1
|
Gupta N, Baker SA, Sanders KM, Griffin CS, Sergeant GP, Hollywood MA, Thornbury KD, Drumm BT. Interstitial cell of Cajal-like cells (ICC-LC) exhibit dynamic spontaneous activity but are not functionally innervated in mouse urethra. Cell Calcium 2024; 123:102931. [PMID: 39068674 DOI: 10.1016/j.ceca.2024.102931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/09/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Urethral smooth muscle cells (USMC) contract to occlude the internal urethral sphincter during bladder filling. Interstitial cells also exist in urethral smooth muscles and are hypothesized to influence USMC behaviours and neural responses. These cells are similar to Kit+ interstitial cells of Cajal (ICC), which are gastrointestinal pacemakers and neuroeffectors. Isolated urethral ICC-like cells (ICC-LC) exhibit spontaneous intracellular Ca2+ signalling behaviours that suggest these cells may serve as pacemakers or neuromodulators similar to ICC in the gut, although observation and direct stimulation of ICC-LC within intact urethral tissues is lacking. We used mice with cell-specific expression of the Ca2+ indicator, GCaMP6f, driven off the endogenous promoter for Kit (Kit-GCaMP6f mice) to identify ICC-LC in situ within urethra muscles and to characterize spontaneous and nerve-evoked Ca2+ signalling. ICC-LC generated Ca2+ waves spontaneously that propagated on average 40.1 ± 0.7 μm, with varying amplitudes, durations, and spatial spread. These events originated from multiple firing sites in cells and the activity between sites was not coordinated. ICC-LC in urethra formed clusters but not interconnected networks. No evidence for entrainment of Ca2+ signalling between ICC-LC was obtained. Ca2+ events in ICC-LC were unaffected by nifedipine but were abolished by cyclopiazonic acid and decreased by an antagonist of Orai Ca2+ channels (GSK-7975A). Phenylephrine increased Ca2+ event frequency but a nitric oxide donor (DEA-NONOate) had no effect. Electrical field stimulation (EFS, 10 Hz) of intrinsic nerves, which evoked contractions of urethral rings and increased Ca2+ event firing in USMC, failed to evoke responses in ICC-LC. Our data suggest that urethral ICC-LC are spontaneously active but are not regulated by autonomic neurons.
Collapse
Affiliation(s)
- Neha Gupta
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Salah A Baker
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Kenton M Sanders
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Caoimhin S Griffin
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Gerard P Sergeant
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Keith D Thornbury
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Bernard T Drumm
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland; Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA.
| |
Collapse
|
2
|
Xu H, Miao F, Fan Y. A bibliometric analysis of diabetic gastroparesis from 1979 to 2024. Front Med (Lausanne) 2024; 11:1445276. [PMID: 39450111 PMCID: PMC11500038 DOI: 10.3389/fmed.2024.1445276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/09/2024] [Indexed: 10/26/2024] Open
Abstract
Objective Gastroparesis is one of the complications of diabetes mellitus, which has a major impact on the quality of life of patients, and the limited therapeutic options currently available make it a public health problem. No bibliometric studies on diabetic gastroparesis have been published to date. Therefore, the aim of this paper is to summarize and analyze the research hotspots for researchers. Methods Research articles related to Diabetic gastroparesis were searched in Web of Science Core Collection (WOSCC), and relevant information was extracted after screening. A comprehensive bibliometric analysis of 699 publications was conducted using Microsoft Excel 2019, Citespace and VOSviewers. Result A total of 699 papers from 738 institutions in 41 countries were retrieved. Publications in this field have increased rapidly since 1979. USA (n = 370) and Mayo Clinical (n = 69) were the most productive country and institution, respectively. Neurogastroenterology and Motility (n = 67) was the most published journal with Parkman, Henry P. (n = 40) having the highest number of articles; Gastroenterology and Mccallum, Richard W. were the most influential journals and authors. Conclusions The research hotspots of Diabetic gastroparesis are mainly focused on treatment modalities and pathological mechanisms. Future research in diabetic gastroparesis will focus on exploring the pathomechanisms, finding long-term effective treatments, and improving patients' quality of life.
