Вы находитесь на странице: 1из 49

Physiol Rev 94: 859 907, 2014

doi:10.1152/physrev.00037.2013

INTERSTITIAL CELLS: REGULATORS OF SMOOTH


MUSCLE FUNCTION
Kenton M. Sanders, Sean M. Ward, and Sang Don Koh

Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada

Sanders KM, Ward SM, Koh SD. Interstitial Cells: Regulators of Smooth Muscle

L
Function. Physiol Rev 94: 859 907, 2014; doi:10.1152/physrev.00037.2013.
Smooth muscles are complex tissues containing a variety of cells in addition to muscle
cells. Interstitial cells of mesenchymal origin interact with and form electrical connec-
tivity with smooth muscle cells in many organs, and these cells provide important
regulatory functions. For example, in the gastrointestinal tract, interstitial cells of Cajal (ICC) and

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


PDGFR cells have been described, in detail, and represent distinct classes of cells with unique
ultrastructure, molecular phenotypes, and functions. Smooth muscle cells are electrically coupled
to ICC and PDGFR cells, forming an integrated unit called the SIP syncytium. SIP cells express a
variety of receptors and ion channels, and conductance changes in any type of SIP cell affect the
excitability and responses of the syncytium. SIP cells are known to provide pacemaker activity,
propagation pathways for slow waves, transduction of inputs from motor neurons, and mechano-
sensitivity. Loss of interstitial cells has been associated with motor disorders of the gut. Interstitial
cells are also found in a variety of other smooth muscles; however, in most cases, the physiological
and pathophysiological roles for these cells have not been clearly defined. This review describes
structural, functional, and molecular features of interstitial cells and discusses their contributions
in determining the behaviors of smooth muscle tissues.

I. INTRODUCTION 859 dant smooth ER, prominent or even complete basal laminae,
II. ICC IN THE GASTROINTESTINAL TRACT 860 and caveolae (318, 376). Interstitial cells can form gap junc-
III. DEVELOPMENT AND PLASTICITY OF ICC 885 tions with each other and with neighboring smooth muscle
IV. INTERSTITIAL CELLS IN THE URINARY... 890 cells and can generate and conduct electrical signals that reg-
V. INTERSTITIAL CELLS IN THE... 894 ulate smooth muscle excitability. Interstitial cells serve as pace-
VI. SUMMARY AND CONCLUSIONS 895 maker cells, propagation pathways for regenerative electrical
events that cannot be propagated actively by smooth muscle
cells, transducers of inputs from motor neurons, and stretch
I. INTRODUCTION receptors. For historical reviews of the morphology and func-
tions of interstitial cells, the reader is referred to a monograph
Interstitial cells is a morphological term denoting a variety
by Lars Thuneberg (369) that reviews more than 200 morpho-
of cells of differing origins and phenotypes occupying spaces
logical studies of muscle-like or fibroblast-like interstitial cells
within the interstitium between the cells most prominent in
and a previous physiological review of electrical rhythmicity in
defining a given tissue. In smooth muscle tissues fibroblasts,
visceral smooth muscles and role of ICC as pacemakers (326).
mast cells, macrophages, and interstitial cells of Cajal meet this
definition. While mainly considered structural or immune cells
Work on interstitial cells in gastrointestinal (GI) muscles has
by many morphologists, interstitial cells have come into prom-
inence because they drive or contribute to the normal func- dominated this field of investigation because there were im-
tions of smooth muscle organs, and remodeling or loss of these portant experimental opportunities that could be exploited.
cells can lead to a variety of motor disorders. This review Interstitial cells in the tunica muscularis of GI muscles have
describes the physiology of the fibroblast-like classes of inter- distinct morphological features (98, 213, 369), mice with mu-
stitial cells, which can include interstitial cells of Cajal (ICC), tations in the protooncogene Kit have reduced populations of
ICC-like cells, Cajal-like cells, fibroblast-like cells and teleo- ICC in specific regions of the GI tract, and Kit mutants develop
cytes in various anatomical descriptions of smooth muscle striking functional phenotypes (45, 167, 250, 378, 404, 417).
tissues (138, 213, 231, 292, 297, 322, 326, 342, 369, 389). Immunolabeling with antibodies against c-Kit has become a
There is a continuum of morphology in this group of cells, standard means for identification of Cajal-like cells (404) in
with some cells having abundant rough endoplasmic reticu- a variety of organs. However, many studies of tissues outside
lum (ER), no basal lamina, no caveolae, and assuming a mor- the gut have encountered difficulties in labeling a distinct class
phology attributed typically to fibroblasts. Other cells assume of interstitial cells (other than mast cells) with this technique,
a more muscle-like appearance with less rough ER, but abun- and Kit mutants neither lacked the cells suspected to be inter-

0031-9333/14 Copyright 2014 the American Physiological Society 859


SANDERS ET AL.

stitial cells nor displayed functional defects. Thus progress in cle cells. ICC and smooth muscle cells express a variety of gap
understanding the functions of interstitial cells in non-GI mus- junction proteins (68, 136, 337), but little is known about the
cles has been slower. The discussion in this review will begin nature and regulation of the gap junctions between interstitial
by reviewing highlights of research on ICC in the GI tract and cells and smooth muscle cells. Gap junctions facilitate commu-
then focus on progress made on this class of cells in other nication between interstitial cells and smooth muscle cells
smooth muscle organs. Recent progress on a second class of (FIGURE 2). Electrical connectivity between interstitial cells
interstitial cells, known for decades in the morphological liter- and smooth muscle cells in GI muscles causes the tunica mus-
ature as fibroblast-like cells, will also be discussed. These cularis to behave, in fact, as a multicellular electrical syncy-
cells are labeled specifically in several smooth muscles by an- tium. We have referred to this functional structure as the
tibodies against platelet-derived growth factor receptor smooth muscle cell/ICC/PDGFR cell (SIP) syncytium (209,
(PDGFR) (174, 207), and this has provided an important 330; see FIGURE 3). Changes in electrical conductances in one
means of accessing these cells in physiological and genomic type of SIP cell affect the electrical excitability of the other
investigations. coupled cells. Spontaneous pacemaker activity generated by
ICC conducts to smooth muscle cells and drives electrical slow
II. ICC IN THE GASTROINTESTINAL TRACT waves and phasic contractions, and neural inputs to ICC and

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


PDGFR cells can conduct to smooth muscle cells and mod-
ulate contractile behavior. The integrated behavior of SIP cells
A. Structural Features of ICC and PDGFR defines what has been referred to classically as myogenic
Cells and Networks regulation of GI motor function. However, with current
knowledge, the term myogenic is too limited to define a major
An important feature of GI interstitial cells (ICC and division of GI regulation, and we suggest replacement of this
PDGFR cells; see FIGURE 1 and TABLE 1 for clarification of term with SIPgenic to refer to the complex and integrated
the terminologies used to describe these cells and their specific regulatory processes operating beneath the level of neural and
anatomical localizations) is electrical coupling to smooth mus- hormonal regulation. It is now recognized that remodeling or

FIGURE 1. AC: c-Kit ICC-MY in mouse,


monkey, and human gastric antrums, re-
spectively. Note similarities in the struc-
tural organization of ICC in all three spe-
cies. DF: c-Kit ICC-MY (D, green) and
PDGFR-MY (fibroblast-like) cells (E, red)
are distinct populations of cells. These pan-
els show cells from murine colon. Merged
images from D and E are shown in F. GI:
intramuscular ICC (ICC-IM; G, c-Kit is green)
in the monkey gastric fundus. ICC-IM form
close associations with enteric motor nerve
processes (nNOS motor neuron processes
are shown in H, red). I: merged image of G
and H. Scale bar in F applies to DF, and
scale bar in I applies to GI. (Authors are
grateful to Dr. Masaaki Kurahashi for im-
ages in DF and Dr. Peter Blair for images in
B and GI.)

860 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

Table 1. Nomenclature and localizations of interstitial cells in the tunica muscularis of the GI tract
PDGFR- PDGFR-
Organ Subregion ICC-MY ICC-IM ICC-SEP ICC-SM ICC-SS MY IM Reference Nos.

Esophagus Skeletal muscle ? ? ? ? 316


Smooth muscle ? ? ? ? ? 73
Lower esophageal ? ? ? ? 408, 432
sphincter
Stomach Fundus CM * 35, 36, 45
LM
Corpus CM * 44
LM *
Antrum CM * 35, 36, 44, 82, 174,
LM * 265, 279, 305
CM *

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


Pylorus ? ? 234, 392, 396, 408
LM
Small intestine Duodenum DMP * ? ? 19, 298
CM *
LM
Jejunum DMP * 36, 167, 174, 317, 376,
CM * 378, 387, 404
LM
Ileum DMP * 44, 100, 113
CM *
LM
Ileo-cecal CM ? ? ? 266
sphincter LM
Colon Proximal CM * 29, 36, 44, 99, 174, 182,
LM 220, 243, 319, 323,
343, 375, 387
Distal CM * 220
LM
Internal anal * ? 61, 62
sphincter

ICC and PDGFR cells are as observed by c-Kit and PDGFR immunoreactivity, respectively. Presence and
relative abundance are denoted by or , respectively. References are representative examples, not an
exhaustive listing; see text for further details. ICC-MY are from a network of cells between the circular and
longitudinal muscle layer and have a pacemaker role in stomach, small bowel, and colon. ICC-MY have been
called ICC-AP or ICC-MP by some authors (see text); however, these terms are misleading because ICC-MY are
not physically within or a component of the myenteric (Auerbachs) plexus. ICC-IM are spindle-shaped intra-
muscular cells lying in muscle bundles between muscle cells and in close apposition to varicose processes of
enteric motor neurons. ICC-IM have also been called ICC-CM or ICC-LM to designate the muscle layer in which
the ICC-IM were observed. ICC-IM are referred to as ICC-DMP in small intestine due to the clustering of these
cells among the nerve process of the deep muscular plexus. Like ICC-IM, the ICC-DMP are closely associated
with varicosities of enteric motor neurons. ICC-SM lie along the submucosal surface of the circular muscle
layer. These have a pacemaker function in the colon. ICC-SS lie at the subserosa of the colon. *Species-
dependent distributions: presence, absence, or low abundance depending on species. Generally found in
larger animals including humans but not in lab rodents.

loss of any component of the SIP syncytium or loss of connec- ascertain the specific functions of each cellular component.
tivity between SIP cells can lead to abnormal motor behaviors Dispersion of the tunica muscularis with proteolytic enzymes
in GI organs. yields a mixture of cells. Smooth muscle cells are abundant and
relatively easy to identify by their shapes, but interstitial cells
While the activity of the SIP syncytium is highly integrated in are more rare and difficult to identify with certainty (typically
GI organs, knowledge about the functions of SIP cells has amounting to 10% of cells within a given volume of muscle)
benefitted from studies of specific cell types. Electrical coupling and display morphologies that overlap with other cells in the
makes it very difficult to deduce the specific functions of one dispersion. Cultures derived from explants or cells dispersed
component in intact tissues. Even with mutant animals lacking from smooth muscle tissues contain a variety of cells, and if the
specific elements of the SIP syncytium, it is still difficult to cells contact each other, gap junctions can form. As in tissues,

Physiol Rev VOL 94 JULY 2014 www.prv.org 861


SANDERS ET AL.

FIGURE 2. Close connections between nerve


varicosity and ICC (C1) and gap junction with
smooth muscle cell in rat gastric antrum (183).
Mitochondria, rough endoplasmic reticulum,
and caveolae (arrowheads) are plentiful in ICC.
Gap junction exists with adjacent smooth muscle
cell (arrow). Very close contact is formed be-
C1 tween nerve varicosity (N) and the ICC. Scale bar
N is 2 m. Inset: higher magnification of the gap
junction between ICC and smooth muscle cell.
Scale bar in inset is 0.2 m. [From Ishikawa et
2 al. (183), with kind permission from Springer
Science and Business Media.]

it again becomes problematic to maintain voltage control in Close connections have also been described between nerve
voltage-clamp studies or to know which cell has generated a varicosities and smooth muscle cells (73, 264, 397), but

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


particular response. Another serious problem is that ionic con- these junctions appear either to be more rare or, at least,
ductances that define the functions of SIP syncytium cells are lost have been deemphasized or gone unmentioned in many
in cell cultures (436). Therefore, it is difficult to see the value of cell morphological reports. The physical proximity of motor
cultures for studies of the native behaviors of SIP cells. neurons to interstitial cells led to much speculation about
the role of these cells in transducing neural inputs; however,
Genetically engineered mice with cell-specific expression of functional innervation is much more controversial and dif-
fluorescent reporters have become important tools for studies ficult to demonstrate (see sect. IIF).
of SIP cells (221, 311, 419, 436). SIP cells from reporter strains
of animals can be identified unequivocally for electrophysio- ICC have an abundance of mitochondria, moderately well-
logical and imaging studies, and fluorescent activated cell sort- developed Golgi, caveolae, and rough and smooth ER. The
ing (FACS) cells can be used to collect purified populations of perinuclear region is often densely packed with mitochon-
cells for comprehensive sequencing of transcriptomes. Expres- dria and cisternae of ER running along the plasma mem-
sion of inducible Cre recombinase in specific SIP cells has been brane. The spaces between ER and the plasma membrane
accomplished and provides a powerful new technique for fu- create tiny volumes in which localized Ca2 transients may
ture studies of interstitial cells (131, 200). regulate plasma membrane ion channels and other func-
tions of interstitial cells, such as pacemaker activity and
Another suggestive anatomical feature of interstitial cells is responses to neurotransmitters. ICC have caveolae and a
their close associations with nerve varicosities, identified orig- basal lamina in many regions of the gut, but these features
inally by electron microscopy (160, 318, 397). In the case of are typically lacking in PDGFR cells. Microtubules and
ICC, this relationship was noted by Santiago Ramn y Cajal thin and intermediate filaments are observed in ICC, but
(47), and he suggested that ICC were a type of peripheral myosin thick filaments are not often found (320). Another
neuron that connects motor neurons to local effector cells, difference between ICC and PDGFR cells is that the lat-
such as smooth muscle cells. We know now that ICC are not ter are typically less electron dense than ICC within the
neurons and of mesodermal origin (238, 379, 409, 430). Close same thin sections. A great abundance of rough ER is com-
associations between neurons and ICC were subjects of note in monly found in PDGFR cells compared with ICC, and
many classical electron microscopic examinations of these this feature was considered fibroblast-like by electron
cells (73, 98, 179, 318). PDGFR cells are also closely asso- microscopists. As mentioned previously, gap junctions are
ciated with terminals of neurons (36, 221, 434); however, the formed between ICC and PDGFR cells and smooth mus-
very close areas of apposition (20 nm) between nerve vari- cle cells (158, 159). While PDGFR cells tend to display
cosities and ICC described in many species have not been similar ultrastructure features in various organs and in dif-
described for PDGFR cells. Close associations between in- ferent species, ICC display a variety of structural and ultra-
tramuscular ICC (ICC-IM) are relatively easy to identify in structural characteristics between species and organs, par-
transmission electron micrographs in experimental animals ticularly in the abundance of caveolae and completeness
and human GI muscles and have been described as synapse- and prominence of their basal laminae (211). However,
like. Close contacts might indicate innervation of ICC by there are not clear differences in ultrastructure that can be
motor neurons. If neurotransmitters are released into regions extrapolated to all regions. Some investigators have de-
of close association, very high concentrations might be scribed ICC as specialized smooth muscle cells (88, 98, 179,
achieved in the tiny postjunctional volumes, and transduction 376). For example, ICC in the deep muscular plexus of the
of neural inputs may tend to be focused at the sites of close small intestine express enteric actin, an actin isoform as-
contact. High junctional concentrations might also increase sociated with GI smooth muscle cells (334), smooth muscle
the rates of metabolism and/or reuptake of neurotransmitters myosin light chains, and sparse distribution of thick fila-
and metabolites. ments (376). In other studies, ICC are described as more

862 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

ICC-IM
NB
PDGFR

*
SMC *

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


B Innervation of SIP syncytium

Interstitial cells

PDGFR+-IM Kit+ (ICC-IM) Varicosity

Enteric motor neuron

SMC

Gap junctions
FIGURE 3. Structures of the SIP syncytium. A: electron micrograph (originally provided by Professor Teru-
masa Komuro) showing three types of cells in close proximity to nerve bundles (NB) and varicosities of enteric
motor neurons (*): smooth muscle cells, ICC, and PDGFR cells. These cells are electrically coupled (gap
junctions not shown in this image), forming an electrical syncytium known as the SIP syncytium. In some cases,
very close contacts (20 nm; black arrow in inset) can be found between nerve varicosities and ICC or smooth
muscle cells. Scale bars are 0.5 m in main image and 0.2 m in inset. B: depiction of SIP syncytium (with
dotted line showing approximate cut of a thin section such as shown in A). Smooth muscle cells, ICC, and
PDGFR cells are arranged around projections of excitatory and inhibitory enteric motor neurons. [Redrawn
from Sanders et al. (329). Copyright 2010 The Authors. Journal compilation Copyright 2010 The Physiological
Society.]

fibroblast-like (66, 113, 212, 309, 314). Cells more like Ultrastructural characteristics are useful for identification
smooth muscle have more complete basal laminae and of ICC and PDGFR cells, but the technical demands of
prominent caveolae; these features are more sparse or lack- transmission electron microscopy (TEM) make this tech-
ing in cells with a fibroblast-like appearance. nique suboptimal for efficient characterizations of intersti-
tial cell distributions and clinical assessments of the state of
Most studies of ICC have been performed on mammalian interstitial cells in human muscles (where frequently ade-
species and limited primarily to common laboratory ani- quate fixation to preserve identifying features is difficult to
mals and humans. Recent, morphological reports describe achieve).
ICC in the GI tract of zebrafish (14). The morphological
features of these cells are similar in description to their 1. Histological and immunomarkers for ICC
mammalian counterpart; however, caveolae were not ob-
served in either ICC or smooth muscle cells. ICC in ze- For many years interstitial cells were difficult to investigate
brafish can also be identified by antibodies for c-Kit. because these cells represent only a fraction of the total cells

Physiol Rev VOL 94 JULY 2014 www.prv.org 863


SANDERS ET AL.

in the tunica muscularis, and there were no specific labels immunocytochemistry (378). Examples of ICC from mouse,
for these cells. Several histological stains were used to high- monkey, and human gastric antral muscles, labeled with anti-
light ICC, such as methylene blue (366, 369), Champy- bodies to c-Kit, are shown in FIGURE 1, AC. Treatment of
Maillet zinc iodide-osmic acid (59, 206, 321), Golgi (silver newborn mice with neutralizing c-Kit antibodies reduced or
impregnation) staining (47, 206), rhodamine 123 (402), abolished the number of c-Kit immunopositive cells, the num-
and NADH diaphorase (95, 420). None of these techniques ber of cells labeled with methylene blue, and cells with ultra-
was highly specific for interstitial cells, and successful stain- structural features of ICC (378). Additional studies showed
ing was accomplished in only some regions of the gut and in that the cells in the region of the myenteric plexus (i.e., in the
only certain species (315). These limitations led one author space between the circular and longitudinal muscle layers) that
to summarize various histological methods for identifying labeled with c-Kit antibodies (ICC-MY) were reduced in intes-
interstitial cells as capricious (58). For many years, the tinal muscles of W/WV mice (167, 404). Mast cells are also
most reliable method for identification of interstitial cells labeled with c-Kit antibodies. However, few mast cells are
was TEM, and a major obstacle in morphological studies present in the tunica muscularis of mice kept in clean animal
was the inability to confirm that cells stained by histological facilities. Labeling with c-Kit antibodies became the standard
techniques and imagined with light microscopy were the technique for study and identification of ICC, and this tech-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


same cells identified by ultrastructural criteria with TEM. nique has been widely exploited in developmental studies,
The ability to image cells with light and electron microscopy pathological evaluations, and molecular and physiological
was an advantage of the NADH diaphorase technique studies during the past two decades. Much progress in our
(420). A study of the dog small intestine explored the nature knowledge of ICC has occurred due to studies made possible
and abundance of intermediate filaments in ICC within the by the use of c-Kit antibodies. c-Kit labeling was also adopted
deep muscular plexus (ICC-DMP) and discovered that vi- for diagnosis of gastrointestinal stromal tumors (GISTs) after
mentin filaments were predominant (376). Vimentin be- GISTs were found to express c-Kit and may develop from ICC
came the first immunolabel for interstitial cells and was due to gain-of-function mutations (discussed further in sect.
used widely in many organs to identify populations of these IIJ) (149).
cells. However, vimentin may not distinguish between c-
Kit (ICC) and PDGFR cells. Progress on ICC has depended on development or novel
application of technology and reagents (TABLE 2). When
Investigators studying hematopoietic cells and developmental c-Kit antibodies directed at extracellular epitopes were
defects in melanocyte and germ cells in Kit (White spotting; W) found to label ICC, we tried immunolabeling of live cells
mutants (49, 118) observed that cells in a variety of tissues after enzymatic dispersion. However, the proteolytic en-
express Kit. To study the functions of c-Kit more broadly, zymes used to disperse cells appear to damage the extracel-
neutralizing antibodies (ACK2) were administered to neonatal lular epitopes. Thus it became desirable to develop reporter
mice (250). These studies found that a population of cells strains with constitutive expression of fluorescent reporters.
within the small intestine expressed c-Kit-like immunoreactiv- Unfortunately, the cell-specific promoters that drive expres-
ity, and neutralizing c-Kit antibody injections produced severe sion of Kit in ICC are unknown, so it was not possible to
defects in intestinal motility described as paralytic ileus. Closer generate a reporter strain with a simple knock-in strategy.
inspection showed that ileal muscles lacked normal phasic The first strain developed utilized endogenous Kit promot-
contractile activity, but instead displayed clusters of arrhyth- ers with a novel green fluorescent protein, ZsGreen,
mic contractions. Responses to agonists (bradykinin and ace- knocked into the first exon of the Kit gene, creating a null
tylcholine) were also altered. Abnormal contractile patterns Kit allele, W(ZsGreen)(418). Heterozygous [W(ZsGreen)/]
were also observed in ileal muscles of W/WV mice. Major had tiny fluorescent dots in Kit expressing cells. This was an
morphological defects were not apparent in enteric nerves or exciting advance, but the expression of ZsGreen was low in
smooth muscle cells of ACK2-treated mice. Immunolabeling most ICC. Pacemaker ICC expressed only a few green dots
demonstrated c-Kit cells in the region surrounding the myen- per cell. Another approach utilized a similar strategy to
teric plexus and within the circular muscle layer. Loss of exploit the endogenous cell-specific promoter and express a
mechanical rhythmicity and reduction in c-Kit cells af- bright green fluorescent protein (copGFP) as the knock-in
ter treatment with neutralizing antibodies suggested that reporter (311, 436). Cells of Kit(copGFP/) mice were highly
the cells expressing c-Kit might be ICC. However, tech- fluorescent and visible in intact muscles and in cell disper-
nical difficulties, including the inability to utilize the an- sions. It should be noted that cells can even be visualized
tibodies for immunocytochemistry or to identify the im- readily in living Kit(copGFP/) mice using a fluorescence mi-
munolabeled cells as ICC with other histological tech- cro-endoscope inserted into the abdomen (unpublished ob-
niques, made it impossible for these authors to identify servations). The reporter strain and cells dispersed from the
the c-Kit cells as ICC definitively (250). tunica muscularis have been used in many subsequent phys-
iological and molecular studies (discussed in sections be-
Positive identification of c-Kit cells in the GI tract was even- low) and offered, for the first time, an efficient means of
tually revealed by colabeling cells with methylene blue and by isolating and purifying living ICC.

864 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

Table 2. Milestones in interstitial cell research in the GI tract


Year(s) Major Investigation and Advances Key Reference Nos.

Before 1985 Morphological studies of ICC; controversies with staining techniques; 47, 98, 113, 179, 309, 314,
EM considered the gold standard for identifying ICC. 369, 434, 435
19801994 Whole muscle experiments; dissection to remove areas rich in 17, 29, 90, 137, 247, 354,
specific ICC populations; use of selective poisons to destroy ICC 355, 370, 402
function.
1989 Isolation of fresh ICC and identification of cells by morphological 235
features; demonstration of intrinsic electrical rhythmicity in
isolated ICC.
1992present Use of c-Kit mutants and c-Kit neutralization to study physiological 45, 167, 250, 377, 378, 400,
functions of ICC. 403, 404
1994present Recognition of Kit immunohistochemistry as specific means to 44, 45, 167, 378, 404
identify and characterize ICC networks.
1994present Demonstration of the role of ICC in electrical rhythmicity. 44, 45, 82, 125, 167, 279,

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


378, 404
1996present Demonstration of the role of ICC in motor neurotransmission. 22, 24, 45, 178, 324, 400,
408
1996present Association of ICC loss or growth with pathophysiological conditions. 205, 249, 252, 279, 282
1998 Discovery that c-Kit is a marker for gastrointestinal stromal tumors 78, 145, 149, 151, 269
(GIST); GIST may arise from gain-of-function mutation in c-Kit of
ICC.
19982009 Culturing of ICC for more detailed physiological studies. 208, 368, 410
2004present Use of flow cytometry to quantify and FACS to purify immunolabeled 52, 53, 161, 276, 277
ICC.
2004present Following activation of interstitial cells and propagation of pacemaker 146, 241243, 283, 423
activity with digital imaging of Ca2 transients.
2005 Demonstration of the role of ICC as stretch receptors. 214, 417
2008 Isolation of progenitor cells capable of generating ICC. 16, 248
2009 Reporter strains of mice in which freshly dispersed interstitial cells 221, 311, 418, 436
can be isolated and positively identified in mixed populations of
cells.
20072009 Recognition of the importance of Ano1 (encoded by Tmem16a) in 52, 122, 172, 436
ICC and its role in pacemaking.
2009 Identification of PDGFR as a specific marker for fibroblast-like cells. 174, 177
2011 Isolation of PDGFR cells using a reporter strain and physiological 221
studies suggesting their role in inhibitory neurotransmission.
2013 Cell-specific iCRE strains that allow inducible knockdowns of genes/ 131, 200
proteins in interstitial cells.

References are representative examples to help the reader into the literature and not meant to be an
exhaustive listing; see text for further details.

Despite the progress made with c-Kit immunohistochemis- trate the muscle layers or, as in human GI muscles, where
try, the search for molecular reagents to identify and ma- they are often abundant.
nipulate ICC populations goes on. A striking advance oc-
curred when antibodies developed from GIST tumors (dis- 2. Histological and immunomarkers for PDGFR
covered on GIST-1; DOG-1) were found to label GIST cells
tumor cells and ICC (93, 415). It was later discovered that
DOG-1 corresponded to the gene product of ANO1 (aka PDGFR cells were referred to as fibroblasts or fibro-
TMEM16a). A gene array screen of transcripts expressed blast-like cells (FLC) by morphologists (213). While TEM
by small intestinal ICC showed Ano1 to be one of the provided a description of the ultrastructural features of
most highly expressed genes in ICC (52), and the gene FLC and close appositions made by these cells to other
product of Ano1, a chloride channel known as ANO1, is cellular constituents of the muscularis, it was difficult to
expressed robustly and exclusively by ICC throughout get a full appreciation of the extent and distribution of
the GI tract (35, 122, 172). An advantage of labeling ICC these cells throughout the GI tract, the major anatomical
in GI muscles with ANO1 antibodies is that mast cells are niches they occupy, and the relationships of these cells to
not labeled, and therefore, this method may be superior other cells from the limited number of images in the
to c-Kit labeling in conditions in which mast cells infil- published literature. Histological techniques were unable

Physiol Rev VOL 94 JULY 2014 www.prv.org 865


SANDERS ET AL.

to identify FLC specifically, and few studies considered a In addition to the obvious advantages of a robust and selec-
role for these cells in regulating the behavior of the tive immunolabel for morphological studies and patholog-
smooth muscle tissues. ical evaluations of PDGFR cells, a mouse engineered to
express a histone 2B-eGFP fusion protein driven off the
Two immunomarkers were initially found to label FLC. endogenous promoter for PDGFR (135) was found to
The discovery that many GIST express c-Kit (149) was ac- have bright eGFP fluorescence in nuclei of cells immuoreac-
companied by reports that GIST cells and ICC also express tive for PDGFR antibodies (221). Cells with eGFP recapit-
CD34 (312). Others reported that CD34 cells were adja- ulated the distribution of PDGFR cells in the gut with
cent to ICC, but represented a distinct population of cells high fidelity. Constitutive labeling of PDGFR cells
(289, 387, 388). Cells with CD34-like immunoreactivity in opened the door to molecular phenotyping and physiolog-
the human small GI tract were identified as c-Kit FLC. ical studies of these cells (as discussed below).
Thus CD34 became a first immunolabel for FLC, but it was
also recognized that CD34 is not a specific label in the gut B. Notes on Nomenclature for Interstitial
wall, and other mesenchymal cell types express this antigen Cells
(289, 387). The guinea pig GI tract was found to express

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


small-conductance Ca2-activated K channels (SK2 and Interstitial cell nomenclature has been somewhat of a bat-
SK3), and SK3 cells were found to be c-Kit cells in the tleground for many years, and it is not yet resolved. Thus it
myenteric region and within the circular muscle layers of GI is important to provide some background on this topic and
muscles (202). Two additional studies reported labeling of a key for readers to understand the varied terminologies
c-Kit cells with antibodies to SK3 in human and mouse GI found in the literature to describe these cells. Cajal de-
muscles (108, 386). SK2 expression was dominant in scribed cells within villi, in the deep muscular plexus of the
smooth muscle cells, but transcripts of SK3 were also found small intestine, and in the region of the myenteric plexus
in these cells (310). These data are consistent with immu- (Auerbachs plexus in the terminology of the day) that had
nohistochemical findings showing punctate SK3 immuno- fusiform cell bodies and long, thin and branching, axonlike
reactivity in smooth muscle cells (202). Thus labeling with processes. The latter may have been the visual cue to Cajal
antibodies to SK3 is also nonspecific under some circum- suggesting that ICC were a class of primitive neuron. When
stances. Cajals staining techniques were repeated in more recent
studies, it appears that neither glial cells nor fibroblast-like
Significant progress occurred when FLC throughout the GI cells (including ICC) displayed this exact morphology. It
tracts of adult animals were found to express PDGFR has been suggested that the cells Cajal described may have
(174, 177). PDGFR cells were shown to be FLC by im- been a chimeric structure consisting of glial cells or intersti-
munocytochemistry and robust expression of SK3. tial cells and contiguous nerve fibers (206). In spite of a
PDGFR cells are distinct from c-Kit ICC and unaffected possible disparity between the structures described by Cajal
in Kit mutants in which ICC are greatly reduced (FIGURE 1, and the cells now identified as ICC, widespread usage of this

DI). PDGFR cells form networks adjacent to ICC in the term in the literature makes altering this terminology coun-
region of the myenteric plexus and are intertwined with ICC terproductive.
and processes of enteric neurons in muscle layers (174,
221). Similar distributions of PDGFR cells and inter- Electron microscopy and histochemical techniques made it
relationships with ICC and enteric motor neurons have clear that ICC occupied several distinctive niches in the GI
tract. Immunolabeling with c-Kit antibodies (e.g., Refs. 35,
been observed in several mammalian species including hu-
44, 183, 377, 378) and ANO1 antibodies (35, 122, 172)
mans (35, 36, 50, 220, 221). Thus PDGFR antibodies
clarified the distribution of ICC in specific locations and
have become a reliable marker for FLC, and most investi-
showed that ICC occupy most of the same locations in all
gators have adopted the terminology of PDGFR cells to
mammals, including humans. Together with functional
refer to these cells. As more is learned about the phenotype studies (described below), certain populations of ICC, dis-
(chemical coding) of this class of interstitial cells, more spe- tributed within specific planes in the tunica muscularis, can
cific terminology may become advantageous, because be grouped and thus linked by more specific terminology.
PDGFR is expressed by many cells in the body and at We have called ICC within the region between the longitu-
various stages of development. At the present time, specific dinal and circular muscle layers, ICC-MY, while others re-
markers to distinguish cells within the region of the myen- fer to these cells as ICC-AP (while Auerbachs plexus was
teric plexus (PDGFR-MY; FIGURE 1, DF) or within the still in common usage) or ICC-MP (since myenteric plexus
muscle bundles of the circular and longitudinal muscle lay- has come into preferential usage). We favor the term
ers (PDGFR-IM; FIGURE 1, GI) have not been identified. ICC-MY because these cells are not a component of the
Developing specific labels for subclasses of PDGFR cells myenteric plexus (as the terms ICC-AP or ICC-MP might
will allow finer resolution of their phenotypes and functions imply). ICC-MY lie between the muscle layers of the myen-
in the future. teron and surround myenteric ganglia, but they are not

866 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

intrinsic to ganglia or the tertiary plexus. Therefore, a label When it became possible to immunolabel fibroblast-like
implying that they are part of the myenteric plexus is mor- cells with PDGFR antibodies (174), it was recognized that
phologically incorrect. Cells within muscle bundles have this population of interstitial cells occupies most of the same
been referred to as intramuscular ICC or ICC-IM. Some anatomical niches as ICC, and therefore, similar terminol-
authors have used specific terms for ICC-IM within the ogy was proposed to designate these cells: 1) PDGFR
circular muscle (ICC-CM) or longitudinal muscle (ICC- cells were utilized because identification by chemical coding
LM); however, no distinctive morphological or functional is more specific than the vague term fibroblast-like cells,
data exist to suggest differences between these ICC. A spe- 2) PDGFR-IM was applied to cells within muscle bun-
cialization of ICC-IM is found in the small intestine in dles, and 3) PDGFR-MY was applied to cells in the plane
which ICC are densely distributed and intimately associated of the myenteric plexus between the circular and longitudi-
with neurons of the deep muscular plexus, and therefore nal muscle layers (FIGURE 1, DI; TABLE 1).
these cells are referred to as ICC-DMP. Cells at the inner
aspect of the circular muscle layer in the colon and more
sparsely in this location in the stomach are in contact with C. Role of Interstitial Cells in Pacemaking
the submucosa and therefore called ICC-SM. Cells along and Generation of Electrical Rhythmicity

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


the submucosal surface of the circular muscle layer are
sometimes referred to as ICC-SMP, but these cells are dis- In many, but not all, regions of the GI tract, motor patterns
tinct and well removed from the submucous plexus, so this are intrinsically phasic in nature. Spontaneous pacemaker
designation is misleading. Cells within the septa that sepa- activity (termed, slow waves; FIGURE 4) drives and times the
rate muscle bundles are referred to as ICC-SEP. TABLE 1 phasic contractile behavior. Slow waves occur in continu-
provides localizations of the different classes of ICC ous rhythmic depolarization/repolarization cycles, and dif-
throughout the GI tract and attempts to also summarize the ferent regions of the bowel display distinct intrinsic slow
alternative terminologies that have been used in the litera- wave frequencies (FIGURE 4, AC). Slow waves also vary in
ture. duration from one to several seconds. Interslow wave trans-

HUMAN MURINE CANINE FIGURE 4. Electrical slow waves recorded from


smooth muscle cells in various regions of the gas-
A Gastric antrum trointestinal (GI) tracts of 3 species. Slow waves
-30
consist of a relatively abrupt upstroke depolarization
followed by a stable or slowly changing plateau
phase. Although frequencies and amplitudes of slow
mV waves vary in different tissues, note the regularity of
duration and frequency in muscles from each region
and species. The activity shown is integrated electri-
-70 cal behavior: a summation of slow waves generated
by ICC, conducted to smooth muscle cells, and re-
1 min sponses of smooth muscle cells. Depolarization of
smooth muscle cells can activate voltage-dependent
B Small intestine Ca2 channels and generate Ca2 action potentials.
0 Ca2 action potentials are elicited in muscles of the
small intestine and colon during periods of enhanced
excitation. Action potentials are not typical in gastric
muscles, except in the terminal portion of the an-
mV
trum and pylorus. Note differences in resting mem-
brane potentials (most negative potentials between
slow waves) in different regions. Regulation of mem-
-55
brane potential sets the excitability of these muscles
10 s 1s 10 s and the response elicited by slow wave activity. Dot-
ted lines were drawn at 40 mV, the approximate
threshold for appreciable activation of L-type Ca2
C Colon currents and excitation-contraction coupling. Note
that slow waves are membrane potential fluctua-
0 tions from negative potentials where open probability
of voltage-dependent Ca2 channels is very low to
potentials where open probabilities increase and
generate Ca2 entry. Thus slow waves are an intrin-
mV
sic mechanism organizing the motor output of GI
muscles into phasic contractions. Propagation of
slow waves in ICC networks generates segmental
-80 and peristaltic contractions. [From Sanders et al.
20 s (331).]

