Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Mar 8.
Published in final edited form as: Gastroenterology. 2008 Nov 1;136(2):630–639. doi: 10.1053/j.gastro.2008.10.031

Changes in the Structure and Function of ICC Networks in ICC Hyperplasia and Gastrointestinal Stromal Tumors

JOONG GOO KWON *, SUNG JIN HWANG *, GRANT W HENNIG *, YULIA BAYGUINOV *, CONOR MCCANN *, HUI CHEN *, FERDINAND ROSSI , PETER BESMER , KENTON M SANDERS *, SEAN M WARD *
PMCID: PMC4782934  NIHMSID: NIHMS619280  PMID: 19032955

Abstract

Background & Aims

Gastrointestinal stromal tumors (GISTs) express the receptor tyrosine kinase c-kit. Approximately 90% of GISTs have gain-of-function mutations in the Kit gene, which leads to its constitutive activation and drives malignant behavior of GISTs. Interstitial cells of Cajal (ICC) express c-kit; however, it is unknown whether uncontrolled hyperplasia of ICC is responsible for GISTs. Here, we sought to determine whether gain-of-function mutations in Kit lead to hyperplasia of all classes of ICC, whether ICC hyperplasia begins before birth, and whether functional defects occur in ICC hyperplasia or the development of GISTs.

Methods

Heterozygous mutant KitV558Δ/+ mice that develop symptoms of human familial GISTs and prematurely die from pathology of the gastrointestinal tract were utilized and compared with wild-type controls. C-kit-immunohistochemistry and intracellular electrical recording of spontaneous and nerve-evoked activity were applied to examine the density and functionality of ICC in these mutants.

Results

There was considerable hyperplasia in all classes of ICC throughout the GI tract of KitV558Δ/+ mice, except for ICC in the deep muscular plexus of the intestine. Spontaneous electrical activity and postjunctional neural responses in hyperplastic ICC tissues appeared normal but were up-regulated in the cecum, where GISTs were commonly found.

Conclusions

Kit gain-of-function leads to hyperplasia of most classes of ICC throughout the GI tract. ICC retain normal pacemaker function and enteric neural responses well after development of hyperplasia.


Interstitial cells of Cajal (ICC) are specialized cells in the gastrointestinal (GI) tract that generate pacemaker activity and mediate enteric motor neurotransmission.13 ICC are essential for motor patterns of the GI tract. ICC express the receptor tyrosine kinase c-kit, and signaling via this pathway is required for development and maintenance of ICC.1,2 When c-kit signaling is disrupted, ICC undergo phenotypic changes4,5 that result in a decreased density of these cells. Reduced numbers of ICC is a pathologic correlate often identified in GI motility disorders.68

Approximately 94% of mesenchymal tumors are positive for c-kit receptors and are defined as gastrointestinal stromal tumors (GISTs).9 Thus, c-kit immunohistochemistry has become a standard for diagnosis of GISTs.1,10 Expression of c-kit and CD34 by GISTs suggested that these tumors are ICC neoplasms because ICC were the only cell-type expressing CD34 and c-kit.9,11 Expression of CD34 by ICC was challenged when it was found that CD34 positive/c-kit negative fibroblast-like cells are distinct from ICC in the human and murine GI tracts.12 Recently, c-kitlowCD44+CD34+ progenitor cells have been identified that could be responsible for postnatal generation of ICC.13 The role of these cells in GISTs has not been evaluated. Taken together, these findings suggest that the cellular origin of GIST is ambiguous.

Human GISTs (80%–90%) have gain-of-function mutations in Kit, particularly in exon 11, encoding the juxtamembrane domain of c-kit.9 These mutations result in ligand-independent activation of c-kit, which increases cellular proliferation and facilitates GIST development. In kindreds with human familial GIST syndrome, a germ-line Kit gain-of-function mutation causes cells expressing c-kit to display ligand-independent activation of c-kit. Thus, ICC outside the margins of neoplastic lesions may undergo hyperplasia, as shown, for example, with the KitT557A familial mutation. ICC hyperplasia is considered a precancerous state.14,15 Murine models, carrying gain-of-function mutations in Kit, yield GI lesions similar to human familial GIST syndrome and provide a tool for investigating the development of GISTs and pathophysiology associated with gain-of-function mutations.1619 One model, based on a mutation found in a human familial GIST, employed introduction of a Kit-activating mutation into Kit,16 KitV558. Heterozygous KitV558Δ/+ mice developed neoplastic lesions in the cecum that were similar to human familial GIST.

Most investigators accept that GISTs develop from gain-of-function mutations in Kit, but the functional consequences of gain-of-function Kit mutations on pre-GIST ICC are unknown. We used immunohistochemistry to study the distribution of c-kit+ cells in the GI tract and characterized functions attributed to ICC (pacemaking and neural responses) in wild-type and KitV558Δ/+ mice. We quantified hyperplasia of c-kit+ cells beyond the site of the GIST in the cecum to determine whether specific classes of ICC are more prone to hyperplasia from a Kit gain-of-function mutation. Our results provide the first comprehensive analysis of ICC structure and function in a murine GIST model with a defined gain-of-function mutation in Kit.

Materials and Methods

Animals

Mutant KitV558Δ/+ mice were generated as previously described.16 Heterozygote males were mated with C57BL/6J females to produce KitV558Δ/+ heterozygotes and homozygote controls. Age-matched mice on the same background as KitV558Δ/+ mice (C57BL/6J) were used as controls. Preterm animals were killed at embryonic day (E) 19 and adult animals between postterm day (P) 40 and P60 for morphologic and electrophysiologic studies. Animals were maintained and experiments performed in accordance with the NIH Guide for Care and Use of Laboratory Animals. All procedures were approved by the Institutional Animal Use and Care Committee at the University of Nevada.