Collapse
Affiliation(s)
| | | | - Yushan Fan
- College of Acupuncture-Moxibustion and Tuina, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
3
|
Hannigan KI, Ni Bhraonain EP, Gould TW, Keef KD, Cobine CA. Modulation of intracellular calcium activity in interstitial cells of Cajal by inhibitory neural pathways within the internal anal sphincter. Am J Physiol Gastrointest Liver Physiol 2024; 327:G382-G404. [PMID: 38860285 PMCID: PMC11427099 DOI: 10.1152/ajpgi.00309.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
The internal anal sphincter (IAS) functions to maintain continence. Previous studies utilizing mice with cell-specific expression of GCaMP6f revealed two distinct subtypes of intramuscular interstitial cells of Cajal (ICC-IM) with differing Ca2+ activities in the IAS. The present study further examined Ca2+ activity in ICC-IM and its modulation by inhibitory neurotransmission. The spatiotemporal properties of Ca2+ transients in Type II ICC-IM mimicked those of smooth muscle cells (SMCs), indicating their joint participation in the "SIP" syncytium. Electrical field stimulation (EFS; atropine present) abolished localized and whole cell Ca2+ transients in Type I and II ICC-IM. The purinergic antagonist MRS2500 did not abolish EFS responses in either cell type, whereas the nitric oxide synthase (NOS) inhibitor NG-nitro-l-arginine (l-NNA) abolished responses in Type I but not Type II ICC-IM. Combined antagonists abolished EFS responses in Type II ICC-IM. In both ICC-IM subtypes, the ability of EFS to inhibit Ca2+ release was abolished by l-NNA but not MRS2500, suggesting that the nitrergic pathway directly inhibits ICC-IM by blocking Ca2+ release from intracellular stores. Since inositol (1,4,5)-trisphosphate receptor-associated cGMP kinase substrate I (IRAG1) is expressed in ICC-IM, it is possible that it participates in the inhibition of Ca2+ release by nitric oxide. Platelet-derived growth factor receptor α (PDGFRα)+ cells but not ICC-IM expressed P2Y1 receptors (P2Y1R) and small-conductance Ca2+-activated K+ channels (SK3), suggesting that the purinergic pathway indirectly blocks whole cell Ca2+ transients in Type II ICC-IM via PDGFRα+ cells. This study provides the first direct evidence for functional coupling between inhibitory motor neurons and ICC-IM subtypes in the IAS, with contractile inhibition ultimately dependent upon electrical coupling between SMCs, ICC, and PDGFRα+ cells via the SIP syncytium.NEW & NOTEWORTHY Two intramuscular interstitial cells of Cajal (ICC-IM) subtypes exist within the internal anal sphincter (IAS). This study provides the first evidence for direct coupling between nitrergic motor neurons and both ICC-IM subtypes as well as indirect coupling between purinergic inputs and Type II ICC-IM. The spatiotemporal properties of whole cell Ca2+ transients in Type II ICC-IM mimic those of smooth muscle cells (SMCs), suggesting that ICC-IM modulate the activity of SMCs via their joint participation in a SIP syncytium (SMCs, ICC, and PDGFRα+ cells).
Collapse
Affiliation(s)
- Karen I Hannigan
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Emer P Ni Bhraonain
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Thomas W Gould
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Kathleen D Keef
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Caroline A Cobine
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| |
Collapse
|
4
|
Ni Bhraonain E, Turner J, Hannigan K, Sanders K, Cobine C. Immunohistochemical characterization of interstitial cells and their relationship to motor neurons within the mouse esophagus. RESEARCH SQUARE 2024:rs.3.rs-4474290. [PMID: 38947055 PMCID: PMC11213231 DOI: 10.21203/rs.3.rs-4474290/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Interstitial cells of Cajal (ICC) and PDGFRα+ cells regulate smooth muscle motility in the gastrointestinal (GI) tract. However, their role(s) in esophageal motility are still unclear. The mouse esophagus has traditionally been described as almost entirely skeletal muscle in nature though ICC have been identified along its entire length. The current study evaluated the distribution of skeletal and smooth muscle within the esophagus using a mouse selectively expressing eGFP in smooth muscle cells (SMCs). The relationship of SMCs to ICC and PDGFRα+ cells was also examined. SMCs declined in density in the oral direction however SMCs represented ~ 25% of the area in the distal esophagus suggesting a likeness to the transition zone observed in humans. ANO1+ intramuscular ICC (ICC-IM) were distributed along the length of the esophagus though like SMCs, declined proximally. ICC-IM were closely associated with SMCs but were also found in regions devoid of SMCs. Intramuscular and submucosal PDGFRα+ cells were densely distributed throughout the esophagus though only intramuscular PDGFRα+ cells within the LES and distal esophagus highly expressed SK3. ICC-IM and PDGFRα+ cells were closely associated with nNOS+, VIP+, VAChT+ and TH+ neurons throughout the LES and distal esophagus. GFAP+ cells resembling intramuscular enteric glia were observed within the muscle and were closely associated with ICC-IM and PDGFRα+ cells, occupying a similar location to c. These data suggest that the mouse esophagus is more similar to the human than thought previously and thus set the foundation for future functional and molecular studies using transgenic mice.