Physiol Rev VOL 94 JULY 2014 www.prv.org 867


SANDERS ET AL.

membrane potentials (typically referred to as resting mem- using a variety of electrophysiological approaches including
brane potential or RMP) of GI smooth muscle cells range voltage clamping via sucrose gap (65, 275, 374). These
between 80 and 40 mV and are generally somewhat studies, and much of the earlier work, were summarized in
negative to the range of potentials required for substantive detail by Professor Tomita (373).
activation of voltage-dependent Ca2 channels. Thus
smooth muscle cells in the SIP syncytium do not generate After development of single-cell voltage-clamp techniques,
Ca2 action potentials at the relatively negative potentials experiments on smooth muscle cells began in several labs,
(RMP) between slow waves. led by Thomas Bolton, Hiroshi Kuriyama, and the team of
Josh Singer and John Walsh (25, 27, 28, 180, 222, 223,
Activation of Ca2 channels provides the switch for excita- 349, 350). None of these studies, or studies performed in
tion-contraction (E-C) coupling in the tunica muscularis. many other labs, recorded intrinsic electrical activity with
Slow waves are oscillations in membrane potential from the characteristics of slow waves from smooth muscle cells
base to peak of 10 60 mV. Muscles with quite negative (96). Smooth muscle cells generated Ca2 action potentials;
resting membrane potentials display larger amplitude slow in some cases these occurred spontaneously, or they could
waves, but these events never overshoot 0 mV. When slow be elicited by depolarization (26). In one study action po-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


waves are recorded from smooth muscle cells, they rise from tentials with plateau components were recorded from co-
RMP at a peak velocity of 1 V/s during the upstroke lonic muscle cells of the dog (293); however, these events
depolarization and then dwell in a plateau phase for vari- were blocked by dihydropyridines, making them dissimilar
able durations. At completion of the plateau phase, the cells to the electrical slow waves of the canine colon (411). The
repolarize to RMP. general conclusion from a multitude of studies was that
smooth muscle cells do not have the ionic apparatus re-
The primary purpose of slow waves is to depolarize smooth quired to generate slow waves or to be the pacemaker cells
muscle cells sufficiently to activate Ca2 influx. Like vascu- in GI muscles. Thus the term myogenic is incorrect. Another
lar muscles, smooth muscle cells of the GI tract express cell type in GI muscles, ICC, was suggested by morpholo-
voltage-dependent (L-type) Ca2 channels (86, 191, 236, gists (98, 369) to be the pacemaker cells because these cells
263). During slow wave depolarizations, the open proba- are electrically coupled to smooth muscle cells. However,
bility of smooth muscle Ca2 channels increases and suffi- physiological verification of this hypothesis was needed.
cient Ca2 entry occurs to trigger excitation-contraction
coupling (281, 391). The rhythmic pattern of slow waves Dissection experiments performed on several regions of the
naturally organizes the contractile activity of GI muscles GI tract suggested that discrete regions of tissue provide
into phasic contractions. In the corpus and antrum of the pacemaker activity for the bulk of the circular and longitu-
stomach, slow wave depolarization alone initiates peristal- dinal muscle layers. Prosser and colleagues (64, 65) noted
tic contractions, and modulation of slow wave amplitude that separation of circular and longitudinal muscles of the
and duration regulates the force of contractions. Depolar- small intestine left circular muscles inactive, but the longi-
ization for several seconds during the plateau phase of slow tudinal muscle layer retained pacemaker activity. They sug-
waves is sufficient for enough Ca2 entry to initiate con- gested that slow waves originate in longitudinal muscles
tractions. In the pyloric sphincter, small bowel, and colon, and are amplified and propagated by circular muscles. Later
slow waves bring membrane potentials of smooth muscle studies, in which specific strata of the circular and longitu-
cells to a threshold for Ca2 action potentials, and trans- dinal muscle layers were isolated, found that slow waves
membrane potential recordings from muscles of these or- were generated in the region between the circular and lon-
gans typically show one or more Ca2 action potentials gitudinal muscle in the stomach and small intestine (17,
superimposed upon the peaks of slow waves (92). In these 137, 185). Similar studies on colon muscles isolated two
regions Ca2 action potentials are the smooth muscle cell pacemaker regions in the dog: the region between the cir-
response to slow wave depolarizations. cular and longitudinal muscle generated a 17 cycle/min elec-
trical rhythm and a thin layer of tissue at the submucosal
The source and mechanism of slow waves in the GI tract surface of the circular muscle layer generated a 6 cycle/min
have been under investigation for decades. Slow waves were rhythm (354, 355). Pacemaker activity extended into the
considered to be nonneurogenic and referred to in the liter- circular muscle layer along the septa that separated the
ature typically as myogenic in origin because blocking circular muscle into bundles (412). ICC networks popu-
nerve action potentials with tetrodotoxin did not abolish lated all of the specialized pacemaker regions (29, 30, 412).
slow wave activity. For many years it was assumed that The next tasks were to disrupt ICC networks and determine
smooth muscle cells generated the spontaneous slow wave effects on slow waves and to isolate ICC to determine
activity. The literature before the mid 1980s contained whether these cells were capable of pacemaker activity.
many studies designed to investigate the mechanism of slow
waves. Some of the more detailed experiments came from Methylene blue can be taken up by ICC in living tissues (a
the laboratories of C. Ladd Prosser and Tadao Tomita, supravital dye), and the cells are damaged after exposure to

868 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

high-intensity illumination (370). Damage to ICC by meth- neous depolarization with characteristics similar to slow
ylene blue exposure was associated with inhibition of slow waves in colonic muscles (235). This was the first direct
wave activity. Methylene blue also has pharmacological evidence that ICC of GI muscles have intrinsic pacemaker
effects on a variety of cells and causes, among other effects, capability.
significant depolarization of smooth muscle cells (328) and
inhibition of nitric oxide synthase (416). Sufficient depolar- 1. ICC as pacemakers in the small intestine
ization (e.g., with elevated external K to the level pro-
duced by methylene blue) blocked slow waves (328). Thus Erratic contractions of intestinal muscles from mice treated
the link between methylene blue labeling of ICC and the with neutralizing c-Kit antibodies and in W/WV mutants
hypothesized pacemaker role of ICC was not clarified by suggested defects in electrical pacemaker activity (250).
experiments using methylene blue (370). Another supravi- Further examination of the intestinal muscles of animals
tal dye, rhodamine 123, accumulates in mitochondria, and treated with c-Kit antibodies showed that ICC-MY, the
ICC were considered to be a potential target for this dye network of ICC in the region identified as the pacemaker
because of their abundance of mitochondria. Rhodamine area in small intestine, were largely missing (378). Loss of
123 is cytotoxic in some cells, so the effect of this dye on ICC correlated with the absence of slow wave activity in

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


pacemaker activity was tested. Rhodamine 123 labeled ICC animals treated with c-Kit antibodies. Small intestinal mus-
of the canine colon and blocked slow wave activity without cles of W/WV animals also lacked most ICC-MY. Electrical
the problem of substantial depolarization as observed with recordings from these mice showed that normal slow wave
methylene blue (402). However, all cells have mitochon- activity occurred in wild-type and heterozygotes, but were
dria, so cytotoxic effects in cells besides ICC may have been absent in small intestinal muscles of W/WV mice (FIGURE 5)
responsible for the effects of rhodamine 123 on pacemaker (167, 404). Additional studies utilizing mutants, such as
activity. Sl/Sld mice, in which the ligand for c-Kit (steel or stem cell
factor) is deficient, also displayed loss of ICC-MY and pace-
Dispersion of colonic smooth muscle tissues yielded smooth maker activity (262, 403). Collectively, these studies, ex-
muscle cells and a few cells with an odd morphology. These ploiting several paradigms of ICC loss (e.g., loss of ICC
cells had multiple short processes and a prominent nuclear after development and developmental failure), established
region. Ultrastructural examination of the cells with pro- the role of ICC as intestinal pacemakers.
cesses showed them to have morphological features com-
mon to ICC (i.e., abundance of mitochondria, caveolae, and Other than the loss of ICC-MY, no other structural or de-
lack of thick filaments). Patch-clamp recordings under cur- velopmental defects were noted in small intestines of c-Kit
rent-clamp conditions showed these cells generated sponta- mutants or animals treated with c-Kit neutralizing antibod-

A B

FIGURE 5. Loss of ICC-MY in small intes-


tine of W/WV mice (404). A: networks of
ICC-MY and ICC-DMP of the small intestine
are shown in this whole-mount labeled with
anti-c-Kit antibodies. B: loss of ICC-MY, but
sparsely labeled ICC-DMP, running parallel
to the circular muscle layer, remain in
W/WV muscles. C: omnipresent slow
wave activity recorded from wild-type mus-
cles. D: loss of slow waves in small intesti-
nal muscles of W/WV mice. Note also the
significant depolarization that occurs in
C D W/WV muscles. Recordings in C and D
-35 were from circular muscle cells.

mV
-55

-65
2s 2s

Physiol Rev VOL 94 JULY 2014 www.prv.org 869


SANDERS ET AL.

ies. It is also important to note that ICC in the region of the nels is reduced, and the intrinsic excitability of smooth mus-
deep muscular plexus (ICC-DMP) appeared normal in cle cells is therefore suppressed between slow waves. Slow
W/WV and Sl/Sld mice. Intestinal motility was altered in waves, superimposed upon the influence of ICC-MY on
c-Kit and stem cell factor mutants, but responses to stimu- membrane potential, organize the generation of muscle ac-
lation of intrinsic enteric motor neurons were retained and tion potentials and phasic contractions into the phasic pat-
smooth muscle tissues responded to neurotransmitters and tern characteristic of the small intestine. The conductances
generated Ca2 action potentials (80, 251, 403). Complete responsible for the negative membrane potentials of ICC
loss of motor activity would prohibit intestinal transit, have not yet been determined.
amounting to a fatal pseudo-obstruction of the bowel.
However, W/WV mice live into adulthood without obvious 2. ICC as gastric pacemakers
loss of nutritional assimilation (56). A normal pattern of
segmentation and peristaltic waves, observed in the small ICC-MY are present in the gastric corpus and antrum of
intestines of wild-type mice, was not present in W/WV mice. W/WV mice, but with application of neutralizing antibod-
Intestinal muscles of these animals displayed action poten- ies, lesions in gastric ICC-MY networks developed after
tials and contractions, but these events were more random 9 40 days in organotypic cultures (279). Slow waves were

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


and less coordinated, leading to weakened propulsive activ- not recorded in regions of muscle where ICC-MY were lost
ity (80). but persisted in areas where ICC networks remained. Elec-
trophysiological recording from guinea pig stomach also
Survival and apparent nutritional sufficiency in W/WV mice supported the hypothesis that ICC-MY are pacemakers in
raises the question of the importance of ICC and slow the stomach (82). When intracellular impalements were
waves. Loss of ICC-MY in W/WV mice resulted in a 10- to made, slow wave activity was recorded from circular
17-mV depolarization in the smooth muscle syncytium smooth muscle cells, positively identified by dye filling dur-
(378, 403, 404). Depolarization alone was not responsible ing recording. More rarely, cells were impaled with much
for loss of slow waves because this level of depolarization, faster and larger amplitude slow wave events (termed driver
as tested with elevated external K, did not block slow potentials by these authors). The cells exhibiting driver po-
waves (378). A second function of ICC-MY, therefore, ap- tentials were between the muscle layers and had morphol-
pears to be setting membrane potential to more negative ogies consistent with cells labeled with c-Kit antibodies.
levels than would be accomplished by the intrinsic ionic Simultaneous recording from ICC and nearby smooth mus-
conductances of smooth muscle cells. Thus ICC-MY in the cle cells showed that the slow waves in ICC preceded the
small intestine shift the membrane potential of the inte- events in smooth muscle cells (FIGURE 6). These investiga-
grated SIP syncytium from a range in which smooth muscle tors also recorded from longitudinal muscle cells, and the
cells generate spontaneous action potentials to more nega- slow waves in these cells were similar in waveform to events
tive potentials in which the open probability of Ca2 chan- in ICC but of reduced amplitude. The slow waves recorded

1
20 mv
2
3 sec

LM

ICC-MY

CM bundle

ICC-IM Enteric motor neuron


FIGURE 6. Propagation of slow waves from ICC-MY to circular smooth muscle cells in the guinea pig stomach
(trace was provided by Professor David Hirst). In the experiment illustrated, an ICC-MY was impaled (electrode
1) and a circular smooth muscle cell was impaled simultaneously. Traces above show slow wave events of large
amplitude in ICC-MY and attenuated amplitude in smooth muscle cell. Note slight delay in activation of slow
wave in smooth muscle cell.

870 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

from longitudinal muscle were termed follower potentials mechanism of slow wave rhythmicity. After a few days in
by these authors. Their results supported the hypothesis primary culture, single cells or networks of cells with
that ICC-MY generated slow waves and these events con- c-Kit immunoreactivity can be identified, and these cells
ducted to both circular and longitudinal muscle layers. generated spontaneous inward currents under voltage-
clamp conditions (208, 368). The inward currents re-
3. ICC as colonic pacemakers
versed 10 20 mV positive to 0 mV, and the currents were
ICC are reduced in the colons of W/WV mice and in genet- not blocked by dihydropyrdines. In current-clamp mode,
ically similar Ws/Ws mutants in rats. However, the loss of spontaneous slow wave-like depolarization transients
ICC in the colon is incomplete, and therefore comparisons were observed that corresponded to the frequency of in-
of activities in wild-type and mutant animals are not as ward currents in voltage-clamp. These events displayed
clear-cut as in other regions of the GI tract. Proximal colons waveforms and mimicked some of the pharmacology of
of wild-type rats and mice display a pattern of slow depo- slow waves in intact muscles; however, the frequency was
larization with superimposed clusters of action potentials lower in cultured cells. Several labs have studied the ionic
(4). Action potential clusters were also observed in colonic conductances expressed and the mechanism of spontane-
muscles of Ws/Ws rats, but these events were irregular and ous electrical rhythmicity in cultured ICC models. The

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


slower in frequency than in wild-type colons. inward current was generally linked to a nonselective
cation conductance; however, one group suggested the
D. Mechanism and Modeling of Pacemaker inward currents were more consistent with a Ca2-acti-
Activity vated Cl conductance on the basis of reversal potentials
and block by SITS (372). Ion channels expressed in cul-
Studies to deduce the slow wave mechanism in whole GI tured c-Kit positive cells and concepts of electrical rhyth-
muscles were frustrated by the inability to record selectively micity were summarized in a previous review (331), and
from ICC. This problem was solved by the elegant work of a mathematical model was developed based on these
groups led by David Hirst and Hikaru Suzuki. As men- ideas (101, 102).
tioned above, Dickens and co-workers (82) recorded from
gastric pacemaker cells and confirmed that the recordings
Problems developed with cultured cell models of ICC. ICC
were made from ICC-MY by filling cells with fluorescent
make up 10% of the cells in the tunica muscularis, so
dyes during recording. Similar recordings of pacemaker ac-
enzymatic dispersion of GI muscles results in a relatively
tivity were made from gastric and small intestinal ICC-MY
small percentage of ICC in the myriad of cells obtained.
(196, 198, 199), and the mechanism of slow waves was
Gene or protein expression studies on these cells are likely
investigated using membrane potential recordings. Experi-
ments using Itpr1/ mice showed that loss of IP3 receptor to be no more specific than expression studies on whole
type 1 blocked gastric slow waves (364). The IP3 receptor muscle extracts. The relative abundance of c-Kit cells after
blocker 2-aminoethoxydiphenyl borate (2-APB), although various periods of time in culture and the effects of growth
not entirely specific, also blocked slow waves in ICC-MY media on retention of c-Kit expression have not been deter-
(196). However, this effect was accompanied by significant mined, so it is unknown whether the c-Kit cell population
depolarization, and sufficient depolarization alone can re- increases or decreases relative to other cells as a function of
duce or block slow wave generation. Treatments with a time in culture. Some investigators have tested growing c-
variety of pharmacological antagonists and ion channel Kit cells on a layer of cells expressing stem cell factor, and
blockers confirmed the general concept that slow waves this promising approach enhanced the number and growth
consist of two components: the initial component (up- of c-Kit cells (308). However, these studies included few
stroke) was sensitive to Ni2 and reduced by extracellular evaluations of the molecular and functional phenotype of
Ca2, and the second (plateau) was sensitive to caffeine, ICC. Most investigations of cultured c-Kit cells have been
cyclopiazonic acid, and Cl channel blockers (155, 196, performed without the benefit of a reporter molecule ex-
198, 199). Reducing extracellular Cl also reduced the pla- pressed in ICC for the purpose of unequivocal identifica-
teau potential. Multiple ionic conductances are at play in tion. Initial labeling with antibodies to confirm the presence
cells of the SIP syncytium. Blocking conductances or Ca2 of c-Kit immunopositive cells (208, 368) is not performed
handling mechanisms in one type of cell or altering ionic on all cells sampled for electrical recording. Cells were se-
gradients might impose effects on ionic mechanisms in cou- lected for recording on the basis of gross morphology. Thus
pled cells. Thus definitive explanations for the pacemaker it is possible that the cells from which molecular evaluations
mechanism were limited until ICC could be isolated for or physiological recordings were made were confused with
single-cell voltage-clamp studies. other cell types in mixed cell populations from which the
1. Studies of pacemaker activity in cultured ICC cultures were grown. Some investigators have performed
purification steps before culturing, such as immunoselec-
c-Kit cells developed in cell culture, and therefore cul- tion or FACS (276); however, tests to certify cell purity have
tured cells were used in many studies to investigate the been rather limited.

Physiol Rev VOL 94 JULY 2014 www.prv.org 871


SANDERS ET AL.

As networks of c-Kit cells develop in culture, they experi- important step in understanding the cellular mechanism of
ence changes in phenotype within a few days. However, few pacemaker activity and a potent indication of the loss of
tests verifying the fidelity of the native phenotype have been function occurring via remodeling of ICC in cell culture.
performed on cultured c-Kit cells. A genomic study of
freshly dispersed ICC from the murine small intestine pro- Production of a reporter strain of mice in which ICC were
duced a catalog of highly expressed genes that could pro- labeled constitutively by expression of a green fluorescent
vide a standard for validation of the ICC phenotype (52). protein (Kit/copGFP) made it possible to identify ICC in
Our own experience with cultured ICC was that the con- mixed cell dispersions of the tunica muscularis and perform
ductances responsible for slow waves in situ were lost experiments soon after enzymatic dispersion (311, 436).
within a few days in primary culture (436). Therefore, we Tmem16a (official name now is Ano1) had previously been
have concluded that the mechanisms of rhythmicity re- identified as one of the most highly expressed genes in ICC
viewed previously (331) may largely be artifacts of cell cul- by a gene array screen of the ICC transcriptome (52), but
ture. This experience should raise caution about conclu- the function of the protein encoded by Tmem16a was un-
sions based on studies of cultured c-Kit cells that are not
known at the time. In 2008 the gene product of Tmem16a
accompanied by careful verification that the mechanisms
was found to be a Ca2-activated Cl channel (Anoctamin

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


and pathways observed are equivalent in the native pheno-
1 or ANO1) (48, 336, 426). Immunohistochemical studies
type. For this reason, little space in this review is devoted to
showed that ANO1 was highly expressed in ICC of several
the many ion channels, agonist responses, and slow wave
mechanisms that have been reported from studies of cul- species, including humans (122, 163, 172).
tured c-Kit cells.
ICC freshly isolated from Kit/copGFP mice display a large-
2. Studies of pacemaker activity in freshly isolated amplitude inward current that was activated by depolariza-
ICC tion and followed a time course similar to the current pre-
viously identified as autonomous current (125). How-
A main concern about the activity recorded from cultured ever, the reversal potential of the current in freshly isolated
ICC-like cells was that they lacked the ability to be paced by ICC indicated that a Cl conductance was responsible for
application of current pulses. This was a fundamental and the large-amplitude inward currents activated by depolar-
important difference between the behavior of cultured cells ization (436). The Cl conductance displayed unusual ac-
and whole muscles, which can be paced (296). Due to skep- tivation properties: a very sharp increase in current was
ticism about the behavior of cultured cells, Akinori Noma noted with depolarizations in the range of 70 and 50
and colleagues (125) sought to study ICC from the myen- mV and a voltage-dependent delay in activation was ob-
teric region (pacemaker region) of the mouse small intes- served at near-threshold potentials. At potentials positive to
tine. These authors developed a unique dispersion tech- the activation threshold, the current developed to maxi-
nique that did not disrupt the anatomical orientation of the mum amplitude and was linearly dependent on the Cl
cells from myenteric plexus region but liberated cells that gradient. The voltage dependence of activation was not fit
could be voltage clamped. ICC-MY, verified by immu- by a Boltzmann function, and therefore, the underlying con-
nolabling of unfixed tissues, had an average whole cell ca- ductance was not likely to be voltage dependent. Activation
pacitance of 25 pF. ICC had resting potentials of 72 mV of the conductance was blocked by Ni2, replacement of
and generated spontaneous transient depolarizations under extracellular Ca2 with Ba2, or exclusion of Ca2 from
current clamp and large inward currents under voltage the extracellular solution. These and other experiments sug-
clamp. The inward currents were termed autonomous cur-
gested that activation of the inward current was due to an
rents, because once these currents were initiated by depo-
initial transient influx of Ca2, and the delay in activation
larization, they displayed an autonomous time course of
with near-threshold depolarizations may have been due to
500 ms. Autonomous currents were attributed to a non-
minimal Ca2 influx at more negative steps. ICC also ex-
selective cation conductance. Autonomous currents dis-
played what appeared to be inactivation properties, but pressed a single-channel, Ca2-activated Cl conductance
inactivation was characteristically different from the of 7.8 pS, which is similar to the conductance of ANO1
Marcovian behavior of voltage-dependent ion channels. It channels expressed in HEK cells (426). ICC currents were
was concluded that activation and deactivation of autono- blocked by niflumic acid with an IC50 of 4.8 M. Under
mous currents depended on intracellular mechanisms that current clamp, ICC discharged spontaneous depolariza-
responded to depolarization and regulated channel open- tions that mimicked the properties of slow waves in intact
ings. It was also shown that replacing the extracellular so- muscles. The spontaneous depolarization events were also
lution with one that was nominally free of Ca2 caused blocked by niflumic acid. As described in the next para-
gradual reduction in the amplitude of autonomous cur- graph, the Ca2-activated Cl conductance appears to be
rents, suggesting that Ca2 stores were an important com- responsible for slow waves in GI muscles, so the current
ponent in activation of autonomous currents. Demonstra- activated in single ICC was referred to as the slow wave
tion of autonomous currents in freshly isolated ICC was an current.

872 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

Experiments on intact muscles suggested that a component activation of a full-amplitude slow wave. STDs are com-
of slow waves was due to a Ca2-activated Cl conduc- monly recorded from cells within intact networks of ICC in
tance (e.g., the plateau phase of slow waves was blocked by situ (155, 196 199, 383) and can be conducted to and
treating tissues with membrane permeable Ca2 buffers, recorded from smooth muscle cells in small strips of muscle
niflumic acid, and by removal of extracellular Cl) (153, (21, 45, 155, 197). STD activity is lost from smooth muscle
198). Cl channel blockers were also found to reduce the recordings when ICC are absent (45). Single ICC also gen-
frequency and block slow waves in murine, monkey, and erate STDs, and these events are due to transient activation
human small intestine and stomach (172). However, it of Ca2-activated Cl currents (436, 437). Stochastic gen-
should be noted that the concentration sensitivity for these eration of STDs in ICC within networks of pacemaker cells
effects varied between organs and species with gastric mus- (155) leads to activation of the voltage-dependent process
cles of monkeys being the most sensitive to niflumic acid. linked to activation of slow wave currents (436). There is
ICC express several splice variants of Ano1 (172), and it is still speculation about the nature of the voltage-dependent
possible that the variable sensitivity to Cl blockers and/or process. Our group has favored voltage-dependent Ca2
the differences in the waveforms of slow waves might be entry, mainly via T-type Ca2 channels, as the voltage-
due to variations in the complement of Ano1 isoforms ex- dependent mechanism (194, 433) because reduced extracel-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


pressed in different regions of the GI tract. The role of luar Ca2, Ni2, and mibefradil can block slow wave gen-
ANO1 channels in slow waves was further tested using eration and slow or block the propagation slow waves in
Tmem16/ (Tmem16atm1Bdh/tm1Bdh) mice (313). Previous intact muscles and generation of slow wave currents in sin-
studies had shown that 80% of mice with congenital de- gle ICC (20, 405, 411, 413, 436). The 1H isoform
activation of ANO1 channels died within a week after birth. (Cacna1h) has been suggested as the T-channels responsible
Experiments were performed, therefore, on newborn ani- for slow wave propagation (120). Others have suggested a
mals and after slow waves developed further in organotypic mechanism involving voltage-dependent activation of IP3
cultures (406). All mice of several litters were assayed for formation or voltage-dependent sensitization of IP3 recep-
electrical slow waves by intracellular recording and then tors (153, 273, 383). More experiments will be needed to
genotyped later for Tmem16atm1Bdh alleles. Newborn
resolve this controversy.
Tmem16a/ mice displayed no slow wave activity while
normal slow wave activity was recorded from mice with one
It is important to note that every type of ICC from which
or both wild-type alleles. After 6 days in organotypic cul-
electrical recordings have been made displays STDs; how-
ture, slow waves developed and increased in amplitude in
ever, only some ICC are capable of generating slow waves
mice with wild-type alleles but were absent in muscles of
(196 199, 436, 437). This suggests that the basic pace-
Tmem16a/ mice. Mice of some litters were allowed to
maker mechanism (i.e., STDs) is an intrinsic feature of ICC;
develop for up to 23 days after birth. No slow waves were
however, some ICC lack the voltage-dependent mecha-
recorded from gastric and small intestinal muscles from
nism(s) required for STDs to summate into slow wave cur-
Tmem16a/ mice, but normal slow waves were recorded
rents or to propagate slow waves actively (see section be-
from siblings with wild-type alleles. These data strongly
low). The simplest explanation for the inability of some
support the hypothesis that Cl channels, encoded by Ano1
(Tmem16a), are responsible for slow waves in GI muscles classes of ICC to generate slow waves is that these cells lack
and justify the term slow wave current to describe the large- the voltage-dependent mechanism(s) responsible for initia-
amplitude inward currents initiated spontaneously and by tion of slow wave currents (see FIGURE 7).
depolarization of single ICC.
Ca2 handling mechanisms that control activation of
3. The clock driving the pacemaker mechanism in ANO1 channels are not well understood at present. It seems
ICC clear that release of Ca2 from Ca2 stores via IP3 receptors
is required for generation of slow waves (364), but sources
Insight into the clock mechanism driving slow waves was of Ca2 for filling of stores, initiation, or amplification of
provided by studies on gastric muscles from the guinea pig Ca2 transients by Ca2 entry or ryanodine receptors, and
(155). The intervals between slow waves were tabulated the role of Na/Ca2 exchange are controversial. The Ca2
and found to vary around a fairly tight mean value, and the sensor that is either intrinsic to ANO1 or provided by an
interval was not dictated by neural inputs to the muscle. accessory protein has also not been identified. Finally, the
The interval between slow waves was tightly linked to the role of excluded volumes that might occur because of close
duration of slow waves, and the probability of initiating the apposition between stores and the plasma membrane are
next slow wave increased from low values immediately af- not understood. At present, our best concept of the slow
ter repolarization of the past slow wave until the next event. wave mechanism is illustrated in FIGURE 7. In spite of many
The increase in excitability was linked to the discharge of uncertain elements, there have been numerous recent at-
small-amplitude spontaneous transient depolarizations tempts at modeling pacemaker behavior in ICC (42, 101,
(STDs, or unitary potentials, as termed by the authors). 102, 384, 428). Simulations of pacemaker activity of ICC
The increase in probability of STDs was associated with are improving, and the models and refinements provided by

Physiol Rev VOL 94 JULY 2014 www.prv.org 873


SANDERS ET AL.

1. Generation of STICs (All ICC)

FIGURE 7. A model of electrical rhyth-


ANO1 micity based on studies of freshly isolated
ICC of the murine small intestine (433,
Plasma membrane 436, 437). Figures show portion of
plasma membrane in close association
Cl Ca2+
with Ca2 release mechanisms of the en-
Ca2+ Excluded doplasmic reticulum (ER). 1: The basic
ATP ADP, Pi volume event of electrical rhythmicity is spontane-
ous transient inward currents (STICs) elic-
RyR IP3R ited by localized release of Ca2 from
stores. IP3-gated channels (IP3R) are the
SERCA main source of Ca2 driving activation of
Ca2+ Ca2+ Ca2-activated Cl channels (ANO1) in the
Ca2+ plasma membrane to generate STICs, but

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


Endoplasmic reticulum ryanodine receptor (RyR) may also contrib-
ute. Ca2 is recovered into stores via ac-
tive Ca2 pumping into the ER. STICs gen-
erate spontaneous transient depolariza-
tions (STDs), the voltage response to the
inward currents. 2: The second phase of
rhythmicity is activation of T-type voltage-
2. Generation of slow wave currents (Pacemaker ICC)
dependent Ca2 channels (VDCC) in re-
Ca2+ Ca2+ sponse to the STDs caused by STICs. Ca2
ANO1 VDCC ANO1 VDCC entry synchronizes Ca2 release in other
regions of membrane, leading to synchro-
nized opening of ANO1 channels through-
out the cell. This generates slow wave cur-
Cl Cl Excluded rents. Depolarization also activates VDCC
Ca2+
volume in adjacent electrically coupled cells, lead-
Ca2+
Ca2+ Ca2+
ATP ADP, Pi
Ca2+
Ca2+ Ca2+
ing to cell-to-cell active propagation of slow
waves in ICC networks. 3: In the 3rd phase
RyR IP3R RyR IP3R of rhythmicity, binding of chronotropic ago-
nists (such as ACh in this example) to G
SERCA protein-coupled receptors (type 3 musca-
rinic receptor, M3, in this example) leads
Ca2+ Ca2+ Ca2+ Ca2+
Ca2+ to IP3 formation and increased frequency
and amplitude of STICs. More frequent
Endoplasmic reticulum STICs and greater amplitude depolariza-
tions increase the likelihood of activation of
whole cell slow wave currents. The steps in
this mechanism are deduced from studies
on ICC of the murine small intestine. Phar-
3. Chronotropic enhancement of pacemaker current (ICC-IM or ICC-MY) macological data suggest that the general-
ized mechanism proposed applies to other
Ca2+ ACh Ca2+ GI muscles (e.g., dependence upon Ca2-
activated Cl currents to generate STICs
ANO1 VDCC M3 ANO1 VDCC and slow wave currents); however, the
mechanism for regulation of frequency is
poorly understood and appears to vary in
Cl Cl ICC from different regions of the GI tract
Ca2+
Ca2+ Ca2+ Excluded and the mechanism for voltage-dependent
ADP, Pi volume Ca2 entry and/or release, necessary for
ATP
Ca2+ IP3 Ca2+ IP3 slow wave propagation, appears to vary.
For example, slow waves recorded from
RyR IP3R RyR IP3R different regions of the murine GI tract and
in other species display a range of sensitiv-
SERCA ities to extracellular Ca2, Ni2, and dihy-
Ca2+ Ca2+ Ca2+ Ca2+ dropyridines. The ionic and molecular ex-
Ca2+ planations for the diversity in the slow wave
Endoplasmic reticulum mechanism and for regulation of frequency
are not fully understood.