Morphologic Analysis and Volume Rendering

Whole mounts and cryostat sections of GI tissues were processed for immunohistochemical analysis of c-kit as previously described.3,4 For double-labeling experiments, cryostat sections were incubated in the c-kit antibody (ACK2)4,5 overnight followed by phosphate-buffered saline (PBS) (0.01 mol/L; 2 hours) before incubation with a rabbit anti-smooth muscle myosin II heavy chain (1:100; Biomedical Technologies, Stoughton, MA). Sections were subsequently washed in PBS and incubated consecutively in Alexa Fluor 594-coupled goat anti-rat 594 and Alexa Fluor 488-coupled goat anti-rabbit (1:1000; for 1 hour each at room temperature).

Tissues and sections were examined with an LSM 510 Meta confocal microscope (Zeiss, Germany) using excitation wavelengths appropriate for Alexa Fluor 488 and 594. Z-series scans were collected using a ×63 objective and 0.25-μm steps. The confocal aperture was set ≤1 Airy unit, and laser power and gain settings were kept the same between specimens. Stacks of images were attenuation corrected and deconvolved (AutoQuantX, Media-Cybernetics, MD) then imported into custom in-house software (Volumetry G6b, Grant W. Herring). The depths of specific layers of ICC (eg, intramuscular ICC [ICC-IM] or myenteric plexus region ICC [ICC-MY]) were recorded. C-kit-immunofluorescent cells were thresholded (background noise inclusion, <1%), and a modified marching cubes algorithm was used to calculate the subvoxel volume between adjacent optical sections throughout the stack. Both the average slice volume (expressed as percentage of the total slice volume) and the maximum slice volume (averaged over 1 μm) were calculated for each defined ICC layer. Three-dimensional surface renderings were constructed and tilted 40° in the x-axis. Double-labeled images were constructed using Zeiss LSM-5 Image Examiner software (Carl Zeiss Microimaging Inc, Thornwood, NY) and converted to .tif files for final processing in Adobe Photoshop 7.0 (Mountain View, CA) and Corel Draw 12.0 (Ontario, Canada).

Electrophysiologic Studies

Gastric fundus and antrum, jejunum, ileum, cecum, and proximal colon were removed and placed in Krebs–Ringer buffer (KRB). Strips of each organ were mounted with circular muscle upward in a recording chamber, and impalements of circular muscle cells were made with microelectrodes (80–120 MΩ). Transmembrane potentials were recorded as described previously.1 Nifedipine (1 μmol/L) was added to reduce contractions and facilitate maintenance of impalements, except where stated. Slow waves are not affected by nifedipine, as shown previously.1,20 Enteric motor neurons were stimulated in muscles strips by electrical field stimulation (EFS; 0.3-ms pulse durations, 1 pulse-20 Hz, train duration of 1 second, 10–15 volts).

Solutions and Drugs

The recording chamber was perfused with oxygenated KRB (in mmol/L): NaCl 118.5; KCl 4.5; MgCl2 1.2; NaHCO3 23.8; KH2PO4 1.2; dextrose 11.0; CaCl2 2.4. The pH of the KRB was 7.3–7.4 when bubbled with 97% O2–3% CO2 at 37 ± 0.5°C. Nifedipine (Sigma Chemical Co.; St Louis, MO) was dissolved in ethanol (10 mmol/L) before further dilution in KRB (1 μmol/L). Atropine, apamin, Nω-nitro-L-arginine (L-NA) and tetrodotoxin (Sigma Chemical Co) were dissolved in deionized H2O before dilution in KRB to desired concentrations.

Analysis of Data

Data are expressed as means ± standard errors of the mean and tested for significance with Student t test. P < .05 was taken as significant. The “n” values refer to number of animals. Several electrical parameters were analyzed: (1) resting membrane potential (RMP); (2) slow wave amplitude; (3) duration of slow wave; and (4) frequency. Responses to EFS were analyzed as excitatory junction potentials (EJPs) or inhibitory junction potentials (IJPs) or phase advancement in slow waves. Changes in slow wave duration to EFS were compared with the average of 5 spontaneous events prior to EFS.

Results

Gross Morphology

KitV558Δ/+ mice and +/+ controls (either E19 or P40–P60) were used. Mice developed enlarged cecums and nodular masses, as previously described.16 The small intestines of these animals were grossly distended for up to 100 mm above the ileal-cecal junction. Below the cecum and above the distended region of small bowel, the remainder of the GI tract was grossly normal in appearance.

c-kit-Positive Cells in KitV558Δ/+ Mutants vs Wild-Type Controls

c-kit-Positive cells in KitV558Δ/+ and +/+ mice were examined using immunofluorescence techniques.

Gastric fundus

In cryostat sections and whole mount preparations of the gastric fundus, c-kit-like immunoreactive (c-kit-LI) cells were observed in the circular (CM) and longitudinal (LM) muscle layers. The cells with c-kit-LI were spindle shaped and dispersed within muscle bundles and ran orthogonal to each other in the CM and LM (Figure 1A and B), as previously described for ICC-IM.3 Density images and calculations of volume of c-kit-positive cells showed that ICC-IM increased significantly in KitV558Δ/+ mice (Supplementary Table 1; see Supplementary material online at www.gastrojournal.org). The muscle layers were also increased in thickness in association with the expansion in c-kit-positive cells (Figure 1B).

Figure 1.

