Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Cell Calcium ; 99: 102472, 2021 11.
Article En | MEDLINE | ID: mdl-34537580

Myenteric interstitial cells of Cajal (ICC-MY) generate and actively propagate electrical slow waves in the stomach. Slow wave generation and propagation are altered in gastric motor disorders, such as gastroparesis, and the mechanism for the gradient in slow wave frequency that facilitates proximal to distal propagation of slow waves and normal gastric peristalsis is poorly understood.  Slow waves depend upon Ca2+-activated Cl- channels (encoded by Ano1). We characterized Ca2+ signaling in ICC-MY in situ using mice engineered to have cell-specific expression of GCaMP6f in ICC. Ca2+ signaling differed in ICC-MY in corpus and antrum. Localized Ca2+ transients were generated from multiple firing sites and were organized into Ca2+ transient clusters (CTCs). Ca2+ transient refractory periods occurred upon cessation of CTCs, but a relatively higher frequency of Ca2+ transients persisted during the inter-CTC interval in corpus than in antrum ICC-MY. The onset of Ca2+ transients after the refractory period was associated with initiation of the next CTC. Thus, CTCs were initiated at higher frequencies in corpus than in antrum ICC-MY. Initiation and propagation of CTCs (and electrical slow waves) depends upon T-type Ca2+ channels, and durations of CTCs relied upon L-type Ca2+ channels. The durations of CTCs mirrored the durations of slow waves. CTCs and Ca2+ transients between CTCs resulted from release of Ca2+ from intracellular stores and were maintained, in part, by store-operated Ca2+ entry. Our data suggest that Ca2+ release and activation of Ano1 channels both initiate and contribute to the plateau phase of slow waves.


Calcium Signaling , Interstitial Cells of Cajal , Animals , Mice , Stomach
2.
J Physiol ; 597(9): 2337-2360, 2019 05.
Article En | MEDLINE | ID: mdl-30843201

KEY POINTS: Electrical pacemaking in gastrointestinal muscles is generated by specialized interstitial cells of Cajal that produce the patterns of contractions required for peristalsis and segmentation in the gut. The calcium-activated chloride conductance anoctamin-1 (Ano1) has been shown to be responsible for the generation of pacemaker activity in GI muscles, but this conclusion is established from studies of juvenile animals in which effects of reduced Ano1 on gastric emptying and motor patterns could not be evaluated. Knocking down Ano1 expression using Cre/LoxP technology caused dramatic changes in in gastric motor activity, with disrupted slow waves, abnormal phasic contractions and delayed gastric emptying; modest changes were noted in the small intestine. Comparison of the effects of Ano1 antagonists on muscles from juvenile and adult small intestinal muscles suggests that conductances in addition to Ano1 may develop with age and contribute to pacemaker activity. ABSTRACT: Interstitial cells of Cajal (ICC) generate slow waves and transduce neurotransmitter signals in the gastrointestinal (GI) tract, facilitating normal motility patterns. ICC express a Ca2+ -activated Cl- conductance (CaCC), and constitutive knockout of the channel protein anoctamin-1 leads to loss of slow waves in gastric and intestinal muscles. These knockout experiments were performed on juvenile mice. However, additional experiments demonstrated significant differences in the sensitivity of gastric and intestinal muscles to antagonists of anoctamin-1 channels. Furthermore, the significance of anoctamin-1 and the electrical and mechanical behaviours facilitated by this conductance have not been evaluated on the motor behaviours of adult animals. Cre/loxP technology was used to generate cell-specific knockdowns of anoctamin-1 in ICC (KitCreERT2/+ ;Ano1tm2jrr/+ ) in GI muscles. The recombination efficiency of KitCreERT was evaluated with an eGFP reporter, molecular techniques and immunohistochemistry. Electrical and contractile experiments were used to examine the consequences of anoctamin-1 knockdown on pacemaker activity, mechanical responses, gastric motility patterns, gastric emptying and GI transit. Reduced anoctamin-1 caused loss of gastric, but not intestinal slow waves. Irregular spike complexes developed in gastric muscles, leading to uncoordinated antral contractions, delayed gastric emptying and increased total GI transit time. Slow waves in intestinal muscles of juvenile mice were more sensitive to anoctamin-1 antagonists than slow waves in adult muscles. The low susceptibility to anoctamin-1 knockdown and weak efficacy of anoctamin-1 antagonists in inhibiting slow waves in adult small intestinal muscles suggest that a conductance in addition to anoctamin-1 may develop in small intestinal ICC with ageing and contribute to pacemaker activity.