874 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

further experimentation will be very useful in planning fu- ing to prove the fidelity of extracellular recording, falls well
ture experiments and in predicting the actions of therapeu- short of a suitable control study for extracellular recording,
tic agents on electrical rhythmicity in the gut. because: 1) dihydropyridines dont block all contractile
movements in many species, 2) no concentration/effect data
for dihydropyridines were provided, 3) movements of the
E. Role of ICC in Propagation of Slow Waves viscera due to respiration and pulsatile blood flow could not
possibly have been absent in a living animal, and 4) more
1. Extracellular electrical recording from visceral rigorous monitoring of movement than visual inspection
smooth muscles would be needed to detect the small movements capable of
eliciting electrical artifacts.
A great deal of the information about GI pacemaker activity
used by gastroenterologists to make diagnoses, biomedical Difficulties in recording authentic electrical activity in GI
scientists in testing hypotheses and drugs, and instructors muscles with extracellular electrodes are likely due to the
for teaching GI physiology has been deduced from studies structure of ICC networks, the fact that ICC capable of
using electrical recordings made with extracellular metal regenerative propagation are buried within muscle layers,

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


electrodes laid against or fastened to the serosal or mucosal and the magnitude and kinetics of transmembrane currents
surfaces of GI organs or applied to the abdomen (i.e., elec- responsible for slow waves. In contrast to the heart, in
trogastrograms). Studies of this sort have been performed which cardiac myocytes actively regenerate action poten-
for almost 100 years; however, a recent report raises ques- tials along a coherent wave front, the gut has a thin and
tions about the validity of results obtained with this method relatively sparse network of ICC generating the transmem-
(18). Activity was recorded from murine gastric sheets in an brane currents responsible for slow waves. The velocity of
attempt to relate electrical activity to the aberrant motility the upstroke phase of slow waves is 100-fold less than the
patterns in mutant mice with dysmotility phenotypes. These upstroke velocity of cardiac action potentials, indicating
studies utilized monopolar or bipolar recording via silver much lower current densities during slow wave depolariza-
electrodes arranged into arrays, as used commonly in many tions. Field potentials resulting from slow waves are far
recent studies (e.g., Refs. 87, 227, 274, 390). Biopotentials weaker than the field potentials generated by cardiac and
detected by these electrodes were monitored by standard skeletal muscles or neurons and may be lost in the noise of
techniques, and it was noted that the frequency of gastric recording.
slow waves was significantly lower than the frequencies
observed when intracellular electrical recording were made 2. Propagation of slow waves in intact muscle strips
from intact gastric sheets (103, 104). When contractions
were blocked by dihydropyridines or wortmannin (both Motility patterns in the gut depend on coordination of mil-
drugs chosen to block contractions downstream of the lions of smooth muscle cells. For example, in gastric motil-
mechanism that generates slow waves), extracellular biopo- ity, slow waves originate in the proximal corpus and peri-
tentials were abolished. Impalement of cells in muscles, staltic contractions spread over many square centimeters of
however, showed that the drugs had no effect on slow wave the stomach. Timing of contractions is accomplished by
amplitude or frequency. By combining imaging of muscle active propagation of slow waves around and along the
movements with extracellular recording, it became appar- tubelike GI organs. GI muscles display clear properties of
ent that very small muscular movements (50 m) were active slow wave propagation, and several studies have
capable of generating voltage transients several tens of mi- characterized propagation using intracellular recordings to
crovolts in amplitude (i.e., equivalent to the events typically determine propagation velocities of slow waves initiated
considered to be slow waves). Thus extracellular electrical from known sites of pacing. These studies have typically
recording from visceral smooth muscles may be largely ar- used muscle strips or sheets from larger animals to obtain
tifacts of contractile movements. In exploring the lengthy cable lengths sufficient to resolve temporal separation of
literature in which extracellular recording has been used to slow waves recorded from two points. In the canine antrum,
monitor the electrical activity of the GI tract, it is apparent for example, slow wave propagation velocity was measured
that controls for movement artifacts have been neglected. at 20 60 mm/s in the circumferential direction (long axis of
Thus it is reasonable to be skeptical about the results of circular muscle fibers) and 10 mm/s in the longitudinal
classic studies that have based major conclusions upon this direction (long axis of longitudinal muscle fibers) (17, 20).
technique. Future investigations using extracellular record- The precise explanation for the anisotropy in propagation
ings will need to demonstrate the validity of these record- velocities is unknown. Slow waves display refractory prop-
ings by control experiments in which recording fidelity is erties, as do excitable events other in excitable cells and cell
maintained after rigorous stabilization of movement. A re- networks. In the canine antrum, the period between slow
cent study claiming maintenance of extracellular recording waves required for complete restoration of slow wave up-
fidelity after infusion of a dihydropyrdine into pig intestine stroke velocity, amplitude, and duration is at least 10 s
in vivo relied only upon visual inspection to ensure that (296), demonstrating the long latencies required for reset-
movement was blocked (10). This study, while clearly seek- ting Ca2 entry/release mechanisms and ion channels re-

Physiol Rev VOL 94 JULY 2014 www.prv.org 875


SANDERS ET AL.

quired for slow wave generation and propagation. In fact, mucosal surface of the circular muscle layer, and active
ion channel recovery from inactivation is categorically propagation of slow waves occurred in this region. Slow
much quicker than the long refractory period of slow wave activity was lost if a thin band of tissue along the
waves, suggesting that other mechanisms, such as reloading submucosal surface (containing pacemaker ICC-SM) was
of intracellular Ca2 stores, is likely to be the rate-limiting removed, and slow waves decayed within a few millimeters
step in slow wave frequency and responsible for long peri- from a region with the pacemaker cell network intact (332).
ods of refractoriness. A similar phenomenon was observed in gastric muscles
treated with neutralizing c-Kit antibodies, slow waves were
ICC have a critical role in slow wave propagation because generated in areas where clusters of ICC were retained, but
these cells provide the pathway for active propagation in GI these events did not propagate to regions devoid of ICC
muscles. Smooth muscle cells lack the ionic apparatus nec- (279). Furthermore, after loss of ICC, gastric muscles could
essary to regenerate slow waves. Thus a coupled network of not be paced by application of current pulses.
ICC is required for coherent propagation of slow waves and
coordination of contractions (FIGURE 8). This point was Recordings of slow waves in ICC and from nearby smooth
demonstrated by dissection experiments in which bits of muscle cells demonstrate how slow waves conduct via elec-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


tissue containing pacemaker cells were removed from mus- trical coupling into smooth muscle cells. Large-amplitude
cles of the canine colon. In intact muscles, slow waves of slow waves were recorded in ICC, and much attenuated
uniform amplitude were recorded from cells along the sub- slow waves were recorded from nearby smooth muscle cells

Single ICC

1.

2. FIGURE 8. Active propagation of slow


waves. 1: The initiating step occurs by lo-
calized Ca2 transients (elevated Ca2 de-
picted by green color in all cells; arrow),
initiating STICs in an ICC. 2: Depolarization
caused by STICs activates Ca2 entry and
ICC network Ca2-induced Ca2 release raising Ca2
throughout the ICC and activating whole cell
slow wave current. 3: Depolarization causes
active propagation of slow waves through the
ICC network (horizontal black arrow shows
direction of propagation), and slow wave cur-
3. rents are activated cell to cell, as the wave-
front spreads. 4: As slow waves propagate
through the ICC network, they conduct pas-
sively into electrically coupled smooth muscle
cells (SMCs). Slow waves depolarize SMCs
and activate L-type Ca2 channels. Ca2 en-
try (green) triggers SMC contraction. Spread
of slow waves in the ICC network leads
ICC network spread of contractions necessary for seg-
coupled to SMCs mental and peristaltic contractions.

876 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

(82, 155, 196, 198, 199). Hirst and colleagues (68) de- elicit depolarization, and initiate contractions; and 4) depo-
scribed the electrical coupling between ICC and smooth larization also initiates active responses in ICC-IM (referred
muscle cells as weak and showed that conduction of slow to as regenerative potentials) which spread rapidly along
waves is similar to conduction of electrotonic potentials the long axis of these cells in circumferential bands (81). In
between these cells. Taken together, these studies suggest order for this mechanism to work, ICC-IM in the corpus
that slow waves, generated by the ICC, conduct passively and antrum must have a voltage-dependent mechanism that
into the smooth muscle syncytium and are not actively re- is primed to provide cell-to-cell regeneration of slow waves.
generated by smooth muscle cells. Thus active (regenera- Cellular studies of ICC-IM have been limited, but a voltage-
tive) propagation of slow waves by ICC-MY is another dependent mechanism intrinsic to ICC-IM has not yet been
central function of these cells. Unlike the heart that is paced identified. Such a mechanism could be present in ICC-IM or
from discrete clusters of pacemaker cells and action poten- in the septal ICC (ICC-SEP) that run between bundles of
tials propagate via the regenerative mechanisms in cardiac smooth muscle cells because small bundles of gastric mus-
myocytes, the gut requires a continuum of ICC for regener- cle, that likely contain both ICC-IM and ICC-SEP, are ca-
ative (active) propagation of slow waves. This feature has pable of regenerative propagation (383).
important consequences in pathophysiological circum-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


stances where loss of some ICC might uncouple regions of There may be significant differences between the ICC-IM in
ICC networks or reduce the safety factor for propagation, different parts of the stomach. In laboratory animals ICC in
thus preventing normal coherent spread of slow waves and fundus muscle bundles do not display voltage-dependent
causing loss of contractile coordination. regenerative mechanisms (21), whereas the ICC within bun-
dles in the distal stomach can generate active responses to
A comment should be included about the form of electrical depolarization or upon break from hyperpolarization (153,
coupling between ICC-MY and smooth muscle cells be- 383). There are clearly many things to learn about the in-
cause some authors have claimed that gap junctions be- teractions between ICC and the specific ionic properties of
tween these cells cannot be found by electron microscopy different classes of ICC before slow wave propagation is
and have proposed alternative methods of electrical cou- fully understood or can be modeled accurately.
pling, including structures called peg-and-socket junctions
3. Ca2 imaging to study mechanism and
(371) or electrical field coupling (360). Whether some form
propagation of pacemaker activity
of ephaptic communication could be effective in conducting
slow waves from ICC-MY to smooth muscle cells seems
Intracellular Ca2 transients are responsible for the spon-
unlikely and has not been demonstrated. Gap junctions be-
taneous transient inward currents (STICs) that generate
tween these cells may be rare and small in size, and these STDs in ICC. Depolarization from STDs triggers global
structures have been observed between ICC-MY and Ca2 transients that activate whole cell slow wave currents
smooth muscle cells in some species (331). Direct electrical (FIGURE 8). Imaging of Ca
2
transients with Ca2 sensitive
measurements have shown that electrical coupling between dyes has been used to characterize propagation of pace-
ICC-MY and smooth muscle cells is weak (68), and this maker activity in networks of ICC in situ (146, 283). ICC
may result from few and small gap junctions. take up Ca2 indicators, such as fluo 4, making it possible
to resolve cyclical Ca2 transients in ICC that correspond to
ICC-IM in corpus and antral muscles also appear to have slow waves, and simultaneous recording of Ca2 transients
regenerative capabilities that can sustain circumferential and slow waves showed 1:1 correlation between these
propagation of slow waves. Hirst and colleagues (152, 154) events. Ca2 transients provide a window and cell-specific
noted that ICC-MY formed a dense network along the detail on pacemaker activity that is not possible with elec-
greater curvature, but this population of ICC was greatly trical recording. Studies using ICC-specific expression of
diminished or missing near the lesser curvature in rodent molecular probes, such as GCaMP, will be a powerful new
stomachs. In spite of the decrease in ICC-MY, slow waves technique for studying slow wave propagation in intact GI
propagated from the greater curvature to the lesser curva- muscles and organs.
ture with little decrease in amplitude (152). These authors
also found that ICC-IM in the corpus were capable of a Ca2 imaging studies on guinea pig and mouse tissues in
relatively fast rate of spontaneous slow wave generation vitro reinforced the concept that slow wave generation is
and suggested that these cells are the source of the higher stochastic in nature (155), because events within a network
frequency slow waves in the corpus. They viewed activation of ICC in sheets of muscle are generated from variable
of the gastric musculature in the following manner: 1) points and propagation within a network occurs via often
ICC-IM generate corporal slow waves, and these events changing pathways (146, 283). Activation of a network of
determine the dominant frequency in the stomach; 2) slow ICC is directed from the first cell to reach threshold, but this
waves from ICC-IM activate slow waves in the ICC-MY position of leadership changes from cycle to cycle. Creation
network, and these events spread down the stomach; 3) of spatiotemporal maps from video images allows visual-
slow waves conduct into bundles of smooth muscle cells, ization of direction and velocity of activity fronts. Ca2

Physiol Rev VOL 94 JULY 2014 www.prv.org 877


SANDERS ET AL.

imaging also reinforced the pacemaker role for ICC-MY showed that electrical coupling between cells was funda-
because Ca2 transients in closely spaced ICC-MY and mental to the organization of propagating Ca2 transients
smooth muscle cells occur sequentially (FIGURE 9), as would (283). Treatment of ICC-MY networks with 18-glycyr-
be expected if slow waves, generated in ICC-MY, initiated rhetinic acid, a gap junction uncoupler, did not block Ca2
depolarization and activation of voltage-dependent Ca2 transients in individual cells, but blocked the spread of tran-
channels in neighboring smooth muscle cells (146, 147, sients through the network. After 18-glycyrrhetinic acid,
283). Average propagation velocities of wave fronts in ICC individual cells paced at their intrinsic frequencies, which
networks in the mouse were between 2 6 mm/s, and there- varied from cell to cell, and failed to be entrained by a
fore consistent with the velocities of slow wave propagation dominant pacemaker frequency in the network. By tabulat-
in murine muscles. The propagation velocity of Ca2 tran- ing the time course of Ca2 transients in multiple cells
sients associated with slow waves greatly exceeds the rates within a network, it was possible to characterize the cellular
of Ca2 waves that result only from diffusion (10 50 m/s) sequence of activation and the time course of cellular tran-
(367), supporting the idea that slow wave propagation de- sients event to event. This analysis showed that the activa-
pends on a voltage-dependent mechanism. tion sequence (i.e., spread of slow waves through a net-
work) varies even when the direction of propagation re-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


The mechanism of propagation was investigated by imag- mains constant. The time lag in activation between adjacent
ing of Ca2 dynamics in ICC networks, and these studies cells also varied from cycle to cycle, likely reflecting the
dynamics of refractoriness and excitability of individual
cells. These properties are consistent with the cablelike
A
properties of a complex two- or three-dimensional network
of excitable cells.

Imaging studies also support the hypothesis that the volt-


LM
age-dependent mechanism for propagation is Ca2 entry
ICC-MY through voltage-dependent, possibly T-type Ca2 channels
(283). Nicardipine did not affect Ca2 transients in ICC-
MY, but greatly reduced or blocked Ca2 transients in
smooth muscle cells. Ni2 (100 M) severely disrupted
0 ms 100 m 64 ms 128 ms 192 ms
Ca2 transients, and mibefradil blocked Ca2 transients
B propagating through ICC networks. Drugs, such as cyclo-
piazonic acid (10 M) and 2-APB (50 M), which affect
intracellular Ca2 handling mechanisms, reduced the fre-
LM quency of ICC-MY Ca2 transients, and ryanodine (10
ICC-MY
M) did not affect frequency but reduced the amplitude of
100 these events. These data are consistent with the idea that
Iu
Ca2 entry entrains the discharge of localized Ca2 release
0.5 s events, generates whole cell Ca2 transients, and facilitates
propagation of slow wave currents in ICC networks.

Ca2+ Transients Imaging has also been used to study pacemaker activity and
C propagation in larger animals and human muscles, and
these studies have allowed generalizations to be made about
Circumferential

100 the role of ICC in pacemaker activity and the mechanism


m
for pacemaking. For example, studies on muscles of the
1s
human jejunum showed Ca2 transients in ICC-MY with
see frames in A characteristics similar to the events in murine muscles
FIGURE 9. Spread of Ca2 transients in the ICC network of murine (241). The Ca2 transients in human ICC-MY were bipha-
small intestine. ICC were loaded with fluo 4. Sequence of video sic in nature, and the initial component was sensitive to
images shows development of Ca2 transient in ICC-MY and spread
Ni2 and mibefradil. Ca2 transients in human jejunal
of wave front from upper left quadrant to whole image. Streaks of
fluorescence are longitudinal smooth muscle cells (LM) that are ICC-MY were not affected by dihydropyridines, but were
above the ICC-MY network. Note that ICC fire Ca2 transients prior reduced or blocked by cyclopiazonic acid and 2-APB. Be-
to events in LM. Ca2 transients in regions of interest in ICC-MY and cause slow waves spread passively (i.e., with decrement)
LM cells are shown as traces as a function of time in the bottom into smooth muscle bundles, electrical events cannot pene-
right of figure. Spatiotemporal maps were constructed from repet-
itive Ca2 transients sweeping through the ICC-MY network. [Re-
trate very far into the depth of thicker muscles of larger
drawn from Hennig et al. (147). Copyright 2010 Blackwell Publish- animals and humans (412). However, ICC lying within the
ing Ltd.] septa (ICC-SEP) between muscle bundles in these tissues

878 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

were found to activate sequentially as a function of distance ICC-MY facilitate coherent propagation of electrical activ-
from ICC-MY in the human small intestine (242). Thus ity which provides organization for muscle depolarization,
propagation of slow waves through ICC-SEP drives depo- Ca2 transients, and contractions (147). When ICC are
larization of smooth muscle cells deep within smooth mus- reduced in number, smooth muscle cells escape from the
cle bundles. discipline of slow waves and revert to intrinsic excitability
mechanisms. After loss of slow waves, smooth muscle cells
There are two pacemaker regions in the canine colon, each generate spontaneous action potentials, and there is little
producing different frequencies of pacemaker events (354). segment-wide regulation of the timing of action potentials.
Interesting differences were observed in the activation and Thus the motor behavior in the absence of ICC-MY differs
propagation of Ca2 transients in ICC in the submucosal considerably from normal motility patterns. Contractions
pacemaker area (ICC-SM), that produce slow waves, and are less circumferential, directional organization of con-
ICC-MY, that produce faster oscillations, called myenteric tractions decreases, and action potentials and contractions
potential oscillations. Rhythmic Ca2 transients (5 8/min) generated from random sites often collide.
spread without decrement through the ICC-SM network,
but Ca2 transients in ICC-MY (16/min) were typically Slow waves can propagate for long distances through ICC

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


unsynchronized in adjacent cells. Excitatory neural inputs networks, providing organization of contractile activity at
synchronized the Ca2 transients in ICC-MY. These find- the organ level. The time constants of the SIP syncytium do
ings demonstrate the diversity of behaviors and pacemaker not favor propagation of action potentials, and therefore,
frequencies found in ICC from different parts of the GI regenerative propagation of action potentials tends to be
tract. Why ICC with different pacemaker frequencies, volt- limited to a small cluster of cells (2). Smaller regions of
age dependencies, and synchronization between cells de- muscle, synchronized only by action potentials, produce
velop in different regions of the GI tract and what factors less organized and weakened contractions observed in small
are responsible for pacemaker diversity are unknown at the intestines of W/WV mice (80). Thus, while muscles and
present time. organs lacking ICC-MY still generate spontaneous electri-
cal activity and contractions, the loss of the rhythmic depo-
4. Relating Ca2 transients in ICC to motor patterns larization-repolarization cycle provided by clock-like slow
of GI organs waves leads to abnormal and less efficient intestinal motil-
ity.
Comparing the activation and propagation of Ca2 tran-
sients in wild-type and W/WV mutants provided a means of
F. Role of Interstitial Cells in
determining the role of ICC-MY in regulating motor pat-
terns in the small intestine (FIGURE 9) (147). In the mouse Neurotransmission
Ca2 transients in ICC-MY precede transients in adjacent
longitudinal muscle fibers by 66 127 ms. The frequency of An early idea proposed by Cajal (47) was that interstitial
Ca2 transients in muscle fibers (30 cpm), variability in the cells are primitive neurons providing connectivity between
intervals between transients (7%), and propagation ve- autonomic nerve fibers and effector tissues. We now know
locities (4 mm/s) in the longitudinal muscle layers of wild- that ICC are not neurons and do not develop from precur-
type mice were similar in flat sheets of tunica muscularis, sors from the neural crest as do the neurons and glia of the
isolated intestinal segments, or exteriorized loops of intes- enteric nervous system (238, 379, 430). However, the idea
tine. In exteriorized loops Ca2 transients were observed that ICC mediate inputs from enteric motor neurons has
around the circumference of the intestine and were con- developed dramatically in recent years. This idea has been a
strained within a band 1.5 mm in length. Ca2 transients controversial topic in ICC research because several investi-
in longitudinal muscle cells preceded longitudinal shorten- gators have reported that contractile responses to nerve
ing and circular contraction by 1 s. Contractions in wild- stimulation are retained in animals lacking some or most of
type mice were stable, most prominent in the longitudinal the ICC thought to mediate neural inputs (ICC-IM). The
axis, and originated at the oral ends of preparations. Force- debate about the role of ICC in neurotransmission was
ful contractions were also observed in muscles of W/WV discussed in recent reviews (126, 329) and is summarized in
mice; however, contractions were abrupt in onset and un- this review. Resolving this controversy is important because
coordinated compared with the contractile patterns of wild- regulating motor input to visceral smooth muscle organs
type mice. The velocity of propagation of Ca2 transients in may provide a means for therapeutic regulation of motor
longitudinal muscles was about six times faster than prop- activity. Understanding the postjunctional mechanisms for
agation in wild-type small intestine, and origination sites of transduction of neural inputs and how these signals are
Ca2 transients were highly unstable. Ca2 transients prop- linked to excitation-contraction coupling are fundamental
agated over smaller areas, and frequent collisions of Ca2 to understanding the reasons for many motor defects in the
transients were apparent. GI tract.

Physiol Rev VOL 94 JULY 2014 www.prv.org 879


SANDERS ET AL.

Some types of ICC (and PDGFR cells, as discussed below) ilar in wild-type and W/WV muscles. The membrane hyper-
are distributed in a manner that would facilitate innervation polarization response to NO donors was greatly reduced in
by motor neurons (FIGURE 1, GI). The cells termed ICC-IM W/WV muscles, but muscle relaxation caused by NO do-
(or ICC-DMP in small intestine) and PDGFR-IM lie along nors was of similar magnitude in wild-type and W/WV mus-
the projections of excitatory and inhibitory motor neurons cles. Thus different mechanisms appear to mediate nitrergic
within bundles of muscle fibers in most regions of the gut. responses in ICC-IM and smooth muscle cells. The impor-
These interstitial cells form gap junctions with smooth mus- tance of one mechanism might be emphasized in wild-type
cle cells. Close associations with varicosities of motor neu- muscles and compensated for by redundant mechanisms in
rons and electrical connectivity with smooth muscle cells muscle cells that develop in the absence of ICC-IM. ICC-IM
are suggestive, but not proof, of a role for these cells in were also greatly reduced in numbers, and inhibitory and
neurotransmission or neuromodulation. Morphologists excitatory junction potentials were reduced in muscles of
noted very close contacts between ICC and varicosities of Steel mutants (Sl/Sld) that lack membrane-bound stem cell
enteric motor neurons with electron microscopy, and Imai- factor (403). Lower esophageal and pyloric sphincter mus-
zumi and Hama (179), for example, suggested that the cles of W/WV mice (408) and small intestinal muscles
interstitial cell may play a role in transmitting stimuli re- treated with neutralizing antibodies to c-Kit (407) also dis-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


ceived from the axon to surrounding smooth muscle cells by played reduced populations of ICC-IM (ICC-DMP in small
an electrotonic response. This comment, and the original intestinal muscles) and reduced postjunctional responses to
idea of Cajal, stimulated further investigation of ICC in stimulation of enteric motor neurons. From these studies, it
neurotransmission. A morphometric study of the lower was reasoned that loss of ICC-IM results in reduced con-
esophageal sphincter performed by Edwin Daniel and Vir- nectivity between enteric motor neurons and smooth mus-
ginia Posey-Daniel revealed an abundance of extremely cle cells, and ICC-IM have an important role in transducing
close contacts (20 nm) between varicosities and ICC and motor neurotransmitters in GI muscles.
far fewer of these contacts between varicosities and smooth
muscle cells (73). These authors suggested that if interca-
2. Close contacts between nerves and effector cells
lation of ICC between motor nerve terminals and smooth
in visceral motor neurotransmission
muscle cells was prevalent, then attempts to define the
characteristics of nonadrenergic, noncholinergic junction
potentials in smooth muscle and compare them with effects There have been many reports about the close connectivity
of putative mediators may be misleading. If the smooth between enteric motor nerve varicosities and ICC (see sect.
muscle junction potential is produced after electrotonic IIA; FIGURES 2 AND 3); however, few quantitative studies
transmission from interstitial cells and its characteristics are on this subject have been provided. Clearly, very close (20
determined by events in the interstitial cells, then there is no nm) connections with varicosities are not exclusive to ICC,
reason to expect coincidence of effects of nerve-released and and a few studies (73, 110, 264) have described similar
exogenously added mediators. The first may act directly on connections between neurons and smooth muscle cells. One
interstitial cells and the second on smooth muscle cells as of these studies, using a morphometric approach, con-
well as on interstitial cells, with the action on smooth mus- cluded that close connections were more prevalent between
cle cells likely to be predominant. This comment turned varicosities and ICC (73). A quantitative understanding of
out to be an important signpost in the course of study of the structural features of motor innervation is important,
enteric motor neurotransmission. because this may provide spatial parameters necessary for
modeling the dynamic profiles of neurotransmitters re-
1. Loss of excitatory and inhibitory leased from motor neurons. Basic questions regarding en-
neurotransmission in muscles of W/WV mice teric motor neurotransmission, such as the nature and mo-
lecular specializations of neural active zones (i.e., neu-
In most regions of the GI tract, ICC-IM are closely associ- rotransmitter release sites), probabilities of vesicle or
ated (20 nm gaps) with varicosities of motor neurons and neurotransmitter release from varicosities per impulse,
form gap junctions with adjacent smooth muscle cells. number of transmitter molecules released upon vesicle fu-
ICC-IM are greatly reduced in numbers in gastric muscles of sion, effects of frequency on types and quantities of neu-
W/WV mice (45). Electrical field stimulation of wild-type rotransmitters released, prejunctional modulation of trans-
muscles evokes excitatory and inhibitory junction poten- mitter release, metabolic and uptake pathways involved in
tials in the fundus, and both nitrergic and cholinergic junc- deactivation of neurotransmitter signals, and dynamics of
tion potentials were reduced significantly in muscles of neurotransmitter concentrations in postjunctional volumes
W/WV mice with reduced ICC-IM (45, 365, 400). Loss of are poorly understood (329). Thus discussions about
these responses was not due to failure of motor neurons to whether volume or synaptic-like neurotransmission
develop and innervate muscle bundles or to release neu- dominate in muscles of the GI tract are speculative at pres-
rotransmitters, and responses were not lost because smooth ent. Even if volume transmission seems likely from rigorous
muscle cells lost sensitivity to ACh and nitric oxide (NO). consideration of morphological details (110), the extent to
Contractile responses to exogenous transmitters were sim- which effective neurotransmitter concentrations reach

880 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

postjunctional receptors depends on the many factors listed trinsic motor neurons, PKC was detected in ICC-DMP.
above. Translocation was verified by showing a shift in PKC from
the soluble to the particulate fractions of extracts of intes-
3. Expression and function of receptors and tinal muscles. Translocation of PKC was also initiated by
effectors of neural responses in interstitial cells phorbol ester, and muscarinic responses and neurally
evoked translocation were blocked by atropine or TTX.
Use of immunohistochemical techniques and isolation of These experiments directly demonstrated innervation of
smooth muscle and interstitial cells has allowed evaluations ICC-DMP by cholinergic neurons.
of the expression of receptors and effects of neurotransmit-
ter responses and comparative responsiveness of specific ICC-DMP also express neurokinin 1 (NK1) receptors (127,
populations of postjunctional cells. In some cases an immu- 178, 357, 361), and binding of agonists to these receptors
nohistochemical approach (i.e., functional immunohisto- on ICC causes internalization (237). NK1 antibodies were
chemistry) can be also useful in determining which cells used to test whether membrane internalization could be
respond to neurotransmitters released from motor neurons elicited by stimulation of enteric motor neurons (178). Ex-
because specific changes in postjunctional signaling pro- citatory motor neurons, which release both ACh and tachy-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


teins (e.g., phosphorylation, generation of second messen- kinins in the GI tract, were shown to be closely associated
gers, and protein translocations) can be determined. with ICC-DMP expressing NK1 receptors. Stimulation of
muscle strips with either exogenous substance P or by elec-
Several studies have shown that soluble guanylate cyclase trical field stimulation led to a shift from diffuse immuno-
(sGC), the physiological receptor for NO, is highly expressed labeling of NK1 receptors around the periphery of cells
in ICC. The inhibitory effects of NO and NO-dependent en-
before stimulation to small granular structures within cells
teric inhibitory neurotransmission in GI muscles are blocked
after stimulation. Internalization of NK1 receptors in re-
by inhibitors of sGC as shown in several studies by the effects
sponse to field stimulation was blocked by a selective NK1
of 1H-(1,2,4)-oxadiazolo-(4,3-)quinoxaline-1-one (ODQ)
antagonist (WIN62577) or TTX.
(106, 164, 429). In the rat small intestine, sGC (-subunit) is
highly expressed in ICC-DMP and colocalized with type 1
Internalization of NK1 receptors was also exploited as a
cGMP-dependent protein kinase (cGK-1) (324). NO syn-
means to separate ICC-DMP from ICC-MY in the small
thase (NOS)-immunoreactive neurons were found adjacent
intestine. Muscles were incubated with substance P labeled
to ICC-DMP. cGK-1 expression was also resolved in
with Oregon green (OG-488) before dispersing cells. Cells
smooth muscle cells, but very low levels of sGC-like immu-
were sorted for c-Kit and then again for Oregon green. Cells
noreactivity were found restricted to the innermost layer of
circular muscle cells. Expression of sGC (- and -subunits with both labels were considered to be ICC-DMP, as con-
probed) is also found in interstitial cells throughout the firmed by fluorescence microscopy. The cells were probed
guinea pig GI tract (175, 176). Both ICC-IM and for numerous genes known to be involved in mediation of
PDGFR cells express these proteins. Expression of postjunctional responses to enteric motor neurotransmit-
sGC- in ICC-IM and PDGFR cells was confirmed by ters. Genes including Chm2 and Chm3 (cholinergic re-
immunocytochemistry. NOS neurons were in close associ- sponses), Tacr1 (neurokinin responses), Gucy1a3 and
ation with cells expressing sGC. As in the rat, smooth mus- Gucy1b3 (nitergic responses), P2ry1 and P2ry4 (purinergic
cle cells show little or no immunoreactivity for sGC. This responses), and Vipr2 (responses to vasoactive intestinal
observation was controlled by clear expression of sGC in peptide) were expressed by ICC-DMP. Further work is nec-
smooth muscle cells of the muscularis mucosae and in blood essary to determine whether receptor expression by intra-
vessels, confirming that the techniques used were capable of muscular ICC in various regions of the GI tract is linked to
detecting sGC in smooth muscle cells. These authors con- ion channels and other effectors that contribute to motor
cluded that sGC expression is low in smooth muscle cells in nerve responses.
the tunica muscularis. Functional immunohistochemical
studies showed that stimulation of nitergic neurons in GI ACh released from motor neurons in the gastric fundus
muscle strips or treatment with NO donors enhanced activates different postjunctional mechanisms than are ac-
cGMP in ICC (348, 431). tivated by muscarinic agonists added to solutions bathing
muscles (33). In this study phosphoproteins involved in
During muscarinic stimulation, PKC isozymes are translo- Ca2 sensitization mechanisms were analyzed before and
cated from the cytoplasm to the membrane to elicit re- after addition of carbachol to bathing solutions or after
sponses. PKC is expressed in the intramuscular ICC of the stimulation of cholinergic motor neurons. Neural release of
murine small intestine (ICC-DMP), and immunohistochem- ACh activated only phosphorylaton of protein kinase C
istry was performed before and after muscarinic stimula- (PKC)-potentiated phosphatase inhibitor protein (CPI-17),
tion of small intestinal muscles (398). The antibody chosen but bath application of carbachol activated phosphoryla-
for these studies did not recognize cytoplasmic PKC, but tion of CPI-17 and myosin phosphatase targeting subunit 1
after exposure to ACh or electrical field stimulation of in- (MYPT1). When acetylcholine esterase was inhibited by

Physiol Rev VOL 94 JULY 2014 www.prv.org 881


SANDERS ET AL.

neostigmine, activation of cholinergic neurons caused phos- resulted in contraction or relaxation under circumstances in
phorylation of CPI-17 and MYPT1. Stimulation of cholin- which relaxation responses were elicited in wild-type mus-
ergic neurons in W/WV mice also caused phosphorylation cles. Tone developed spontaneously in wild-type muscles,
of CPI-17 and MYPT1. These data suggested that ACh but phasic contractions were noted in W mutants. Noncho-
released from neurons is compartmentalized, possibly linergic excitatory responses appeared elevated in Ws/Ws
within the tiny volumes created by the close apposition of rat muscles. Differences in motor patterns and variability in
nerve varicosities and ICC. Due to this compartmentaliza- the dominance of excitatory or inhibitory regulation in dif-
tion, ACh released from neurons does not appear to reach ferent animals indicate remodeling of neuromuscular re-
smooth muscle receptors linked to activation of Rho kinase sponses in animals with mutant W alleles.
and MYPT1 phosphorylation. Inhibiting ACh metabolism
or reducing contacts between ICC and motor neurons and Postjunctional nitrergic responses were also reported to be
the small junctional volumes formed as a result of these normal, or at least preserved, in a study of neuromuscular
contacts appears to enhance the availability of ACh and responses in the lower esophageal sphincter (LES) of W/WV
exposure of the neurotransmitter to smooth muscle recep- mice (432). Some animals displayed nitrergic responses
tors. while muscles of other animals of the same genotype did

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


not. It was also reported that ICC-IM were retained in some
Taken together, there is significant evidence suggesting di- regions of the LES; however, the presence or absence of cells
rect innervation of ICC by enteric motor neurons. Studies labeled with c-Kit antibodies did not appear to correlate
summarized above show spatial proximity of ICC to nerve with nitrergic responses. Although some muscles displayed
varicosities, loss of postjunctional responses in the absence nitrergic components during inhibitory junction potentials,
of ICC, expression of receptors for enteric motor neu- most of the electrical responses displayed in this paper (e.g.,
rotransmitters, translocation of signaling molecules, and see FIGS. 3 6 of Ref. 432) were similar to the responses of
internalization of receptors in ICC upon nerve stimulation. LES muscles from W/WV mice (e.g., reduced nitrergic com-
Of course, parallel neurotransmission to other SIP syncy- ponents of IJPs and absence of membrane STDs) that had
tium cells is quite possible; smooth muscle cells and been described previously (408).
PDGFR cells also lie in close proximity to varicosities of
motor neurons. The involvement of PDGFR cells will be
In other studies, responses monitored by manometry were
discussed below. At this point it seems important to note
compared in the LES of W/WV and nNOS/ mice (352).
that there is little evidence in the literature of direct motor
These authors reasoned that if ICC-IM are required for
innervation of smooth muscle cells before removal of ICC.
nitrergic responses, then pressure responses in the LES of
W/WV and nNOS/ mice should be very similar. How-
4. Studies reporting normal neuromuscular
ever, the motor behaviors of W/WV and nNOS/ mice
responses in W mutants
were different: tone was elevated in the LES of nNOS/
There are reports in the literature demonstrating sustained mice and relaxation responses were abnormal; tone was
responses to nitrergic and cholinergic neurotransmission in greatly depressed in W/WV mice and relaxation responses
W/WV mice and Ws/Ws rats (165, 432). These findings have were considered normal. This study seems to have neglected
led some authors to conclude that ICC are not important or previous reports showing that ICC-IM are involved in me-
necessary for enteric motor neurotransmission. However, a diating responses to excitatory and inhibitory neural inputs
better question to ask might be whether responses to neu- (45, 400). Therefore, one might predict that responses in
rotransmitters and other bioagonists are normal in the W/WV and nNOS/ mice would be quite different. W/WV
absence of ICC. Even though contractile responses to neu- mice would be expected to have reduced LES tone due to
rotransmitters are apparent in the absence of ICC, it is clear reduced cholinergic input. If only nitrergic inputs are com-
that responses differ from wild-type responses. In the case promised, as in nNOS/ mice, LES tone may be elevated
of the gastric fundus, loss of ICC led to recruitment of a and relaxation responses would be greatly decreased. It
Ca2 sensitization mechanism not normally activated by should also be noted that it is problematic to compare the
ACh released from neurons (33), thus increasing the gain on magnitudes of responses in muscles with different levels of
smooth muscle contractile responses. This may help pre- basal tone, and the conclusions that nitrergic responses
serve (or even amplify) cholinergic contractile responses in were normal in the LES and pyloric sphincter of W/WV mice
muscles lacking ICC-IM, but it would also tend to alter were based on such comparisons (351, 352).
responses to all other physiological agonists, such as para-
crine substances and hormones. Such an effect might ad- Lesions in ICC are not homogeneous throughout the bowel
versely affect integrated motor responses in the stomach. in W mutants (45, 404). At present, the relationship be-
tween loss of ICC and loss of motor function has not been
Ws/Ws rat fundus muscles developed significantly less quantified, and in the case of inhibitory regulation of con-
spontaneous tone than wild-type rats, and responses to tractions, the relationship might be very complicated. For
nerve stimulation were variable (165). Nerve stimulation example, regulation of contraction by membrane potential

882 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

is highly nonlinear and might be accomplished in the gut by ylate cyclase (Gucy1b3) was targeted in ICC, smooth mus-
small membrane potential changes. Membrane potentials cle cells or in both types of cells (131). Knocking down
of GI muscles, at rest in tonic muscles or at the peaks of slow soluble guanylate cyclase (sGC) in fundus smooth muscle
waves in phasic muscles, lie near or within the range of cells caused reduction in responses to a bath-applied NO
potentials in which the open probability of L-type Ca2 donor, but responses were not greatly affected with knock-
channels is steeply dependent on voltage. Therefore, small down of sGC in ICC. Responses to NO released from neu-
hyperpolarization responses can greatly reduce the open rons was not greatly affected in mice with cell-specific
probability of Ca2 channels and reduce Ca2 entry to knockdown of sGC in smooth muscle cells or in ICC, but
levels that do not sustain contraction. In fact, typical hyper- knocking down sGC in both cell types blocked most of the
polarization responses in wild-type muscles take membrane nitrergic response in the fundus. These authors attributed
potential far negative to the range in which the open prob- nitrergic responses, therefore, to combined effects on ICC
ability of Ca2 channels is sufficient to elicit contraction. and smooth muscle cells, suggesting parallel neurotransmis-
Therefore, hyperpolarization responses might be greatly at- sion to both cell types.
tenuated before contractile responses are inhibited. Regions
of the gut with reduced ICC or reduced ICC function might Animal models in which specific genes can be inactivated

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


not show motor effects without a substantial reduction in selectively in one type of cell will be powerful tools for
ICC. Studies have been conducted on muscles of W mutants future exploration of the role of interstitial cells in enteric
from regions of the gut with partial reduction in ICC. For motor functions. One must be cautious in interpreting the
example, ICC were reduced but not abolished in colons of responses of iCre-dependent gene inactivation models,
Ws/Ws rats (4). Nitrergic components of IJPs were recorded however, because this approach seldom results in quantita-
from approximately half of cells impaled for electrical re- tive inactivation of floxed alleles, particularly when crosses
cording, and nitrergic responses were reduced or not re- are made with homozygous mice with two floxed alleles in
solved in the remaining cells. Tabulation of ICC at or near the targeted cells (15). In such cases, recombination of both
each impalement was not provided, so it is unknown floxed alleles must occur for knockout of gene transcription
whether loss-of-function correlated with a localized reduc- in a given cell. Leaving one functional allele results in partial
tion in ICC. Other studies of this type were reviewed previ- depletion of gene products, as in heterozygotes. It is well
ously (329). It should also be noted that within a given known that heterozygotes often display functions equiva-
region of muscle (e.g., gastric fundus) there is variation in lent to the wild-type and are frequently used as control
the numbers of c-Kit cells from animal to animal. Thus animals in gene inactivation studies. Assessment of the de-
studies in the future using W mutants should include assess- gree of gene inactivation in the cells targeted for expression
ments of ICC populations in regions from which neural of Cre or iCre is important information to include in future
responses are recorded. studies using this technology.