Collapse
|
5
|
Llorente C. The Imperative for Innovative Enteric Nervous System-Intestinal Organoid Co-Culture Models: Transforming GI Disease Modeling and Treatment. Cells 2024; 13:820. [PMID: 38786042 PMCID: PMC11119846 DOI: 10.3390/cells13100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/29/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
This review addresses the need for innovative co-culture systems integrating the enteric nervous system (ENS) with intestinal organoids. The breakthroughs achieved through these techniques will pave the way for a transformative era in gastrointestinal (GI) disease modeling and treatment strategies. This review serves as an introduction to the companion protocol paper featured in this journal. The protocol outlines the isolation and co-culture of myenteric and submucosal neurons with small intestinal organoids. This review provides an overview of the intestinal organoid culture field to establish a solid foundation for effective protocol application. Remarkably, the ENS surpasses the number of neurons in the spinal cord. Referred to as the "second brain", the ENS orchestrates pivotal roles in GI functions, including motility, blood flow, and secretion. The ENS is organized into myenteric and submucosal plexuses. These plexuses house diverse subtypes of neurons. Due to its proximity to the gut musculature and its cell type complexity, there are methodological intricacies in studying the ENS. Diverse approaches such as primary cell cultures, three-dimensional (3D) neurospheres, and induced ENS cells offer diverse insights into the multifaceted functionality of the ENS. The ENS exhibits dynamic interactions with the intestinal epithelium, the muscle layer, and the immune system, influencing epithelial physiology, motility, immune responses, and the microbiome. Neurotransmitters, including acetylcholine (ACh), serotonin (5-HT), and vasoactive intestinal peptide (VIP), play pivotal roles in these intricate interactions. Understanding these dynamics is imperative, as the ENS is implicated in various diseases, ranging from neuropathies to GI disorders and neurodegenerative diseases. The emergence of organoid technology presents an unprecedented opportunity to study ENS interactions within the complex milieu of the small and large intestines. This manuscript underscores the urgent need for standardized protocols and advanced techniques to unravel the complexities of the ENS and its dynamic relationship with the gut ecosystem. The insights gleaned from such endeavors hold the potential to revolutionize GI disease modeling and treatment paradigms.
Collapse
Affiliation(s)
- Cristina Llorente
- Department of Medicine, University of California San Diego, MC0063, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Sanders KM, Drumm BT, Cobine CA, Baker SA. Ca 2+ dynamics in interstitial cells: foundational mechanisms for the motor patterns in the gastrointestinal tract. Physiol Rev 2024; 104:329-398. [PMID: 37561138 PMCID: PMC11281822 DOI: 10.1152/physrev.00036.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/29/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal (GI) tract displays multiple motor patterns that move nutrients and wastes through the body. Smooth muscle cells (SMCs) provide the forces necessary for GI motility, but interstitial cells, electrically coupled to SMCs, tune SMC excitability, transduce inputs from enteric motor neurons, and generate pacemaker activity that underlies major motor patterns, such as peristalsis and segmentation. The interstitial cells regulating SMCs are interstitial cells of Cajal (ICC) and PDGF receptor (PDGFR)α+ cells. Together these cells form the SIP syncytium. ICC and PDGFRα+ cells express signature Ca2+-dependent conductances: ICC express Ca2+-activated Cl- channels, encoded by Ano1, that generate inward current, and PDGFRα+ cells express Ca2+-activated K+ channels, encoded by Kcnn3, that generate outward current. The open probabilities of interstitial cell conductances are controlled by Ca2+ release from the endoplasmic reticulum. The resulting Ca2+ transients occur spontaneously in a stochastic manner. Ca2+ transients in ICC induce spontaneous transient inward currents and spontaneous transient depolarizations (STDs). Neurotransmission increases or decreases Ca2+ transients, and the resulting depolarizing or hyperpolarizing responses conduct to other cells in the SIP syncytium. In pacemaker ICC, STDs activate voltage-dependent Ca2+ influx, which initiates a cluster of Ca2+ transients and sustains activation of ANO1 channels and depolarization during slow waves. Regulation of GI motility has traditionally been described as neurogenic and myogenic. Recent advances in understanding Ca2+ handling mechanisms in interstitial cells and how these mechanisms influence motor patterns of the GI tract suggest that the term "myogenic" should be replaced by the term "SIPgenic," as this review discusses.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| | - Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Caroline A Cobine
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Salah A Baker
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| |
Collapse
|
7
|
Mandarino FV, Testoni SGG, Barchi A, Azzolini F, Sinagra E, Pepe G, Chiti A, Danese S. Imaging in Gastroparesis: Exploring Innovative Diagnostic Approaches, Symptoms, and Treatment. Life (Basel) 2023; 13:1743. [PMID: 37629600 PMCID: PMC10455809 DOI: 10.3390/life13081743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/08/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Gastroparesis (GP) is a chronic disease characterized by upper gastrointestinal symptoms, primarily nausea and vomiting, and delayed gastric emptying (GE), in the absence of mechanical GI obstruction. The underlying pathophysiology of GP remains unclear, but factors contributing to the condition include vagal nerve dysfunction, impaired gastric fundic accommodation, antral hypomotility, gastric dysrhythmias, and pyloric dysfunction. Currently, gastric emptying scintigraphy (GES) is considered the gold standard for GP diagnosis. However, the overall delay in GE weakly correlates with GP symptoms and their severity. Recent research efforts have focused on developing treatments that address the presumed underlying pathophysiological mechanisms of GP, such as pyloric hypertonicity, with Gastric Peroral Endoscopic Myotomy (G-POEM) one of these procedures. New promising diagnostic tools for gastroparesis include wireless motility capsule (WMC), the 13 carbon-GE breath test, high-resolution electrogastrography, and the Endoluminal Functional Lumen Imaging Probe (EndoFLIP). Some of these tools assess alterations beyond GE, such as muscular electrical activity and pyloric tone. These modalities have the potential to characterize the pathophysiology of gastroparesis, identifying patients who may benefit from targeted therapies. The aim of this review is to provide an overview of the current knowledge on diagnostic pathways in GP, with a focus on the association between diagnosis, symptoms, and treatment.