Figure 1

Hyperplasia of ICC in the stomachs of KitV558Δ/+ mice. In each panel (A–D), cryostat cross sections (Aa–Da), whole mounts (Ab–Db), and volume-rendered micrographs (Ac–Dc; see Materials and Methods section) are shown. In each cryostat cross section (Aa–Da), c-kit-LI is labeled in red to distinguish intramuscular ICC (ICC-IM) and ICC of the myenteric plexus region (ICC-MY) from smooth muscle cells (SMC) that are labeled with smooth muscle myosin II heavy chain (SMMHC) in green. Panels Aa–c show the gastric fundus of +/+ mice. ICC-IM are present in both circular (ICC-IMC; CM) and longitudinal (ICC-IML; LM) muscle layers. Panels Ba–c show the gastric fundus from KitV558Δ/+ mice. Panels Ca–c show ICC-IM within the circular layer and ICC-MY in the gastric antrum of +/+ mice, and panels Da–c show ICC-IM and ICC-MY in the gastric antrum of KitV558Δ/+ mice. Scale bars at bottom apply to panels above.

Gastric antrum

Two populations of ICC were observed in the antrum, as previously reported.3 Spindle-shaped ICC-IM were interspersed within the CM and ran parallel to the CM fibers (Figure 1C and D). ICC-MY were present in the intermuscular plane between CM and LM (Figure 1C). c-kit-Positive cells were greatly increased in KitV558Δ/+ mice (Figure 1D; Supplementary Table 1) to an extent that the ICC-MY developed a mat-like appearance with few intercellular spaces.

Small intestine

In samples of jejunum and ileum, c-kit-positive cells formed an anastomosing network of cells between CM and LM (ICC-MY) and within the deep muscular plexus (DMP) (ICC-DMP) near the submucosal surface of the CM (Figure 2A). ICC-DMP formed a fine network with elongated cell bodies and lateral processes that contacted adjacent ICC-DMP (Figure 2A).

Figure 2.

Figure 2

ICC hyperplasia in small intestine of KitV558Δ/+ mice. Cryostat cross sections (Aa–Ba), whole mounts (Ab–Bb), and volume-rendered micrographs (Ac–Bc) are shown. In cryostat cross sections (Aa–Ba), c-kit-LI is labeled in red to distinguish ICC of the deep muscular plexus (ICC-DMP) and ICC-MY from SMC labeled with SMMHC in green. Panel A shows images from +/+ mice, and panel B shows images from KitV558Δ/+ mice. Scale bars at bottom apply to panels above.

Partial obstruction of the small intestine leads to hypertrophy of the bowel and loss of ICC above the obstruction.21 The small intestines of KitV558Δ/+ mice were obstructed because of the masses in the cecum and greatly distended; however, c-kit-positive cells were abundant and increased in density (Supplementary Table 1). As in the stomach, ICC-MY developed a diffuse, mat-like appearance (Figure 2B). Although there was significant enhancement in ICC-MY, significant hyperplasia was not observed in ICC-DMP (Supplementary Table 1).

Cecum

c-kit-Positive cells formed networks within the CM and LM in the cecum and fine cellular processes contacted adjacent ICC-IM, forming a network. A second population of ICC generally shorter in length and possessing a dendritic morphology with numerous spiny projections was also present. ICC typically ran parallel to CM, but a few cells had no obvious orientation (Figure 3A). In many cases, cecums of KitV558Δ/+ mice were hard and nodular, and whole mount preparations were not possible. Cryostat sections of these tissues were used to inspect cells with c-kit-LI. c-kit-Positive cells in these tissues formed a mass with no morphologic identity as ICC (Figure 3B; Supplementary Table 1).

Figure 3.

Figure 3

Hyperplasia of ICC in the cecum and colon of KitV558Δ/+ mice. Panels A–D show cryostat cross sections (Aa–Da), whole mounts (Ab–Db), and volume-rendered micrographs (Ac–Dc). In cryostat cross sections (Aa–Da), c-kit-LI is labeled in red (ICC-IM and ICC-MY) and ICC along the submucosal surface of the circular muscle (ICC-SM) and SMMHC is labeled in green to identify smooth muscle cells. Panels Aa–c show the cecum of +/+ mice. Panels Ba–c show images from the cecum of KitV558Δ/+ mice. Panels Ca–c show images from the proximal colon of +/+ mice, and Panels Da–c show images from the proximal colon of KitV558Δ/+ mice. Scale bars at bottom apply to panels above.

Proximal colon

c-kit-Positive cells were located along the submucosal surface of CM (ICC-SM), within CM and LM (ICC-IM), and between CM and LM (ICC-MY; Figure 3C).12,22 ICC-SM were orientated parallel to the muscle fibers of CM and ICC-IM orientated parallel to muscle fibers of CM and LM and contacted adjacent ICC with thin processes. ICC-MY possessed multiple fine processes that formed an anastomosing network with adjacent ICC-MY (Figure 3C). c-kit-Positive cells were increased in density in KitV558Δ/+ mice (Figure 3D; Supplementary Table 1). ICC-MY were increased in number to where it was hard to distinguish individual cells.

Distribution in ICC Before Birth in KitV558Δ/+ Fetuses

c-kit-Positive cells emerge at approximately E12.5 during normal development and form a network of ICC-MY by E17.4 We examined E19 muscles to determine whether the hyperplasia of c-kit-positive cells develops before birth in KitV558Δ/+ mice. As in adults, c-kit-positive cells were increased in density throughout the GI tracts of fetal tissues. Therefore, gain-of-function of c-kit in KitV558Δ/+ mice results in hyperplasia of ICC prior to birth, and the ICC population continues to expand after birth (Figure 4).

Figure 4.