5. Cell specific inactivation of neurotransmitter


6. PDGFR cells in enteric motor
receptors or effectors
neurotransmission
Two recent reports have utilized the Cre/loxP recombina-
tion approach to target activation or deactivation of specific ICC are not the only cells in the postjunctional environ-
genes in ICC. c-KitCreERT2 mice were generated to target ment that respond to enteric motor neurotransmitters.
inducible activation of iCre recombinase in ICC (200). By PDGFR cells are also closely associated with varicos-
crossing these mice with a strain with conditional expres- ities of motor neurons and intermingled with ICC around
sion of diphtheria toxin A, it was possible to reduce ICC in nerve processes (174, 177, 221). Isolation of colonic
adult mice. Depletion of ICC caused significant distur- PDGFR cells from Pdgfratm11(EGFP)Sor/J, a reporter
bances in GI motility. Membrane potentials of small intes- strain with eGFP expressed in cell nuclei driven off the
tinal muscles were depolarized, slow wave activity was dis- endogenous Pdgfra promoter (135), allowed positive selec-
rupted, and there were significant slowing of gastric emp- tion of cells for molecular and functional studies (13, 221).
tying and intestinal transit. Mice with disrupted ICC Cells from colon and stomach sorted by FACS displayed
exhibited no excitatory or inhibitory junction potentials in high expression of Pdgfra, P2ry1, and Kcnn3 (encoding
the small intestine, suggesting that reducing the number of SK3 channels). Under voltage clamp, PDGFR cells dia-
ICC effectively denervated the smooth muscle. In the colon, lyzed with 500 nM Ca2 displayed robust outward currents
excitatory junction potentials were greatly reduced by loss that reversed at approximately EK. The outward currents
of ICC, but effects on inhibitory junction potentials were were blocked by apamin. Held at physiological resting po-
less affected. Targeted deletion of Prkg1, which encodes a tentials, some cells generated spontaneous transient out-
major effector for nitrergic neurotransmission, PKG1, ward currents (STOCs) that were blocked by apamin. Sin-
caused significant reduction in nitrergic responses and de- gle-channel studies showed that PDGFR cells express 10
layed colonic transit. In another study using ICC and pS Ca2-activated K channels, which is consistent with
smooth muscle specific iCre strains, the 1 subunit of guan- expression of SK3.

Physiol Rev VOL 94 JULY 2014 www.prv.org 883


SANDERS ET AL.

Purines, such as ATP, -nicotinamide adenine dinucleotide within 100 nm of nerve varicosities in GI muscles, and each
(-NAD), and ADP, activated Ca2-dependent K currents of these cells expresses specialized receptors and effectors
under voltage clamp, and these responses were blocked by that respond to enteric motor neurotransmitters. Binding of
apamin and the P2Y1 receptor antagonist MRS2500 (221). neurotransmitter molecules to ICC and PDGFR cell re-
In current-clamp mode, purines activated rapid responses ceptors activate or modulate ionic conductances, as de-
that hyperpolarized cells to EK. In contrast, responses of scribed above, and these cells are electrically coupled to
colonic smooth muscle cells held at physiological resting smooth muscle cells by gap junctions. Thus the neuroeffec-
potentials were minimal, or small inward currents were tor junction in GI muscles is more accurately conceptual-
activated. Smooth muscle cells had small depolarization ized as the SIP syncytium, composed of electrically coupled
responses to ATP in current clamp. These results show that smooth muscle cells, ICC, and PDGFR cells (FIGURE 3)
PDGFR cells mimic the purinergic neural responses of (209, 330). Changes in the conductance of any type of SIP
whole colonic muscles (i.e., hyperpolarizations inhibited by cell can affect the input resistance of the syncytium. To-
apamin and MRS2500; Refs. 116, 173), and these re- gether SIP cells not only determine the integrated excitabil-
sponses do not occur in smooth muscle cells at physiological ity of muscle layers and responses to neuotransmitters, but
potentials. ICC also show little or no response to purines it is quite likely that responses to hormones and paracrine

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


that can explain purinergic responses in situ. Thus mediators are also integrated through the responses of the
PDGFR cells are the most likely postjunctional cells to SIP syncytium. Classically, terms such as myogenic,
mediate purinergic motor neurotransmission. PDGFR neurogenic, and neuroeffector junction used to de-
cells may also contribute to tonic inhibition in the colon. scribe different regulatory mechanisms in GI tissues have
PDGFR cells generate STOCs, and this could provide a not been inclusive of the important responses elicited by
net stabilizing effect on the excitability of smooth muscle interstitial cells.
cells. It should also be noted that PDGFR cells express
sGC (175, 176), but a role for NO/cGMP-dependent mech-
anisms in these cells has not been identified. G. ICC as Stretch Receptors in GI Muscles

Activation of SK3 currents in PDGFR cells would require The walls of GI organs go through dramatic distortions
dynamic cytoplasmic Ca2 signaling. The occurrence of while filling and during the digestion and movement of food
STOCs in these cells suggests that periodic, localized Ca2 and wastes. Stretch-dependent mechanisms have been de-
release from intracellular stores is an important mechanism. tected in neurons (84, 219, 358, 359) and smooth muscle
Ca2 signaling mechanisms were investigated in PDGFR cells (97), but interstitial cells may also have an important
cells of gastric fundus muscles in situ (13). Cells in fundus sensory role in transducing mechanical events and pro-
muscles of Pdgfratm11(EGFP)Sor/J mice were loaded with fluo viding immediate feedback affecting motility. Freshly iso-
4 and identified by nuclear eGFP. Spontaneous localized lated ICC from human intestine displayed a Na current in
Ca2 transients were observed in PDGFR cells. Purines patch-clamp experiments (362). The conductance was
(ATP, ADP, UTP, and -NAD) and the P2Y1 agonist MRS- blocked by QX-314 but not by nifedipine or Ni2. Tran-
2365 enhanced Ca2 transients, and these responses were scripts for SCN5A, which encode subunits of cardiac
blocked by MRS-2500, a specific P2Y1 antagonist. ADP, TTX-resistant Na channels, were found by single-cell
MRS-2365, and -NAD failed to elicit Ca2 transients in PCR. Stretch of intestinal muscle increased the frequency of
cells of P2ry1/ mice, but responses to ATP persisted. slow waves, and QX-314 or lidocaine reduced slow wave
Phospholipase C (U-73122), IP3 receptor (2-APB), ryano- frequency. These data suggest that pacemaker ICC express
dine receptor (ryanodine), and SERCA pump (cyclopia- a mechanosensitive Na conductance that regulates slow
zonic acid and thapsigargin) inhibitors blocked Ca2 tran- wave frequency.
sients evoked by purines. Activation and regulation of Ca2
transients are likely to mediate the openings of SK3 chan- Another study investigated the chronotropic effects of
nels, generation of STOCs and responses to purines, and stretch in the murine gastric antrum (417). Slow waves were
purinergic inhibitory junction potentials in GI muscles. recorded from antral muscles during computerized applica-
tion of muscle stretch at a rate of 6.0 m/s to a maximum
7. Neuromuscular transmission and integration in tension of 5 mN. This increased muscle length by 27%,
the SIP syncytium which is within what might be expected physiologically
during antral contractions. Stretch of antral muscles caused
As discussed above, the extent to which motor neurotrans- 4 mV of depolarization and increased slow wave frequency
mission is volume-like or more focalized (e.g., synaptic- from 2.5 to 3.6 cpm. The magnitude of the response to
like due to close appositions between varicosities and stretch was positively related to the rate of muscle stretch.
postjunctional cells or restrictions on transmitter diffusion Stretch responses were blocked by reduced extracellular
due to metabolism or uptake) is controversial at present. Ca2 and by Ni2, but unaffected by nifedipine. Responses
What seems not debatable, based on current knowledge, is to stretch were not neurogenic because they were not
that at least three postjunctional cell types are clustered blocked by TTX. Acceleration of antral slow waves dis-

884 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

rupted corpus to antrum slow wave propagation temporar- that have a characteristic electrophysiology signature of
ily. Stretch responses were not recorded in muscles of after-hyperpolarization (AH) following action poten-
W/WV mice, suggesting that ICC-IM may mediate stretch- tials. Therefore, IPANs are also known as AH neurons.
dependent chronotropic effects. It was also found that in- Identified AH neurons were described as making close con-
domethacin blocked stretch-dependent chronotropic re- tacts with ICC-MY, and depolarization of the neurons cor-
sponses, and these responses were also not present in mice related with modulation of Ca2 transients in ICC-MY
lacking cyclooxygenase-2 (Ptgs2/). These findings sug- (438). These authors concluded that direct inputs from AH
gest that stretch may activate phospholipase A2 and gener- neurons regulate ICC-MY function.
ate arachidonic acid. Synthesis of PGE2, a product of
COX-2, may drive stretch-dependent responses in ICC-IM,
since this prostanoid has been shown to have positive chro- III. DEVELOPMENT AND PLASTICITY OF
notropic effects in antral muscles of several species (103, ICC
327, 333).
A. Role of c-Kit in Development of ICC
Networks

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


H. Interactions With Afferent Nerve Fibers

There are at least two types of mechanoreceptor structures ICC are of mesodermal origin (238, 379, 430) and de-
arising from extrinsic afferent neurons in smooth muscles of rived from cells within the gut. Cells expressing tran-
GI organs: intraganglionic laminar endings (IGLEs) and scripts of Kit appear on about embryonic day 9 (E9) in
intramuscular arrays (IMAs). The morphology and intercel- mice (280), and c-Kit protein expression is detected by
lular associations of IMAs have been carefully described in immunohistochemistry at E12 (379). Initially c-Kit cells
recent years; however, the function of these structures is still form a layer of undifferentiated mesenchymal cells
poorly understood (294). IMAs arborize into a complex around the periphery of the intestine (at E12). c-Ret
array of terminal processes that run in parallel to the cells, which are precursors of enteric neurons and glia,
smooth muscle fibers and are associated with ICC-IM in the form a distinct layer at the inner surface of the c-Kit
stomach (32, 295). IMAs are also associated with three cells. Circular muscle cells are apparent at E15, but at
species of efferent fibers, distinguished by antibodies to this stage the intestine lacks a longitudinal muscle layer.
nNOS (inhibitory motor neurons), vesicular acetylcholine Within the circular muscle layer, smooth muscle anti-
transporter (excitatory motor neurons), and tyrosine hy- gens, such as enteric actin and desmin, are prominent,
droxylase (sympathetic neurons). Calcitonin gene-related and the cells have typical ultrastructural features such as
peptide (CGRP)-immunopositive afferent fibers (presumed thin and thick myofilaments, intermediate filaments,
to be collaterals of visceral afferent neurons) are also asso- dense bodies, caveolae, and well-developed endoplasmic
ciated with IMAs (294). The varicosities of IMAs form reticula. c-Kit cells are largely immunopositive for vi-
synaptic-like contacts with ICC and display vesicles and mentin, but a few cells begin to develop immunoreactiv-
prejunctional densities (166, 294). The ICC-IMA complex ity for desmin at E15, suggesting that cells of the longi-
has been likened to a functional correlate of the muscle tudinal muscle layer may develop from c-Kit precursors
spindle organ of striated muscles. The function of ICC in (379). Others reached a similar conclusion by showing
relation to IMAs has not been clarified. There is evidence with in situ hybridization that cells at the periphery of the
that vagal afferents can engage in axon reflexes (414), small intestine that expressed Kit mRNA coexpressed
which is accomplished by branches of peripheral axons that myosin heavy chain transcripts (203). By E18, a clear
contain both sensory and effector functions without the longitudinal muscle layer is formed and c-Kit cells de-
benefit of any synapses or an integration center. In the case veloped processes and formed a lose network in the re-
of ICC-IMA complexes, the numerous lamellar varicosities gion of the myenteric plexus (ICC-MY). Longitudinal
of IMA neurites distributed around ICC and close associa- muscle cells express desmin, and ICC retain expression of
tions with CGRP-immunopositive processes may, in fact, vimentin. The other population of ICC in the small bowel
innervate ICC, providing an integrative sensorimotor func- (ICC-DMP) is not found in murine embryos and develops
tion to supplement the main sensory activities of afferent within the first week after birth.
neurons. Physiological tests of this hypothesis have not been
reported. The heterogeneity of ICC lesions in W/WV mice (404) led to
speculation that c-Kit is not required for development of all
There is also a report of innervation of ICC-MY by intrinsic ICC. Some populations of ICC fail to develop with the loss
primary afferent neurons (IPANs) in the small intestine. of function in W/WV mutants, but other populations of ICC
IPANs are myenteric neurons that send processes to the develop, albeit usually in reduced density and with reduced
lamina propria to sense the mechanical and chemical state immunoreactivity to c-Kit antibodies. These observations
of the gut lumen. These neurons provide the afferent limb of suggest that ICC from different regions of the GI tract and
intrinsic GI reflexes (109). IPANs are Dogiel type II neurons even within specific regions have different sensitivities to

Physiol Rev VOL 94 JULY 2014 www.prv.org 885


SANDERS ET AL.

c-Kit signaling. Another possible difference might be in sur- require signaling via c-Kit for development (23, 306, 404),
vivability of different ICC in the absence of c-Kit. One study so one explanation for their localization within muscles
investigated the development of ICC in animals with Kit might be that they are concentrated near cells expressing
alleles deactivated by knock-in of LacZ into the Kit locus. stem cell factor, the ligand for c-Kit. Experiments on trans-
Homozygotes for the transgene (KitLacZ/) expressed c-Kit genic animals with lacZ linked to the promoter for stem cell
and ICC normally, but KitLacZ/LacZ mice lacked c-Kit but factor showed that enteric neurons express stem cell factor
cells identified as ICC were as abundant as in KitLacZ/ mice (380), but others have reported that smooth muscle cells
(31). It is possible, however, that the cells identified as ICC also express stem cell factor transcripts and protein (161,
were precursors for ICC identified in development by E15 238, 422). Expression of stem cell factor by neurons might
(379). Whether the cells identified as ICC (at about E18) direct positioning of ICC, because either nerve cell bodies or
were functional was not determined. Another group inves- processes occupy most of the same niches as ICC. However,
tigated the distribution of ICC in Wbanded (Wbd) mice (203). it is unclear how smooth muscle expression of stem cell
Wbd contains a genomic rearrangement of chromosome 5 factor might direct the localization of ICC with such precise
that results in inversion of the c-kit gene and ectopic expres- patterning. In fact, experimental evidence suggests that neu-
sion of Kit (204). These mice displayed normal distributions rons, and thus trophic factors expressed by neurons, are not

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


of cells identified as ICC at D5, but adult animals lacked obligatory for development or localization of ICC, because
ICC. The authors concluded that ICC did not require sig- ICC developed in muscles taken for culture from mamma-
naling via c-Kit for development, but functional c-Kit was lian and bird intestines before colonization of the muscles
required for maintenance of the ICC phenotype as animals with neural crest cells (238, 430). Functional ICC also de-
matured. Identification of cells as ICC was performed by veloped in c-Ret/ mice that lack enteric neurons below
methylene blue staining, but there was no verification that the proximal stomach (409). Therefore, significant ques-
the cells labeled with methylene blue were ICC. Electrical tions remain about the signals and sources of signals direct-
recordings failed to demonstrate slow wave activity in Wbd/ ing the positioning, architecture, and connectivity of inter-
Wbd mice, suggesting that the ICC pacemaker phenotype stitial cell networks.
never developed. Thus the question is whether ICC devel-
oped in Wbd/Wbd mice or whether another cell type, labeled Significant development of ICC occurs in mice during the
by methylene blue, was identified as ICC in these mutants. postnatal period. At P0, ICC-MY form a loose network
For example, it has never been determined whether meth- with cells displaying a few short and branching processes.
ylene blue labels ICC exclusively or whether PDGFR The density of ICC-MY at P0 was 750 mm2. The de-
cells are also labeled by this dye. mands of maintaining the structure of ICC networks as
bowel diameter expands require proliferation of ICC. An
The question of the role of c-Kit in development of func- age-dependent proliferation of ICC-MY, estimated to be
tional ICC during the late embryonic period was addressed 15-fold from neonates to adulthood, maintained the rel-
in another study by treating muscles with neutralizing c-Kit ative density of these cells in adult animals (259). The pro-
antibodies from E15 through birth. Functional ICC develop liferative potential of ICC was studied by incorporation of
in small intestinal segments in organotypic cultures started 5-bromo-2-deoxyuridine (BrdU). A large number of cells
at E15; however, inclusion of c-Kit neutralizing antibodies within ICC networks were labeled with BrdU shortly after
blocked development of c-Kit positive cells and pacemaker birth, but the number of cells incorporating BrdU, 24 h after
activity (406). It was also observed that ICC-MY and slow loading, decreased as a function of age. Cells with short
waves developed normally in WV/ and in W/ heterozy- processes, but apparently already integrated into ICC-MY
gotes, but fail to develop between E17 and birth (postnatal networks, incorporated BrdU within 1 h after injections,
day 0; P0) in W/WV embryos (23). Similar results, including and pairs of cells with BrdU were noted after 24 h. It was
loss of ICC and failure of pacemaker development, oc- difficult to find proliferating cells after P24. The fraction of
curred in muscles treated for several days with the tyrosine total cells that incorporated BrdU was also determined by
kinase inhibitor imatinib mesylate (STI157). These data daily injections, and this number increased to 64% by P24.
support the concept that ICC precursors cells might emerge The expansion of ICC-MY appeared to be due to prolifer-
in animals with compromised c-Kit function, but subse- ating cells within ICC networks (259), as opposed to pro-
quent development into mature and functional ICC re- liferation from a rare population of KitlowCD44
quires c-Kit signaling. CD34IGF-1 cells, proposed by others as the progenitors
of ICC (248). It should be noted that most cells within the
The reasons why ICC develop within specific planes in the ICC-MY network were CD44, and some cells showed
tunica muscularis (e.g., ICC-MY and ICC-SM) or in asso- weak labeling with antibodies to CD34 and insulin-like
ciation with specific structures, such as enteric neurons growth factor I (IGF-I) (259). These findings tend to suggest
(ICC-DMP or ICC-IM), are poorly understood, but the pat- that KitCD44CD34IGF-1 cells are normal compo-
terns of ICC localization are conserved, with minor varia- nents of ICC-MY networks and do not necessarily represent
tions, in mammalian species and fish (14, 210, 307). ICC a specialized population of ICC progenitors.

886 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

Early in human development (7 wk), c-Kit cells can be less reliable than immunohistochemical techniques (see
found along the entire length of the esophagus in the region sect. IIA1).
of the myenteric plexus, but this distribution changes with
age such that ICC were far more abundant in the distal Consistent with the role of ICC as pacemakers, there is a
one-third versus the proximal esophagus by weeks 1316 proximal-to-distal delay in the developmental of pace-
(300). After 20 wk, ICC were extremely rare in the proxi- maker activity (406). Slow waves were recorded from the
mal (skeletal muscle) portion of the esophagus and had proximal GI tract (stomach and antrum) during the late
developed into spindle-shaped ICC-IM in the circular and embryonic period (as described above), but developed after
longitudinal muscle layers in the distal third (smooth mus- birth in the ileum and colon. Slow wave activity with clus-
cle) portion of the esophagus. The middle third of the ters of action potentials superimposed, characteristic of the
esophagus displayed ICC in septa between smooth and skel- proximal colon, developed in colonic muscles by P6-P8;
etal muscle fibers. ICC developed over approximately the however, this activity was infrequent, with long periods of
same window of time in the stomach, forming first a layer of quiescence between events. Regular, rhythmic slow waves
c-Kit precursors around the periphery of the organ adja- and action potential clusters were not fully developed in the
cent to the developing myenteric plexus (301). By 20 wk, proximal colon until about P10.

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


ICC can be found in all of the adult niches: ICC-MY, ICC-
IM, and ICC-SEP. ICC develop in the human midgut and Development of ICC and slow waves occurs unfettered in
hindgut somewhat later than in the proximal GI tract. At 9 organotypic cultures of GI muscles (23, 406), and this ap-
wk, c-Kit labeling is present at low levels within the circular proach has been utilized to correlate the timing of ICC-MY
muscle (393). By week 11, c-Kit labeling is present along the development and the development of electrical rhythmicity
inner and outer aspects of myenteric ganglia. At 14 wk, ICC and to evaluate the impact of c-Kit signaling on develop-
displayed an approximately adultlike distribution. In the ment. Inclusion of neutralizing c-Kit antibodies in muscles
distal colon, c-Kit cells emerged during weeks 10 11. of neonatal animals abolished ICC networks and blocked
Two parallel bands of cells were found within the myenteric pacemaker activity within a few days of exposure. At birth,
region between the circular and longitudinal muscle layers as observed in animals, ICC were much more susceptible to
and near the submucosal edge of the circular muscle (299). effects of neutralizing c-Kit antibodies than after developing
The proximal to distal delay in the development of ICC in in animals for several days before being put into organ
the human GI tract is also a feature of the development of culture (406). Longer periods (e.g., 30 days) of exposure
ICC and pacemaker activity in the mouse (379, 406). to c-Kit antibodies were required to lesion ICC-MY net-
works and block pacemaker activity in muscles from ma-
ture (P20) animals. Since the dependence on c-Kit signal-
B. Development of Electrical Rhythmicity in ing for initial development of ICC-MY was challenged by
ICC others (31, 203), experiments were also performed to deter-
mine whether functional ICC developed with c-Kit blocked
or disabled genetically. ICC-MY and slow waves developed
A lineage decision is made at about E15 in the murine in jejunal muscles of wild-type, W/, and WV/ mice by
intestine, and cells that continue to express c-Kit develop E18, but failed to develop in muscles of W/WV muscles.
pacemaker activity (379). Electrophysiological studies of ICC-MY failed to develop in jejunal muscles removed from
jejunal muscles during the late embryonic period found no mice at E17 (i.e., before slow waves develop) and cultured
electrical slow waves at E17, but small-amplitude fluctua- with neutralizing c-Kit antibodies (23). Similar results were
tions in membrane potentials (STDs) were observed. Net- obtained if muscles were put into organ culture after slow
works of ICC were well-articulated at E17, and multiple waves developed (E18-P0). Imatinib mesylate (STI157), a
processes linking cells together were observed (23, 379, c-Kit inhibitor, had no acute effect on slow waves, but ex-
406). At E18, slow waves, resistant to block by dihydro- posure to this compound blocked postnatal growth and
pyridines (a signature feature of slow waves vs. action development of ICC-MY and slow waves. Thus c-Kit sig-
potentials), were recorded by intracellular microelectrode naling during development is requisite for establishment
techniques (23). The organization, frequency, and ampli- and maintenance of the pacemaker function of ICC.
tude of slow waves continued to develop through birth and
the first week after birth, reaching mature amplitude and
frequencies by about P10. Other investigators set the onset C. Fate of ICC When c-Kit Is Blocked
of electrical rhythmicity in the proximal small intestine later
in development, finding weak electrical activity in unfed Loss of ICC has been observed in human GI muscles from
neonates and strong reduction in the electrical activity by patients with a variety of motility disorders (11, 43, 143,
verapamil (246). These authors concluded that ICC-MY 168, 205, 385). Dysmotilities seem an obvious consequence
were not functional at birth, and pacemaker activity and of defective ICC or loss of ICC due to the important func-
slow waves did not develop until about P2. ICC-MY were tions of these cells. In spite of the clinical significance, the
identified with methylene blue in this study, but this dye is factors responsible for ICC loss in patients are poorly un-

Physiol Rev VOL 94 JULY 2014 www.prv.org 887


SANDERS ET AL.

derstood. Attempts have been made to develop animal these mice, and it was concluded that signaling through
models of ICC loss with the idea of establishing the cause PI3-kinase is not required for development and function of
and effect of ICC loss in motility disorders and to study the ICC. There are two means of activating PI3-kinase, how-
plasticity of ICC. ever, by direct binding of PI-3kinase to the phorphorylated
tyrosine residue of c-Kit and indirectly by binding to the
ICC are vulnerable to block of c-Kit signaling in neonatal tyrosine phosphorylated accessory protein GAB2 (271).
and adult animals; however, the time required for c-Kit Thus the Y719F mutation in Kit may not block all PI3-
block increases from the postnatal period to adulthood. kinase signaling.
Neutralizing c-Kit antibodies injected into neonatal mice
and for several days after birth cause loss of ICC within a ICC-MY networks and slow wave activity can be reestab-
week (250, 378), and all classes of ICC are affected. The lished after removal of conditions or reagents that block
c-Kit inhibitor imatinib mesylate also lesions ICC networks c-Kit. This point will be more completely illustrated in the
in neonatal murine intestinal muscles in organ culture (23). next section, but model studies of this concept have been
In adult guinea pigs, imatinib caused a time-dependent re- performed on muscle strips in organ culture. As above,
duction in ICC-MY of the small intestine, shortening of cell treatment of P0 muscles with neutralizing c-Kit antibodies

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


processes, and generalized thinning of the network (258). or imatinib mesylate for 3 days caused loss or dramatic
The number of ICC-MY was reduced by about one-third reduction in ICC-MY and loss of pacemaker activity. After
after 16 days of treatment. ICC-DMP were also affected, removal of these reagents, ICC networks and slow waves
but effects were less dramatic and slower in onset. No evi- were restored within 9 days (23).
dence of apoptosis was noted in ICC after treatment with
imatinib; instead, remaining cells coexpressed -smooth D. Animal Models of ICC Loss and
muscle actin, suggesting that ICC-MY undergo a pheno- Pathophysiology
typic change toward a smooth muscle or myofibroblast phe-
notype when c-Kit is blocked. BruD incorporation was ob- In addition to Kit mutants and animals in which c-Kit is
served in ICC-MY after withdrawal of imatinib, and pairs blocked, ICC loss has been reported in several animal mod-
of cells with highly developed processes that were well- els of GI motility disorders. These are important because
incorporated in ICC-MY networks demonstrated that cell parallel examples of ICC loss occur in similar or equivalent
division of mature ICC was at least partially responsible for diseases in human patients. It should be noted that if a
repopulation of ICC-MY networks after ending c-Kit common factor is responsible for ICC loss in various disease
blockade (258). states, it has not been identified. Furthermore, a debate
exists about whether functional ICC are lost through cell
Downstream cell signaling initiated by stem cell factor bind- death or by remodeling of the ICC phenotype. Examples of
ing to c-Kit includes contributions from phosphatidylinosi- animal models with predictable, and in some cases reversal,
tol 3-kinase (PI3-kinase), phospholipase C- (PLC-), defects in ICC are described below.
phospholipase D, SRC family kinases, p21ras GTPase-acti-
vating protein, and mitogen-activated protein kinase 1. Bowel obstruction
(MAPK) (244). There are multiple classes of PI3-kinase, but
growth factor receptors typically activate class IA. Binding Partial obstruction of the bowel leads to marked distension,
of stem cell factor leads to autophosphorylation and hypertrophy, and hyperplasia of smooth muscle cells, and
dimerization of c-Kit and generation of high-affinity bind- thickening of the muscle layers of the bowel proximal to the
ing sites for signaling molecules, including PI3-kinase. obstruction (111, 112). Studies of a rat model of partial
Phosphorylation of c-Kit leads to binding of the p85 regu- obstruction showed remodeling of enteric neurons and re-
latory subunit of PI3-kinase and subsequent activation of duced numbers of ICC in the hypertrophic region of bowel
the catalytic subunit p110. The effects of PI3-kinase inhib- (91). In mice, partial obstruction resulted in hypertrophy in
itors, wortmannin and LY 294002, were tested on develop- a long segment of small intestine and a lesion in ICC net-
ment of ICC and pacemaker activity in mice. The amplitude works that was nearly complete a few millimeters above the
of slow waves increases after birth, and this development obstruction but decreased in severity over 100 mm of bowel
also occurs in organotypic cultures (406). Inclusion of wort- (51). In specific planes of the tunica muscularis normally
mannin or LY 294002 in organ cultures of intestinal mus- occupied by ICC, cells with ultrastructural features more
cles taken from animals at birth caused loss of ICC-MY and typical of fibroblasts were observed, but there was no evi-
blocked slow waves within a few days (401). Pacemaker dence of cell death or necrosis (e.g., shrinking, condensed or
activity was also blocked in adult muscles by PI3-kinase fragmented nuclei, indicative of apoptosis, or cell swelling
inhibitors, but up to a month of exposure was required to or bursting) was observed in any region where ICC were
develop these effects. Another study employed a transgenic lost. Slow waves were greatly reduced in amplitude or ab-
mouse with a tyrosine 719 to phenylalanine (Y719F) mu- sent in the section of bowel with the most extensive loss of
tation in Kit (119). No loss of ICC function was observed in ICC and responses to neural inputs were also greatly re-

888 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

duced in these regions. ICC, slow wave generation, and The reasons for loss of ICC in NOD/LtJ mice have been
neural responses recovered in 1 mo if the obstruction was investigated. Reduced IGF-I and insulin signaling, and not
relieved. hyperglycemia, were found to be responsible for the loss of
ICC in diabetes (162). Expression of insulin and IGF-I re-
2. Diabetes ceptors were localized to enteric neurons and smooth mus-
cle cells, and transcripts for these receptors were not re-
solved in ICC purified by FACS (161). The same cells that
Reduction in ICC in diabetes and the importance of this expressed IGF-I and insulin receptors also expressed stem
phenomenon in diabetic gastroparesis has been a topic of cell factor. Loss of stem cell factor was found to be the cause
interest since defects in ICC networks were noted in a of ICC loss in diabetic tissues. Therefore, signaling via IGF-I
mouse model of type I diabetes. Non-obese diabetic mice and insulin does not occur directly through ICC, but ICC
(NOD/LtJ) mice were shown to have delayed gastric emp- maintenance depends on these growth factor receptors, as
tying and reduced and arrhythmic slow wave activity in the they are important in maintaining expression of stem cell
antrum (278). Fundus muscles from NOD/LtJ mice were factor in cells adjacent to ICC.
less responsive to activation by enteric motor neurons,

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


which might be associated with reduced gastric accommo- Type II diabetes is by far the most prominent form in human
dation during ingestion of food. Functional defects were patients. C57BL/KsJdb/db mice, a monogenic model of type
associated with reduced ICC in the antrum and loss of close II diabetes, were examined for gastric emptying and damage
connectivity between motor neurons and ICC in the fundus. to ICC networks (421). c-Kit cell density and expression
Another study confirmed the link between loss of ICC and of stem cell factor were also evaluated. Compared with
development of delayed gastric emptying, and showed that db/ mice, db/db had delayed gastric emptying as well as
loss of Kit expression, the marker for ICC, was the most prolonged gut transit times, and contractile activity was
consistent correlate for defects in gastric emptying (57). irregular in frequency. Cells with c-Kit immunoreactivity
These authors noted that hemeoxygenase-1 (HO1) expres- were reduced in antrum, small intestine, and colon, as was
sion, which normally is expressed at low levels in the tunica expression of stem cell factor. These authors concluded that
muscularis, increased as diabetes developed. Mice that loss of ICC contributes to the motility defects in type II
maintained expression of HO1 were protected from devel- diabetes.
opment of delayed gastric emptying, and mice that failed to
maintain levels of HO1 developed emptying delays. It was Reduced ICC numbers have also been reported in samples
suggested that the benefit of HO1 in diabetes may be to of antral muscles from full thickness gastric biopsies from
protect ICC against enhanced oxidative stress. An exciting human patients symptomatic of diabetic and nondiabetic
finding was that induction of HO1 expression by hemin gastroparesis (105, 132, 133, 184). An interesting finding in
rescued mice that had developed delayed gastric emptying. human samples was a shift in splice variants of ANO1 in
Resident macrophages, which lie in close proximity to ICC, ICC from patients with gastroparesis (252). A novel splice
were identified as the cells with enhanced expression of variant that lacked exons 1 and 2 and a portion of exon 3 of
HO1 in diabetes. The protective effects of HO1 on gastric ANO1 was observed in patients gastroparesis. ANO1 en-
codes the ion channel responsible for pacemaker currents in
emptying appear to be due to the production of carbon
ICC (see sect. IID2), and a shift in splice variant expression
monoxide (CO) (190). Inhalation of CO by NOD/LtJ mice
might affect the kinetics or frequency of slow waves. Ex-
with delayed gastric emptying, reduced oxidative stress, en-
pression of the unique splice variant from diabetic tissues in
hanced Kit expression, and reversed delayed gastric empty-
HEK cells resulted in decreased conductance and slower
ing.
kinetics than displayed by full-length ANO1 channels.
The density of ICC in the distal stomach was also reduced in 3. Surgical resection of the bowel
streptozotocin-induced diabetes in rats, and loss of connec-
tivity between nerves and ICC-IM was observed in the fun- Acute inflammatory responses to surgery, such as bowel
dus (395). These changes were associated with decreased resection, have also been linked to adverse effects on ICC
gastric emptying in diabetic animals. Others have reported and their associated functions. Resection of murine small
similar finding for streptozotocin-induced diabetes in rats intestine caused loss of c-Kit-like immunoreactivity, loss of
and found significant loss of ICC in the antrum (267). These slow waves, attenuated neural responses, and abnormal re-
authors also found an association between retention of ICC sponsiveness to carbachol (425). The loss of c-Kit immuno-
and upregulation of HO1 expression. For example, HO1 reactivity was shown to be due to loss of ICC via electron
expression was enhanced in the corpus and ICC networks microscopy. The lesion in ICC and loss of function de-
were essentially unchanged in that region. Antral ICC were creased as a function of distance (5 cm) above and below the
depressed, and this was associated with no enhancement in point of the anastomosis. Loss of ICC was acute and par-
expression of HO1. Loss of ICC was reversed by induction tially recovered within 24 h of surgery. Recovery was more
of HO1 expression by cobalt protoporphyrin. rapid when muscles were treated with an iNOS inhibitor,

Physiol Rev VOL 94 JULY 2014 www.prv.org 889


SANDERS ET AL.