Anoctamin-1/metabolism , Gastrointestinal Motility , Intestine, Small/physiology , Muscle, Smooth/metabolism , Stomach/physiology , Animals , Anoctamin-1/genetics , Calcium Channel Blockers/pharmacology , Interstitial Cells of Cajal/metabolism , Intestine, Small/cytology , Intestine, Small/growth & development , Mice , Mice, Inbred C57BL , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Nifedipine/pharmacology , Stomach/cytology , Stomach/growth & development
3.
Diabetes ; 68(3): 637-647, 2019 03.
Article En | MEDLINE | ID: mdl-30626609

Patients with diabetes often develop gastrointestinal motor problems, including gastroparesis. Previous studies have suggested this gastric motor disorder was a consequence of an enteric neuropathy. Disruptions in interstitial cells of Cajal (ICC) have also been reported. A thorough examination of functional changes in gastric motor activity during diabetes has not yet been performed. We comprehensively examined the gastric antrums of Lepob mice using functional, morphological, and molecular techniques to determine the pathophysiological consequences in this type 2 diabetic animal model. Video analysis and isometric force measurements revealed higher frequency and less robust antral contractions in Lepob mice compared with controls. Electrical pacemaker activity was reduced in amplitude and increased in frequency. Populations of enteric neurons, ICC, and platelet-derived growth factor receptor α+ cells were unchanged. Analysis of components of the prostaglandin pathway revealed upregulation of multiple enzymes and receptors. Prostaglandin-endoperoxide synthase-2 inhibition increased slow wave amplitudes and reduced frequency of diabetic antrums. In conclusion, gastric pacemaker and contractile activity is disordered in type 2 diabetic mice, and this appears to be a consequence of excessive prostaglandin signaling. Inhibition of prostaglandin synthesis may provide a novel treatment for diabetic gastric motility disorders.


Diabetes Mellitus, Type 2/metabolism , Prostaglandins/metabolism , Animals , Blood Glucose/metabolism , Blood Glucose Self-Monitoring , Cyclooxygenase 2/metabolism , Diabetes Mellitus, Experimental/metabolism , Electrophysiology , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Real-Time Polymerase Chain Reaction , Signal Transduction
4.
J Physiol ; 596(9): 1549-1574, 2018 05 01.
Article En | MEDLINE | ID: mdl-29430647

KEY POINTS: Enteric neurotransmission is essential for gastrointestinal (GI) motility, although the cells and conductances responsible for post-junctional responses are controversial. The calcium-activated chloride conductance (CaCC), anoctamin-1 (Ano1), was expressed by intramuscular interstitial cells of Cajal (ICC-IM) in proximal stomach and not resolved in smooth muscle cells (SMCs). Cholinergic nerve fibres were closely apposed to ICC-IM. Conductances activated by cholinergic stimulation in isolated ICC-IM and SMCs were determined. A CaCC was activated by carbachol in ICC-IM and a non-selective cation conductance in SMCs. Responses to cholinergic nerve stimulation were studied. Excitatory junction potentials (EJPs) and mechanical responses were evoked in wild-type mice but absent or greatly reduced with knockout/down of Ano1. Drugs that block Ano1 inhibited the conductance activated by carbachol in ICC-IM and EJPs and mechanical responses in tissues. The data of the present study suggest that electrical and mechanical responses to cholinergic nerve stimulation are mediated by Ano1 expressed in ICC-IM and not SMCs. ABSTRACT: Enteric motor neurotransmission is essential for normal gastrointestinal (GI) motility. Controversy exists regarding the cells and ionic conductance(s) that mediate post-junctional neuroeffector responses to motor neurotransmitters. Isolated intramuscular ICC (ICC-IM) and smooth muscle cells (SMCs) from murine fundus muscles were used to determine the conductances activated by carbachol (CCh) in each cell type. The calcium-activated chloride conductance (CaCC), anoctamin-1 (Ano1) is expressed by ICC-IM but not resolved in SMCs, and CCh activated a Cl- conductance in ICC-IM and a non-selective cation conductance in SMCs. We also studied responses to nerve stimulation using electrical-field stimulation (EFS) of intact fundus muscles from wild-type and Ano1 knockout mice. EFS activated excitatory junction potentials (EJPs) in wild-type mice, although EJPs were absent in mice with congenital deactivation of Ano1 and greatly reduced in animals in which the CaCC-Ano1 was knocked down using Cre/loxP technology. Contractions to cholinergic nerve stimulation were also greatly reduced in Ano1 knockouts. SMCs cells also have receptors and ion channels activated by muscarinic agonists. Blocking acetylcholine esterase with neostigmine revealed a slow depolarization that developed after EJPs in wild-type mice. This depolarization was still apparent in mice with genetic deactivation of Ano1. Pharmacological blockers of Ano1 also inhibited EJPs and contractile responses to muscarinic stimulation in fundus muscles. The data of the present study are consistent with the hypothesis that ACh released from motor nerves binds muscarinic receptors on ICC-IM with preference and activates Ano1. If metabolism of acetylcholine is inhibited, ACh overflows and binds to extrajunctional receptors on SMCs, eliciting a slower depolarization response.