Collapse
Affiliation(s)
- Francesco Vito Mandarino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (S.G.G.T.); (A.B.); (F.A.); (S.D.)
| | - Sabrina Gloria Giulia Testoni
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (S.G.G.T.); (A.B.); (F.A.); (S.D.)
| | - Alberto Barchi
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (S.G.G.T.); (A.B.); (F.A.); (S.D.)
| | - Francesco Azzolini
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (S.G.G.T.); (A.B.); (F.A.); (S.D.)
| | - Emanuele Sinagra
- Gastroenterology & Endoscopy Unit, Fondazione Istituto G. Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy;
| | - Gino Pepe
- Department of Nuclear Medicine, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (G.P.); (A.C.)
| | - Arturo Chiti
- Department of Nuclear Medicine, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (G.P.); (A.C.)
| | - Silvio Danese
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (S.G.G.T.); (A.B.); (F.A.); (S.D.)
| |
Collapse
|
8
|
Soliman H, Gourcerol G. Targeting the pylorus in gastroparesis: From physiology to endoscopic pyloromyotomy. Neurogastroenterol Motil 2023; 35:e14529. [PMID: 36594414 PMCID: PMC10077918 DOI: 10.1111/nmo.14529] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/28/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND The pylorus plays a key role in the control of gastric content outflow. Impairment of pyloric physiology has been observed in gastroparesis, particularly when associated with diabetes mellitus or opioid intake or after antireflux surgery. New tools have been developed to identify pyloric dysfunction in routine care, including functional luminal impedance planimetry (FLIP). As such, a new therapeutic strategy targeting the pylorus, namely endoscopic pyloromyotomy (G-POEM), has received increasing attention and emerged as a promising treatment for gastroparesis. PURPOSE The present review details the involvement of the pyloric pathophysiology in gastroparesis, as well as clinical results of G-POEM according to the current literature.
Collapse
Affiliation(s)
- Heithem Soliman
- Physiology Department, INSERM UMR 1073, CIC-CRB 1404, Centre Hospitalo-Universitaire de Rouen, Rouen, France.,Département d'Hépato-Gastro-Entérologie, Hôpital Louis Mourier, AP-HP Nord, Université de Paris Cité, Colombes, France
| | - Guillaume Gourcerol
- Physiology Department, INSERM UMR 1073, CIC-CRB 1404, Centre Hospitalo-Universitaire de Rouen, Rouen, France
| |
Collapse
|
9
|
"M1/M2" Muscularis Macrophages Are Associated with Reduction of Interstitial Cells of Cajal and Glial Cells in Achalasia. Dig Dis Sci 2022; 68:1260-1268. [PMID: 36346489 PMCID: PMC10102055 DOI: 10.1007/s10620-022-07734-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/10/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND AND AIMS Several studies showed muscularis macrophages (MMφ) are associated with GI motility disorders. The purpose of this study was to preliminary explore the association between MMφ and achalasia. METHODS Tissue samples of the lower esophageal sphincter (LES) high-pressure zone were obtained from 27 achalasia patients and 10 controls. Immunohistochemistry for MMφ, interstitial cells of Cajal (ICC), neuronal nitric oxide synthase (nNOS), and glial cells were conducted. Histological characteristics were compared between groups, and correlation analysis was performed. RESULTS Fewer ICC was found in achalasia compared with controls (P = 0.018), and the level of M1 macrophages was higher than that in controls no matter in terms of the number or the proportion of M1(P = 0.026 for M1 and 0.037 for M1/MMφ). Statistical differences were found between two groups in terms of proportion of M2 and ratio of M1 to M2 (P = 0.048 for M2/ MMφ and < 0.001 for M1/M2). For the correlation analysis, significant correlations were detected between levels of nNOS, ICC, and glial cells in patients with achalasia (P = 0.026 for nNOS and ICC, 0.001 for nNOS and glial cells, 0.019 for ICC and glial cells). There were significant correlations between M2/MMφ and levels of ICC (P = 0.019), glial cells (P = 0.004), and nNOS (P = 0.135). CONCLUSION Patients with achalasia had a higher level of M1/M2 ratio in LES and significant correlations were found between M2/MMφ and numbers of ICC and glial cells, which suggested that MMφ were probably associated with occurrence and development of achalasia.