6
L-N -(1-iminoethyl)lysine hydrochloride (L-NIL). Animals survival. Thus gain-of-function mutations in c-Kit in the
treated preoperatively with iNOS inhibitors experienced re- progenitor cells could yield continuous output of malignant
duced pacemaker activity only near the site of the anasto- GIST cells that is not controlled by blocking the tyrosine
mosis, and transgenic animals lacking iNOS were protected kinase activity of c-Kit (16). It was suggested that Kitlow
against most of the postsurgical defects in ICC networks Cd44 Cd34 ICC stem cells are the source of GIST cells.
and pacemaker activity (424). These are c-Kit independent stem cells that are not suscep-
tible to therapeutic approaches designed to inhibit c-Kit.

E. Cancer
IV. INTERSTITIAL CELLS IN THE URINARY
Kit is a protooncogene encoding a receptor tyrosine kinase. TRACT
Gain-of-function mutations in the juxtamembrane domain
of Kit are found in 75 80% of GISTs, the most common Smooth muscle tissues of the urinary tract generate sponta-
form of human sarcoma, and the idea that GISTs derive neous phasic contractions and tone. The intrinsic electrical
from ICC has been considered (149, 150, 195, 272). Jux- and contractile activities appear to be generated by special-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


tamembrane domain mutations discovered in GISTs render ized pacemaker cells. Electrical and contractile activities are
c-Kit constitutively active, and expression of the mutated also regulated by neurotransmitters, and interstitial cells
isoforms in cells resulted in unregulated growth (149). Im- may be involved in transduction of neural inputs. Electrical
munostaining with antibodies to c-Kit has been an impor- activity generated by specialized cells may propagate to
tant advance in categorizing mesenchymal tumors, and re- smooth muscle cells, control membrane excitability, and
alizing that c-Kit drives the growth of GISTs has afforded a generate phasic contractile activity. Two types of special-
therapeutic approach to controlling GISTs. Activating mu- ized cells have been reported: c-Kit and c-Kit cells. Re-
tations in PDGFR have also been identified in GISTs. Thus cently, PDGFR cells have been shown to be at least part
it is also possible that cells of the PDGFR cell lineage of the c-Kit interstitial cell population. In the following
could contribute to development of GISTs. Inhibition of sections, the distributions and physiological and patho-
c-Kit or PDGFR with tyrosine kinase inhibitors reduces physiological roles suggested for interstitial cells of the up-
progression of GISTs, but these drugs need to be maintained per and lower urinary tract are discussed.
for indefinite periods for effectiveness. This has suggested
that sustained growth of GISTs may be due to proliferation
of cancer stem cells that are not dependent on signaling via A. Renal Pelvis and Ureter
c-Kit for survival.
Pacemaker activity is generated in the most proximal re-
Mice with a familial mutation in c-Kit, KitV558/, develop gions of pyeloureteric tissues, and this initiates peristaltic
GISTs and die prematurely from pathology of the GI tract. contractions that spread toward the distal ureter. The cells
These mice were used to determine the fate of ICC and ICC and ionic mechanisms responsible for pacemaker activity in
functions during GIST development (224). Most classes of the upper urinary tract have received considerable study.
ICC developed marked hyperplasia throughout the GI Two populations of specialized cells were observed in the
tracts of KitV558/ mice. Normal pacemaker activity and renal pelvis, and their ability to generate pacemaker activity
responses to enteric motor neurons were conserved in tis- has been investigated (232). Short, spindle-shaped cells, re-
sues during development of GISTs. If GISTs develop from ferred to as atypical smooth muscle cells (ASMC), are lo-
mature ICC, one would think that hyperplasia of these cells cated in the proximal region of ureteropelvic junction.
would result in enhanced frequency and amplitude of STICs These cells generate spontaneous electrical rhythmicity
and slow waves due to an expansion in the pacemaker pop- (123, 124, 201). Another type of cell, referred to as ICC-like
ulation. The fact that rhythmicity was not enhanced in cells (ICC-LC), has also been described. c-Kit immunoposi-
KitV558/ mice suggests that either the hyperplasia associ- tive cells have been reported in the upper urinary tract (74,
ated with development of GISTs is accompanied by reduc- 231, 260, 261, 286, 356), but c-Kit immmunoreactivity is
tion in the spontaneous rhythmicity or that GISTs develop not consistent in all species (e.g., ICC-LC in guinea pig
from c-Kit cells that are not spontaneously active (i.e., pyelouretic system were not labeled by c-Kit antibodies;
immature ICC or ICC precursor cells). Ref. 201). ICC-LC are mainly distributed in more distal
regions of the ureteropelvic junction. ICC-LC may be in-
Although the lineage of ICC has been suggested to be the volved in generation of pyelrouteric pacemaker activity
source of GISTs, the exact cells from which GISTs are de- (201, 232). Both ASMC (13pF) and ICC-LC (17 pF) are
rived is controversial (312). Cells characterized as Kitlow smaller than SMC (20 pF) based on cell capacitance mea-
have been suggested as progenitor cells for ICC (248). These surements (181).
cells also express CD44, CD34, insulin receptors, and
IGF-I. Signaling via c-Kit might be necessary for differenti- ASMC and ICC-LC cells display high (10 40/min) and low
ation of the progenitor cells into ICC, but not for their (13/min) frequency of Ca2 transients, respectively (228,

890 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

229). Based on the frequency of Ca2 transients and spon- been used typically to identify interstitial cells in the blad-
taneous transient depolarizations (STD), the dominant der. Vimentin cells have been reported in the suburothe-
pacemaker cells may be ASMC. Spontaneous Ca2 tran- lium of guinea pig (76, 129, 215, 302) and human bladders
sients and STDs were inhibited in ASMC when extracellular (189, 268, 363), and there are reports of c-Kit cells in the
Ca2 was reduced or by application of cyclopiazonic acid suburothelium of the human (189, 287, 288, 344) and pig
(CPA) and 2-APB (230). Ryanodine (up to 100 M) slightly bladder (260). Some studies have noted that not all vimen-
reduced Ca2 transients but failed to block STDs in ASMC tin cells are c-Kit (75, 256). Thus at least two classes of
(229, 230). Reduced extracellular Na also decreased STDs interstitial cells may exist in the bladder. It should also be
in ASMC, and these events were not affected by Cl chan- noted that mast cells are also c-Kit and can be found in the
nel blocking drugs or replacement of extracellular Cl suburothelium and in detrusor muscles. However, mast
(230). The data suggested that pacemaker activity in ASMC cells are typically rounded and of a less fusiform shape than
is due to openings of cation-selective channels stimulated by the c-Kit cells described in bladder tissues (189, 287).
release of Ca2 from IP3 receptor-operated stores. Muscarinic receptor type 3 (M3) is expressed specifically by
suburethral interstitial cells, but the significance of these
A recent report, using eYFP-SMA transgenic mice, receptors in interstitial cells is unknown (129, 130).

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


showed that ASMC, isolated from smooth muscle cells by
the absence of an A-type K current and STOCs, displayed Some investigators have referred to the interstitial cells of
two types of inward conductances that yielded STICs and the urothelium as myofibroblasts. These cells express vi-
large inward currents (LICs) (181). LICs were blocked by mentin and connexin 43 and are in proximity to unmyeli-
replacement of the external Na with TEA (181). Thus nated nerves, suggesting possible neural interactions (107).
LICs might arise from activation of nonselective cation Vimentin myofibroblasts exhibit STICs that are mediated
channels (NSCC), and these events may be responsible for by activation of a CaCC. ATP, acting via P2Y6 receptors,
STDs in intact muscles. The importance of STICs, the in- increased intracellular Ca2 and activated CaCC (107). De-
volvement of a Ca2-activated Cl condutance (CaCC), veloping strains of animals expressing reporters or finding
and how STICs might link to the generation of LICs are not immunoreactive proteins and antibodies directed toward
clear at present.
extracellular epitopes will be important advances for un-
equivocal identification of bladder myofibroblasts in future
ICC-LC have also been shown to generate STICs and LICs
studies.
(232, 233). The conductances responsible for STICs and
LICs were not affected by Cl channel blockers. The cur-
Immunohistochemical evidence for c-Kit cells in the blad-
rents reversed close to 0 mV, suggesting the currents were
der detrusor muscles has been reported in human (189, 287,
due to NSCC conductances (232). Current clamp (I 0)
288, 344), pig (260), dog (12), guinea pig (72, 75, 256, 268,
experiments suggested that NSCC are responsible for STDs
399), rat (79), and mouse (255). Transmission electron mi-
in ICC-LC (233), and spontaneous generation of currents
croscopy (TEM) identified a prominent network of detrusor
may be due to Ca2 release via IP3 or ryanodine receptors.
interstitial cells (CD34) in human detrusor (304). These
In another study, interstitial cells immunonegative for
cells are described as bipolar with slender dendritic process,
smooth muscle actin (most likely ICC-LC) displayed
Xe991-sensitive, Kv7 currents, spontaneous transient out- abundant mitochondria, rough endoplasmic recticulum,
ward currents due to activation of large conductance Ca2- Golgi apparatus, and intermediate filaments. The cells were
activated K channels, and niflumic acid-sensitive STICs found at the peripheral edges of muscle bundles and occa-
(possibly CaCC) (181). These findings were supported by sionally could be found within muscle bundles. They were
immunostaining for ANO1 and KV7.5 immunostaining. considered to be similar to the ICC of the GI tract. How-
How ASMC and ICC-LC interact with smooth muscle cells ever, these cells were immunonegative for CD117 (c-Kit).
to generate peristaltic contractions in the renal pelvis and The number of interstitial cells has been reported to increase
pyelouretic junction has not been fully clarified. in overactive bladder generated in animal models of partial
obstruction or in patients with overactive bladder (34, 193,
216). Bladder contractions are induced by activation of
B. Bladder cholinergic M3 receptors (8, 55, 335), and smooth muscle
cells make close contacts with and are apparently directly
Much has been written about interstitial cells in the urinary innervated by motor neurons (114, 115). Interstitial cells
bladder since vimentin interstitial cells in the detrusor are also found in close proximity to cholinergic nerves (63,
muscle were shown to display cGMP-like immunoreactivity 189, 256), so these cells might have neuromodulatory func-
(353). Two general classes of interstitial cells have been tions in the bladder (128, 186). It should be noted that
described on the basis of their histological distributions: interstitial cells immnopositive for c-Kit are not observed
suburothelial and detrusor interstitial cells. Suburothelial consistently in the bladder. Some investigators have found
interstitial cells lie beneath the urothelium and are cGMP- few cells immunopositive for c-Kit in the detrusor and subu-
immunopositive (121). Vimentin immunoreactivity has rothelium, and the few cells that are c-Kit appear to be

Physiol Rev VOL 94 JULY 2014 www.prv.org 891


SANDERS ET AL.

mast cells in mice (207, 225, 245) and humans (303). Thus
the presence and significance of c-Kit interstitial cells in
A D
the bladder is controversial at present.

1. Functional observations on cells identified as


interstitial cells

Far less is known about the functional role of c-Kit inter-


stitial cells in detrusor muscles than in GI muscles. Studies
of these cells have relied on identification by morphological
characteristics, which are not definitive in dispersed cell
populations. Nevertheless, branched cells, thought to be B E
interstitial cells, were compared with smooth muscle cells.
The branched cells expressed large-conductance Ca2-acti-
vated K channels (BK), TEA-sensitive delayed rectifier K

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


channels (Kv), KCNQ channels, hyperpolarization-acti-
vated cyclic nucleotide-gated channels (HCN), L-type Ca2
channels, and T-type Ca2 channels (7, 79, 144, 186, 253,
254). A T-type Ca2 channel blocker inhibited spontaneous
phasic contractions in detrusor strips and decreased intra-
cellular Ca2 transients in detrusor interstitial cells, sug-
gesting that T-type Ca2 channels could be a Ca2 entry C F
source in generating spontaneous activity (79). Application
of carbachol increased Ca2 transients and phasic contrac-
tions through the M3-Gq/11-PLC pathway in the branched
cells of detrusor muscles, suggesting these cells may respond
to cholinergic neurotransmission in situ (186, 256, 382).
High concentrations of the c-Kit inhibitor imatinib abol-
ished action potential firing and significantly decreased in-
tracellular Ca2 transients in interstitial cells and muscle
contractions (79, 217). Thus it was suggested that a tyrosine
kinase inhibitor might be a useful target for treating symp-
FIGURE 10. Interstitial cells in bladder and oviduct. AC: PDGFR
toms of bladder overactivity (34, 218). However, imatinib cells in the detrusor muscle of cynomolgus monkey (Macaca fascicu-
has effects on Ca2 channels in smooth muscles, so the laris) bladder. A and B: cryostat cross sections through the detrusor
acute effects of this drug may not be through inhibition of wall. At least two populations of PDGFR cells were observed: cells
c-Kit (140). within (arrowheads) and surrounding detrusor muscle bundles (ar-
rows). PDGFR cells also appeared to bridge between adjacent mus-
cle bundles (*). C: flat-profile cryostat section through the detrusor.
There have been suggestions that bladder dysfunction in PDGFR cells located within (arrowheads) and between (arrows) mus-
diabetes and spinal cord injury could be related to changes cle bundles are present. DF: ICC and PDGFR cells within the mouse
in interstitial cell distribution (54, 187). However, it must and cynomolgus monkey oviduct. A and B: ICC within the myosalpinx of
be stated that a role of c-Kit cells in normal or diseased mouse (D) and monkey (E) form an anastomosing network of cells
bladder tissues is far from established. (arrows). These cells have been shown to provide electrical pacemaker
function for propulsive contractions in the oviduct (see text for details).
F: double labeling of PDGFR cells in a cross section of the isthmus
2. PDGFR cells in detrusor muscles region of the mouse oviduct. Tissue comes from Pdgfratm11(EGFP)Sor/J
mice that have constitutive expression of eGFP in nuclei driven off the
Pdgfra promoter. The tissues were double-labeled with antibodies
Recently, another type of interstitial cell was identified in against PDGFR. Thus the soma and cytoplasmic processes of
mouse, guinea pig, and human bladders (207, 268). These PDGFR cells are labeled with antibody (red), and nuclei of PDGFR
cells express PDGFR and therefore have been referred to cells are green. A dense population of these cells were observed within
as PDGFR cells, as in GI muscles (FIGURE 10, AC). the myosalpinx (arrowheads) and within the submucosa and endosal-
pinx (arrows) of the oviduct. The specific functions of these cells have
PDGFR cells are distributed throughout the bladder not been determined. Scale bars are as indicated in each panel. (Au-
wall, including the lamina propria and detrusor muscle. thors are grateful to Dr. Rose Ellen Dixon for images shown in D and E.)
These cells have a spindle-shaped or stellate morphology
and often possess multiple processes, possibly forming net-
works. PDGFR cells surround and are located between A mouse strain expressing eGFP in PDGFR cells (135)
smooth muscle bundles, and they are often in close proxim- has been used to identify PDGFR cells in cell dispersions
ity to intramural nerve fibers. and for purification of cells by FACS (207). PDGFR cells

892 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

are highly enriched in Pdgfra and Kcnn3 (SK3 channel) and smooth muscle cells. The exact nature of the interstitial
transcripts in relation to the whole detrusor cell population. cells in the urethra (i.e., whether mainly c-Kit or
SK3 protein was also observed in PDGFR cells by immu- PDGFR) is unclear (286, 282).
nohistochemistry but was not resolved in smooth muscle
cells. SK channel modulators, CyPPA and SKA-31, hyper- Urethral smooth muscles exhibit STDs and slow wave-like
polarized PDGFR cells and activated SK currents under events. The electrical events are due to activation of CaCC
voltage clamp (239). Similar responses were not observed in in response to release of Ca2 from intracellular Ca2
SMCs held at membrane potentials simulating physiologi- stores (142). Ca2 transients occur in interstitial cells in
cal potentials. The single-channel conductance of SK chan- intact urethral muscles; however, these events were of lower
nels in PDGFR cells was 10 pS, and intracellular Ca2 frequency and longer duration than Ca2 transients in
activated the channels. PDGFR cells displayed STOCs at smooth muscle cells. Ca2 transients in interstitial cells
potentials positive to 60 mV, and these events were inhib- were inhibited by CPA, ryanodine, and 2-APB (141), as also
ited by apamin. found in isolated interstitial cells (see below).

Doxycycline-induced suppression of SK3 expression in Isolated urethral interstitial cells generate spontaneous elec-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


bladder muscles was associated with a decrease in SK cur- trical activity, but smooth muscle cells from the urethra are
rent density in bladder smooth muscle cells and increased normally electrically quiescent. The spontaneous electrical
contractions of detrusor muscle strips (148). Cystometric activity in interstitial cells appears to be due to intracellular
evaluations found that reduced SK3 channel expression Ca2 oscillations that generate pacemaker currents. CaCC
caused a marked increase in nonvoiding contractions. were activated by Ca2 release from IP3 and ryanodine
Taken together, these studies indicate that SK3 channels are receptors on Ca2 stores (67, 156, 339). Mitochondria also
important regulators of bladder muscle excitability. How- regulate the activity of CaCC in interstitial cells, and elec-
ever, the current density of SK channels in smooth muscle tron transport chain blockers, rotenone and antimycin A,
cells is very low relative to PDGFR cells (239). Thus it is and protonophore uncouplers, FCCP and CCCP, inhibited
possible that the effects of suppressing SK3 channel expres- intracellular Ca2 waves and abolished STICs in these cells.
sion could be mediated by loss of this conductance in The evidence for functional expression of CaCC in urethral
PDGFR cells. As in the GI tract, robust expression of
interstitial cells is strong from studies of isolated cells, but a
SK3 channels in PDGFR cells may be an important
recent report suggested that Ano1 is expressed in smooth
means of controlling bladder excitability during the filling
muscle cells, but not in interstitial cells (325). Thus deter-
phase.
mining the molecular species responsible for CaCC in ure-
thral interstitial cells will be a goal for the future.
Recently, the SK conductance in PDGFR cells was shown
to be activated by purines acting on P2Y1 receptors (240).
Urethral smooth muscle cells express L-type and T-type
P2Y receptors (mainly P2ry1) were highly expressed in
Ca2 channels (41, 157), but expression of voltage-depen-
PDGFR cells. ATP also induced significant hyperpolar-
dent Ca2 channels in interstitial cells has not been deter-
ization responses in PDGFR cells under current clamp.
mined. The Na-Ca2 exchanger type 3 (NCX3) has been
MRS2365, a P2Y1 agonist, mimicked the effects of ATP,
and MRS2500, an antagonist, inhibited ATP-activated SK suggested as an important contributor of Ca2 influx in
currents. ATP responses were largely abolished in urethral interstitial cells. Lowering extracellular Na en-
PDGFR cells of P2ry1/ mice. These data demonstrate hanced Ca2 oscillations, and NCX inhibitors, KB-R7943
that purines activate SK currents and would tend to stabilize and SEA0400, decreased Ca2 oscillations and STICs. Thus
excitability in bladder muscles, thus providing an explana- the Ca2 influx mode of NCX may drive or regulate spon-
tion for purinergic relaxation of detrusor muscles (37, 38, taneous Ca2 release events and electrical rhythmicity (40).
46, 257). How NCX3 is switched to reverse mode in interstitial cells
is not fully understood, but might occur by localized in-
creases in intracellular Na due to actions of other trans-
C. Urethra porters or cation channels.

Urethra smooth muscles also display spontaneous electrical Urethral interstitial cells may also have a role in transducing
activity. Branched cells from rabbit urethra have been iso- or modulating neural inputs from autonomic motor neu-
lated and studied by voltage-clamp and Ca2 imaging (188, rons. Adrenergic neurotransmission affects urethral tone
338). The branched cells were identified as interstitial cells via postjunctional 1 adrenoceptors (39). Norepinephrine
on the basis of vimentin expression, absence of smooth (NE) activated STICs in interstitial cells, and this is blocked
muscle myosin, and presence of a discontinuous basal lam- by 1 antagonists, a phospholipase C inhibitor, IP3 receptor
ina, sparse rough endoplasmic reticulum, abundant mito- antagonists, and CaCC blockers (341). NO is a prominent
chondria, and a well-developed smooth ER. Significant dif- inhibitory neurotransmitter in the urethra (9), and inhibi-
ferences were observed in the behaviors of branched cells tory effects are mediated through activation of protein ki-

Physiol Rev VOL 94 JULY 2014 www.prv.org 893


SANDERS ET AL.

nase G (285). NO donors and membrane-permeable cGMP ward currents or action potentials in contrast to myocytes.
analogs inhibited STICs in urethral interstitial cells by re- Outward currents were elicited by depolarization of the
ducing intracellular Ca2 waves (340). NO may also target ICC-like cells. The numbers of ICC-like cells increase in
cyclic nucleotide-gated channels (CNG), which are ex- pregnancy, and the suggestion was made that the cells might
pressed in urethral interstitial cells (381). help to stabilize uterine excitability during the later stages of
pregnancy (89, 427).
V. INTERSTITIAL CELLS IN THE
REPRODUCTIVE TRACT B. Oviduct

A. Uterus Oviducts, or fallopian tubes in humans, are smooth muscle-


lined tubular organs that transport ova from ovary to uterus
The uterus has extraordinary requirements to accomplish (284). Several stages of the reproductive process occur
the tasks of implantation and embryogenesis. The hypertro- within the oviduct and are subject to physiological regula-
phy that accommodates the growth of an embryo and main- tion by a variety of processes. The four main functions of

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


tenance of a low level of smooth muscle excitability facili- the oviduct include 1) transport of the ovum from the ovary
tate uterine function during pregnancy. Several studies have to the site of fertilization. After delivery to the oviduct,
reported the presence of ICC or ICC-like interstitial cells in oocytes are propelled down the tube, assisted by peristaltic
the human uterus (60, 89, 170, 171, 290, 345), but the role contractions and oviductal fluid moved by ciliated epithelial
of these cells in uterine contractile activity and maintenance cells, into the ampulla where fertilization takes place (134).
of pregnancy is poorly understood. 2) The oviduct aids in transport of spermatozoa from the
site of deposition to the site of fertilization within the am-
A few studies have attempted to study the role of c-Kit pulla. Delivery of sperm to the site of fertilization is also
ICC-like cells in the uterus by testing the effects of imatinib thought to involve muscular movements of the oviduct (3).
mesylate (5, 169). This drug caused concentration-depen- 3) The oviduct provides a suitable environment for the egg.
dent (100 M) inhibition of spontaneous uterine contrac- Secretions along the oviduct provide a suitable environment
tions and contractions initiated by elevated external K. It and maintain the viability of the egg during the descent
is possible that the effects of imatinib were mediated by a through the oviduct (71). 4) Transportation of the fertilized
tyrosine kinase independent signaling pathway since the ovum (embryo) to the uterus where implantation and fur-
concentrations required to affect contractions were much ther development occurs. After fertilization, the developing
greater than concentrations required to inhibit c-Kit (69). morula descends through the isthmus of the oviduct rela-
When acute and sustained application of imatinib was stud- tively rapidly, followed by successful implantation within
ied in GI muscles, it was found that chronic application for the endometrium of the uterus (70).
several days was required to affect c-Kit populations of
cells and inhibit electrical rhythmicity (23, 139). ICC net- Video imaging and spatiotemporal mapping showed that
works were lost from the small intestines after at least 3 cumulus oocyte complex (COC) movements are due to the
days of imatinib treatment (35 M), and this was similar contractile activity of the myosalpinx, and ciliary beating
to the time course of the effects of neutralizing c-Kit anti- stirs the fluid environment that provides lubrication and
bodies on ICC and pacemaker activity. In studies of gastric viability to the egg and sperm (1, 192). Thus the source of
antrum, imatinib (10 M) was shown to reduce spontane- peristaltic contractions in oviduct is important to under-
ous contractions, intracellular Ca2 transients, and basal standing the physiology of this organ. Morphological stud-
[Ca2]i levels without affecting pacemaker activity (139). ies using light and electron microscopy identified ICC-like
Thus imatinib may generally suppress spontaneous con- cells in the fallopian tube, and it was postulated the cells
tractions of smooth muscles by blocking L-type Ca2 chan- might provide pacemaker activity (290, 291, 346). These
nels. studies described elongated c-Kit cells with dendritic pro-
cesses located within the circular muscle layer and also be-
Studies of cells isolated from uterine muscles and identified tween the circular and longitudinal muscle layers in all seg-
as interstitial cells are limited. Cells distinct from myocytes, ments of the oviduct. The elongated c-Kit cells were dis-
based on morphology and ultrastructure, were obtained tinct from the rounded and unbranched mast cells also
from pregnant human and rat uterus (89, 427). The ICC- detected within the muscularis. c-Kit cells were also de-
like cells were multipolar with spiderlike projections and tected beneath the epithelium in a layer that was 10 15 m
enlarged nuclear regions, but c-Kit immunoreactivity was thick. The ICC-like cells represented 9 and 7% of total
not resolved (89). The cells were noncontractile and con- cell population in the lamina propria and muscularis, re-
tained no myofilaments, but numerous mitochondria, cave- spectively (291). Although no direct functional evidence
olae, and intermediate filaments were observed and the cells was presented, based on physiological studies in the GI
were immunopositive for vimentin (89). The cells had rest- tract, it was proposed that the ICC-like cells might provide
ing membrane potentials of 58 mV and displayed no in- pacemaker activity and/or mediate neurotransmission.

894 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

ICC have also been described in oviducts of mouse and hen Ca2 influx through L-type Ca2 channels is a crucial fac-
(85, 117). In mice and monkeys, ICC, termed ICC-OVI, are tor for intracellular Ca2 oscillations. However, an early
found within the myosalpinx and organized into an exten- study reported that the STDs were not affected by nifedi-
sive network throughout the oviduct (Figure 10, D and E). pine (94) in the same species. CPA, caffeine, or reduced
Functional studies demonstrated the importance of ICC- extracellular Ca2 also abolished spontaneous Ca2 tran-
OVI in oviduct motility. Selective removal of ICC-OVI us- sients, suggesting that intracellular Ca2 stores are involved
ing a c-Kit neutralizing antibody resulted in loss of ICC, in the generation of Ca2 transients in interstitial cells
electrical slow waves, and propulsive contractions of the (226).
oviduct (85). It is not known whether removal of ICC-OVI
leads to loss of egg propulsion and infertility as these studies Interstitial cells in the prostate are associated with sympa-
were performed in neonatal animals where ICC-OVI phe- thetic neurons, and the interstitial cells express 1-adreno-
notype is susceptible to neutralizing antibody (23). ceptors and the gap junction protein connexin 43 as dem-
PDGFR cells are also widely distributed in the oviduct onstrated by immunohistochemistry (394). Norepinephrine
(FIGURE 10F). evoked inward currents in isolated cells identified as inter-
stitial cells, and these responses were blocked by phentol-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


Mice infected with Chlamydia muridarum develop dilated amine and prazosin, suggesting the responses to norepi-
oviducts, pyosalpinx, and loss of spontaneous contractile nephrine were mediated by 1-adrenoceptors. These au-
activity (85). Morphological inspection showed disruption thors suggested that interstitial cells may be targets for
of ICC-OVI networks, and electrophysiological recordings adrenergic regulation of contractions in prostatic ducts.
showed loss of pacemaker activity without change in basal
smooth muscle membrane potential. Although spontaneous Interestingly, GISTs have shown a relationship with pros-
activity was absent in infected mice, membrane depolariza- tatic GIST, either primarily or as a result of metastasis (6,
tions, similar in amplitude and duration to slow waves, 77, 83). Thus expression of CD117 (c-Kit) might be useful
could be evoked by electrical field stimulation, suggesting for the diagnosis of GIST-like tumors in the prostate.
the oviducts remained excitable. Chlamydia-induced infec-
tion of oviducts was associated with increased expression of
inducible nitric oxide synthase (Nos2) and prostaglandin VI. SUMMARY AND CONCLUSIONS
synthase 2 (Ptgs2) in stellate-shaped, macrophage-like cells
that lie in close proximity to ICC-OVI in the oviduct. A Smooth muscle tissues contain a variety of interstitial cells.
correlation was shown to exist between damage to ICC- These cells provide important regulatory controls in normal
OVI and loss of pacemaker activity and increased Nos2 and pathophysiological responses of smooth muscles, and
expression. It was suggested that loss of ICC-OVI could be we are just beginning to understand the full range of behav-
a major contributing factor to the development of tubal iors imposed by interstitial cells on the mechanical perfor-
factor infertility. mance of smooth muscles. In the GI tract, several basic
regulatory functions have been attributed to interstitial cells
and a mechanistic understanding of these cells is develop-
C. Prostate ing. Electrical coupling between smooth muscle cells and
interstitial cells is a key structural feature that plugs inter-
Movement of prostatic contents from the peripheral acini stitial cells into smooth muscle cells, forming the SIP syncy-
into the prostatic ducts is driven by myogenic activity in the tium. ICC and PDGFR cells express unique conduc-
stromal wall (94). Interstitial cells, immunopositive for c- tances and generate spontaneous transient inward and out-
Kit, were identified in guinea pig prostate stroma, and these ward currents, respectively, that modulate the excitability
cells were in close contact with smooth muscle cells and of the SIP syncytium. ICC are pacemaker cells, generating
varicose axon bundles (94). Examination of tissues with electrical slow waves that conduct to smooth muscle cells
electron microscopy showed typical smooth muscle cells and provide the basis for the phasic contractions of muscle
and interstitial cells with more electron-dense cytoplasm, strips and segmental and peristaltic contractions of GI or-
ovoid nuclei, an abundance of mitochondria, rough ER, gans. ICC and PDGFR cells are innervated and contrib-
some caveolae, and an incomplete basal lamina. Thick fila- ute to the transduction of neural inputs from enteric motor
ments were not observed in these cells. These are general neurons, and they appear to be specialized, generating
ultrastructural features of the ICC of the GI tract. Intersti- responses to specific neurotransmitters. ICC have also been
tial cells have also been reported in human prostate tissues shown to have a role in mechanotransduction; however, the
(347). A slow wave-like electrical event drives phasic con- specific mechanosensitive responses of each cell type in the
tractions of prostate smooth muscles, and these events may SIP syncytium and the contributions of each cell to inte-
be driven by the c-Kit interstitial cells (270). grated mechanosensitive responses of GI organs is not fully
understood at present. More research is needed to under-
Prostate interstitial cells generate spontaneous Ca2 tran- stand the complete molecular apparatus required for devel-
sients that were abolished by nicardipine, suggesting that opment and maintenance of pacemaker activity in ICC. A

Physiol Rev VOL 94 JULY 2014 www.prv.org 895


SANDERS ET AL.

complete description of the ion channels and transporters notypes at play in smooth muscle tissues should be included
responsible for generating and sustaining pacemaker activ- in studies of cultured smooth muscles and tissue engineering
ity may provide ideas for drugs that can normalize gastric protocols to better understand the fates of cell types that are
dysrhythmias or improve slow colonic transit. Therapeutic important to the performance of native muscles.
benefits might also be realized by understanding the
postjunctional mechanisms involved in neuro- and mecha- A better understanding of the roles of interstitial cells in
notransduction. non-GI muscles is necessary. Studies of interstitial cells in
many smooth muscle organs have stalled somewhat after
It is very difficult to deduce the role of specific cell types in the initial morphological descriptions of these cells. It is
tissues as complicated as smooth muscles, particularly tempting to speculate that similar cells will be widespread in
when the cells are electrically coupled. Functional studies smooth muscle tissues and have important functions in reg-
on muscle strips and expression studies on whole muscle ulating smooth muscle excitability or in mediation of re-
extracts do not reveal the cells that express specific genes or sponses to neurotransmitters. However, the lack of pheno-
are responsible for specific functions. Having fluorescent types in various smooth muscle tissues of animals with Kit
reporters expressed in SIP cells makes it possible to isolate mutations suggests that c-Kit cells may not be present or

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


and purify each cellular component. Studies focused on the important in some smooth muscle organs. For example, the
cellular components are building data bases about the presence and functional significance of c-Kit interstitial
unique molecular and functional features of each class of cells, other than mast cells, in bladder detrusor muscles, is
cell in the SIP syncytium. The integrated output and re- questionable. Use of imatinib mesylate is a poor method of
sponses of the SIP syncytium are fundamental to GI motil- testing the function of interstitial cells because the concen-
ity. We suggest that the classic term myogenic to describe trations needed for acute effects have nonspecific effects on
the subneural, subhormonal level of motor regulation in the voltage-dependent Ca2 channels and/or the contractile
gut is limited and not inclusive of the important behaviors mechanism of smooth muscle cells. These effects are down-
and regulation provided by interstitial cells. Thus the term stream from generation of pacemaker activity and/or trans-
SIPgenic is suggested as a replacement for myogenic, or duction of neural responses by interstitial cells. Effects of
to be used as an intermediate term, to incorporate the inte- tyrosine kinase inhibitors on interstitial cells are likely to
grative functions and full range of activities and responses require long-term treatments to block the signaling func-
of smooth muscle cells and interstitial cells of the SIP syn- tions of c-Kit or PDGFR. New studies examining the pres-
cytium. ence of PDGFR cells should be performed in various
types of smooth muscles. Finding these cells in the bladder
Based on the anatomical localization and morphologies of has provided novel hypotheses about the role of interstitial
interstitial cells, there are subclasses of SIP cells. Therefore, cells in this organ. Strains of mice with reporters expressed
better vital cellular markers or reporters driven by promot- in interstitial cells should be used in studies of non-GI mus-
ers of genes expressed in specific subclasses will be needed cles. These experiments will give the opportunity to make
for efficient molecular and functional subclassifications of unequivocal identification of cells purported to be intersti-
interstitial cells. Questions about why slow waves are prop- tial cells and provide better cell preparations for determina-
agated actively by ICC-MY but not by ICC-IM of the fun- tion of interstitial cell functions.
dus, why interstitial cells develop in specific anatomical
niches, and why there are differential sensitivities to inacti- Considerable additional work is needed to understand the de-
vation of c-Kit will require a finer level of granularity in velopment and plasticity of interstitial cells. Many studies have
future studies of SIP cells. made associations between failures of interstitial cells to de-
velop or loss of cells in adults and motor dysfunction, but the
Tissue engineering efforts have considered growing smooth factor(s) responsible for developmental failures or interstitial
muscle tissues and organs for reconstructive therapies. cell loss have not been identified. One intriguing lead is that
Maintenance of the smooth muscle phenotype is a continu- many of the pathological conditions that result in loss of ICC
ing obstacle in these efforts, as smooth muscle cells rediffer- are associated with inflammatory responses. Thus it is possible
entiate readily when isolated from the native extracellular that these cells are victims of inflammatory mediators. At pres-
matrix. Another issue to consider in tissue engineering, and ent, there is little information about the response of interstitial
indeed in studies of cultured smooth muscle cells, is the cells to paracrine substances, hormones, and/or cytokines. It is
impact of losing or abnormal growth or development of possible that responses or loss of responses to one of these
interstitial cells in culture. Few studies of cultured smooth factors during the development of pathological conditions
muscles have evaluated the content or status of interstitial could negatively impact interstitial cells, causing cell death or
cells and either loss- or gain-of-function or loss of growth remodeling of the phenotype. Now that it is possible to purify
factor support might impact the behavior of smooth muscle each cellular component of the SIP syncytium, studies can be
cells in culture or in cultured muscle tissues. Expression designed to understand cellular mechanisms and cellular re-
studies that include cellular markers for the many cell phe- sponses to pathological challenges. Deep sequencing of tran-

896 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

scriptomes of each type of SIP cell will yield important insights REFERENCES
into the molecular personalities of these cells and predict the
stimuli to which the cells will respond. Having cell-specific 1. Abe H. The mammalian oviductal epithelium: regional variations in cytological and
iCRE strains will make it possible to test these hypotheses functional aspects of the oviductal secretory cells. Histol Histopathol 11: 743768,
1996.
without total reliance on pharmacology. Fate mapping studies
using molecular markers will make it possible to finally deter- 2. Abe Y, Tomita T. Cable properties of smooth muscle. J Physiol 196: 87100, 1968.
mine what happens to interstitial cells in response to patholog- 3. Adams CE. Rate of sperm transport in the female reproductive tract of the rabbit. J
ical challenges. If remodeling of the interstitial cell phenotype Endocrinol 13: xxixxii, 1956.
is responsible for the disappearance of interstitial cells in 4. Alberti E, Mikkelsen HB, Wang XY, Diaz M, Larsen JO, Huizinga JD, Jimenez M.
pathophysiological states, then it may be possible to learn the Pacemaker activity and inhibitory neurotransmission in the colon of Ws/Ws mutant
signaling that controls remodeling and loss of interstitial cell rats. Am J Physiol Gastrointest Liver Physiol 292: G1499 G1510, 2007.
functions and reestablishment of interstitial cells after patho- 5. Allix S, Reyes-Gomez E, Aubin-Houzelstein G, Noel D, Tiret L, Panthier JJ, Bernex F.
logical conditions are resolved. Detailed knowledge of SIP cell Uterine contractions depend on KIT-positive interstitial cells in the mouse: genetic
transcriptomes may help identify biomarkers that will provide and pharmacological evidence. Biol Reprod 79: 510 517, 2008.
noninvasive tests to assess the health of specific types of SIP 6. Anagnostou E, Miliaras D, Panagiotakopoulos V. Diagnosis of gastrointestinal stromal

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


cells in disease. As in most areas of biomedical research, mo- tumor (GIST) on transurethral resection of the prostate: a case report and review of
lecular technologies have made rapid progress on the physiol- the literature. Int J Surg Pathol 19: 632 636, 2011.

ogy and pathophysiology of interstitial cells possible. New 7. Anderson UA, Carson C, McCloskey KD. KCNQ currents and their contribution to
dimensions of understanding of the significance of interstitial resting membrane potential and the excitability of interstitial cells of Cajal from the
cells in the behavior of smooth muscle organs will result from guinea pig bladder. J Urol 182: 330 336, 2009.

these endeavors. 8. Andersson KE, Arner A. Urinary bladder contraction and relaxation: physiology and
pathophysiology. Physiol Rev 84: 935986, 2004.