Acetylcholine/metabolism , Interstitial Cells of Cajal/physiology , Myocytes, Smooth Muscle/physiology , Stomach/physiology , Synaptic Transmission , Animals , Anoctamin-1/physiology , Chloride Channels/physiology , Electric Stimulation , Gastric Fundus/cytology , Gastric Fundus/physiology , Interstitial Cells of Cajal/cytology , Mice , Mice, Knockout , Muscle Contraction , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Stomach/cytology
5.
PLoS One ; 12(2): e0171262, 2017.
Article En | MEDLINE | ID: mdl-28152551

Serum response factor (SRF) transcriptionally regulates expression of contractile genes in smooth muscle cells (SMC). Lack or decrease of SRF is directly linked to a phenotypic change of SMC, leading to hypomotility of smooth muscle in the gastrointestinal (GI) tract. However, the molecular mechanism behind SRF-induced hypomotility in GI smooth muscle is largely unknown. We describe here how SRF plays a functional role in the regulation of the SMC contractility via myotonic dystrophy protein kinase (DMPK) and L-type calcium channel CACNA1C. GI SMC expressed Dmpk and Cacna1c genes into multiple alternative transcriptional isoforms. Deficiency of SRF in SMC of Srf knockout (KO) mice led to reduction of SRF-dependent DMPK, which down-regulated the expression of CACNA1C. Reduction of CACNA1C in KO SMC not only decreased intracellular Ca2+ spikes but also disrupted their coupling between cells resulting in decreased contractility. The role of SRF in the regulation of SMC phenotype and function provides new insight into how SMC lose their contractility leading to hypomotility in pathophysiological conditions within the GI tract.


Calcium Channels, L-Type/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Myotonin-Protein Kinase/physiology , Serum Response Factor/physiology , Animals , Blotting, Western , Female , Male , Mice , Mice, Knockout , Microscopy, Confocal , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/ultrastructure , Polymerase Chain Reaction , Proteomics , Tamoxifen/pharmacology
6.
J Neurogastroenterol Motil ; 20(3): 294-317, 2014 Jul 31.
Article En | MEDLINE | ID: mdl-24948131

Smooth muscle layers of the gastrointestinal tract consist of a heterogeneous population of cells that include enteric neurons, several classes of interstitial cells of mesenchymal origin, a variety of immune cells and smooth muscle cells (SMCs). Over the last number of years the complexity of the interactions between these cell types has begun to emerge. For example, interstitial cells, consisting of both interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor alpha-positive (PDGFRα(+)) cells generate pacemaker activity throughout the gastrointestinal (GI) tract and also transduce enteric motor nerve signals and mechanosensitivity to adjacent SMCs. ICC and PDGFRα(+) cells are electrically coupled to SMCs possibly via gap junctions forming a multicellular functional syncytium termed the SIP syncytium. Cells that make up the SIP syncytium are highly specialized containing unique receptors, ion channels and intracellular signaling pathways that regulate the excitability of GI muscles. The unique role of these cells in coordinating GI motility is evident by the altered motility patterns in animal models where interstitial cell networks are disrupted. Although considerable advances have been made in recent years on our understanding of the roles of these cells within the SIP syncytium, the full physiological functions of these cells and the consequences of their disruption in GI muscles have not been clearly defined. This review gives a synopsis of the history of interstitial cell discovery and highlights recent advances in structural, molecular expression and functional roles of these cells in the GI tract.