Figure 4

c-kit Immunohistochemistry of wholemount preparations from KitV558Δ/+ and +/+ E19 fetuses. Panels A and B show confocal images of ICC-IM from the fundus of KitV558Δ/+ mice, and panel C shows ICC-IM from the fundus of an E19 +/+ fetus. Panels D and E show images of ICC-IM and ICC-MY from the antrums of KitV558Δ/+ E19 embryos and panel F from a +/+ E19 fetus. Marked hyperplasia of ICC-IM and ICC-MY occurs prior to birth. Scale bar (100 μm) applies to all panels.

Functional Changes in the GI Tracts of KitV558Δ Mice vs Wild-Type Controls

Gastric fundus and antrum

RMP of fundus CM averaged −41.1 ± 0.6 mV in wild-type mice. An ongoing discharge of spontaneous transient depolarizations (unitary potentials) was superimposed upon resting potentials, as previously described (Figure 5A; n = 14)3,23 RMPs of antral muscles averaged +64.1 ± 1.0 mV, and electrical slow waves (27.4 ± 3.0 mV in amplitude, 8.9 ± 0.6 seconds) occurred at a frequency of 3.7 ± 0.3 cycles min−1 (n = 14; Figure 5B).

Figure 5.

Figure 5

Intracellular electrical activity of gastric smooth muscle of +/+ and KitV558Δ/+ mice. Panel A shows basal activity and postjunctional neural responses to electrical field stimulation (EFS) in +/+ and KitV558Δ/+ mice. Basal electrical activity was characterized by ongoing unitary potential discharge. Responses to EFS (arrowheads; 10 Hz, 0.3 ms; 1 s) were biphasic IJPs that were largely blocked by L-NNA (100 μmol/L; dashed line). EJPs (arrows) were blocked by atropine (1 μmol/L) and were unmasked by L-NNA. After L-NNA and atropine, only purinergic IJPs were observed (blocked by apamin; not shown). Panel B shows spontaneous electrical activity and responses to EFS in antrums of +/+ and KitV558Δ/+ mice. Slow waves were similar in +/+ and KitV558Δ/+ mice. Neural responses (EFS at arrowheads) in +/+ and KitV558Δ/+ mice were not significantly different (eg, transient IJP that phase advanced but attenuated the amplitude of the next slow wave). L-NNA (100 μmol/L) largely blocked IJPs (not shown).

The electrical activity of gastric muscles of KitV558Δ/+ mice was similar to wild-type muscles. RMPs of fundus CM averaged −41.1 ± 3.8 mV (n = 3), and ongoing unitary potentials were observed (Figure 5A). Antral muscles of KitV558Δ/+ mice had RMPs of −64.2 ± 1.7 mV (n = 7), and slow waves (28.2 ± 4.4 mV in amplitude and 7.5 ± 0.8 seconds) occurred 5.4 ± 0.4 cycles min−1 (Figure 5B). There was no statistical difference in slow waves in wild-type and KitV558Δ/+ mice.

EFS (1–20 Hz for 1 second) caused frequency-dependent IJPs in fundus muscles (Figure 5A). IJPs were reduced by L-NNA (100 μmol/L), unmasking an EJP that was inhibited by atropine (1 μmol/L). Blocking the EJP revealed a fast transient IJP that was apamin-sensitive (30 nmol/L; not shown). Postjunctional neural responses were similar in fundus muscles of KitV558Δ/+ mice. EFS caused IJPs averaging 8.3 ± 2.3 mV in amplitude and 2.7 ± 0.2 seconds in duration. L-NNA (100 μmol/L) reduced the amplitude and duration of IJPs and unmasked an EJP that was reduced by atropine (Figure 5A).

Antral muscles responded to EFS by generating IJPs averaging 8.9 ± 1.2 mV and 1.8 ± 0.1s (Figure 5B). The next slow wave was phase-advanced upon termination of the IJP. KitV558Δ/+ mice also responded to EFS by generating IJPs that averaged 7.3 ± 1.4 mV in amplitude and 1.8 ± 0.2 seconds in duration. IJPs were largely blocked in antral muscles of wild-type and KitV558Δ/+ mice by L-NNA (not shown).

Small intestine

RMPs of jejunal CM cells of wild-type muscles averaged −59.2 ± 0.6 mV, and slow waves (21.3 ± 2.6 mV in amplitude and 1.2 ± 0.1 seconds in duration) occurred at 35.4 ± 1.4 cycles min−1 (Figure 6A). In ileum, RMPs averaged −60.9 ± 0.3 mV, and slow waves 21.7 ± 1.3 mV in amplitude and 1.3 ± 0.1 seconds in duration occurred at 30.2 ± 0.7 cycles min−1 (Figure 6A). RMP of jejunual CM of KitV558Δ/+ mice averaged −61.0 ± 0.8 mV, and slow waves (22.1 ± 2.5 mV in amplitude and 1.3 ± 0.2 seconds) occurred at 32.7 ± 2.1 cycles min−1 (Figure 6B). RMP of ileal CM of KitV558Δ/+ mice averaged −60.9 ± 1.1 mV, and slow waves (29.3 ± 1.8 mV in amplitude and 1.3 ± 0.1 seconds) occurred at 29.3 ± 1.8 cycles min−1 (Figure 6B). The pacemaker activity of jejunums and ileums of wild-type and KitV558Δ/+ mice was not statistically different.

Figure 6.

Figure 6

Intracellular electrical recordings from small intestines of +/+ and KitV558Δ/+ mice. Panels A and B show slow waves from jejunums and ileums of +/+ and KitV558Δ/+ mice. Panels C and D show neural responses (EFS at arrowheads; 10 Hz, 0.3 ms; 1 s) from ileums of +/+ and KitV558Δ/+ mice. Responses consisted of a transient inhibitory junction potential (IJP) and inhibition of slow wave amplitude for several cycles following EFS. Inhibitory effects of EFS were largely blocked by L-NNA (100 μmol/L). Responses to EFS were not different in +/+ and KitV558Δ/+ mice.

