. However, since the anti-c-Kit antibody for mammals does not work for chickens, cellular mechanisms by which ICCs are regulated have largely been unexplored. Here, we report a newly raised polyclonal antibody against the chicken c-Kit protein. The specificity of the antibody was validated by both Western blotting analyses and immunocytochemistry. Co-immunostaining with the new antibody and anti-α smooth muscle actin (αSMA) antibody successfully visualized ICCs in the chicken developing hindgut in the circular muscle- and longitudinal muscle layers: as previously shown in mice, common progenitors of ICCs and smooth muscle cells at early stages were double positive for αSMA and c-Kit, and at later stages, differentiated ICCs and smooth muscle cells exhibited only c-Kit and αSMA, respectively. A novel ICC population was also found that radially extended from the submucosal layer to circular muscle layer. Furthermore, the new antibody delineated individual ICCs in a cleared hindgut. The antibody newly developed in this study will facilitate the study of peristaltic movement in chicken embryos.
... The interstitial cells of Cajal constitute a subset of myofibroblasts that exist in the muscularis propria of the intestine and arise from c-kit-positive stem cells. 21 Therefore, we sought c-kit staining in the myofibroblasts of the lamina propria, hoping to understand the origin of these subepithelial stromal cells. In all the tissue sections studied, c-kit expression was seen only in a few inflammatory cells (mast cells) in the lamina propria. ...
Article
Context.—Myofibroblasts are distinct cells with characteristics of both smooth muscle cells and fibroblasts. Through their ability to secrete cytokines, chemokines, prostaglandins, growth factors, and matrix components, they are thought to play critical roles in inflammation, growth, repair, and neoplasia. Objective.—The goal of this study was to identify the distinct cell populations of the lamina propria of normal colon and colorectal polyps. Design.—We studied the expression of α-smooth muscle actin (αSMA), smooth muscle myosin (SMM), desmin, vimentin, and c-kit by intestinal mesenchymal (stromal) cells in the normal colonic mucosa (n = 5), as well as in hyperplastic polyps (n = 5), sporadic colorectal adenomas (n = 47), and adenomas from patients with familial polyposis (n = 36). Results.—In the normal colonic mucosa, the pericryptal stromal cells were αSMA+, SMM+, desmin−, and vimentin+, defining them as myofibroblasts. In contrast, cells of the muscularis mucosae were αSMA+, SMM+, desmin+, and vimentin−, defining them as smooth muscle cells. α-Smooth muscle actin also highlighted direct connections between the muscularis mucosae and the pericryptal myofibroblasts, and vimentin immunostaining showed a network of connections between the αSMA+ pericryptal myofibroblasts and the αSMA− fibroblasts in the interstitium. In all hyperplastic polyps and adenomatous polyps, the interstitial stromal cells (fibroblasts) now also express αSMA and form a syncytium of αSMA+ networklike connections throughout the lamina propria. Stromal cells of sporadic adenomas demonstrated the same immunohistochemical staining characteristics displayed by adenomas from patients with familial polyposis and by hyperplastic polyps. Conclusions.—These findings indicate that in normal colon, αSMA− fibroblasts are the predominant cell type in the lamina propria. However, the pericryptal (subepithelial) stromal cells are a distinct cell type (αSMA+ myofibroblast) that is immunophenotypically different from muscularis mucosae smooth muscle cells and are connected to the interstitial, nonpericryptal fibroblasts with which they exist as a network throughout the lamina propria of the normal colon. Furthermore, in both hyperplastic and neoplastic polyps, there are changes in nonpericryptal fibroblasts from vimentin+, αSMA−, and SMM− to vimentin+, αSMA+, and SMM+; thus, the interstitial fibroblasts are replaced by myofibroblasts. The factors that cause these changes and the origin of the myofibroblasts need to be determined to clarify the biology of colorectal tumorigenesis.