7.
Cell Tissue Res ; 350(2): 199-213, 2012 Nov.
Article En | MEDLINE | ID: mdl-22864981

Kit immunohistochemistry and confocal reconstructions have provided detailed 3-dimensional images of ICC networks throughout the gastrointestinal (GI) tract. Morphological criteria have been used to establish that different classes of ICC exist within the GI tract and physiological studies have shown that these classes have distinct physiological roles in GI motility. Structural studies have focused predominately on rodent models and less information is available on whether similar classes of ICC exist within the GI tracts of humans or non-human primates. Using Kit immunohistochemistry and confocal imaging, we examined the 3-dimensional structure of ICC throughout the GI tract of cynomolgus monkeys. Whole or flat mounts and cryostat sections were used to examine ICC networks in the lower esophageal sphincter (LES), stomach, small intestine and colon. Anti-histamine antibodies were used to distinguish ICC from mast cells in the lamina propria. Kit labeling identified complex networks of ICC populations throughout the non-human primate GI tract that have structural characteristics similar to that described for ICC populations in rodent models. ICC-MY formed anastomosing networks in the myenteric plexus region. ICC-IM were interposed between smooth muscle cells in the stomach and colon and were concentrated within the deep muscular plexus (ICC-DMP) of the intestine. ICC-SEP were found in septal regions of the antrum that separated circular muscle bundles. Spindle-shaped histamine(+) mast cells were found in the lamina propria throughout the GI tract. Since similar sub-populations of ICC exist within the GI tract of primates and rodents and the use of rodents to study the functional roles of different classes of ICC is warranted.


Gastrointestinal Tract/cytology , Interstitial Cells of Cajal/cytology , Animals , Female , Gastrointestinal Tract/metabolism , Humans , Immunohistochemistry , Interstitial Cells of Cajal/metabolism , Macaca fascicularis , Male
9.
J Physiol ; 590(8): 1957-72, 2012 Apr 15.
Article En | MEDLINE | ID: mdl-22371476

Activation of enteric inhibitory motor neurons causes inhibitory junctional potentials (IJPs) and muscle relaxation in mammalian gastrointestinal (GI) muscles, including humans. IJPs in many GI muscles are bi-phasic with a fast initial hyperpolarization (fIJP) due to release of a purine neurotransmitter and a slower hyperpolarization component (sIJP) due to release of nitric oxide. We sought to characterize the nature of the post-junctional receptor(s) involved in transducing purinergic neural inputs in the murine colon using mice with genetically deactivated P2ry1. Wild-type mice had characteristic biphasic IJPs and pharmacological dissection confirmed that the fIJP was purinergic and the sIJP was nitrergic. The fIJP was completely absent in P2ry1(−/−) mice and the P2Y1 receptor antagonist MRS2500 had no effect on electrical activity or responses to electrical field stimulation of intrinsic nerves in these mice. Contractile experiments confirmed that purinergic responses were abolished in P2ry1(−/−) mice. Picospritzing of neurotransmitter candidates (ATP and its primary metabolite, ADP) and ß-NAD (and its primary metabolite, ADP-ribose, ADPR) caused transient hyperpolarization responses in wild-type colons, but responses to ß-NAD and ADPR were completely abolished in P2ry1(−/−) mice. Hyperpolarization and relaxation responses to ATP and ADP were retained in colons of P2ry1(−/−) mice. Video imaging revealed that transit of fecal pellets was significantly delayed in colons from P2ry1(−/−) mice. These data demonstrate the importance of purinergic neurotransmission in regulating colonic motility and confirm pharmacological experiments suggesting that purinergic neurotransmission is mediated via P2Y1 receptors.


Colon/physiology , Motor Neurons/metabolism , Neuromuscular Junction/metabolism , Neurotransmitter Agents/metabolism , Receptors, Purinergic P2Y1/metabolism , Animals , Colon/drug effects , Colon/metabolism , Electric Stimulation/methods , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Gastrointestinal Transit/drug effects , Gastrointestinal Transit/physiology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Motor Neurons/drug effects , Muscle Relaxation/drug effects , Muscle Relaxation/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Neuromuscular Junction/drug effects , Nitric Oxide/metabolism , Purinergic P2 Receptor Antagonists/pharmacology , Purines/metabolism , Synaptic Transmission/drug effects
10.
Gastroenterology ; 138(3): 1068-78.e1-2, 2010 Mar.
Article En | MEDLINE | ID: mdl-19917283