Collapse
|
10
|
Filho FC, Silva JDP, Petri C, Almendra JSL, de Sousa ÍA, Cavalcanti SMG, Silva BA, Formiga Melo MF, Cavalcanti PMDS. Pharmacological evidence that GABA-induced relaxation of rat proximal duodenum longitudinal muscle depends on NKCC cotransporter activity and Ca 2+ influx. Can J Physiol Pharmacol 2022; 100:728-740. [PMID: 35880679 DOI: 10.1139/cjpp-2021-0639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
γ-Aminobutyric acid (GABA) is the main inhibitory neurotransmitter in adult central nervous system (CNS) synapses, but it excites immature CNS neurons as well as neurons in the myenteric plexus. The present work aimed to determine whether GABA-induced nonadrenergic, noncholinergic (NANC) neuronal-mediated relaxation of the rat duodenum is dependent on the activity of Na+ K+ Cl- cotransporters (NKCC) and requires calcium influx. In the presence of guanethidine (3 µmol/L), atropine (3 µmol/L), and indomethacin (1 µmol/L), relaxations induced by GABA (100 µmol/L), KCl (5-10 mmol/L) and electrical field stimulation (1-8 Hz, 2 ms, 60 V), but not those induced by bradykinin (10-100 nmol/L) were abolished by lidocaine (300 µmol/L). However, only GABA-induced relaxations were reduced in a concentration-dependent manner by the NKCC1/2 inhibitors bumetanide (0.1-1 µmol/L) and furosemide (1-10 µmol/L). GABA-induced NANC neuronal relaxation was abolished by bicuculline (30 µmol/L) and inhibited by N-nitroarginine methyl ester (l-NAME, 300 µmol/L). The ω-conotoxin GVIA (1 µmol/L), which acts exclusively on neuronal CaV2 channels, but not on smooth muscle voltage-gated Ca2+ CaV1 channels, and nonselective blockers of these channels (verapamil 100 nmol/L and ruthenium red 10 µmol/L), reduced GABA-induced relaxations. These results showed that the activation of GABAA receptors induces NANC nitrergic neuronal relaxations in the rat duodenum, which depend on NKCC activity and CaV2 channel activation, suggesting that this phenomenon results from neuronal depolarization promoted by Cl- efflux through GABAA receptors, with subsequent Ca2+ influx and nitric oxide release.
Collapse
Affiliation(s)
- Francisco Chagas Filho
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Janyerson Dannys Pereira Silva
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Caio Petri
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - João Santos Lima Almendra
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Ícaro Araújo de Sousa
- Biophysics and Physiology Department, 64049-550, Health Sciences Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | - Bagnólia A Silva
- Pharmacological Sciences Department, 58051-900, Health Sciences Center, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | - Margareth Fátima Formiga Melo
- Pharmacological Sciences Department, 58051-900, Health Sciences Center, Federal University of Paraíba, João Pessoa, Paraíba, Brazil
| | | |
Collapse
|
11
|
Drumm BT, Cobine CA, Baker SA. Insights on gastrointestinal motility through the use of optogenetic sensors and actuators. J Physiol 2022; 600:3031-3052. [PMID: 35596741 DOI: 10.1113/jp281930] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/13/2022] [Indexed: 11/08/2022] Open
Abstract
The muscularis of the gastrointestinal (GI) tract consists of smooth muscle cells (SMCs) and various populations of interstitial cells of Cajal (ICC), platelet-derived growth factor receptor α+ (PDGFRα+ ) cells, as well as excitatory and inhibitory enteric motor nerves. SMCs, ICC and PDGFRα+ cells form an electrically coupled syncytium, which together with inputs from the enteric nervous system (ENS) regulate GI motility. Early studies evaluating Ca2+ signalling behaviours in the GI tract relied upon indiscriminate loading of tissues with Ca2+ dyes. These methods lacked the means to study activity in specific cells of interest without encountering contamination from other cells within the preparation. Development of mice expressing optogenetic sensors (GCaMP, RCaMP) has allowed visualization of Ca2+ signalling behaviours in a cell specific manner. Additionally, availability of mice expressing optogenetic modulators (channelrhodopsins or halorhodospins) has allowed manipulation of specific signalling pathways using light. GCaMP expressing animals have been used to characterize Ca2+ signalling behaviours of distinct classes of ICC and SMCs throughout the GI musculature. These findings illustrate how Ca2+ signalling in ICC is fundamental in GI muscles, contributing to tone in sphincters, pacemaker activity in rhythmic muscles and relaying enteric signals to SMCs. Animals that express channelrhodopsin in specific neuronal populations have been used to map neural circuitry and to examine post junctional neural effects on GI motility. Thus, optogenetic approaches provide a novel means to examine the contribution of specific cell types to the regulation of motility patterns within complex multi-cellular systems. Abstract Figure Legends Optogenetic activators and sensors can be used to investigate the complex multi-cellular nature of the gastrointestinal (GI tract). Optogenetic activators that are activated by light such as channelrhodopsins (ChR2), OptoXR and halorhodopsinss (HR) proteins can be genetically encoded into specific cell types. This can be used to directly activate or silence specific GI cells such as various classes of enteric neurons, smooth muscle cells (SMC) or interstitial cells, such as interstitial cells of Cajal (ICC). Optogenetic sensors that are activated by different wavelengths of light such as green calmodulin fusion protein (GCaMP) and red CaMP (RCaMP) make high resolution of sub-cellular Ca2+ signalling possible within intact tissues of specific cell types. These tools can provide unparalleled insight into mechanisms underlying GI motility and innervation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bernard T Drumm