Article
Full-text available
Interstitial cells of Cajal (ICCs) are pacemaker cells of gastrointestinal motility that generate and transmit electrical slow waves to smooth muscle cells in the gut wall, thus inducing phasic contractions and coordinated peristalsis. Traditionally, tyrosine-protein kinase Kit (c-kit), also known as CD117 or mast/stem cell growth factor receptor, has been used as the primary marker of ICCs in pathology specimens. More recently, the Ca ²⁺ -activated chloride channel, anoctamin-1, has been introduced as a more specific marker of ICCs. Over the years, various gastrointestinal motility disorders have been described in infants and young children in which symptoms of functional bowel obstruction arise from ICC-related neuromuscular dysfunction of the colon and rectum. The current article provides a comprehensive overview of the embryonic origin, distribution, and functions of ICCs, while also illustrating the absence or deficiency of ICCs in pediatric patients with Hirschsprung disease intestinal neuronal dysplasia, isolated hypoganglionosis, internal anal sphincter achalasia, and congenital smooth muscle cell disorders such as megacystis microcolon intestinal hypoperistalsis syndrome.
Article
The gut peristaltic movement, a wave‐like propagation of a local contraction, is important for the transportation and digestion of ingested materials. Among three types of cells, the enteric nervous system (ENS), smooth muscle cells, and interstitial cells of Cajal (ICCs), the ICCs have been thought to act as a pacemaker, and therefore it is important to decipher the cellular functions of ICCs for the understanding of gut peristalsis. c‐Kit, a tyrosine kinase receptor, has widely been used as a marker for ICCs. Most studies with ICCs have been conducted in mammals using commercially available anti‐c‐Kit antibody. Recently, the chicken embryonic gut has emerged as a powerful model to study the gut peristalsis. However, since the anti‐c‐Kit antibody for mammals does not work for chickens, cellular mechanisms by which ICCs are regulated have largely been unexplored. Here, we report a newly raised polyclonal antibody against the chicken c‐Kit protein. The specificity of the antibody was validated by both Western blotting analyses and immunocytochemistry. Co‐immunostaining with the new antibody and anti‐α smooth muscle actin (αSMA) antibody successfully visualized ICCs in the chicken developing hindgut in the circular muscle‐ and longitudinal muscle layers: as previously shown in mice, common progenitors of ICCs and smooth muscle cells at early stages were double positive for αSMA and c‐Kit, and at later stages, differentiated ICCs and smooth muscle cells exhibited only c‐Kit and αSMA, respectively. A novel ICC population was also found that radially extended from the submucosal layer to circular muscle layer. Furthermore, the new antibody delineated individual ICCs in a cleared hindgut. The antibody newly developed in this study will facilitate the study of peristaltic movement in chicken embryos.
Article
Full-text available
A discovery that the protooncogene encoding the receptor tyrosine kinase, c-kit, is allelic with the Dominant white spotting (W) locus establishes that c-kit plays a functional role in the development of three cell lineages, melanocyte, germ cell, and hematopoietic cell which are defective in W mutant mice. Recent analyses of c-kit expression in various tissues of mouse, however, have demonstrated that c-kit is expressed in more diverse tissues which are phenotypically normal in W mutant mice. Thus, whether or not c-kit expressed outside the three known cell lineages plays a functional role is one of the important questions needing answering in order to fully elucidate the role of c-kit in the development of the mouse. Here, we report that some of the cells in smooth muscle layers of developing intestine express c-kit. Blockade of its function for a few days postnatally by an antagonistic anti-c-kit monoclonal antibody (mAb) results in a severe anomaly of gut movement, which in BALB/c mice produces a lethal paralytic ileus. Physiological analysis indicates that the mechanisms required for the autonomic pacing of contraction in an isolated gut segment are defective in the anti-c-kit mAb-treated mice, W/Wv mice and even W/+ mice. These findings suggest that c-kit plays a crucial role in the development of a component of the pacemaker system that is required for the generation of autonomic gut motility.