BACKGROUND & AIMS: Interstitial cells of Cajal (ICC) express the receptor tyrosine kinase, KIT, the receptor for stem cell factor. In the gastrointestinal (GI) tract, ICC are pacemaker cells that generate spontaneous electrical slow waves, and mediate inputs from motor neurons. Absence or loss of ICC are associated with GI motility disorders, including those consequent of diabetes. Studies of ICC have been hampered by the low density of these cells and difficulties in recognizing these cells in cell dispersions. METHODS: Kit(+/copGFP) mice harboring a copepod super green fluorescent protein (copGFP) complementary DNA, inserted at the Kit locus, were generated. copGFP(+) ICC from GI muscles were analyzed using confocal microscopy and flow cytometry. copGFP(+) ICC from the jejunum were purified by a fluorescence-activated cell sorter and validated by cell-specific markers. Kit(+/copGFP) mice were crossbred with diabetic Lep(+/ob) mice to generate compound Kit(+/copGFP);Lep(ob/ob) mutant mice. copGFP(+) ICC from compound transgenic mice were analyzed by confocal microscopy. RESULTS: copGFP in Kit(+/copGFP) mice colocalized with KIT immunofluorescence and thus was predominantly found in ICC. In other smooth muscles, mast cells were also labeled, but these cells were relatively rare in the murine GI tract. copGFP(+) cells from jejunal muscles were Kit(+) and free of contaminating cell-specific markers. Kit(+/copGFP);Lep(ob/ob) mice displayed ICC networks that were dramatically disrupted during the development of diabetes. CONCLUSIONS: Kit(+/copGFP) mice offer a powerful new model to study the function and genetic regulation of ICC phenotypes. Isolation of ICC from animal models will help determine the causes and responses of ICC to therapeutic agents.


Diabetes Mellitus, Type 2/metabolism , Gastrointestinal Diseases/metabolism , Interstitial Cells of Cajal/metabolism , Intestinal Mucosa/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Animals , Biomarkers/metabolism , Cell Separation/methods , Cells, Cultured , Crosses, Genetic , Diabetes Mellitus, Type 2/pathology , Female , Flow Cytometry , Fluorescent Antibody Technique , Gastrointestinal Diseases/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Interstitial Cells of Cajal/pathology , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Phenotype , Proto-Oncogene Proteins c-kit/genetics
11.
J Physiol ; 587(Pt 20): 4887-904, 2009 Oct 15.
Article En | MEDLINE | ID: mdl-19687122

Interstitial cells of Cajal (ICC) generate pacemaker activity (slow waves) in gastrointestinal (GI) smooth muscles, but the mechanism(s) of pacemaker activity are controversial. Several conductances, such as Ca(2+)-activated Cl() channels (CaCC) and non-selective cation channels (NSCC) have been suggested to be involved in slow wave depolarization. We investigated the expression and function of a new class of CaCC, anoctamin 1 (ANO1), encoded by Tmem16a, which was discovered to be highly expressed in ICC in a microarray screen. GI muscles express splice variants of the Tmem16a transcript in addition to other paralogues of the Tmem16a family. ANO1 protein is expressed abundantly and specifically in ICC in all regions of the murine, non-human primate (Macaca fascicularis) and human GI tracts. CaCC blocking drugs, niflumic acid and 4,4-diisothiocyano-2,2-stillbene-disulfonic acid (DIDS) reduced the frequency and blocked slow waves in murine, primate, human small intestine and stomach in a concentration-dependent manner. Unitary potentials, small stochastic membrane depolarizations thought to underlie slow waves, were insensitive to CaCC blockers. Slow waves failed to develop by birth in mice homozygous for a null allele of Tmem16a (Tmem16a(tm1Bdh)(/tm1Bdh)) and did not develop subsequent to birth in organ culture, as in wildtype and heterozygous muscles. Loss of function of ANO1 did not inhibit the development of ICC networks that appeared structurally normal as indicated by Kit antibodies. These data demonstrate the fundamental role of ANO1 in the generation of slow waves in GI ICC.


Gastrointestinal Motility/physiology , Gastrointestinal Tract/physiology , Interstitial Cells of Cajal/physiology , Membrane Proteins/metabolism , Muscle, Smooth/physiology , Neoplasm Proteins/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Animals , Anoctamin-1 , Chloride Channels , Cyclooxygenase Inhibitors/pharmacology , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/cytology , Gastrointestinal Tract/drug effects , Gene Expression Regulation , Humans , Immunohistochemistry , Interstitial Cells of Cajal/cytology , Interstitial Cells of Cajal/drug effects , Macaca fascicularis , Membrane Proteins/genetics , Mice , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Neoplasm Proteins/genetics , Niflumic Acid/pharmacology , RNA/analysis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
...