EFS of small intestinal muscles caused frequency-dependent IJPs (eg, 8.6 ± 1.0 mV at 10 Hz) and attenuation of several slow waves following EFS (Figure 6C). IJPs and effects on slow waves were reduced by L-NNA (100 μmol/L), although a transient, apamin-sensitive IJP persisted after L-NNA (Figure 6C). EFS of small intestinal muscles from KitV558Δ/+ mice also caused L-NNA-sensitive IJPs (eg, 6.8 ± 1.0 mV at 10 Hz) and attenuation of several slow waves after EFS (Figure 6D).

Cecum

CM from wild-type cecums had RMPs of −55.0 ± 0.9 mV and was electrically quiescent (Figure 7A). As described above, cecums of KitV558Δ/+ mice varied in gross morphology; some contained nodular masses, and others were fluid filled and distended. We were unable to make impalements in cells of tissues with nodular masses. RMP of CM in distended cecums averaged −55.9 ± 0.9 mV. In contrast to wild-type muscles, 33% of cecums of KitV558Δ/+ mice generated spontaneous slow waves (2 muscles of 9 mice) or spike complexes (1 of 9; Figure 7B). Slow waves and spike complexes were blocked by nifedipine (1 μmol/L; Figure 7C).

Figure 7.

Figure 7

Electrical activities of cecum of +/+ and KitV558Δ/+ mice. Panel A shows the electrical activity of the circular muscle layer of a control animal. Panel B shows normal and spontaneous action potentials or slow waves in the cecums of KitV558Δ/+ mice in which spontaneous electrical activity consisted of small “unitary potentials.” Spontaneous action potentials or slow wave activity developed in the cecums of KitV558Δ/+ mice in which solid tumors had not yet formed. Action potentials and slow wave-like depolarizations were inhibited by nifedipine (1 μmol/L; panel C).

EFS of wild-type cecum muscles consisted of transient IJPs that were insensitive to L-NNA (100 μmol/L). Neural responses of KitV558Δ/+ cecums also consisted of L-NNA-insensitive IJPs. In contrast to wild-type muscles, rebound excitation often occurred at the break of IJPs (neural responses not shown).

Proximal colon

RMPs of CM from wild-type colonic muscles averaged −53.0 ± 1.0 mV (n = 20 animals). Spontaneous spike complexes (30.0 ± 3.0 mV) occurred at 2.7 ± 0.3 cycles min−1, as previously reported.22 Each complex consisted of 13.8 ± 1.2 action potentials (Figure 8A). Proximal colon CM of KitV558Δ/+ mice had RMPs of −51.0 ± 3.0 mV (n = 10 animals). Spontaneous activity of KitV558Δ/+ colons was not significantly different than wild-type mice (eg, spike complexes of 35.4 ± 5.2 mV occurred at 2.3 ± 0.3 cycles min−1; Figure 8B).

Figure 8.

Figure 8

Electrical activity and neural responses in proximal colons of +/+ and KitV558Δ/+ mice. Panels A and B show that spontaneous electrical activity in the proximal colon was similar in +/+ and mutant animals. Panels C and D show neural responses in +/+ and KitV558Δ/+ mice. EFS (single pulse, 0.3-ms duration; arrowheads) caused biphasic IJPs, characterized by a rapid transient component followed by a sustained component (dashed lines). The sustained component was blocked by L-NNA (100 μmol/L). Atropine (1 μmol/L) in the presence of L-NNA had little effect on responses.

EFS (single pulse, 0.3 ms) elicited biphasic IJPs (first and second components were 19.7 ± 2.3 mV and 12.0 ± 1.0 mV and 0.8 ± 0.03 and 8.1 ± 0.3 seconds, respectively; Figure 8C). Similar responses were observed in CM of KitV558Δ/+ colons (eg, first and second components were 17.5 ± 0.1 mV and 10.7 ± 1.3 mV and 0.8 ± 0.03 and 7.6 ± 0.3 seconds, respectively; Figure 8D; n = 10; P > .05).

Discussion

We characterized and quantified the density of c-kit+ cells in a GIST model with many of the features of human familial GIST, including hyperplasia of ICC beyond the margins of the primary tumor.16,17 Extensive hyperplasia of all of the major classes of ICC was observed in the stomach and colon (ICC-IM and ICC-MY) but only ICC-MY, and not ICC-DMP, in the small intestine. Surprisingly, in spite of remarkable expansion in ICC mass, few demonstrable functional differences were observed in KitV558Δ/+ tissues (pacemaker activity and enteric motor neuron responses). In the cecum, where primary tumors developed, abnormal spontaneous electrical rhythmicity developed in some muscles before nodular masses were found.

Enhancement in ICC was not matched by increased neural responses in KitV558Δ/+ mice. ICC-IM are innervated by enteric motor neurons but do not require interactions with neurons for development. ICC developed normally in GDNF−/− mice that lack enteric neurons distal to the proximal stomach.24 Likewise, when ICC-IM are absent, varicose processes of enteric motor neurons are distributed normally within GI muscles.3 Synaptic contacts form between enteric motor neurons and ICC-IM during normal development,25 and major inhibitory and excitatory neurotransmitters, nitric oxide and acetylcholine, appear to be largely confined to these junctions.3,26 Therefore, increasing the number of ICC-IM per se may not impact postjunctional neural responses unless this is accompanied by parallel enhancement in nerve terminals or synaptic contacts.