Article
The role of interstitial cells of Cajal (ICC) is difficult to determine because these cells are not easily identified by light microscopy, and there are no compounds available to specifically lesion ICC. Ultrastructural studies have shown an abundance of mitochondria in ICC. Therefore, we have used rhodamine 123, a fluorescent dye that is specifically accumulated by mitochondria, to identify ICC in canine proximal colon. This technique provided good discrimination between ICC and smooth muscle cells, but enteric neurons were labeled with rhodamine 123. This compound has cytotoxic properties in some cells. Therefore, we treated intact muscle strips with rhodamine 123 while recording intracellular electrical activity from circular muscle cells. Uptake of rhodamine 123 by ICC was associated with an alteration in electrical rhythmicity. These data suggest that rhodamine 123 may be a useful tool for visualizing and perhaps chemically lesioning ICC.
Article
Interstitial cells of Cajal (ICC) of various morphologies have been described in the gastrointestinal (GI) tracts of mammals. Different classes of ICC are likely to have different functional roles. ICC of the mouse GI tract have been shown to express c-kit, a proto-oncogene that codes for a receptor tyrosine kinase. We have studied the distribution of ICC within the guinea pig GI tract using antibodies to c-Kit protein and immunohistochemical techniques. c-Kit-like immunoreactivity revealed at least 6 types of ICC: (1) intramuscular ICC (IC-IM1) that lie within the muscle layers of the esophagus, stomach, and cecum, (2) ICC within the myenteric plexus region (IC-MY1) in the corpus, antrum, small intestine, and colon,(3) ICC that populate the deep muscular plexus of the small intestine (IC-DMP), (4) ICC at the submucosal surface of the circular muscle layer in the colon (IC-SM), (5) stellate ICC that are closely associated with the myenteric plexus (IC-MY2) and orientated toward the longitudinal muscle layer in the colon, and (6) branching intramuscular ICC (IC-IM2) in the proximal colon within the circular and longitudinal muscle layers. c-Kit immunohistochemistry appears to be an excellent and selective technique for labeling ICC of the guinea-pig GI tract. Labeling of these cells at the light-microscopic level provides an opportunity for characterizing the distribution, density, organization, and relationship between ICC and other cell types.
Article
A subpopulation of interstitial cells (ICs) are interposed between nerve terminals and smooth muscle cells in the gastrointestinal tract and may participate in neuromuscular transmission. These cells appear to be targets for NO released from enteric inhibitory nerves and respond to exogenous NO with: (i) an elevation in cGMP levels; (ii) an increase in intracellular Ca2+; (iii) and release of a diffusible substance that has tentatively been identified as NO. For the latter to be possible, ICs must express a constitutive isoform of NOS. This study characterized the expression of NOS-like immunoreactivity (NOS-LI) in ICs of the canine colon using 3 antibodies raised against the 2 known constitutive forms of NOS (i.e., neural (nNOS) and endothelial (eNOS) isoforms). Antibodies raised against eNOS and an antibody raised against rat cerebellar nNOS labeled ICs along the submucosal surface of the circular muscle layer (IC-SM), along the surface of septa that separate the circular muscle into fiber bundles (IC-SM), and in the myenteric region between the circular and longitudinal muscle layers (IC-MY). Another antibody raised against rat cerebellar nNOS failed to label ICs. Cultured IC-SM also expressed NOS-LI, suggesting that this feature of the IC phenotype survives culture conditions. Arteriolar endothelial cells in the canine colon were labeled with the same 2 antibodies that labeled ICs, suggesting there are significant structural similarities between NO synthases in ICs and endothelial cells. The data suggest that IC-SM and IC-MY express a constitutive form of NOS. Synthesis of NO by ICs may influence electrical rhythmicity and may serve to amplify and even propagate enteric inhibitory neurotransmission.