- Smooth Muscle Research Centre, Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland.,Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Caroline A Cobine
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Salah A Baker
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
12
|
Drumm BT, Hannigan KI, Lee JY, Rembetski BE, Baker SA, Koh SD, Cobine CA, Sanders KM. Ca 2+ signalling in interstitial cells of Cajal contributes to generation and maintenance of tone in mouse and monkey lower esophageal sphincters. J Physiol 2022; 600:2613-2636. [PMID: 35229888 DOI: 10.1113/jp282570] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 02/15/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The lower esophageal sphincter (LES) generates contractile tone preventing reflux of gastric contents into the esophagus. LES smooth muscle cells (SMCs) display depolarized membrane potentials facilitating activation of L-type Ca2+ channels. Interstitial cells of Cajal (ICC) express Ca2+ -activated Cl- channels encoded by Ano1 in mouse and monkey LES. Ca2+ signaling in ICC activates ANO1 currents in ICC. ICC displayed spontaneous Ca2+ transients in mice from multiple firing sites in each cell and no entrainment of Ca2+ firing between sites or between cells. Inhibition of ANO1 channels with a specific antagonist caused hyperpolarization of mouse LES and inhibition of tone in monkey and mouse LES muscles. Our data suggest a novel mechanism for LES tone in which Ca2+ transient activation of ANO1 channels in ICC generates depolarizing inward currents that conduct to SMCs to activate L-type Ca2+ currents, Ca2+ entry and contractile tone. ABSTRACT The lower esophageal sphincter (LES) generates tone and prevents reflux of gastric contents. LES smooth muscle cells (SMCs) are relatively depolarized, facilitating activation of Cav 1.2 channels to sustain contractile tone. We hypothesised that intramuscular interstitial cells of Cajal (ICC-IM), through activation of Ca2+ -activated-Cl- channels (ANO1), set membrane potentials of SMCs favorable for activation of Cav 1.2 channels. In some gastrointestinal muscles, ANO1 channels in ICC-IM are activated by Ca2+ transients, but no studies have examined Ca2+ dynamics in ICC-IM within the LES. Immunohistochemistry and qPCR were used to determine expression of key proteins and genes in ICC-IM and SMCs. These studies revealed that Ano1 and its gene product, ANO1 are expressed in c-Kit+ cells (ICC-IM) in mouse and monkey LES clasp muscles. Ca2+ signaling was imaged in situ, using mice expressing GCaMP6f specifically in ICC (Kit-KI-GCaMP6f). ICC-IM exhibited spontaneous Ca2+ transients from multiple firing sites. Ca2+ transients were abolished by CPA or caffeine but were unaffected by tetracaine or nifedipine. Maintenance of Ca2+ transients depended on Ca2+ influx and store reloading, as Ca2+ transient frequency was reduced in Ca2+ free solution or by Orai antagonist. Spontaneous tone of LES muscles from mouse and monkey was reduced ∼80% either by Ani9, an ANO1 antagonist or by the Cav 1.2 channel antagonist nifedipine. Membrane hyperpolarisation occurred in the presence of Ani9. These data suggest that intracellular Ca2+ activates ANO1 channels in ICC-IM in the LES. Coupling of ICC-IM to SMCs drives depolarization, activation of Cav 1.2 channels, Ca2+ entry and contractile tone. Abstract figure legend Proposed mechanism for generation of contractile tone in the lower esophageal sphincter (LES). Interstitial cells of Cajal (ICC) in the LES generate spontaneous, stochastic Ca2+ transients via Ca2+ release from the endoplasmic reticulum (ER). The Ca2+ transients activate ANO1 Cl- channels causing Cl- efflux (inward current). ANO1 currents have a depolarizing effect on ICC (+++s inside membrane) and this conducts through gap junctions (GJ) to smooth muscle cells (SMCs). Input from thousands of ICC results in depolarized membrane potentials (-40 to -50 mV) which is within the window current range for L-type Ca2+ channels. Activation of these channels causes Ca2+ influx, activation of contractile elements (CE) and development of tonic contraction. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA.,Smooth Muscle Research Centre, Dundalk Institute of Technology, Ireland
| | - Karen I Hannigan
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Ji Yeon Lee
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Benjamin E Rembetski
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Salah A Baker
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Sang Don Koh
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Caroline A Cobine
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Kenton M Sanders
- Department of Physiology & Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
13
|
Abstract