It is harder to reconcile the seeming lack of effect of hyperplasia in most regions on pacemaker activity. Pacemaker activity and active propagation of slow waves are attributed to ICC-MY.1,2,2729 Thus, an increase in ICC-MY as observed in the antrum, small bowel, and colon might be expected to enhance pacemaker frequency or increase the magnitude of pacemaker currents and increase slow wave amplitude. Except for the emergence of spontaneous activity in the cecum, significant effects on slow wave behavior were not observed. During hyperplasia, ICC-MY possibly undergo down-regulation of the pacemaker mechanism or electrical coupling. An evaluation of the pacemaker activity of individual ICC will be needed to determine whether down-regulation of pacemaker function is a feature of pre-GIST hyperplasia.

Resting potentials of wild-type cecum muscles showed electrical quiescence, but recordings from KitV558Δ/+ cecums before nodular GISTs developed revealed 2 atypical patterns of electrical activity: slow wave activity, more typical of the small intestine, and spike complexes, more typical of the colon. Unlike the slow waves of small intestine, the emergent slow waves in the cecum were blocked by nifedipine. A dramatic rearrangement of ICC and development of novel properties accompanied the hyperplasia of c-kit+ cells in the cecum. Mutant mice that developed neoplasms displayed spindle-shaped c-kit+ cells that differed from ICC in cecums of animals before solid tumors developed. It is unknown whether c-kit+ cells of solid tumors retained ICC function because we were unable to impale cells in these tumors.

KitV558Δ/+ mice develop GISTs and eventually die from GI pathology,16 possibly because of an obstruction caused by the mass that develops in the cecum. The observed distension of the small intestine is consistent with the formation of a partial obstruction. Partial obstructions of the small bowel result in distention for up to 100 mm above the obstruction,21 and the appearance of the bowel is grossly similar to the small bowels of KitV558Δ/+ mice after formation of solid tumors in the cecum. In the previous study, ICC were greatly reduced above the site of obstruction.21 Loss of ICC was accompanied by loss of electrical slow waves and postjunctional neural responses. These findings contrast to the current study in which ICC were present (normal numbers in the DMP and expanded density in the myenteric region) and electrical slow waves and neural responses were intact in the distended small intestines of KitV558Δ/+ mice. At present, the reasons for loss of ICC in the obstructed intestine are not fully understood, but c-kit ligand expression decreases in bowel obstruction (Hwang SJ and Ward SM, unpublished observations), and this might explain the loss of ICC in these tissues.30 Reduced c-kit ligand would not be expected to affect ICC in KitV558Δ/+ mice because KitV558Δ is a gain-of-function in which c-kit is activated in the absence of ligand binding.16

The onset of ICC hyperplasia in KitV558Δ/+ mice occurred prior to birth. Previous studies demonstrated that c-kit signaling prior to birth is required for establishment of the ICC phenotype31 and development of functional ICC.5,31 Others have argued that c-kit is only important for development and maintenance of the ICC phenotype after birth.32,33 Data from the present study clearly show the significance of c-kit signaling for ICC during the late prenatal period because hyperplasia of ICC networks developed in KitV558Δ/+ mice before birth.

GISTs were proposed to arise from proliferation of ICC because these were the only cells found to express c-kit and CD34 in the gut.11,34 This idea was later challenged because c-kit and CD34 were found to be expressed by discrete populations of cells in the human and murine GI tract,35 and doubts developed about the cellular origin of GIST. Recently, a rare population of putative stem cells (c-kitlowCD44+CD34+) was described that may be responsible for replacement of ICC because of normal turnover and the source of cells for GISTs.13 c-kit+lowCD34+CD44+ cells required stimulation by insulin-like growth factor-I for proliferation and secondary stimulation by c-kitL to develop the ICC phenotype.13 Constitutive activation of c-kit in KitV558Δ/+ mice might preclude the need for c-kit-L for proliferation of ICC progenitor cells. Stimulation by insulin-like growth factor-I or other growth factors in situ might expand the progenitor cell population throughout most regions of the GI tract normally populated by ICC. It should be noted that hyperplasia of c-kit+/ICC began before birth, so abnormal expansion of this population likely results from overstimulation of early developmental pathways for ICC rather than postnatal activation of a specialized pool of adult stem cells.

We investigated previously the emergence of c-kit-positive cells and cells with the morphologic and functional phenotypes of ICC in the murine small intestine. These experiments showed that c-kit-positive cells line the exterior of the gut at E15 and develop ICC-like morphology and function by E17.31 We also found that early c-kit+ precursor cells undergo a lineage decision at approximately E15 and develop into either mature ICC or longitudinal muscle cells. We concluded that the lineage decision might depend on c-kit signaling, such that cells receiving c-kitL stimulation from opposing cells developed into mature ICC, but cells without direct contact to c-kitL developed into longitudinal muscles cells. The present study argues against this hypothesis, however, because c-kit is constitutively active in KitV558Δ/+ and longitudinal muscle developed in these mice (although it was thin and penetrated by c-kit-positive cells in some images). These data suggest that a positive signal (besides the lack of c-kit signaling) might be necessary for early c-kit-positive precursors to develop into a smooth muscle phenotype.36

In summary, KitV558Δ/+ mice develop neoplastic lesions in the cecum that lead to obstruction, as evidenced by distension of the small intestine. KitV558Δ/+ mice display dramatic hyperplasia of most classes of ICC, from the stomach to the colon. ICC-DMP of the small intestine, however, were not increased in numbers or density. Despite marked hyperplasia of ICC, functional characteristics are retained, and few differences were noted in the electrical behavior or responses to enteric motor neurons in KitV558Δ/+ mice. An exception was the emergence of electrical rhythmicity in the cecum. Hyperplasia of ICC develops before birth in KitV558Δ/+ mice, suggesting that c-kit signaling in the fetus regulates the density and normal distribution of ICC.