Article
Interstitial cells of Cajal (ICC) have been linked to the generation of intestinal pacemaker activity, but their role in in vivo motor dysfunction is unclear. In this study, we investigated the hypothesis that ICC play a role in the generation of distention-induced peristalsis using W/Wv mice that lack ICC associated with Auerbach's plexus. Radiological observations were made of the movement of contrast fluid through the proximal small intestine. Electrical activities were recorded in the external muscle layers. In addition, intraluminal pressure changes were recorded in isolated intestinal segments. In control mice, after gavage of 0.5 mL of barium sulfate in the stomach, the contrast fluid moved through the proximal small intestine in peristaltic waves at approximately 47 times a minute, propagating aborally at approximately 2 cm/s. Electrical slow waves and intraluminal pressure waves were synchronized at similar frequencies and propagation velocities. In W/Wv mice, such regular peristaltic waves were not observed. Action potentials and contractions appeared random, and contents moved back and forth in an irregular manner. The net propulsive effect of contractile activity in W/Wv mutant mice was much weaker than that in controls. Slow wave controlled peristalsis occurs in the normal proximal small intestine upon gastric emptying of a semiliquid. This motor pattern is absent in W/Wv mice that lack ICC.
Article
The c-kit proto-oncogene, the gene at the mouse W developmental locus, is one of a substantial group of genes that appear to encode cell surface receptors but for which the ligands are unknown. We have characterized the kit ligand by a generally applicable approach: the receptor extracellular domain was genetically fused to placental alkaline phosphatase, producing a soluble receptor affinity reagent with an enzyme tag that could be easily and sensitively traced. This fusion protein, APtag-KIT, was used to demonstrate a specific binding interaction (KD = 3 x 10(-8) M) with a ligand on 3T3 fibroblast lines. In situ staining showed labeling over the whole surface of the 3T3 cells, but not extending to adjacent nonexpressing cells. These findings provide direct molecular evidence that the kit ligand can exist as a cell surface protein. Binding was not detected on 3T3 fibroblasts carrying the steel (Sl) mutation, confirming the biological significance of the binding activity and demonstrating that mutations at the Sl locus affect the expression or structure of the kit ligand.
Article
We report the purification and N-terminal amino acid sequence of a novel mast cell growth factor, termed MGF, from the supernatants of a murine stromal cell line. A panel of interleukin 3-dependent cell lines were screened for responsiveness to partially purified MGF in [3H]thymidine incorporation assays; proliferative stimulation of these cells in response to MGF correlated with expression of mRNA for the c-kit protooncogene. MGF was shown to be a ligand for c-kit by cross-linking 125I-labeled MGF to c-kit-expressing cells with subsequent immunoprecipitation of the complex with antiserum specific for the C-terminus of c-kit. This establishes MGF as a ligand for the c-kit protein.
Article
Evidence showing that interstitial cells of Cajal have important regulatory functions in the gut musculature is accumulating. In the current study, the ultrastructure of the deep muscular plexus and associated interstial cells of Cajal in human small intestine were studied to provide a reference for identification and further physiological or pathological studies. The deep muscular plexus was sandwiched between a thin inner layer of smooth muscle (one to five cells thick) and the bulk of the circular muscle. Interstitial cells of Cajal in this region very much resembled smooth muscle cells (with a continuous basal lamina, caveolae, intermediate filaments, dense bodies, dense bands, and a well-developed subsurface smooth endoplasmic reticulum), but the arrangement of organelles was clearly different, and cisternae of granular endoplasmic reticulum were abundant. Interstitial cells of Cajal were distinguished from fibroblasts or macrophages in the region. They ramified in the inner zone of the outer division of circular muscle, penetrated the inner-most circular layer, and were also found at the submucosal border. They were in close, synapselike contact with nerve terminals of the deep muscular plexus, and only few gap junctions with other interstitial cells of Cajal or with the musculature were observed. Compared with interstitial cells of Cajal from other mammals, those associated with the deep muscular plexus in the human small intestine more closely resemble smooth muscle cells, and their organization appears more diffuse; however, the ultrastructure and organization of interstitial cells of Cajal is compatible with modulatory actions on the circular muscle also in humans.