Gastroparesis is characterized by symptoms suggestive of, and objective evidence of, delayed gastric emptying in the absence of mechanical obstruction. This review addresses the normal emptying of solids and liquids from the stomach and details the myogenic and neuromuscular control mechanisms, including the specialized function of the pyloric sphincter, that result in normal emptying, based predominantly on animal research. A clear understanding of fundamental mechanisms is necessary to comprehend derangements leading to gastroparesis, and additional research on human gastric muscles is needed. The section on pathophysiology of gastroparesis considers neuromuscular diseases that affect nonsphincteric gastric muscle, disorders of the extrinsic neural control, and pyloric dysfunction that lead to gastroparesis. The potential cellular basis for gastroparesis is attributed to the effects of oxidative stress and inflammation, with increased pro-inflammatory and decreased resident macrophages, as observed in full-thickness biopsies from patients with gastroparesis. Predominant diagnostic tests involving measurements of gastric emptying, the use of a functional luminal imaging probe, and high-resolution antral duodenal manometry in characterizing the abnormal motor functions at the gastroduodenal junction are discussed. Management is based on supporting nutrition; dietary interventions, including the physical reduction in particle size of solid foods; pharmacological agents, including prokinetics and anti-emetics; and interventions such as gastric electrical stimulation and pyloromyotomy. These are discussed briefly, and comment is added on the potential for individualized treatments in the future, based on optimal gastric emptying measurement and objective documentation of the underlying pathophysiology causing the gastroparesis.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV
| |
Collapse
|
14
|
Otsuka Y, Bai X, Tanaka Y, Ihara E, Chinen T, Ogino H, Ogawa Y. Involvement of interstitial cells of Cajal in nicotinic acetylcholine receptor-induced relaxation of the porcine lower esophageal sphincter. Eur J Pharmacol 2021; 910:174491. [PMID: 34506779 DOI: 10.1016/j.ejphar.2021.174491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 08/27/2021] [Accepted: 09/03/2021] [Indexed: 11/15/2022]
Abstract
The interstitial cells of Cajal (ICCs) play an important role in coordinated gastrointestinal motility. The present study aimed to elucidate whether or how ICCs are involved in the lower esophageal sphincter (LES) relaxation induced by stimulation of the nicotinic acetylcholine receptor. The application of 1,1-dimethyl-4-phenyl-piperazinium (DMPP; a nicotinic acetylcholine receptor agonist) induced a transient relaxation in the circular smooth muscle of the porcine LES. DMPP-induced relaxation was abolished by not only 1 μM tetrodotoxin but also the inhibition of ICC activity by pretreatment with 100 μM carbenoxolone (a gap junction inhibitor), pretreatment with 100 μM CaCCinh-A01 (an anoctamin-1 blocker acting as a calcium-activated chloride channel inhibitor), and pretreatment with Cl--free solution. However, pretreatment with 100 μM Nω-nitro-L-arginine methyl ester had little effect on DMPP-induced relaxation. Furthermore, DMPP-induced relaxation was inhibited by pretreatment with 1 mM suramin, a purinergic P2 receptor antagonist, but not by 1 μM VIP (6-28), a vasoactive intestinal peptide (VIP) receptor antagonist. Stimulation of the purinergic P2 receptor with adenosine triphosphate (ATP) induced relaxation, which was abolished by the inhibition of ICC activity by pretreatment with CaCCinh-A01. In conclusion, membrane hyperpolarization of the ICCs via the activation of anoctamin-1 plays a central role in DMPP-induced relaxation. ATP may be a neurotransmitter for inhibitory enteric neurons, which stimulate the ICCs. The ICCs act as the interface of neurotransmission of nicotinic acetylcholine receptor in order to induce LES relaxation.
Collapse
Affiliation(s)
- Yoshihiro Otsuka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Xiaopeng Bai
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshimasa Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eikichi Ihara
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan; Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takatoshi Chinen
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Haruei Ogino
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
15
|
Radenkovic G, Petrovic V, Zivanovic D, Stoiljkovic N, Sokolovic D, Zivkovic N, Radenkovic D, Velickov A, Jovanovic J. Interstitial Cells of Cajal and Neural Structures in the Human Fetal Appendix. J Neurogastroenterol Motil 2021; 27:127-133. [PMID: 33380557 PMCID: PMC7786081 DOI: 10.5056/jnm20100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 11/20/2022] Open
Abstract
Background/Aims The interstitial cells of Cajal (ICC) are located within and around the digestive tract's muscle layers. They function as intestinal muscle pacemakers and aid in the modification of enteric neurotransmission. The appendix's unique position requires an appropriate contraction pattern of its muscular wall to adequately evacuate its contents. We investigated the development and distribution of nervous structures and ICC in the human fetal appendix. Methods Specimens were exposed to anti-c-kit (CD117) antibodies to investigate ICC differentiation. Enteric plexuses were examined using anti-neuron-specific enolase, and the differentiation of smooth muscle cells was studied with anti-desmin antibodies. Results During weeks 13-14, numerous myenteric plexus ganglia form an almost uninterrupted sequence throughout the body and apex of the appendix. Fewer ganglia were present at the submucosal border of the circular muscle layer and within this layer. A large number of ganglia appear within the circular and longitudinal muscle layers in a later fetal period. The first ICC subtypes noted were of the myenteric plexus and the submucous plexus. In the later fetal period, the number of intramuscular ICC markedly rises, and this subtype becomes predominant. Conclusions The ICC and nervous structure distribution in the human fetal appendix are significantly different from all other parts of the small and large intestine. The organization of ICC and the enteric nervous system provides the basis for the specific contraction pattern of the muscular wall of the appendix.