Supplementary Material

Supplementary Data

Acknowledgments

Supported by NIH grants DK57236 and DK41315, by grant P01 DK41315 for the Morphology Core Laboratory, an equipment grant (1S10-RR16871), and a grant from the Nevada Cancer Institute (NVCI) (to S.M.W.); image processing in a Core Laboratory was funded by COBRE (P20-RR018751, to G.H.).

Abbreviations used in this paper

EFS

electrical field stimulation

EJPs

excitatory junction potentials

GIST

gastrointestinal stromal tumors

ICC

interstitial cells of Cajal

IJPs

inhibitory junction potentials

RMP

resting membrane potential

Footnotes

Supplementary Data

Note: To access the supplementary material accompanying this article, visit the online version of Gastroenterology at www.gastrojournal.org, and at doi: 10.1053/j.gastro.2008.10.031.

References

  • 1.Ward SM, Burns AJ, Torihashi S, et al. Mutation of the proto-oncogene c-kit blocks development of interstitial cells and electrical rhythmicity in murine intestine. J Physiol. 1994;480:91–97. doi: 10.1113/jphysiol.1994.sp020343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Huizinga JD, Thuneberg L, Kluppel M, et al. W/kit gene required for interstitial cells of Cajal and for intestinal pacemaker activity. Nature. 1995;373:347–349. doi: 10.1038/373347a0. [DOI] [PubMed] [Google Scholar]
  • 3.Burns AJ, Lomax AE, Torihashi S, et al. Interstitial cells of Cajal mediate inhibitory neurotransmission in the stomach. Proc Natl Acad Sci U S A. 1996;93:12008–12013. doi: 10.1073/pnas.93.21.12008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Torihashi S, Ward SM, Nishikawa S-I, et al. c-kit-Dependent development of interstitial cells and electrical activity in the murine gastrointestinal tract. Cell Tiss Res. 1995;280:97–111. doi: 10.1007/BF00304515. [DOI] [PubMed] [Google Scholar]
  • 5.Beckett EA, Ro S, Bayguinov Y, et al. Kit signaling is essential for development and maintenance of interstitial cells of Cajal and electrical rhythmicity in the embryonic gastrointestinal tract. Dev Dyn. 2007;236:60–72. doi: 10.1002/dvdy.20929. [DOI] [PubMed] [Google Scholar]
  • 6.Sanders KM, Ordog T, Koh SD, et al. Development and plasticity of interstitial cells of Cajal. Neurogastroenterol Motil. 1999;11:311–338. doi: 10.1046/j.1365-2982.1999.00164.x. [DOI] [PubMed] [Google Scholar]
  • 7.Vanderwinden JM, Rumessen JJ. Interstitial cells of Cajal in human gut and gastrointestinal disease. Microsc Res Tech. 1999;47:344–360. doi: 10.1002/(SICI)1097-0029(19991201)47:5<344::AID-JEMT6>3.0.CO;2-1. [DOI] [PubMed] [Google Scholar]
  • 8.Farrugia G. Interstitial cells of Cajal in health and disease. Neurogastroenterol Motil. 2008;20(Suppl 1):54–63. doi: 10.1111/j.1365-2982.2008.01109.x. [DOI] [PubMed] [Google Scholar]
  • 9.Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998;279:577–580. doi: 10.1126/science.279.5350.577. [DOI] [PubMed] [Google Scholar]
  • 10.Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal tumors: a consensus approach. Hum Pathol. 2002;33:459–465. doi: 10.1053/hupa.2002.123545. [DOI] [PubMed] [Google Scholar]
  • 11.Kindblom LG, Remotti HE, Aldenborg F, et al. Gastrointestinal pacemaker cell tumor (GIPACT): gastrointestinal stromal tumors show phenotypic characteristics of the interstitial cells of Cajal. Am J Pathol. 1998;152:1259–1269. [PMC free article] [PubMed] [Google Scholar]
  • 12.Vanderwinden JM, Rumessen JJ, Bernex F, et al. Distribution and ultrastructure of interstitial cells of Cajal in the mouse colon, using antibodies to Kit and Kit(W-lacZ) mice. Cell Tissue Res. 2000;302:155–170. doi: 10.1007/s004419900170. [DOI] [PubMed] [Google Scholar]
  • 13.Lorincz A, Redelman D, Horváth VJ, et al. Progenitors of interstitial cells of Cajal in the postnatal murine stomach. Gastroenterology. 2008;134:1083–1093. doi: 10.1053/j.gastro.2008.01.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Robson ME, Glogowski E, Sommer G, et al. Pleomorphic characteristics of a germ-line KIT mutation in a large kindred with gastrointestinal stromal tumors, hyperpigmentation, and dysphagia. Clin Cancer Res. 2004;10:1250–1254. doi: 10.1158/1078-0432.ccr-03-0110. [DOI] [PubMed] [Google Scholar]
  • 15.Hirota S, Okazaki T, Kitamura Y, et al. Cause of familial and multiple gastrointestinal autonomic nerve tumors with hyperplasia of interstitial cells of Cajal is germline mutation of the c-kit gene. Am J Surg Pathol. 2000;24:326–327. doi: 10.1097/00000478-200002000-00045. [DOI] [PubMed] [Google Scholar]
  • 16.Sommer G, Agosti V, Ehlers I, et al. Gastrointestinal stromal tumors in a mouse model by targeted mutation of the Kit receptor tyrosine kinase. Proc Natl Acad Sci U S A. 2003;100:6706–6711. doi: 10.1073/pnas.1037763100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rossi F, Ehlers I, Agosti V, et al. Oncogenic Kit signaling and therapeutic intervention in a mouse model of gastrointestinal stromal tumor. Proc Natl Acad Sci U S A. 2006;103:12843–12848. doi: 10.1073/pnas.0511076103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Rubin BP, Antonescu CR, Scott-Browne JP, et al. A knock-in mouse model of gastrointestinal stromal tumor harboring kit K641E. Cancer Res. 2005;65:6631–6639. doi: 10.1158/0008-5472.CAN-05-0891. [DOI] [PubMed] [Google Scholar]