ACKNOWLEDGMENTS 9. Andersson KE, Wein AJ. Pharmacology of the lower urinary tract: basis for current
and future treatments of urinary incontinence. Pharmacol Rev 56: 581 631, 2004.
We thank the many post-docs and students that have 10. Angeli TR, Du P, Paskaranandavadivel N, Janssen PW, Beyder A, Lentle RG, Bissett IP,
worked in our labs on interstitial cells. We are eternally Cheng LK, OGrady G. The bioelectrical basis and validity of gastrointestinal extracel-
grateful to Nancy Horowitz and Yulia Bayguinov for their lular slow wave recordings. J Physiol 591: 4567 4579, 2013.

technical support on all projects related to interstitial cells. 11. Antonucci A, Fronzoni L, Cogliandro L, Cogliandro RF, Caputo C, De Giorgio R,
We are especially grateful to Drs. Masaaki Kurahashi, Peter Pallotti F, Barbara G, Corinaldesi R, Stanghellini V. Chronic intestinal pseudo-obstruc-
Blair, and Rose Ellen Dixon for images used in FIGURES 1 tion. World J Gastroenterol 14: 29532961, 2008.

AND 10; Dr. Grant Hennig for FIGURE 9; and Professor Wei 12. Arrighi S, Bosi G, Groppetti D, Cremonesi F. Identification of C-kit-positive interstitial
Yan for helpful discussions about gene inactivation studies. cells in the dog lower urinary tract and relationship with smooth muscle and nerves.
Hypotheses for a likely pacemaker role. Vet Med Int 2010: 17, 2010.
We are also most grateful for the insightful comments and
collaborations with Professor David Hirst and Dr. Gerard 13. Baker SA, Hennig GW, Salter AK, Kurahashi M, Ward SM, Sanders KM. Distribution
Sergeant regarding the pacemaker mechanism in ICC. and Ca2 signaling of fibroblast-like (PDGFR) cells in the murine gastric fundus. J
Physiol 591: 6193 6208, 2013.

Address for reprint requests and other correspondence: K. 14. Ball ER, Matsuda MM, Dye L, Hoffmann V, Zerfas PM, Szarek E, Rich A, Chitnis AB,
Sanders, Dept. of Physiology and Cell Biology, Univ. of Stratakis CA. Ultra-structural identification of interstitial cells of Cajal in the zebrafish
Danio rerio. Cell Tissue Res 349: 483 491, 2012.
Nevada School of Medicine, Reno, NV 89557 (e-mail:
ksanders@medicine.nevada.edu). 15. Bao J, Ma HY, Schuster A, Lin YM, Yan W. Incomplete cre-mediated excision leads to
phenotypic differences between Stra8-iCre; Mov10l1 and Stra8-iCre; Mov10l1 mice.
Genesis 110, 2013.
GRANTS
16. Bardsley MR, Horvath VJ, Asuzu DT, Lorincz A, Redelman D, Hayashi Y, Popko LN,
Young DL, Lomberk GA, Urrutia RA, Farrugia G, Rubin BP, Ordog T. Kitlow stem
We are extremely grateful to the National Institutes of cells cause resistance to Kit/platelet-derived growth factor alpha inhibitors in murine
Health (via the IDeA Program and the National Institute of gastrointestinal stromal tumors. Gastroenterology 139: 942952, 2010.
Diabetes and Digestive and Kidney Diseases) for funding 17. Bauer AJ, Publicover NG, Sanders KM. Origin and spread of slow waves in canine
from R37 DK40569 and R01 DK91336 (to K. M. Sanders), gastric antral circular muscle. Am J Physiol Gastrointest Liver Physiol 249: G800 G806,
R01 DK57236 and DK078736 (to S. M. Ward), R01 1985.
DK98388 (to S. D. Koh), as well as P01 DK41315 and P20 18. Bayguinov O, Hennig GW, Sanders KM. Movement based artifacts may contaminate
GM103513 (shared by K. M. Sanders, S. M. Ward, and extracellular electrical recordings from GI muscles. Neurogastroenterol Motil 23:
S. D. Koh). 1029 1042, 2011.

19. Bayguinov O, Vogalis F, Morris B, Sanders KM. Patterns of electrical activity and neural
responses in canine proximal duodenum. Am J Physiol Gastrointest Liver Physiol 263:
DISCLOSURES G887G894, 1992.

20. Bayguinov O, Ward SM, Kenyon JL, Sanders KM. Voltage-gated Ca2 currents are
No conflicts of interest, financial or otherwise, are declared necessary for slow-wave propagation in the canine gastric antrum. Am J Physiol Cell
by the authors. Physiol 293: C1645C1659, 2007.

Physiol Rev VOL 94 JULY 2014 www.prv.org 897


SANDERS ET AL.

21. Beckett EA, Bayguinov YR, Sanders KM, Ward SM, Hirst GD. Properties of unitary 43. Burns AJ. Disorders of interstitial cells of Cajal. J Pediatr Gastroenterol Nutr 45 Suppl 2:
potentials generated by intramuscular interstitial cells of Cajal in the murine and S103106, 2007.
guinea-pig gastric fundus. J Physiol 559: 259 269, 2004.
44. Burns AJ, Herbert TM, Ward SM, Sanders KM. Interstitial cells of Cajal in the guinea-
22. Beckett EA, Horiguchi K, Khoyi M, Sanders KM, Ward SM. Loss of enteric motor pig gastrointestinal tract as revealed by c-Kit immunohistochemistry. Cell Tissue Res
neurotransmission in the gastric fundus of Sl/Sl(d) mice. J Physiol 543: 871 887, 2002. 290: 1120, 1997.

23. Beckett EA, Ro S, Bayguinov Y, Sanders KM, Ward SM. Kit signaling is essential for 45. Burns AJ, Lomax AE, Torihashi S, Sanders KM, Ward SM. Interstitial cells of Cajal
development and maintenance of interstitial cells of Cajal and electrical rhythmicity in mediate inhibitory neurotransmission in the stomach. Proc Natl Acad Sci USA 93:
the embryonic gastrointestinal tract. Dev Dyn 236: 60 72, 2007. 12008 12013, 1996.

24. Beckett EA, Takeda Y, Yanase H, Sanders KM, Ward SM. Synaptic specializations exist 46. Burnstock G, Satchell DG, Smythe A. A comparison of the excitatory and inhibitory
between enteric motor nerves and interstitial cells of Cajal in the murine stomach. J effects of non-adrenergic, non-cholinergic nerve stimulation and exogenously applied
Comp Neurol 493: 193206, 2005. ATP on a variety of smooth muscle preparations from different vertebrate species. Br
J Pharmacol 46: 234 242, 1972.
25. Benham CD, Bolton TB. Patch-clamp studies of slow potential-sensitive potassium
channels in longitudinal smooth muscle cells of rabbit jejunum. J Physiol 340: 469 486, 47. Cajal SR. Histologie du Systeme Nerveux de lHomme et des Vertebres. Paris: Maloine,
1983. 1911.

26. Benham CD, Bolton TB, Denbigh JS, Lang RJ. Inward rectification in freshly isolated 48. Caputo A, Caci E, Ferrera L, Pedemonte N, Barsanti C, Sondo E, Pfeffer U, Ravazzolo
single smooth muscle cells of the rabbit jejunum. J Physiol 383: 461 476, 1987. R, Zegarra-Moran O, Galietta LJ. TMEM16A, a membrane protein associated with

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


calcium-dependent chloride channel activity. Science 322: 590 594, 2008.
27. Benham CD, Bolton TB, Lang RJ. Acetylcholine activates an inward current in single
mammalian smooth muscle cells. Nature 316: 345347, 1985. 49. Chabot B, Stephenson DA, Chapman VM, Besmer P, Bernstein A. The proto-onco-
gene c-kit encoding a transmembrane tyrosine kinase receptor maps to the mouse W
28. Benham CD, Bolton TB, Lang RJ, Takewaki T. Calcium-activated potassium channels locus. Nature 335: 88 89, 1988.
in single smooth muscle cells of rabbit jejunum and guinea-pig mesenteric artery. J
Physiol 371: 45 67, 1986. 50. Chan F, Liu Y, Sun H, Li X, Shang H, Fan D, An J, Zhou D. Distribution and possible
role of PDGF-AA and PDGFR-alpha in the gastrointestinal tract of adult guinea pigs.
29. Berezin I, Huizinga JD, Daniel EE. Interstitial cells of Cajal in the canine colon: a special Virchows Arch 457: 381388, 2010.
communication network at the inner border of the circular muscle. J Comp Neurol
273: 4251, 1988. 51. Chang IY, Glasgow NJ, Takayama I, Horiguchi K, Sanders KM, Ward SM. Loss of
interstitial cells of Cajal and development of electrical dysfunction in murine small
30. Berezin I, Huizinga JD, Daniel EE. Structural characterization of interstitial cells of bowel obstruction. J Physiol 536: 555568, 2001.
Cajal in myenteric plexus and muscle layers of canine colon. Can J Physiol Pharmcol 68:
1419 1431, 1990. 52. Chen H, Ordog T, Chen J, Young DL, Bardsley MR, Redelman D, Ward SM, Sanders
KM. Differential gene expression in functional classes of interstitial cells of Cajal in
31. Bernex F, De Sepulveda P, Kress C, Elbaz C, Delouis C, Panthier JJ. Spatial and murine small intestine. Physiol Genomics 31: 492509, 2007.
temporal patterns of c-kit-expressing cells in WlacZ/ and WlacZ/WlacZ mouse
embryos. Development 122: 30233033, 1996. 53. Chen H, Redelman D, Ro S, Ward SM, Ordog T, Sanders KM. Selective labeling and
isolation of functional classes of interstitial cells of Cajal of human and murine small
32. Berthoud HR, Powley TL. Vagal afferent innervation of the rat fundic stomach: mor- intestine. Am J Physiol Cell Physiol 292: C497C507, 2007.
phological characterization of the gastric tension receptor. J Comp Neurol 319: 261
276, 1992. 54. Chen W, Jiang C, Jin X, Shen W, Song B, Li L. Roles of stem cell factor on loss of
interstitial cells of Cajal in bladder of diabetic rats. Urology 78: 14431446, 2011.
33. Bhetwal BP, Sanders KM, An C, Trappanese DM, Moreland RS, Perrino BA. Ca2
sensitization pathways accessed by cholinergic neurotransmission in the murine gas- 55. Chess-Williams R, Chapple CR, Yamanishi T, Yasuda K, Sellers DJ. The minor popu-
tric fundus. J Physiol 591: 29712986, 2013. lation of M3-receptors mediate contraction of human detrusor muscle in vitro. J Auton
Pharmacol 21: 243248, 2001.
34. Biers SM, Reynard JM, Doore T, Brading AF. The functional effects of a c-kit tyrosine
inhibitor on guinea-pig and human detrusor. BJU Int 97: 612 616, 2006. 56. Chi MM, Powley TL. c-Kit mutant mouse behavioral phenotype: altered meal patterns
and CCK sensitivity but normal daily food intake and body weight. Am J Physiol Regul
35. Blair PJ, Bayguinov Y, Sanders KM, Ward SM. Interstitial cells in the primate gastroin- Integr Comp Physiol 285: R1170 R1183, 2003.
testinal tract. Cell Tissue Res 350: 199 213, 2012.
57. Choi KM, Gibbons SJ, Nguyen TV, Stoltz GJ, Lurken MS, Ordog T, Szurszewski JH,
36. Blair PJ, Bayguinov Y, Sanders KM, Ward SM. Relationship between enteric neurons Farrugia G. Heme oxygenase-1 protects interstitial cells of Cajal from oxidative stress
and interstitial cells in the primate gastrointestinal tract. Neurogastroenterol Motility and reverses diabetic gastroparesis. Gastroenterology 135: 20552064, 2008.
24: e437 449, 2012.
58. Christensen J. A commentary on the morphological identification of interstitial cells of
37. Boland B, Himpens B, Paques C, Casteels R, Gillis JM. ATP induced-relaxation in the Cajal in the gut. J Auton Nerv Syst 37: 75 88, 1992.
mouse bladder smooth muscle. Br J Pharmacol 108: 749 753, 1993.
59. Christensen J, Rick GA, Soll DJ. Intramural nerves and interstitial cells revealed by the
38. Bolego C, Pinna C, Abbracchio MP, Cattabeni F, Puglisi L. The biphasic response of rat Champy-Maillet stain in the opossum esophagus. J Auton Nerv Syst 19: 137151, 1987.
vesical smooth muscle to ATP. Br J Pharmcol 114: 15571562, 1995.
60. Ciontea SM, Radu E, Regalia T, Ceafalan L, Cretoiu D, Gherghiceanu M, Braga RI,
39. Brading AF, McCoy R, Dass N. alpha1-adrenoceptors in urethral function. Eur Urol 36 Malincenco M, Zagrean L, Hinescu ME, Popescu LM. C-kit immunopositive interstitial
Suppl 1: 74 79, 1999. cells (Cajal-type) in human myometrium. J Cell Mol Med 9: 407 420, 2005.

40. Bradley E, Hollywood MA, Johnston L, Large RJ, Matsuda T, Baba A, McHale NG, 61. Cobine CA, Hennig GW, Bayguinov YR, Hatton WJ, Ward SM, Keef KD. Interstitial
Thornbury KD, Sergeant GP. Contribution of reverse Na-Ca2 exchange to spon- cells of Cajal in the cynomolgus monkey rectoanal region and their relationship to
taneous activity in interstitial cells of Cajal in the rabbit urethra. J Physiol 574: 651 661, sympathetic and nitrergic nerves. Am J Physiol Gastrointest Liver Physiol 298: G643
2006. G656, 2010.

41. Bradley JE, Anderson UA, Woolsey SM, Thornbury KD, McHale NG, Hollywood MA. 62. Cobine CA, Hennig GW, Kurahashi M, Sanders KM, Ward SM, Keef KD. Relationship
Characterization of T-type calcium current and its contribution to electrical activity in between interstitial cells of Cajal, fibroblast-like cells and inhibitory motor nerves in
rabbit urethra. Am J Physiol Cell Physiol 286: C1078 C1088, 2004. the internal anal sphincter. Cell Tissue Res 344: 1730, 2011.

42. Buist ML, Corrias A, Poh YC. A model of slow wave propagation and entrainment 63. Collins C, Klausner AP, Herrick B, Koo HP, Miner AS, Henderson SC, Ratz PH.
along the stomach. Ann Biomed Eng 38: 30223030, 2010. Potential for control of detrusor smooth muscle spontaneous rhythmic contraction by

898 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

cyclooxygenase products released by interstitial cells of Cajal. J Cell Mol Med 13: 85. Dixon RE, Hwang SJ, Hennig GW, Ramsey KH, Schripsema JH, Sanders KM, Ward SM.
3236 3250, 2009. Chlamydia infection causes loss of pacemaker cells and inhibits oocyte transport in the
mouse oviduct. Biol Reprod 80: 665 673, 2009.
64. Connor JA, Kreulen D, Prosser CL, Weigel R. Interaction between longitudinal and
circular muscle in intestine of cat. J Physiol 273: 665 689, 1977. 86. Droogmans G, Callewaert G. Ca2-channel current and its modification by the dihy-
dropyridine agonist BAY k 8644 in isolated smooth muscle cells. Pflgers Arch 406:
65. Connor JA, Prosser CL, Weems WA. A study of pace-maker activity in intestinal 259 265, 1986.
smooth muscle. J Physiol 240: 671701, 1974.
87. Du P, OGrady G, Egbuji JU, Lammers WJ, Budgett D, Nielsen P, Windsor JA, Pullan
66. Cook RD, Burnstock G. The ultrastructure of Auerbachs plexus in the guinea-pig. II. AJ, Cheng LK. High-resolution mapping of in vivo gastrointestinal slow wave activity
Non-neuronal elements. J Neurocytol 5: 195206, 1976. using flexible printed circuit board electrodes: methodology and validation. Ann
Biomed Eng 37: 839 846, 2009.
67. Cotton KD, Hollywood MA, McHale NG, Thornbury KD. Ca2 current and Ca2-
activated chloride current in isolated smooth muscle cells of the sheep urethra. J 88. Duchon G, Henderson R, Danniel EE. Circular muscle layers in the small intestine. In:
Physiol 505: 121131, 1997. Proceedings of the International Symposium of Gastrointestinal Motility, edited by Daniel
EE. Vancouver, Canada: Mitchell, 1974, p. 635 646.
68. Cousins HM, Edwards FR, Hickey H, Hill CE, Hirst GD. Electrical coupling between
the myenteric interstitial cells of Cajal and adjacent muscle layers in the guinea-pig 89. Duquette RA, Shmygol A, Vaillant C, Mobasheri A, Pope M, Burdyga T, Wray S.
gastric antrum. J Physiol 550: 829 844, 2003. Vimentin-positive, c-kit-negative interstitial cells in human and rat uterus: a role in
pacemaking? Biol Reprod 72: 276 283, 2005.
69. Cretoiu SM, Simionescu AA, Caravia L, Curici A, Cretoiu D, Popescu LM. Complex

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


effects of imatinib on spontaneous and oxytocin-induced contractions in human non- 90. Durdle NG, Kingma YJ, Bowes KL, Chambers MM. Origin of slow waves in the canine
colon. Gastroenterology 84: 375382, 1983.
pregnant myometrium. Acta Physiol Hung 98: 329 338, 2011.
91. Ekblad E, Sjuve R, Arner A, Sundler F. Enteric neuronal plasticity and a reduced
70. Croxatto HB. Physiology of gamete and embryo transport through the fallopian tube.
number of interstitial cells of Cajal in hypertrophic rat ileum. Gut 42: 836 844, 1998.
Reprod Biomed Online 4: 160 169, 2002.
92. El-Sharkawy TY, Daniel EE. Electrical activity of small intestinal smooth muscle and its
71. Croxatto HB, Ortiz ME. Egg transport in the fallopian tube. Gynecol Invest 6: 215225,
temperature dependence. Am J Physiol 229: 1268 1276, 1975.
1975.
93. Espinosa I, Lee CH, Kim MK, Rouse BT, Subramanian S, Montgomery K, Varma S,
72. Cunningham RM, Larkin P, McCloskey KD. Ultrastructural properties of interstitial Corless CL, Heinrich MC, Smith KS, Wang Z, Rubin B, Nielsen TO, Seitz RS, Ross DT,
cells of Cajal in the guinea pig bladder. J Urol 185: 11231131, 2011. West RB, Cleary ML, van de Rijn M. A novel monoclonal antibody against DOG1 is a
sensitive and specific marker for gastrointestinal stromal tumors. Am J Surg Pathol 32:
73. Daniel EE, Posey-Daniel V. Neuromuscular structures in opossum esophagus:
210 218, 2008.
role of interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 246: G305
G315, 1984. 94. Exintaris B, Klemm MF, Lang RJ. Spontaneous slow wave and contractile activity of the
guinea pig prostate. J Urol 168: 315322, 2002.
74. David SG, Cebrian C, Vaughan ED Jr, Herzlinger D. c-kit and ureteral peristalsis. J Urol
173: 292295, 2005. 95. Fang S, Christensen J. Distribution of NADPH diaphorase in intramural plexuses of cat
and opossum esophagus. J Auton Nerv Syst 46: 123133, 1994.
75. Davidson RA, McCloskey KD. Morphology and localization of interstitial cells in the
guinea pig bladder: structural relationships with smooth muscle and neurons. J Urol 96. Farrugia G. Ionic conductances in gastrointestinal smooth muscles and interstitial cells
173: 13851390, 2005. of Cajal. Annu Rev Physiol 61: 45 84, 1999.

76. De JR, Grol S, van Koeveringe GA, van Kerrebroeck PE, de VJ, Gillespie JI. The 97. Farrugia G, Holm AN, Rich A, Sarr MG, Szurszewski JH, Rae JL. A mechanosensitive
localization of cyclo-oxygenase immuno-reactivity (COX I-IR) to the urothelium and calcium channel in human intestinal smooth muscle cells. Gastroenterology 117: 900
to interstitial cells in the bladder wall. J Cell Mol Med 13: 3069 3081, 2009. 905, 1999.

77. De la Roza G, Naqvi A, Clark K. Gastrointestinal stromal tumors presenting as a 98. Faussone Pellegrini MS, Cortesini C, Romagnoli P. [Ultrastructure of the tunica mus-
prostatic mass. Can J Urol 16: 4502 4506, 2009. cularis of the cardial portion of the human esophagus and stomach, with special
reference to the so-called Cajals interstitial cells]. Ital J Anat Embryol 82: 157177,
78. Demetri GD, von Mehren M, Blanke CD, Van den Abbeele AD, Eisenberg B, Roberts 1977.
PJ, Heinrich MC, Tuveson DA, Singer S, Janicek M, Fletcher JA, Silverman SG, Silber-
man SL, Capdeville R, Kiese B, Peng B, Dimitrijevic S, Druker BJ, Corless C, Fletcher 99. Faussone-Pellegrini MS, Pantalone D, Cortesini C. Smooth muscle cells, interstitial
CD, Joensuu H. Efficacy and safety of imatinib mesylate in advanced gastrointestinal cells of Cajal and myenteric plexus interrelationships in the human colon. Acta Anat
stromal tumors. N Engl J Med 347: 472 480, 2002. 139: 31 44, 1990.

100. Faussone-Pellegrini MS, Vannucchi MG, Alaggio R, Strojna A, Midrio P. Morphology of


79. Deng J, He P, Zhong X, Wang Q, Li L, Song B. Identification of T-type calcium channels
the interstitial cells of Cajal of the human ileum from foetal to neonatal life. J Cell Mol
in the interstitial cells of Cajal in rat bladder. Urology 80: 1389.e14727, 2012.
Med 11: 482 494, 2007.
80. Der-Silaphet T, Malysz J, Hagel S, Larry Arsenault A, Huizinga JD. Interstitial cells of
101. Faville RA, Pullan AJ, Sanders KM, Koh SD, Lloyd CM, Smith NP. Biophysically based
Cajal direct normal propulsive contractile activity in the mouse small intestine. Gas-
mathematical modeling of interstitial cells of Cajal slow wave activity generated from
troenterology 114: 724 736, 1998.
a discrete unitary potential basis. Biophys J 96: 4834 4852, 2009.
81. Dickens EJ, Edwards FR, Hirst GD. Selective knockout of intramuscular interstitial
102. Faville RA, Pullan AJ, Sanders KM, Smith NP. A biophysically based mathematical
cells reveals their role in the generation of slow waves in mouse stomach. J Physiol 531: model of unitary potential activity in interstitial cells of Cajal. Biophys J 95: 88 104,
827 833, 2001. 2008.
82. Dickens EJ, Hirst GD, Tomita T. Identification of rhythmically active cells in guinea-pig 103. Forrest AS, Hennig GW, Jokela-Willis S, Park CD, Sanders KM. Prostaglandin regula-
stomach. J Physiol 514: 515531, 1999. tion of gastric slow waves and peristalsis. Am J Physiol Gastrointest Liver Physiol 296:
G1180 G1190, 2009.
83. Dickson BC, Srigley JR, Pollett AF, Blackstein ME, Honey JD, Juco JW. Rectal gastro-
intestinal stromal tumor mimicking a primary prostatic lesion. Can J Urol 15: 4112 104. Forrest AS, Ordog T, Sanders KM. Neural regulation of slow-wave frequency in the
4114, 2008. murine gastric antrum. Am J Physiol Gastrointest Liver Physiol 290: G486 G495, 2006.

84. Dickson EJ, Spencer NJ, Hennig GW, Bayguinov PO, Ren J, Heredia DJ, Smith TK. An 105. Forster J, Damjanov I, Lin Z, Sarosiek I, Wetzel P, McCallum RW. Absence of the
enteric occult reflex underlies accommodation and slow transit in the distal large interstitial cells of Cajal in patients with gastroparesis and correlation with clinical
bowel. Gastroenterology 132: 19121924, 2007. findings. J Gastrointest Surg 9: 102108, 2005.

Physiol Rev VOL 94 JULY 2014 www.prv.org 899


SANDERS ET AL.

106. Franck H, Sweeney KM, Sanders KM, Shuttleworth CW. Effects of a novel guanylate 128. Gray SM, McGeown JG, McMurray G, McCloskey KD. Functional innervation of
cyclase inhibitor on nitric oxide-dependent inhibitory neurotransmission in canine Guinea-pig bladder interstitial cells of cajal subtypes: neurogenic stimulation evokes in
proximal colon. Br J Pharmacol 122: 12231229, 1997. situ calcium transients. PloS One 8: e53423, 2013.

107. Fry CH, Sui GP, Kanai AJ, Wu C. The function of suburothelial myofibroblasts in the 129. Grol S, Essers PB, van Koeveringe GA, Martinez-Martinez P, de VJ, Gillespie JI. M(3)
bladder. Neurourol Urodyn 26: 914 919, 2007. muscarinic receptor expression on suburothelial interstitial cells. BJU Int 104: 398
405, 2009.
108. Fujita A, Takeuchi T, Jun H, Hata F. Localization of Ca2-activated K channel, SK3,
in fibroblast-like cells forming gap junctions with smooth muscle cells in the mouse 130. Grol S, Nile CJ, Martinez-Martinez P, van KG, de WS, de VJ, Gillespie JI. M3 muscarinic
small intestine. J Pharmacol Sci 92: 35 42, 2003. receptor-like immunoreactivity in sham operated and obstructed guinea pig bladders.
J Urol 185: 1959 1966, 2011.
109. Furness JB. The enteric nervous system and neurogastroenterology. Nat Rev Gastro-
enterol 9: 286 294, 2012. 131. Groneberg D, Konig P, Lies B, Jager R, Seidler B, Klein S, Saur D, Friebe A. Cell-
Specific deletion of nitric oxide-sensitive guanylyl cyclase reveals a dual pathway for
110. Gabella G. Cells of visceral smooth muscles. J Smooth Muscle Res 48: 6595, 2012. nitrergic neuromuscular transmission in the murine fundus. Gastroenterology 145:
188 196, 2013.
111. Gabella G. Hypertrophy of intestinal smooth muscle. Cell Tissue Res 163: 199 214,
1975. 132. Grover M, Bernard CE, Pasricha PJ, Lurken MS, Faussone-Pellegrini MS, Smyrk TC,
112. Gabella G. Hypertrophy of visceral smooth muscle. Anat Embryol 182: 409 424, Parkman HP, Abell TL, Snape WJ, Hasler WL, McCallum RW, Nguyen L, Koch KL,
1990. Calles J, Lee L, Tonascia J, Unalp-Arida A, Hamilton FA, Farrugia G, Consortium

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


NGCR. Clinical-histological associations in gastroparesis: results from the Gastropa-
113. Gabella G. Innervation of the intestinal muscular coat. J Neurocytol 1: 341362, 1972. resis Clinical Research Consortium. Neurogastroenterol Motil 24: 531539, e249,
2012.
114. Gabella G. The structural relations between nerve fibres and muscle cells in the
urinary bladder of the rat. J Neurocytol 24: 159 187, 1995. 133. Grover M, Farrugia G, Lurken MS, Bernard CE, Faussone-Pellegrini MS, Smyrk TC,
Parkman HP, Abell TL, Snape WJ, Hasler WL, Unalp-Arida A, Nguyen L, Koch KL,
115. Gabella G, Uvelius B. Urinary bladder of rat: fine structure of normal and hypertrophic Calles J, Lee L, Tonascia J, Hamilton FA, Pasricha PJ, Consortium NGCR. Cellular
musculature. Cell Tissue Res 262: 6779, 1990. changes in diabetic and idiopathic gastroparesis. Gastroenterology 140: 15751585,
2011.
116. Gallego D, Hernandez P, Clave P, Jimenez M. P2Y1 receptors mediate inhibitory
purinergic neuromuscular transmission in the human colon. Am J Physiol Gastrointest 134. Halbert SA, Tam PY, Blandau RJ. Egg transport in the rabbit oviduct: the roles of cilia
Liver Physiol 291: G584 G594, 2006. and muscle. Science 191: 10521053, 1976.

117. Gandahi JA, Chen SF, Yang P, Bian XG, Chen QS. Ultrastructural identification of 135. Hamilton TG, Klinghoffer RA, Corrin PD, Soriano P. Evolutionary divergence of
interstitial cells of Cajal in hen oviduct. Poult Sci 91: 1410 1417, 2012. platelet-derived growth factor alpha receptor signaling mechanisms. Mol Cell Biol 23:
4013 4025, 2003.
118. Geissler EN, Ryan MA, Housman DE. The dominant-white spotting (W) locus of the
mouse encodes the c-kit proto-oncogene. Cell 55: 185192, 1988. 136. Hanani M, Farrugia G, Komuro T. Intercellular coupling of interstitial cells of cajal in
the digestive tract. Int Rev Cytol 242: 249 282, 2005.
119. Gibbons SJ, Rich A, Distad MA, Miller SM, Schmalz PF, Szurszewski JH, Sha L, Blume-
Jensen P, Farrugia G. Kit/stem cell factor receptor-induced phosphatidylinositol 3- 137. Hara Y, Kubota M, Szurszewski JH. Electrophysiology of smooth muscle of the small
kinase signalling is not required for normal development and function of interstitial intestine of some mammals. J Physiol 372: 501520, 1986.
cells of Cajal in mouse gastrointestinal tract. Neurogastroenterol Motil 15: 643 653,
2003. 138. Harhun MI, Gordienko DV, Povstyan OV, Moss RF, Bolton TB. Function of interstitial
cells of Cajal in the rabbit portal vein. Circ Res 95: 619 626, 2004.
120. Gibbons SJ, Strege PR, Lei S, Roeder JL, Mazzone A, Ou Y, Rich A, Farrugia G. The
alpha1H Ca2 channel subunit is expressed in mouse jejunal interstitial cells of Cajal 139. Hashitani H, Hayase M, Suzuki H. Effects of imatinib mesylate on spontaneous elec-
and myocytes. J Cell Mol Med 13: 4422 4431, 2009. trical and mechanical activity in smooth muscle of the guinea-pig stomach. Br J Phar-
macol 154: 451 459, 2008.
121. Gillespie JI, Markerink-van IM, de VJ. Endogenous nitric oxide/cGMP signalling in the
guinea pig bladder: evidence for distinct populations of sub-urothelial interstitial cells. 140. Hashitani H, Lang RJ. Functions of ICC-like cells in the urinary tract and male genital
Cell Tissue Res 325: 325332, 2006. organs. J Cell Mol Med 14: 1199 1121, 2010.