  • 19.Nakai N, Ishikawa T, Nishitani A, et al. A mouse model of a human multiple GIST family with KIT-Asp820Tyr mutation generated by a knock-in strategy. J Pathol. 2008;214:302–311. doi: 10.1002/path.2296. [DOI] [PubMed] [Google Scholar]
  • 20.Suzuki H, Hirst GD. Regenerative potentials evoked in circular smooth muscle of the antral region of guinea-pig stomach. J Physiol. 1999;517:563–573. doi: 10.1111/j.1469-7793.1999.0563t.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Chang IY, Glasgow NJ, Takayama I, et al. Loss of interstitial cells of Cajal and development of electrical dysfunction in murine small bowel obstruction. J Physiol. 2001;536:555–568. doi: 10.1111/j.1469-7793.2001.0555c.xd. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ward SM, Gershon MD, Keef K, et al. Interstitial cells of Cajal and electrical activity in ganglionic and aganglionic colons of mice. Am J Physiol. 2002;283:G445–D456. doi: 10.1152/ajpgi.00475.2001. [DOI] [PubMed] [Google Scholar]
  • 23.Beckett EA, Bayguinov YR, Sanders KM, et al. Properties of unitary potentials generated by intramuscular interstitial cells of Cajal in the murine and guinea-pig gastric fundus. J Physiol. 2004;559:259–269. doi: 10.1113/jphysiol.2004.063016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ward SM, Ordög T, Bayguinov JR, et al. Development of interstitial cells of Cajal and pacemaking in mice lacking enteric nerves. Gastroenterology. 1999;117:584–594. doi: 10.1016/s0016-5085(99)70451-8. [DOI] [PubMed] [Google Scholar]
  • 25.Beckett EA, Takeda Y, Yanase H, et al. Synaptic specializations exist between enteric motor nerves and interstitial cells of Cajal in the murine stomach. J Comp Neurol. 2005;493:193–206. doi: 10.1002/cne.20746. [DOI] [PubMed] [Google Scholar]
  • 26.Ward SM, Beckett EA, Wang X, et al. Interstitial cells of Cajal mediate cholinergic neurotransmission from enteric motor neurons. J Neurosci. 2000;20:1393–1403. doi: 10.1523/JNEUROSCI.20-04-01393.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ordög T, Ward SM, Sanders KM. Interstitial cells of Cajal generate electrical slow waves in the murine stomach. J Physiol. 1999;518:257–269. doi: 10.1111/j.1469-7793.1999.0257r.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hirst GD, Ward SM. Interstitial cells: involvement in rhythmicity and neural control of gut smooth muscle. J Physiol. 2003;550:337–346. doi: 10.1113/jphysiol.2003.043299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sanders KM, Koh SD, Ward SM. Interstitial cells of Cajal as pacemakers in the gastrointestinal tract. Annu Rev Physiol. 2006;68:307–343. doi: 10.1146/annurev.physiol.68.040504.094718. [DOI] [PubMed] [Google Scholar]
  • 30.Horváth VJ, Vittal H, Lörincz A, et al. Reduced stem cell factor links smooth myopathy and loss of interstitial cells of cajal in murine diabetic gastroparesis. Gastroenterology. 2006;130:759–770. doi: 10.1053/j.gastro.2005.12.027. [DOI] [PubMed] [Google Scholar]
  • 31.Torihashi S, Ward SM, Sanders KM. Development of c-kit-positive cells and the onset of electrical rhythmicity in murine small intestine. Gastroenterology. 1997;12:144–155. doi: 10.1016/s0016-5085(97)70229-4. [DOI] [PubMed] [Google Scholar]
  • 32.Klüppel M, Huizinga JD, Malysz J, et al. Developmental origin and Kit-dependent development of the interstitial cells of cajal in the mammalian small intestine. Dev Dyn. 1998;211:60–71. doi: 10.1002/(SICI)1097-0177(199801)211:1<60::AID-AJA6>3.0.CO;2-5. [DOI] [PubMed] [Google Scholar]
  • 33.Bernex F, De Sepulveda P, Kress C, et al. Spatial and temporal patterns of c-kit-expressing cells in WlacZ/+ and WlacZ/WlacZ mouse embryos. Development. 1996;122:3023–3033. doi: 10.1242/dev.122.10.3023. [DOI] [PubMed] [Google Scholar]
  • 34.Sircar K, Hewlett BR, Huizinga JD, et al. Interstitial cells of Cajal as precursors of gastrointestinal stromal tumors. Am J Surg Pathol. 1999;23:377–389. doi: 10.1097/00000478-199904000-00002. [DOI] [PubMed] [Google Scholar]
  • 35.Vanderwinden JM, Rumessen JJ, De Laet MH, et al. CD34 immunoreactivity and interstitial cells of Cajal in the human and mouse gastrointestinal tract. Cell Tissue Res. 2000;302:145–153. doi: 10.1007/s004410000264. [DOI] [PubMed] [Google Scholar]
  • 36.Kurahashi M, Niwa Y, Cheng J, et al. Platelet-derived growth factor signals play critical roles in differentiation of longitudinal smooth muscle cells in mouse embryonic gut. Neurogastroenterol Motil. 2008;20:521–531. doi: 10.1111/j.1365-2982.2007.01055.x. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

RESOURCES