122. Gomez-Pinilla PJ, Gibbons SJ, Bardsley MR, Lorincz A, Pozo MJ, Pasricha PJ, Van de 141. Hashitani H, Suzuki H. Properties of spontaneous Ca2 transients recorded from
Rijn M, West RB, Sarr MG, Kendrick ML, Cima RR, Dozois EJ, Larson DW, Ordog T, interstitial cells of Cajal-like cells of the rabbit urethra in situ. J Physiol 583: 505519,
Farrugia G. Ano1 is a selective marker of interstitial cells of Cajal in the human and 2007.
mouse gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 296: G1370
G1381, 2009. 142. Hashitani H, Van Helden DF, Suzuki H. Properties of spontaneous depolarizations in
circular smooth muscle cells of rabbit urethra. Br J Pharmacol 118: 16271632, 1996.
123. Gosling JA, Dixon JS. Morphologic evidence that rhe renal calyx and pelvis control
ureteric activity in the rabbit. Am J Anat 130: 393 408, 1971. 143. Hasler WL. Gastroparesis: pathogenesis, diagnosis and management. Nat Rev Gastro-
enterol Hepatol 8: 438 453, 2011.
124. Gosling JA, Dixon JS. Species variation in the location of upper urinary tract pacemaker
cells. Invest Urol 11: 418 423, 1974. 144. He P, Deng J, Zhong X, Zhou Z, Song B, Li L. Identification of a hyperpolarization-
activated cyclic nucleotide-gated channel and its subtypes in the urinary bladder of the
125. Goto K, Matsuoka S, Noma A. Two types of spontaneous depolarizations in the rat. Urology 79: 14111413, 2012.
interstitial cells freshly prepared from the murine small intestine. J Physiol 559: 411
422, 2004. 145. Heinrich MC, Corless CL, Duensing A, McGreevey L, Chen CJ, Joseph N, Singer S,
Griffith DJ, Haley A, Town A, Demetri GD, Fletcher CD, Fletcher JA. PDGFRA
126. Goyal RK, Chaudhury A. Mounting evidence against the role of ICC in neurotrans- activating mutations in gastrointestinal stromal tumors. Science 299: 708 710, 2003.
mission to smooth muscle in the gut. Am J Physiol Gastrointest Liver Physiol 298:
G10 G13, 2010. 146. Hennig GW, Hirst GD, Park KJ, Smith CB, Sanders KM, Ward SM, Smith TK. Propa-
gation of pacemaker activity in the guinea-pig antrum. J Physiol 556: 585599, 2004.
127. Grady EF, Baluk P, Bohm S, Gamp PD, Wong H, Payan DG, Ansel J, Portbury AL,
Furness JB, McDonald DM, Bunnett NW. Characterization of antisera specific to 147. Hennig GW, Spencer NJ, Jokela-Willis S, Bayguinov PO, Lee HT, Ritchie LA, Ward SM,
NK1, NK2, and NK3 neurokinin receptors and their utilization to localize receptors in Smith TK, Sanders KM. ICC-MY coordinate smooth muscle electrical and mechanical
the rat gastrointestinal tract. J Neurosci 16: 6975 6986, 1996. activity in the murine small intestine. Neurogastroenterol Motil 22: e138 151, 2010.

900 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

148. Herrera GM, Pozo MJ, Zvara P, Petkov GV, Bond CT, Adelman JP, Nelson MT. 168. Huizinga JD, Zarate N, Farrugia G. Physiology, injury, and recovery of interstitial cells
Urinary bladder instability induced by selective suppression of the murine small con- of Cajal: basic and clinical science. Gastroenterology 137: 1548 1556, 2009.
ductance calcium-activated potassium (SK3) channel. J Physiol 551: 893903, 2003.
169. Hutchings G, Deprest J, Nilius B, Roskams T, De Ridder D. The effect of imatinib
149. Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, Kawano K, mesylate on the contractility of isolated rabbit myometrial strips. Gynecol Obstet Invest
Hanada M, Kurata A, Takeda M, Muhammad Tunio G, Matsuzawa Y, Kanakura Y, 62: 79 83, 2006.
Shinomura Y, Kitamura Y. Gain-of-function mutations of c-kit in human gastrointes-
tinal stromal tumors. Science 279: 577580, 1998. 170. Hutchings G, Gevaert T, Deprest J, Roskams T, Van Lommel A, Nilius B, De Ridder D.
Immunohistochemistry using an antibody to unphosphorylated connexin 43 to iden-
150. Hirota S, Isozaki K, Nishida T, Kitamura Y. Effects of loss-of-function and gain-of- tify human myometrial interstitial cells. Reprod Biol Endocrinol 6: 43, 2008.
function mutations of c-kit on the gastrointestinal tract. J Gastroenterol 35 Suppl 12:
7579, 2000. 171. Hutchings G, Williams O, Cretoiu D, Ciontea SM. Myometrial interstitial cells and the
coordination of myometrial contractility. J Cell Mol Med 13: 4268 4282, 2009.
151. Hirota S, Ohashi A, Nishida T, Isozaki K, Kinoshita K, Shinomura Y, Kitamura Y.
Gain-of-function mutations of platelet-derived growth factor receptor alpha gene in 172. Hwang SJ, Blair PJ, Britton FC, ODriscoll KE, Hennig G, Bayguinov YR, Rock JR, Harfe
gastrointestinal stromal tumors. Gastroenterology 125: 660 667, 2003. BD, Sanders KM, Ward SM. Expression of anoctamin 1/TMEM16A by interstitial cells
of Cajal is fundamental for slow wave activity in gastrointestinal muscles. J Physiol 587:
152. Hirst GD, Beckett EA, Sanders KM, Ward SM. Regional variation in contribution of 4887 4904, 2009.
myenteric and intramuscular interstitial cells of Cajal to generation of slow waves in
mouse gastric antrum. J Physiol 540: 10031012, 2002. 173. Hwang SJ, Blair PJ, Durnin L, Mutafova-Yambolieva V, Sanders KM, Ward SM. P2Y1
purinoreceptors are fundamental to inhibitory motor control of murine colonic ex-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


153. Hirst GD, Bramich NJ, Teramoto N, Suzuki H, Edwards FR. Regenerative component citability and transit. J Physiol 590: 19571972, 2012.
of slow waves in the guinea-pig gastric antrum involves a delayed increase in [Ca2]i
and Cl channels. J Physiol 540: 907919, 2002. 174. Iino S, Horiguchi K, Horiguchi S, Nojyo Y. c-Kit-negative fibroblast-like cells express
platelet-derived growth factor receptor alpha in the murine gastrointestinal muscu-
154. Hirst GD, Edwards FR. Electrical events underlying organized myogenic contractions lature. Histochem Cell Biol 131: 691702, 2009.
of the guinea pig stomach. J Physiol 576: 659 665, 2006.
175. Iino S, Horiguchi K, Nojyo Y. Interstitial cells of Cajal are innervated by nitrergic
155. Hirst GD, Edwards FR. Generation of slow waves in the antral region of guinea-pig nerves and express nitric oxide-sensitive guanylate cyclase in the guinea-pig gastroin-
stomach: a stochastic process. J Physiol 535: 165180, 2001. testinal tract. Neuroscience 152: 437 448, 2008.

156. Hollywood MA, Sergeant GP, McHale NG, Thornbury KD. Activation of Ca2-acti- 176. Iino S, Horiguchi K, Nojyo Y, Ward SM, Sanders KM. Interstitial cells of Cajal contain
vated Cl current by depolarizing steps in rabbit urethral interstitial cells. Am J Physiol signalling molecules for transduction of nitrergic stimulation in guinea pig caecum.
Cell Physiol 285: C327C333, 2003. Neurogastroenterol Motil 21: 542550, 2009.

157. Hollywood MA, Woolsey S, Walsh IK, Keane PF, McHale NG, Thornbury KD. T- and 177. Iino S, Nojyo Y. Immunohistochemical demonstration of c-Kit-negative fibroblast-like
L-type Ca2 currents in freshly dispersed smooth muscle cells from the human prox- cells in murine gastrointestinal musculature. Arch Histol Cytol 72: 107115, 2009.
imal urethra. J Physiol 550: 753764, 2003.
178. Iino S, Ward SM, Sanders KM. Interstitial cells of Cajal are functionally innervated by
158. Horiguchi K, Komuro T. Ultrastructural characterization of interstitial cells of Cajal in excitatory motor neurones in the murine intestine. J Physiol 556: 521530, 2004.
the rat small intestine using control and Ws/Ws mutant rats. Cell Tissue Res 293:
277284, 1998. 179. Imaizumi M, Hama K. An electron microscopic study on the interstitial cells of the
gizzard in the love-bird (Uroloncha domestica). Z Zellforsch Mik Ana 97: 351357,
159. Horiguchi K, Komuro T. Ultrastructural observations of fibroblast-like cells forming 1969.
gap junctions in the W/W(nu) mouse small intestine. J Auton Nerv Syst 80: 142147,
2000. 180. Inoue R, Kitamura K, Kuriyama H. Two Ca-dependent K-channels classified by the
application of tetraethylammonium distribute to smooth muscle membranes of the
160. Horiguchi K, Sanders KM, Ward SM. Enteric motor neurons form synaptic-like junc- rabbit portal vein. Pflgers Arch 405: 173179, 1985.
tions with interstitial cells of Cajal in the canine gastric antrum. Cell Tissue Res 311:
299 313, 2003. 181. Iqbal J, Tonta MA, Mitsui R, Li Q, Kett M, Li J, Parkington HC, Hashitani H, Lang RJ.
Potassium and ANO1/TMEM16A chloride channel profiles distinguish atypical and
161. Horvath VJ, Vittal H, Lorincz A, Chen H, Almeida-Porada G, Redelman D, Ordog T. typical smooth muscle cells from interstitial cells in the mouse renal pelvis. Br J Phar-
Reduced stem cell factor links smooth myopathy and loss of interstitial cells of cajal in macol 165: 2389 2408, 2012.
murine diabetic gastroparesis. Gastroenterology 130: 759 770, 2006.
182. Ishikawa K, Komuro T. Characterization of the interstitial cells associated with the
162. Horvath VJ, Vittal H, Ordog T. Reduced insulin and IGF-I signaling, not hyperglycemia, submuscular plexus of the guinea-pig colon. Anat Embryol 194: 49 55, 1996.
underlies the diabetes-associated depletion of interstitial cells of Cajal in the murine
stomach. Diabetes 54: 1528 1533, 2005. 183. Ishikawa K, Komuro T, Hirota S, Kitamura Y. Ultrastructural identification of the
c-kit-expressing interstitial cells in the rat stomach: a comparison of control and
163. Huang F, Rock JR, Harfe BD, Cheng T, Huang X, Jan YN, Jan LY. Studies on expression Ws/Ws mutant rats. Cell Tissue Res 289: 137143, 1997.
and function of the TMEM16A calcium-activated chloride channel. Proc Natl Acad Sci
USA 106: 2141321418, 2009. 184. Iwasaki H, Kajimura M, Osawa S, Kanaoka S, Furuta T, Ikuma M, Hishida A. A defi-
ciency of gastric interstitial cells of Cajal accompanied by decreased expression of
164. Huber A, Trudrung P, Storr M, Franck H, Schusdziarra V, Ruth P, Allescher HD. neuronal nitric oxide synthase and substance P in patients with type 2 diabetes mel-
Protein kinase G expression in the small intestine and functional importance for litus. J Gastroenterol 41: 1076 1087, 2006.
smooth muscle relaxation. Am J Physiol Gastrointest Liver Physiol 275: G629 G637,
1998. 185. Jimenez M, Borderies JR, Vergara P, Wang Y, Daniel EE. Slow waves in circular muscle
of porcine ileum: structural and electrophysiological studies. Am J Physiol Gastrointest
165. Huizinga JD, Liu LW, Fitzpatrick A, White E, Gill S, Wang XY, Zarate N, Krebs L, Choi Liver Physiol 276: G393G406, 1999.
C, Starret T, Dixit D, Ye J. Deficiency of intramuscular ICC increases fundic muscle
excitability but does not impede nitrergic innervation. Am J Physiol Gastrointest Liver 186. Johnston L, Carson C, Lyons AD, Davidson RA, McCloskey KD. Cholinergic-induced
Physiol 294: G589 G594, 2008. Ca2 signaling in interstitial cells of Cajal from the guinea pig bladder. Am J Physiol
Renal Physiol 294: F645F655, 2008.
166. Huizinga JD, Reed DE, Berezin I, Wang XY, Valdez DT, Liu LW, Diamant NE. Survival
dependency of intramuscular ICC on vagal afferent nerves in the cat esophagus. Am J 187. Johnston L, Cunningham RM, Young JS, Fry CH, McMurray G, Eccles R, McCloskey
Physiol Regul Integr Comp Physiol 294: R302R310, 2008. KD. Altered distribution of interstitial cells and innervation in the rat urinary bladder
following spinal cord injury. J Cell Mol Med 16: 15331543, 2012.
167. Huizinga JD, Thuneberg L, Kluppel M, Malysz J, Mikkelsen HB, Bernstein A. W/kit
gene required for interstitial cells of Cajal and for intestinal pacemaker activity. Nature 188. Johnston L, Sergeant GP, Hollywood MA, Thornbury KD, McHale NG. Calcium
373: 347349, 1995. oscillations in interstitial cells of the rabbit urethra. J Physiol 565: 449 461, 2005.

Physiol Rev VOL 94 JULY 2014 www.prv.org 901


SANDERS ET AL.

189. Johnston L, Woolsey S, Cunningham RM, OKane H, Duggan B, Keane P, McCloskey 210. Komuro T. Atlas of Interstitail Cells of Cajal in the Gastrointestinal Tract. Dordrecht:
KD. Morphological expression of KIT positive interstitial cells of Cajal in human blad- Springer, 2012, p. .
der. J Urol 184: 370 377, 2010.
211. Komuro T. Comparative morphology of interstitial cells of Cajal: ultrastructural char-
190. Kashyap PC, Choi KM, Dutta N, Linden DR, Szurszewski JH, Gibbons SJ, Farrugia G. acterization. Microsc Res Tech 47: 267285, 1999.
Carbon monoxide reverses diabetic gastroparesis in NOD mice. Am J Physiol Gastro-
212. Komuro T. Three-dimensional observation of the fibroblast-like cells associated with
intest Liver Physiol 298: G1013G1019, 2010.
the rat myenteric plexus, with special reference to the interstitial cells of Cajal. Cell
191. Katzka DA, Morad M. Properties of calcium channels in guinea-pig gastric myocytes. Tissue Res 255: 343351, 1989.
J Physiol 413: 175197, 1989.
213. Komuro T, Seki K, Horiguchi K. Ultrastructural characterization of the interstitial cells
192. Killian GJ. Evidence for the role of oviduct secretions in sperm function, fertilization of Cajal. Arch Histol Cytol 62: 295316, 1999.
and embryo development. Anim Reprod Sci 82 83: 141153, 2004.
214. Kraichely RE, Farrugia G. Mechanosensitive ion channels in interstitial cells of Cajal
193. Kim SO, Oh BS, Chang IY, Song SH, Ahn K, Hwang EC, Oh KJ, Kwon D, Park K. and smooth muscle of the gastrointestinal tract. Neurogastroenterol Motil 19: 245
252, 2007.
Distribution of interstitial cells of Cajal and expression of nitric oxide synthase after
experimental bladder outlet obstruction in a rat model of bladder overactivity. Neur- 215. Kubota Y, Hashitani H, Shirasawa N, Kojima Y, Sasaki S, Mabuchi Y, Soji T, Suzuki H,
ourol Urodynam 30: 1639 1645, 2011. Kohri K. Altered distribution of interstitial cells in the guinea pig bladder following
bladder outlet obstruction. Neurourol Urodyn 27: 330 340, 2008.
194. Kim YC, Koh SD, Sanders KM. Voltage-dependent inward currents of interstitial cells

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


of Cajal from murine colon and small intestine. J Physiol 541: 797 810, 2002. 216. Kubota Y, Hashitani H, Shirasawa N, Kojima Y, Sasaki S, Mabuchi Y, Soji T, Suzuki H,
Kohri K. Altered distribution of interstitial cells in the guinea pig bladder following
195. Kitamura Y, Hirota S, Nishida T. Molecular pathology of c-kit proto-oncogene and
bladder outlet obstruction. Neurourol Urodyn 27: 330 340, 2008.
development of gastrointestinal stromal tumors. Ann Chir Gynaecol 87: 282286,
1998. 217. Kubota Y, Kajioka S, Biers SM, Yokota E, Kohri K, Brading AF. Investigation of the
effect of the c-kit inhibitor Glivec on isolated guinea-pig detrusor preparations. Auton
196. Kito Y, Fukuta H, Suzuki H. Components of pacemaker potentials recorded from the Neurosci 115: 64 73, 2004.
guinea pig stomach antrum. Pflgers Arch 445: 202217, 2002.
218. Kubota Y, Kojima Y, Shibata Y, Imura M, Sasaki S, Kohri K. Role of KIT-positive
197. Kito Y, Sanders KM, Ward SM, Suzuki H. Interstitial cells of Cajal generate spontane- interstitial cells of Cajal in the urinary bladder and possible therapeutic target for
ous transient depolarizations in the rat gastric fundus. Am J Physiol Gastrointest Liver overactive bladder. Adv Urol 2011: 816342, 2011.
Physiol 297: G814 G824, 2009.
219. Kunze WA, Furness JB, Bertrand PP, Bornstein JC. Intracellular recording from my-
198. Kito Y, Suzuki H. Properties of pacemaker potentials recorded from myenteric inter- enteric neurons of the guinea-pig ileum that respond to stretch. J Physiol 506: 827
stitial cells of Cajal distributed in the mouse small intestine. J Physiol 553: 803 818, 842, 1998.
2003.
220. Kurahashi M, Nakano Y, Hennig GW, Ward SM, Sanders KM. Platelet-derived growth
199. Kito Y, Ward SM, Sanders KM. Pacemaker potentials generated by interstitial cells of factor receptor alpha-positive cells in the tunica muscularis of human colon. J Cell Mol
Cajal in the murine intestine. Am J Physiol Cell Physiol 288: C710 C720, 2005. Med 16: 13971404, 2012.

200. Klein S, Seidler B, Kettenberger A, Sibaev A, Rohn M, Feil R, Allescher HD, Vander- 221. Kurahashi M, Zheng H, Dwyer L, Ward SM, Don Koh S, Sanders KM. A functional role
winden JM, Hofmann F, Schemann M, Rad R, Storr MA, Schmid RM, Schneider G, Saur for the fibroblast-like cells in gastrointestinal smooth muscles. J Physiol 589: 697
D. Interstitial cells of Cajal integrate excitatory and inhibitory neurotransmission with 710, 2011.
intestinal slow-wave activity. Nat Commun 4: 1630, 2013.
222. Kuriyama H, Kitamura K. Electrophysiological aspects of regulation of precapillary
201. Klemm MF, Exintaris B, Lang RJ. Identification of the cells underlying pacemaker vessel tone in smooth muscles of vascular tissues. J Cardiovasc Pharmcol 7 Suppl 3:
activity in the guinea-pig upper urinary tract. J Physiol 519: 867 884, 1999. S119 128, 1985.

202. Klemm MF, Lang RJ. Distribution of Ca2-activated K channel (SK2 and SK3) im- 223. Kuriyama H, Kitamura K, Itoh T, Inoue R. Physiological features of visceral smooth
munoreactivity in intestinal smooth muscles of the guinea-pig. Clin Exp Pharmacol muscle cells, with special reference to receptors and ion channels. Physiol Rev 78:
Physiol 29: 18 25, 2002. 811920, 1998.

203. Kluppel M, Huizinga JD, Malysz J, Bernstein A. Developmental origin and Kit-depen- 224. Kwon JG, Hwang SJ, Hennig GW, Bayguinov Y, McCann C, Chen H, Rossi F, Besmer
dent development of the interstitial cells of cajal in the mammalian small intestine. Dev P, Sanders KM, Ward SM. Changes in the structure and function of ICC networks in
ICC hyperplasia and gastrointestinal stromal tumors. Gastroenterology 136: 630 639,
Dyn 211: 60 71, 1998.
2009.
204. Kluppel M, Nagle DL, Bucan M, Bernstein A. Long-range genomic rearrangements
225. Lagou M, de VJ, Kirkwood TB, Hedlund P, Andersson KE, Gillespie JI, Drake MJ.
upstream of Kit dysregulate the developmental pattern of Kit expression in W57
Location of interstitial cells and neurotransmitters in the mouse bladder. BJU Int 97:
and Wbanded mice and interfere with distinct steps in melanocyte development.
13321337, 2006.
Development 124: 6577, 1997.
226. Lam M, Shigemasa Y, Exintaris B, Lang RJ, Hashitani H. Spontaneous Ca2 signaling of
205. Knowles CH, Farrugia G. Gastrointestinal neuromuscular pathology in chronic con-
interstitial cells in the guinea pig prostate. J Urol 186: 2478 2486, 2011.
stipation. Best Pract Res Clin Gastroenterol 25: 4357, 2011.
227. Lammers WJ, al-Kais A, Singh S, Arafat K, el-Sharkawy TY. Multielectrode mapping of
206. Kobayashi S, Furness JB, Smith TK, Pompolo S. Histological identification of the slow-wave activity in the isolated rabbit duodenum. J Appl Physiol 74: 1454 1461,
interstitial cells of Cajal in the guinea-pig small intestine. Arch Histol Cytol 52: 267286, 1993.
1989.
228. Lang RJ, Hashitani H, Tonta MA, Bourke JL, Parkington HC, Suzuki H. Spontaneous
207. Koh BH, Roy R, Hollywood MA, Thornbury KD, McHale NG, Sergeant GP, Hatton electrical and Ca2 signals in the mouse renal pelvis that drive pyeloureteric peristal-
WJ, Ward SM, Sanders KM, Koh SD. Platelet-derived growth factor receptor-alpha sis. Clin Exp Pharmcol Physiol 37: 509 515, 2010.
cells in mouse urinary bladder: a new class of interstitial cells. J Cell Mol Med 16:
691700, 2012. 229. Lang RJ, Hashitani H, Tonta MA, Parkington HC, Suzuki H. Spontaneous electrical and
Ca2 signals in typical and atypical smooth muscle cells and interstitial cell of Cajal-
208. Koh SD, Sanders KM, Ward SM. Spontaneous electrical rhythmicity in cultured inter- like cells of mouse renal pelvis. J Physiol 583: 1049 1068, 2007.
stitial cells of cajal from the murine small intestine. J Physiol 513: 203213, 1998.
230. Lang RJ, Hashitani H, Tonta MA, Suzuki H, Parkington HC. Role of Ca2 entry and
209. Koh SD, Ward SM, Sanders KM. Ionic conductances regulating the excitability of Ca2 stores in atypical smooth muscle cell autorhythmicity in the mouse renal pelvis.
colonic smooth muscles. Neurogastroenterol Motil 24: 705718, 2012. Br J Pharmacol 152: 1248 1259, 2007.

902 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

231. Lang RJ, Klemm MF. Interstitial cell of Cajal-like cells in the upper urinary tract. J Cell 252. Mazzone A, Bernard CE, Strege PR, Beyder A, Galietta LJ, Pasricha PJ, Rae JL, Park-
Mol Med 9: 543556, 2005. man HP, Linden DR, Szurszewski JH, Ordog T, Gibbons SJ, Farrugia G. Altered
expression of Ano1 variants in human diabetic gastroparesis. J Biol Chem 286: 13393
232. Lang RJ, Tonta MA, Zoltkowski BZ, Meeker WF, Wendt I, Parkington HC. Py- 13403, 2011.
eloureteric peristalsis: role of atypical smooth muscle cells and interstitial cells of
Cajal-like cells as pacemakers. J Physiol 576: 695705, 2006. 253. McCloskey KD. Calcium currents in interstitial cells from the guinea-pig bladder. BJU
Int 97: 1338 1343, 2006.
233. Lang RJ, Zoltkowski BZ, Hammer JM, Meeker WF, Wendt I. Electrical characteriza-
tion of interstitial cells of Cajal-like cells and smooth muscle cells isolated from the 254. McCloskey KD. Characterization of outward currents in interstitial cells from the
mouse ureteropelvic junction. J Urol 177: 15731580, 2007. guinea pig bladder. J Urol 173: 296 301, 2005.

234. Langer JC, Berezin I, Daniel EE. Hypertrophic pyloric stenosis: ultrastructural abnor- 255. McCloskey KD, Anderson UA, Davidson RA, Bayguinov YR, Sanders KM, Ward SM.
malities of enteric nerves and the interstitial cells of Cajal. J Pediatr Surg 30: 1535 Comparison of mechanical and electrical activity and interstitial cells of Cajal in urinary
1543, 1995. bladders from wild-type and W/Wv mice. Br J Pharmacol 156: 273283, 2009.

235. Langton P, Ward SM, Carl A, Norell MA, Sanders KM. Spontaneous electrical activity 256. McCloskey KD, Gurney AM. Kit positive cells in the guinea pig bladder. J Urol 168:
of interstitial cells of Cajal isolated from canine proximal colon. Proc Natl Acad Sci USA 832 836, 2002.
86: 7280 7284, 1989. 257. McMurray G, Dass N, Brading AF. Purinoceptor subtypes mediating contraction and
relaxation of marmoset urinary bladder smooth muscle. Br J Pharmcol 123: 1579
236. Langton PD, Burke EP, Sanders KM. Participation of Ca currents in colonic electrical
1586, 1998.

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


activity. Am J Physiol Cell Physiol 257: C451C460, 1989.
258. Mei F, Han J, Huang Y, Jiang ZY, Xiong CJ, Zhou DS. Plasticity of interstitial cells of
237. Lavin ST, Southwell BR, Murphy R, Jenkinson KM, Furness JB. Activation of neurokinin
cajal: a study in the small intestine of adult guinea pigs. Anat Rec 292: 985993, 2009.
1 receptors on interstitial cells of Cajal of the guinea-pig small intestine by substance
P. Histochem Cell Biol 110: 263271, 1998. 259. Mei F, Zhu J, Guo S, Zhou DS, Han J, Yu B, Li SF, Jiang ZY, Xiong CJ. An age-dependent
proliferation is involved in the postnatal development of interstitial cells of Cajal in the
238. Lecoin L, Gabella G, Le Douarin N. Origin of the c-kit-positive interstitial cells in the
small intestine of mice. Histochem Cell Biol 131: 4353, 2009.
avian bowel. Development 122: 725733, 1996.
260. Metzger R, Neugebauer A, Rolle U, Bohlig L, Till H. C-Kit receptor (CD117) in the
239. Lee H, Koh BH, Peri LE, Sanders KM, Koh SD. Functional expression of SK channels porcine urinary tract. Pediatr Surg Int 24: 6776, 2008.
in murine detrusor PDGFRalpha cells. J Physiol 591: 501513, 2013.
261. Metzger R, Schuster T, Till H, Franke FE, Dietz HG. Cajal-like cells in the upper
240. Lee H, Koh BH, Peri LE, Sanders KM, Koh SD. Purinergic inhibitory regulation of urinary tract: comparative study in various species. Pediatr Surg Int 21: 169 174, 2005.
murine detrusor muscles mediated by PDGFRalpha interstitial cells. J Physiol. In
press. 262. Mikkelsen HB, Malysz J, Huizinga JD, Thuneberg L. Action potential generation, Kit
receptor immunohistochemistry and morphology of steel-Dickie (Sl/Sld) mutant
241. Lee HT, Hennig GW, Fleming NW, Keef KD, Spencer NJ, Ward SM, Sanders KM, mouse small intestine. Neurogastroenterol Motil 10: 1126, 1998.
Smith TK. The mechanism and spread of pacemaker activity through myenteric in-
terstitial cells of Cajal in human small intestine. Gastroenterology 132: 18521865, 263. Mitra R, Morad M. Ca2 and Ca2-activated K currents in mammalian gastric
2007. smooth muscle cells. Science 229: 269 272, 1985.

242. Lee HT, Hennig GW, Fleming NW, Keef KD, Spencer NJ, Ward SM, Sanders KM, 264. Mitsui R, Komuro T. Direct and indirect innervation of smooth muscle cells of rat
Smith TK. Septal interstitial cells of Cajal conduct pacemaker activity to excite muscle stomach, with special reference to the interstitial cells of Cajal. Cell Tissue Res 309:
bundles in human jejunum. Gastroenterology 133: 907917, 2007. 219 227, 2002.

243. Lee HT, Hennig GW, Park KJ, Bayguinov PO, Ward SM, Sanders KM, Smith TK. 265. Mitsui R, Komuro T. Distribution and ultrastructure of interstitial cells of Cajal in the
Heterogeneities in ICC Ca2 activity within canine large intestine. Gastroenterology gastric antrum of wild-type and Ws/Ws rats. Anat Embryol 206: 453 460, 2003.
136: 2226 2236, 2009.
266. Miyamoto-Kikuta S, Ezaki T, Komuro T. Distribution and morphological characteris-
244. Lennartsson J, Ronnstrand L. Stem cell factor receptor/c-Kit: from basic science to tics of the interstitial cells of Cajal in the ileocaecal junction of the guinea-pig. Cell
clinical implications. Physiol Rev 92: 1619 1649, 2012. Tissue Res 338: 29 35, 2009.

267. Mogami S, Suzuki H, Tsugawa H, Fukuhara S, Hibi T. Impaired heme oxygenase-1


245. Lies B, Groneberg D, Friebe A. Correlation of cellular expression with function of
induction in the gastric antrum induces disruption of the interstitial cells of Cajal
NO-sensitive guanylyl cyclase in the murine lower urinary tract. J Physiol 591: 5365
network in a rat model of streptozotocin-induced diabetes. Neurogastroenterol Motil
5375, 2013.
25: -609 e465, 2013.
246. Liu LW, Thuneberg L, Huizinga JD. Development of pacemaker activity and interstitial
268. Monaghan KP, Johnston L, McCloskey KD. Identification of PDGFRalpha positive
cells of Cajal in the neonatal mouse small intestine. Dev Dyn 213: 271282, 1998.
populations of interstitial cells in human and guinea pig bladders. J Urol 188: 639 647,
247. Liu LW, Thuneberg L, Huizinga JD. Selective lesioning of interstitial cells of Cajal by 2012.
methylene blue and light leads to loss of slow waves. Am J Physiol Gastrointest Liver
269. Nakahara M, Isozaki K, Hirota S, Miyagawa J, Hase-Sawada N, Taniguchi M, Nishida T,
Physiol 266: G485G496, 1994. Kanayama S, Kitamura Y, Shinomura Y, Matsuzawa Y. A novel gain-of-function muta-
tion of c-kit gene in gastrointestinal stromal tumors. Gastroenterology 115: 1090
248. Lorincz A, Redelman D, Horvath VJ, Bardsley MR, Chen H, Ordog T. Progenitors of
1095, 1998.
interstitial cells of cajal in the postnatal murine stomach. Gastroenterology 134: 1083
1093, 2008. 270. Nguyen DT, Dey A, Lang RJ, Ventura S, Exintaris B. Contractility and pacemaker cells
in the prostate gland. J Urol 185: 347351, 2011.
249. Lyford GL, He CL, Soffer E, Hull TL, Strong SA, Senagore AJ, Burgart LJ, Young-Fadok
T, Szurszewski JH, Farrugia G. Pan-colonic decrease in interstitial cells of Cajal in 271. Nishida K, Wang L, Morii E, Park SJ, Narimatsu M, Itoh S, Yamasaki S, Fujishima M,
patients with slow transit constipation. Gut 51: 496 501, 2002. Ishihara K, Hibi M, Kitamura Y, Hirano T. Requirement of Gab2 for mast cell devel-
opment and KitL/c-Kit signaling. Blood 99: 1866 1869, 2002.
250. Maeda H, Yamagata A, Nishikawa S, Yoshinaga K, Kobayashi S, Nishi K, Nishikawa S.
Requirement of c-kit for development of intestinal pacemaker system. Development 272. Nishida T, Hirota S, Taniguchi M, Hashimoto K, Isozaki K, Nakamura H, Kanakura Y,
116: 369 375, 1992. Tanaka T, Takabayashi A, Matsuda H, Kitamura Y. Familial gastrointestinal stromal
tumours with germline mutation of the KIT gene. Nat Genet 19: 323324, 1998.
251. Malysz J, Thuneberg L, Mikkelsen HB, Huizinga JD. Action potential generation in the
small intestine of W mutant mice that lack interstitial cells of Cajal. Am J Physiol 273. Nose K, Suzuki H, Kannan H. Voltage dependency of the frequency of slow waves in
Gastrointest Liver Physiol 271: G387G399, 1996. antrum smooth muscle of the guinea-pig stomach. Jpn J Physiol 50: 625 633, 2000.

Physiol Rev VOL 94 JULY 2014 www.prv.org 903


SANDERS ET AL.

274. OGrady G, Du P, Cheng LK, Egbuji JU, Lammers WJ, Windsor JA, Pullan AJ. Origin 295. Powley TL, Wang XY, Fox EA, Phillips RJ, Liu LW, Huizinga JD. Ultrastructural evi-
and propagation of human gastric slow-wave activity defined by high-resolution map- dence for communication between intramuscular vagal mechanoreceptors and inter-
ping. Am J Physiol Gastrointest Liver Physiol 299: G585G592, 2010. stitial cells of Cajal in the rat fundus. Neurogastroenterol Motil 20: 69 79, 2008.

275. Ohba M, Sakamoto Y, Tomita T. The slow wave in the circular muscle of the guinea- 296. Publicover NG, Sanders KM. Effects of frequency on the wave form of propagated
pig stomach. J Physiol 253: 505516, 1975. slow waves in canine gastric antral muscle. J Physiol 371: 179 189, 1986.

276. Ordog T, Redelman D, Horowitz NN, Sanders KM. Immunomagnetic enrichment of 297. Pucovsky V, Moss RF, Bolton TB. Non-contractile cells with thin processes resem-
interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 286: G351G360, 2004. bling interstitial cells of Cajal found in the wall of guinea-pig mesenteric arteries. J
Physiol 552: 119 133, 2003.
277. Ordog T, Redelman D, Horvath VJ, Miller LJ, Horowitz B, Sanders KM. Quantitative
analysis by flow cytometry of interstitial cells of Cajal, pacemakers, and mediators of 298. Radenkovic G. Two patterns of development of interstitial cells of Cajal in the human
neurotransmission in the gastrointestinal tract. Cytometry 62: 139 149, 2004. duodenum. J Cell Mol 16: 185192, 2012.

278. Ordog T, Takayama I, Cheung WK, Ward SM, Sanders KM. Remodeling of networks 299. Radenkovic G, Abramovic M. Differentiation of interstitial cells of Cajal in the human
of interstitial cells of Cajal in a murine model of diabetic gastroparesis. Diabetes 49: distal colon. Cells Tissues Organs 196: 463 469, 2012.
17311739, 2000.
300. Radenkovic G, Ilic I, Zivanovic D, Vlajkovic S, Petrovic V, Mitrovic O. C-kit-immuno-
279. Ordog T, Ward SM, Sanders KM. Interstitial cells of cajal generate electrical slow positive interstitial cells of Cajal in human embryonal and fetal oesophagus. Cell Tissue
waves in the murine stomach. J Physiol 518: 257269, 1999. Res 340: 427 436, 2010.

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


280. Orr-Urtreger A, Avivi A, Zimmer Y, Givol D, Yarden Y, Lonai P. Developmental 301. Radenkovic G, Savic V, Mitic D, Grahovac S, Bjelakovic M, Krstic M. Development of
expression of c-kit, a proto-oncogene encoded by the W locus. Development 109: c-kit immunopositive interstitial cells of Cajal in the human stomach. J Cell Mol Med 14:
911923, 1990. 11251134, 2010.

281. Ozaki H, Stevens RJ, Blondfield DP, Publicover NG, Sanders KM. Simultaneous mea- 302. Rahnamai MS, van Koeveringe GA, Essers PB, de Wachter SG, de VJ, van Kerre-
surement of membrane potential, cytosolic Ca2, and tension in intact smooth mus- broeck PE, Gillespie JI. Prostaglandin receptor EP1 and EP2 site in guinea pig bladder
cles. Am J Physiol Cell Physiol 260: C917C925, 1991. urothelium and lamina propria. J Urol 183: 12411247, 2010.

282. Pardi DS, Miller SM, Miller DL, Burgart LJ, Szurszewski JH, Lennon VA, Farrugia G. 303. Rasmussen H, Hansen A, Smedts F, Rumessen JJ, Horn T. CD34-positive interstitial
Paraneoplastic dysmotility: loss of interstitial cells of Cajal. Am J Gastroenterol 97: cells of the human detrusor. Acta Path Micro Im 115: 1260 1266, 2007.
1828 1833, 2002.
304. Rasmussen H, Rumessen JJ, Hansen A, Smedts F, Horn T. Ultrastructure of Cajal-like
283. Park KJ, Hennig GW, Lee HT, Spencer NJ, Ward SM, Smith TK, Sanders KM. Spatial interstitial cells in the human detrusor. Cell Tissue Res 335: 517527, 2009.
and temporal mapping of pacemaker activity in interstitial cells of Cajal in mouse ileum
305. Rhee PL, Lee JY, Son HJ, Kim JJ, Rhee JC, Kim S, Koh SD, Hwang SJ, Sanders KM, Ward
in situ. Am J Physiol Cell Physiol 290: C1411C1427, 2006.
SM. Analysis of pacemaker activity in the human stomach. J Physiol 589: 6105 6118,
284. Pauerstein CJ, Woodruff JD, Zachary AS. Factors influencing physiologic activities in 2011.
the fallopian tube; the anatomy, physiology, and pharmacology of tubal transport.
306. Rich A, Gordon S, Brown C, Gibbons SJ, Schaefer K, Hennig G, Farrugia G. Kit
Obstet Gynecol Surv 23: 215243, 1968. signaling is required for development of coordinated motility patterns in zebrafish
gastrointestinal tract. Zebrafish 10: 154 160, 2013.
285. Persson K, Pandita RK, Aszodi A, Ahmad M, Pfeifer A, Fassler R, Andersson KE.
Functional characteristics of urinary tract smooth muscles in mice lacking cGMP 307. Rich A, Leddon SA, Hess SL, Gibbons SJ, Miller S, Xu X, Farrugia G. Kit-like immu-
protein kinase type I. Am J Physiol Regul Integr Comp Physiol 279: R1112R1120, 2000. noreactivity in the zebrafish gastrointestinal tract reveals putative ICC. Dev Dyn 236:
903911, 2007.
286. Pezzone MA, Watkins SC, Alber SM, King WE, de Groat WC, Chancellor MB, Fraser
MO. Identification of c-kit-positive cells in the mouse ureter: the interstitial cells of 308. Rich A, Miller SM, Gibbons SJ, Malysz J, Szurszewski JH, Farrugia G. Local presentation
Cajal of the urinary tract. Am J Physiol Renal Physiol 284: F925F929, 2003. of Steel factor increases expression of c-kit immunoreactive interstitial cells of Cajal in
culture. Am J Physiol Gastrointest Liver Physiol 284: G313G320, 2003.
287. Piaseczna PA, Rolle U, Solari V, Puri P. Interstitial cells of Cajal in the human normal
urinary bladder and in the bladder of patients with megacystis-microcolon intestinal 309. Richardson KC. Electronmicroscopic observations on Auerbachs plexus in the rabbit,
hypoperistalsis syndrome. BJU Int 94: 143146, 2004. with special reference to the problem of smooth muscle innervation. Am J Anat 103:
99 135, 1958.
288. Piaseczna-Piotrowska AM, Dzieniecka M, Kulig A, Danilewicz M, Chilarski A. Differ-
ent distribution of c-kit positive interstitial cells of Cajal-like in childrens urinary 310. Ro S, Hatton WJ, Koh SD, Horowitz B. Molecular properties of small-conductance
bladders. Folia Histochem Cytobiol 49: 431 435, 2011. Ca2-activated K channels expressed in murine colonic smooth muscle. Am J Physiol
Gastrointest Liver Physiol 281: G964 G973, 2001.
289. Pieri L, Vannucchi MG, Faussone-Pellegrini MS. Histochemical and ultrastructural
characteristics of an interstitial cell type different from ICC and resident in the muscle 311. Ro S, Park C, Jin J, Zheng H, Blair PJ, Redelman D, Ward SM, Yan W, Sanders KM. A
coat of human gut. J Cell Mol Med 12: 1944 1955, 2008. model to study the phenotypic changes of interstitial cells of Cajal in gastrointestinal
diseases. Gastroenterology 138: 1068 1078 e10611062, 2010.
290. Popescu LM, Ciontea SM, Cretoiu D. Interstitial Cajal-like cells in human uterus and
fallopian tube. Ann NY Acad Sci 1101: 139 165, 2007. 312. Robinson TL, Sircar K, Hewlett BR, Chorneyko K, Riddell RH, Huizinga JD. Gastro-
intestinal stromal tumors may originate from a subset of CD34-positive interstitial
291. Popescu LM, Ciontea SM, Cretoiu D, Hinescu ME, Radu E, Ionescu N, Ceausu M, cells of Cajal. Am J Pathol 156: 11571163, 2000.
Gherghiceanu M, Braga RI, Vasilescu F, Zagrean L, a Ardeleanu C. Novel type of
interstitial cell (Cajal-like) in human fallopian tube. J Cell Mol Med 9: 479 523, 2005. 313. Rock JR, Futtner CR, Harfe BD. The transmembrane protein TMEM16A is required
for normal development of the murine trachea. Dev Biol 321: 141149, 2008.
292. Popescu LM, Faussone-Pellegrini MS. TELOCYTES: a case of serendipity: the winding
way from Interstitial Cells of Cajal (ICC), via Interstitial Cajal-Like Cells (ICLC) to 314. Rogers DC, Burnstock G. The interstitial cell and its place in the concept of the
TELOCYTES. J Cell Mol Med 14: 729 740, 2010. autonomic ground plexus. J Comp Neurol 126: 255284, 1966.

293. Post JM, Hume JR. Ionic basis for spontaneous depolarizations in isolated smooth 315. Rumessen JJ. Identification of interstitial cells of Cajal. Significance for studies of human
muscle cells of canine colon. Am J Physiol Cell Physiol 263: C691C699, 1992. small intestine and colon. Dan Med Bull 41: 275293, 1994.

294. Powley TL, Phillips RJ. Vagal intramuscular array afferents form complexes with in- 316. Rumessen JJ, de Kerchove dExaerde A, Mignon S, Bernex F, Timmermans JP, Schiff-
terstitial cells of Cajal in gastrointestinal smooth muscle: analogues of muscle spindle mann SN, Panthier JJ, Vanderwinden JM. Interstitial cells of Cajal in the striated
organs? Neuroscience 186: 188 200, 2011. musculature of the mouse esophagus. Cell Tissue Res 306: 114, 2001.

904 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

317. Rumessen JJ, Mikkelsen HB, Qvortrup K, Thuneberg L. Ultrastructure of interstitial 339. Sergeant GP, Hollywood MA, McHale NG, Thornbury KD. Spontaneous Ca2 acti-
cells of Cajal in circular muscle of human small intestine. Gastroenterology 104: 343 vated Cl currents in isolated urethral smooth muscle cells. J Urol 166: 11611166,
350, 1993. 2001.

318. Rumessen JJ, Mikkelsen HB, Thuneberg L. Ultrastructure of interstitial cells of Cajal 340. Sergeant GP, Johnston L, McHale NG, Thornbury KD, Hollywood MA. Activation of
associated with deep muscular plexus of human small intestine. Gastroenterology 102: the cGMP/PKG pathway inhibits electrical activity in rabbit urethral interstitial cells of
56 68, 1992. Cajal by reducing the spatial spread of Ca2 waves. J Physiol 574: 167181, 2006.

319. Rumessen JJ, Peters S, Thuneberg L. Light- and electron microscopical studies of 341. Sergeant GP, Thornbury KD, McHale NG, Hollywood MA. Characterization of nor-
interstitial cells of Cajal and muscle cells at the submucosal border of human colon. Lab epinephrine-evoked inward currents in interstitial cells isolated from the rabbit ure-
Invest 68: 481 495, 1993. thra. Am J Physiol Cell Physiol 283: C885C894, 2002.

320. Rumessen JJ, Thuneberg L. Interstitial cells of Cajal in human small intestine. Ultra- 342. Sergeant GP, Thornbury KD, McHale NG, Hollywood MA. Interstitial cells of Cajal in
structural identification and organization between the main smooth muscle layers. the urethra. J Cell Mol Med 10: 280 291, 2006.
Gastroenterology 100: 14171431, 1991. 343. Serio R, Huizinga JD, Barajas-Lopez C, Daniel EE. Interstitial cells of Cajal and slow
wave generation in canine colonic circular muscle. J Auton Nerv Syst 30 Suppl: S141
321. Rumessen JJ, Thuneberg L. Plexus muscularis profundus and associated interstitial
S143, 1990.
cells. I. Light microscopical studies of mouse small intestine. Anat Rec 203: 115127,
1982. 344. Shafik A, El-Sibai O, Shafik AA, Shafik I. Identification of interstitial cells of Cajal in
human urinary bladder: concept of vesical pacemaker. Urology 64: 809 813, 2004.

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


322. Rumessen JJ, Vanderwinden JM. Interstitial cells in the musculature of the gastroin-
testinal tract: Cajal and beyond. Int Rev Cytol 229: 115208, 2003. 345. Shafik A, El-Sibai O, Shafik I. Identification of c-kit-positive cells in the uterus. Int J
Gynaecol Obstet 87: 254 255, 2004.
323. Rumessen JJ, Vanderwinden JM, Rasmussen H, Hansen A, Horn T. Ultrastructure of
interstitial cells of Cajal in myenteric plexus of human colon. Cell Tissue Res 337: 346. Shafik A, Shafik AA, El Sibai O, Shafik IA. Specialized pacemaking cells in the human
197212, 2009. Fallopian tube. Mol Hum Reprod 11: 503505, 2005.

324. Salmhofer H, Neuhuber WL, Ruth P, Huber A, Russwurm M, Allescher HD. Pivotal 347. Shafik A, Shafik I, el-Sibai O. Identification of c-kit-positive cells in the human prostate:
role of the interstitial cells of Cajal in the nitric oxide signaling pathway of rat small the interstitial cells of Cajal. Arch Androl 51: 345351, 2005.
intestine. Morphological evidence. Cell Tissue Res 305: 331340, 2001.
348. Shuttleworth CW, Xue C, Ward SM, de Vente J, Sanders KM. Immunohistochemical
325. Sancho M, Garcia-Pascual A, Triguero D. Presence of the Ca2-activated chloride localization of 3,5-cyclic guanosine monophosphate in the canine proximal colon:
channel anoctamin 1 in the urethra and its role in excitatory neurotransmission. Am J responses to nitric oxide and electrical stimulation of enteric inhibitory neurons.
Physiol Renal Physiol 302: F390 F400, 2012. Neuroscience 56: 513522, 1993.

326. Sanders KM. A case for interstitial cells of Cajal as pacemakers and mediators of 349. Sims SM, Singer JJ, Walsh JV Jr. Cholinergic agonists suppress a potassium current in
neurotransmission in the gastrointestinal tract. Gastroenterology 111: 492515, 1996. freshly dissociated smooth muscle cells of the toad. J Physiol 367: 503529, 1985.

327. Sanders KM. Role of prostaglandins in regulating gastric motility. Am J Physiol Gastro- 350. Singer JJ, Walsh JV. Large conductance Ca-activated K channels in smooth muscle cell
intest Liver Physiol 247: G117G126, 1984. membrane: reduction in unitary currents due to internal na ions. Biophys J 45: 68 70,
1984.
328. Sanders KM, Burke EP, Stevens RJ. Effects of methylene blue on rhythmic activity and
membrane potential in the canine proximal colon. Am J Physiol Gastrointest Liver Physiol 351. Sivarao DV, Mashimo H, Goyal RK. Pyloric sphincter dysfunction in nNOS/ and
256: G779 G784, 1989. W/Wv mutant mice: animal models of gastroparesis and duodenogastric reflux. Gas-
troenterology 135: 1258 1266, 2008.
329. Sanders KM, Hwang SJ, Ward SM. Neuroeffector apparatus in gastrointestinal smooth
352. Sivarao DV, Mashimo HL, Thatte HS, Goyal RK. Lower esophageal sphincter is acha-
muscle organs. J Physiol 588: 4621 4639, 2010.
lasic in nNOS(/) and hypotensive in W/W(v) mutant mice. Gastroenterology 121:
330. Sanders KM, Koh SD, Ro S, Ward SM. Regulation of gastrointestinal motility-insights 34 42, 2001.
from smooth muscle biology. Nat Rev Gastroenterol Hepatol 9: 633 645, 2012.
353. Smet PJ, Jonavicius J, Marshall VR, de VJ. Distribution of nitric oxide synthase-immu-
331. Sanders KM, Koh SD, Ward SM. Interstitial cells of cajal as pacemakers in the gastro- noreactive nerves and identification of the cellular targets of nitric oxide in guinea-pig
intestinal tract. Annu Rev Physiol 68: 307343, 2006. and human urinary bladder by cGMP immunohistochemistry. Neuroscience 71: 337
348, 1996.
332. Sanders KM, Stevens R, Burke E, Ward SW. Slow waves actively propagate at sub-
354. Smith TK, Reed JB, Sanders KM. Interaction of two electrical pacemakers in muscu-
mucosal surface of circular layer in canine colon. Am J Physiol Gastrointest Liver Physiol
laris of canine proximal colon. Am J Physiol Cell Physiol 252: C290 C299, 1987.
259: G258 G263, 1990.
355. Smith TK, Reed JB, Sanders KM. Origin and propagation of electrical slow waves in
333. Sanders KM, Szurszewski JH. Does endogenous prostaglandin affect gastric antral
circular muscle of canine proximal colon. Am J Physiol Cell Physiol 252: C215C224,
motility? Am J Physiol Gastrointest Liver Physiol 241: G191G195, 1981.
1987.
334. Sawtell NM, Lessard JL. Cellular distribution of smooth muscle actins during mam-
356. Solari V, Piotrowska AP, Puri P. Altered expression of interstitial cells of Cajal in
malian embryogenesis: expression of the alpha-vascular but not the gamma-enteric
congenital ureteropelvic junction obstruction. J Urol 170: 2420 2422, 2003.
isoform in differentiating striated myocytes. J Cell Biol 109: 2929 2937, 1989.
357. Southwell BR, Woodman HL, Murphy R, Royal SJ, Furness JB. Characterisation of
335. Schneider T, Hein P, Michel MC. Signal transduction underlying carbachol-induced substance P-induced endocytosis of NK1 receptors on enteric neurons. Histochem
contraction of rat urinary bladder. I. Phospholipases and Ca2 sources. J Pharmacol Cell Biol 106: 563571, 1996.
Exp Ther 308: 4753, 2004.
358. Spencer NJ, Hennig GW, Smith TK. Stretch-activated neuronal pathways to longitu-
336. Schroeder BC, Cheng T, Jan YN, Jan LY. Expression cloning of TMEM16A as a dinal and circular muscle in guinea pig distal colon. Am J Physiol Gastrointest Liver Physiol
calcium-activated chloride channel subunit. Cell 134: 1019 1029, 2008. 284: G231G241, 2003.

337. Seki K, Komuro T. Immunocytochemical demonstration of the gap junction proteins 359. Spencer NJ, Smith TK. Mechanosensory S-neurons rather than AH-neurons appear to
connexin 43 and connexin 45 in the musculature of the rat small intestine. Cell Tissue generate a rhythmic motor pattern in guinea-pig distal colon. J Physiol 558: 577596,
Res 306: 417 422, 2001. 2004.

338. Sergeant GP, Hollywood MA, McCloskey KD, Thornbury KD, McHale NG. Spe- 360. Sperelakis N. Electric field model: an alternative mechanism for cell-to-cell propaga-
cialised pacemaking cells in the rabbit urethra. J Physiol 526: 359 366, 2000. tion in cardiac muscle and smooth muscle. Neurogastroenterol Motil 3: 64 83, 1991.

Physiol Rev VOL 94 JULY 2014 www.prv.org 905


SANDERS ET AL.

361. Sternini C, Su D, Gamp PD, Bunnett NW. Cellular sites of expression of the neuro- 382. Vahabi B, McKay NG, Lawson K, Sellers DJ. The role of c-kit-positive interstitial cells
kinin-1 receptor in the rat gastrointestinal tract. J Comp Neurol 358: 531540, 1995. in mediating phasic contractions of bladder strips from streptozotocin-induced dia-
betic rats. BJU Int 107: 1480 1487, 2011.
362. Strege PR, Ou Y, Sha L, Rich A, Gibbons SJ, Szurszewski JH, Sarr MG, Farrugia G.
Sodium current in human intestinal interstitial cells of Cajal. Am J Physiol Gastrointest 383. Van Helden DF, Imtiaz MS, Nurgaliyeva K, von der Weid P, Dosen PJ. Role of calcium
Liver Physiol 285: G1111G1121, 2003. stores and membrane voltage in the generation of slow wave action potentials in
guinea-pig gastric pylorus. J Physiol 524: 245265, 2000.
363. Sui GP, Rothery S, Dupont E, Fry CH, Severs NJ. Gap junctions and connexin expres-
sion in human suburothelial interstitial cells. BJU Int 90: 118 129, 2002. 384. Van Helden DF, Laver DR, Holdsworth J, Imtiaz MS. Generation and propagation of
gastric slow waves. Clin Exp Pharmacol Physiol 37: 516 524, 2010.
364. Suzuki H, Takano H, Yamamoto Y, Komuro T, Saito M, Kato K, Mikoshiba K. Prop-
erties of gastric smooth muscles obtained from mice which lack inositol trisphosphate 385. Vanderwinden JM, Rumessen JJ. Interstitial cells of Cajal in human gut and gastroin-
receptor. J Physiol 525: 105111, 2000. testinal disease. Microsc Res Tech 47: 344 360, 1999.

365. Suzuki H, Ward SM, Bayguinov YR, Edwards FR, Hirst GD. Involvement of intramus- 386. Vanderwinden JM, Rumessen JJ, de Kerchove dExaerde A Jr, Gillard K, Panthier JJ, de
Laet MH, Schiffmann SN. Kit-negative fibroblast-like cells expressing SK3, a Ca2-
cular interstitial cells in nitrergic inhibition in the mouse gastric antrum. J Physiol 546:
activated K channel, in the gut musculature in health and disease. Cell Tissue Res 310:
751763, 2003.
349 358, 2002.
366. Suzuki N, Prosser CL, Dahms V. Boundary cells between longitudinal and circular
387. Vanderwinden JM, Rumessen JJ, De Laet MH, Vanderhaeghen JJ, Schiffmann SN.
layers: essential for electrical slow waves in cat intestine. Am J Physiol Gastrointest Liver
CD34 immunoreactivity and interstitial cells of Cajal in the human and mouse gastro-

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


Physiol 250: G287G294, 1986.
intestinal tract. Cell Tissue Res 302: 145153, 2000.
367. Thomas AP, Bird GS, Hajnoczky G, Robb-Gaspers LD, Putney JW Jr. Spatial and
388. Vanderwinden JM, Rumessen JJ, De Laet MH, Vanderhaeghen JJ, Schiffmann SN.
temporal aspects of cellular calcium signaling. FASEB J 10: 15051517, 1996.
CD34 cells in human intestine are fibroblasts adjacent to, but distinct from, inter-
stitial cells of Cajal. Lab Invest 79: 59 65, 1999.
368. Thomsen L, Robinson TL, Lee JC, Farraway LA, Hughes MJ, Andrews DW, Huizinga
JD. Interstitial cells of Cajal generate a rhythmic pacemaker current. Nat Med 4: 389. Vannucchi MG, Traini C, Manetti M, Ibba-Manneschi L, Faussone-Pellegrini MS. Te-
848 851, 1998. locytes express PDGFRalpha in the human gastrointestinal tract. J Cell Mol Med 17:
1099 1108, 2013.
369. Thuneberg L. Interstitial cells of Cajal: intestinal pacemaker cells? Adv Anat Embryol Cell
Biol 71: 1130, 1982. 390. Ver Donck L, Lammers WJ, Moreaux B, Smets D, Voeten J, Vekemans J, Schuurkes JA,
Coulie B. Mapping slow waves and spikes in chronically instrumented conscious dogs:
370. Thuneberg L, Johansen V, Rumessen JJ, Anderson BG. Interstitial cells of Cajal: selec-
implantation techniques and recordings. Med Biol Eng Comput 44: 170 178, 2006.
tive uptake of methylene blue inhibits slow wave activity. In: Gastrointestinal Motility,
edited by Roman C. Lancaster, UK: MTP Press Limited, 1984, p. 495502. 391. Vogalis F, Publicover NG, Hume JR, Sanders KM. Relationship between calcium cur-
rent and cytosolic calcium in canine gastric smooth muscle cells. Am J Physiol Cell
371. Thuneberg L, Peters S. Toward a concept of stretch-coupling in smooth muscle. I. Physiol 260: C1012C1018, 1991.
Anatomy of intestinal segmentation and sleeve contractions. Anat Rec 262: 110 124,
2001. 392. Vogalis F, Ward SM, Sanders KM. Correlation between electrical and morphological
properties of canine pyloric circular muscle. Am J Physiol Gastrointest Liver Physiol 260:
372. Tokutomi N, Maeda H, Tokutomi Y, Sato D, Sugita M, Nishikawa S, Nishikawa S, G390 G398, 1991.
Nakao J, Imamura T, Nishi K. Rhythmic Cl current and physiological roles of the
intestinal c-kit-positive cells. Pflgers Arch 431: 169 177, 1995. 393. Wallace AS, Burns AJ. Development of the enteric nervous system, smooth muscle
and interstitial cells of Cajal in the human gastrointestinal tract. Cell Tissue Res 319:
373. Tomita T. Electrical activity (spikes and slow waves) in gastrointestinal smooth mus- 367382, 2005.
cles. In: Smooth Muscle: An Assessment of Current Knowledge, edited by Bulbring E,
Brading A. F., Tomita T. London: Edward Arnold, 1981, p. 127156. 394. Wang JP, Ding GF, Wang QZ. Interstitial cells of Cajal mediate excitatory sympathetic
neurotransmission in guinea pig prostate. Cell Tissue Res 352: 479 486, 2013.
374. Tomita T. Electrophysiology of mammalian smooth muscle. Prog Biophys Mol Biol 30:
185203, 1975. 395. Wang XY, Huizinga JD, Diamond J, Liu LW. Loss of intramuscular and submuscular
interstitial cells of Cajal and associated enteric nerves is related to decreased gastric
375. Torihashi S, Gerthoffer WT, Kobayashi S, Sanders KM. Identification and classification emptying in streptozotocin-induced diabetes. Neurogastroenterol Motil 21: e1092
of interstitial cells in the canine proximal colon by ultrastructure and immunocyto- e1095, 2009.
chemistry. Histochemistry 101: 169 183, 1994.
396. Wang XY, Lammers WJ, Bercik P, Huizinga JD. Lack of pyloric interstitial cells of Cajal
376. Torihashi S, Kobayashi S, Gerthoffer WT, Sanders KM. Interstitial cells in deep mus- explains distinct peristaltic motor patterns in stomach and small intestine. Am J Physiol
cular plexus of canine small intestine may be specialized smooth muscle cells. Am J Gastrointest Liver Physiol 289: G539 G549, 2005.
Physiol Gastrointest Liver Physiol 265: G638 G645, 1993.
397. Wang XY, Sanders KM, Ward SM. Intimate relationship between interstitial cells of
377. Torihashi S, Nishi K, Tokutomi Y, Nishi T, Ward S, Sanders KM. Blockade of kit cajal and enteric nerves in the guinea-pig small intestine. Cell Tissue Res 295: 247256,
signaling induces transdifferentiation of interstitial cells of cajal to a smooth muscle 1999.
phenotype. Gastroenterology 117: 140 148, 1999.
398. Wang XY, Ward SM, Gerthoffer WT, Sanders KM. PKC-epsilon translocation in
enteric neurons and interstitial cells of Cajal in response to muscarinic stimulation. Am
378. Torihashi S, Ward SM, Nishikawa S, Nishi K, Kobayashi S, Sanders KM. c-kit-depen-
J Physiol Gastrointest Liver Physiol 285: G593G601, 2003.
dent development of interstitial cells and electrical activity in the murine gastrointes-
tinal tract. Cell Tissue Res 280: 97111, 1995. 399. Wang Y, Fang Q, Lu Y, Song B, Li W, Li L. Effects of mechanical stretch on interstitial
cells of Cajal in guinea pig bladder. J Surg Res 164: e213 e219, 2010.
379. Torihashi S, Ward SM, Sanders KM. Development of c-Kit-positive cells and the onset
of electrical rhythmicity in murine small intestine. Gastroenterology 112: 144 155, 400. Ward SM, Beckett EA, Wang X, Baker F, Khoyi M, Sanders KM. Interstitial cells of
1997. Cajal mediate cholinergic neurotransmission from enteric motor neurons. J Neurosci
20: 13931403, 2000.
380. Torihashi S, Yoshida H, Nishikawa S, Kunisada T, Sanders KM. Enteric neurons ex-
press Steel factor-lacZ transgene in the murine gastrointestinal tract. Brain Res 738: 401. Ward SM, Brennan MF, Jackson VM, Sanders KM. Role of PI3-kinase in the develop-
323328, 1996. ment of interstitial cells and pacemaking in murine gastrointestinal smooth muscle. J
Physiol 516: 835 846, 1999.
381. Triguero D, Sancho M, Garcia-Flores M, Garcia-Pascual A. Presence of cyclic nucle-
otide-gated channels in the rat urethra and their involvement in nerve-mediated 402. Ward SM, Burke EP, Sanders KM. Use of rhodamine 123 to label and lesion interstitial
nitrergic relaxation. Am J Physiol Renal Physiol 297: F1353F1360, 2009. cells of Cajal in canine colonic circular muscle. Anat Embryol 182: 215224, 1990.

906 Physiol Rev VOL 94 JULY 2014 www.prv.org


INTERSTITIAL CELLS IN SMOOTH MUSCLES

403. Ward SM, Burns AJ, Torihashi S, Harney SC, Sanders KM. Impaired development of 421. Yamamoto T, Watabe K, Nakahara M, Ogiyama H, Kiyohara T, Tsutsui S, Tamura S,
interstitial cells and intestinal electrical rhythmicity in steel mutants. Am J Physiol Cell Shinomura Y, Hayashi N. Disturbed gastrointestinal motility and decreased interstitial
Physiol 269: C1577C1585, 1995. cells of Cajal in diabetic db/db mice. J Gastroenterol Hepatol 23: 660 667, 2008.

404. Ward SM, Burns AJ, Torihashi S, Sanders KM. Mutation of the proto-oncogene c-kit 422. Yamataka A, Kato Y, Tibboel D, Murata Y, Sueyoshi N, Fujimoto T, Nishiye H, Miyano
blocks development of interstitial cells and electrical rhythmicity in murine intestine. T. A lack of intestinal pacemaker (c-kit) in aganglionic bowel of patients with Hirsch-
J Physiol 480: 9197, 1994. sprungs disease. J Pediatr Surg 30: 441 444, 1995.

405. Ward SM, Dixon RE, de Faoite A, Sanders KM. Voltage-dependent calcium entry 423. Yamazawa T, Iino M. Simultaneous imaging of Ca2 signals in interstitial cells of Cajal
underlies propagation of slow waves in canine gastric antrum. J Physiol 561: 793 810, and longitudinal smooth muscle cells during rhythmic activity in mouse ileum. J Physiol
2004. 538: 823 835, 2002.

406. Ward SM, Harney SC, Bayguinov JR, McLaren GJ, Sanders KM. Development of 424. Yanagida H, Sanders KM, Ward SM. Inactivation of inducible nitric oxide synthase
electrical rhythmicity in the murine gastrointestinal tract is specifically encoded in the protects intestinal pacemaker cells from postoperative damage. J Physiol 582: 755
tunica muscularis. J Physiol 505: 241258, 1997. 765, 2007.

407. Ward SM, McLaren GJ, Sanders KM. Interstitial cells of Cajal in the deep muscular 425. Yanagida H, Yanase H, Sanders KM, Ward SM. Intestinal surgical resection disrupts
plexus mediate enteric motor neurotransmission in the mouse small intestine. J Physiol electrical rhythmicity, neural responses, and interstitial cell networks. Gastroenterol-
573: 147159, 2006. ogy 127: 1748 1759, 2004.

408. Ward SM, Morris G, Reese L, Wang XY, Sanders KM. Interstitial cells of Cajal mediate 426. Yang YD, Cho H, Koo JY, Tak MH, Cho Y, Shim WS, Park SP, Lee J, Lee B, Kim BM,

Downloaded from http://physrev.physiology.org/ by 10.220.33.5 on November 8, 2017


enteric inhibitory neurotransmission in the lower esophageal and pyloric sphincters. Raouf R, Shin YK, Oh U. TMEM16A confers receptor-activated calcium-dependent
Gastroenterology 115: 314 329, 1998. chloride conductance. Nature 455: 1210 1215, 2008.

409. Ward SM, Ordog T, Bayguinov JR, Horowitz B, Epperson A, Shen L, Westphal H, 427. Yoshino M, Wang SY, Kao CY. Sodium and calcium inward currents in freshly disso-
Sanders KM. Development of interstitial cells of Cajal and pacemaking in mice lacking ciated smooth myocytes of rat uterus. J Gen Physiol 110: 565577, 1997.
enteric nerves. Gastroenterology 117: 584 594, 1999.
428. Youm JB, Kim N, Han J, Kim E, Joo H, Leem CH, Goto G, Noma A, Earm YE. A
410. Ward SM, Ordog T, Koh SD, Baker SA, Jun JY, Amberg G, Monaghan K, Sanders KM. mathematical model of pacemaker activity recorded from mouse small intestine.
Pacemaking in interstitial cells of Cajal depends upon calcium handling by endoplasmic Philos Trans R Soc A 364: 11351154, 2006.
reticulum and mitochondria. J Physiol 525: 355361, 2000.
429. Young HM, Ciampoli D, Johnson PJ, Stebbing MJ. Inhibitory transmission to the lon-
411. Ward SM, Sanders KM. Dependence of electrical slow waves of canine colonic gitudinal muscle of the mouse caecum is mediated largely by nitric oxide acting via
smooth muscle on calcium gradient. J Physiol 455: 307319, 1992. soluble guanylyl cyclase. J Auton Nerv Syst 61: 103108, 1996.

412. Ward SM, Sanders KM. Pacemaker activity in septal structures of canine colonic 430. Young HM, Ciampoli D, Southwell BR, Newgreen DF. Origin of interstitial cells of
circular muscle. Am J Physiol Gastrointest Liver Physiol 259: G264 G273, 1990. Cajal in the mouse intestine. Dev Biol 180: 97107, 1996.

413. Ward SM, Sanders KM. Upstroke component of electrical slow waves in canine 431. Young HM, McConalogue K, Furness JB, De Vente J. Nitric oxide targets in the
colonic smooth muscle due to nifedipine-resistant calcium current. J Physiol 455: guinea-pig intestine identified by induction of cyclic GMP immunoreactivity. Neuro-
321337, 1992. science 55: 583596, 1993.

414. Wei JY, Adelson DW, Tache Y, Go VL. Centrifugal gastric vagal afferent unit activities: 432. Zhang Y, Carmichael SA, Wang XY, Huizinga JD, Paterson WG. Neurotransmission in
another source of gastric efferent control. J Anton Nerv Syst 52: 8397, 1995. lower esophageal sphincter of W/Wv mutant mice. Am J Physiol Gastrointest Liver
Physiol 298: G14 G24, 2010.
415. West RB, Corless CL, Chen X, Rubin BP, Subramanian S, Montgomery K, Zhu S, Ball
CA, Nielsen TO, Patel R, Goldblum JR, Brown PO, Heinrich MC, van de Rijn M. The 433. Zheng H, Park KS, Koh SD, Sanders KM. Expression and function of a T-type Ca2
novel marker, DOG1, is expressed ubiquitously in gastrointestinal stromal tumors conductance in interstitial cells of Cajal of the murine small intestine. Am J Physiol Cell
irrespective of KIT or PDGFRA mutation status. Am J Pathol 165: 107113, 2004. Physiol 306: C705C713, 2014.

416. Wolin MS, Cherry PD, Rodenburg JM, Messina EJ, Kaley G. Methylene blue inhibits 434. Zhou DS, Komuro T. The cellular network of interstitial cells associated with the deep
vasodilation of skeletal muscle arterioles to acetylcholine and nitric oxide via the muscular plexus of the guinea pig small intestine. Anat Embryol 186: 519 527, 1992.
extracellular generation of superoxide anion. J Pharmacol Exp Ther 254: 872 876,
1990. 435. Zhou DS, Komuro T. Interstitial cells associated with the deep muscular plexus of the
guinea-pig small intestine, with special reference to the interstitial cells of Cajal. Cell
417. Won KJ, Sanders KM, Ward SM. Interstitial cells of Cajal mediate mechanosensitive Tissue Res 268: 205216, 1992.
responses in the stomach. Proc Natl Acad Sci USA 102: 1491314918, 2005.
436. Zhu MH, Kim TW, Ro S, Yan W, Ward SM, Koh SD, Sanders KM. A Ca2-activated
418. Wouters M, Smans K, Vanderwinden JM. WZsGreen/: a new green fluorescent Cl conductance in interstitial cells of Cajal linked to slow wave currents and pace-
protein knock-in mouse model for the study of KIT-expressing cells in gut and cere- maker activity. J Physiol 587: 4905 4918, 2009.
bellum. Physiol Genomics 22: 412 421, 2005.
437. Zhu MH, Sung IK, Zheng H, Sung TS, Britton FC, ODriscoll K, Koh SD, Sanders KM.
419. Xin HB, Deng KY, Rishniw M, Ji G, Kotlikoff MI. Smooth muscle expression of Cre Muscarinic activation of Ca2-activated Cl current in interstitial cells of Cajal. J
recombinase and eGFP in transgenic mice. Physiol Genomics 10: 211215, 2002. Physiol 589: 4565 4582, 2011.

420. Xue C, Ward SM, Shuttleworth CW, Sanders KM. Identification of interstitial cells in 438. Zhu YF, Wang XY, Lowie BJ, Parsons S, White L, Kunze W, Pawelka A, Huizinga JD.
canine proximal colon using NADH diaphorase histochemistry. Histochemistry 99: Enteric sensory neurons communicate with interstitial cells of Cajal to affect pace-
373384, 1993. maker activity in the small intestine. Pflgers Arch. In press.

Physiol Rev VOL 94 JULY 2014 www.prv.org 907

Вам также может понравиться