Collapse
Affiliation(s)
- Goran Radenkovic
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, Serbia
| | - Vladimir Petrovic
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, Serbia
| | | | - Nenad Stoiljkovic
- Department of Physiology, Faculty of Medicine, University of Nis, Serbia
| | - Dusan Sokolovic
- Department of Biochemistry, Faculty of Medicine, University of Nis, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Faculty of Medicine, University of Nis, Serbia
| | - Dina Radenkovic
- Guy's and St Thomas' Hospital and King's College London, London, UK
| | - Aleksandra Velickov
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, Serbia
| | | |
Collapse
|
16
|
Camilleri M, Sanders KM. Opiates, the Pylorus, and Gastroparesis. Gastroenterology 2020; 159:414-421. [PMID: 32389662 DOI: 10.1053/j.gastro.2020.04.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada
| |
Collapse
|
17
|
Drumm BT, Rembetski BE, Huynh K, Nizar A, Baker SA, Sanders KM. Excitatory cholinergic responses in mouse colon intramuscular interstitial cells of Cajal are due to enhanced Ca 2+ release via M 3 receptor activation. FASEB J 2020; 34:10073-10095. [PMID: 32539213 DOI: 10.1096/fj.202000672r] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022]
Abstract
Colonic intramuscular interstitial cells of Cajal (ICC-IM) are associated with cholinergic varicosities, suggesting a role in mediating excitatory neurotransmission. Ca2+ release in ICC-IM activates Ano1, a Ca2+ -activated Cl- conductance, causing tissue depolarization and increased smooth muscle excitability. We employed Ca2+ imaging of colonic ICC-IM in situ, using mice expressing GCaMP6f in ICC to evaluate ICC-IM responses to excitatory neurotransmission. Expression of muscarinic type 2, 3 (M2 , M3 ), and NK1 receptors were enriched in ICC-IM. NK1 receptor agonists had minimal effects on ICC-IM, whereas neostigmine and carbachol increased Ca2+ transients. These effects were reversed by DAU 5884 (M3 receptor antagonist) but not AF-DX 116 (M2 receptor antagonist). Electrical field stimulation (EFS) in the presence of L-NNA and MRS 2500 enhanced ICC-IM Ca2+ transients. Responses were blocked by atropine or DAU 5884, but not AF-DX 116. ICC-IM responses to EFS were ablated by inhibiting Ca2+ stores with cyclopiazonic acid and reduced by inhibiting Ca2+ influx via Orai channels. Contractions induced by EFS were reduced by an Ano1 channel antagonist, abolished by DAU 5884, and unaffected by AF-DX 116. Colonic ICC-IM receive excitatory inputs from cholinergic neurons via M3 receptor activation. Enhancing ICC-IM Ca2+ release and Ano1 activation contributes to excitatory responses of colonic muscles.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA.,Department of Life & Health Science, Dundalk Institute of Technology, Dundalk, Ireland
| | - Benjamin E Rembetski
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Kaitlin Huynh
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Aqeel Nizar
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Salah A Baker
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, USA
| |
Collapse
|
18
|
Patel YA, Pasricha PJ. Enteric Neuromodulation for the Gut and Beyond. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a034355. [PMID: 30858329 DOI: 10.1101/cshperspect.a034355] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The small intestine is the longest organ in the human body, spanning a length of ∼5 m and compartmentalized into three distinct regions with specific roles in maintenance of comprehensive homeostasis. Along its length exists as a unique and independent system-called the enteric nervous system (ENS)-which coordinates the multitude of functions continuously around the clock. Yet, with so many vital roles played, the functions, relationships, and roles of the small intestine and ENS remain largely elusive. This fundamental hole in the physiology of the small intestine and ENS introduces a substantial number of challenges when attempting to create bioelectronic approaches for treatment of various disorders originating in the small intestine. Here, we review existing therapeutic options for modulating the small intestine, discuss fundamental gaps that must be addressed, and highlight novel methods and approaches to consider for development of bioelectronic approaches aiming to modulate the small intestine.
Collapse
Affiliation(s)
- Yogi A Patel
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland 21205
| | - Pankaj J Pasricha
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland 21205.,Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
19
|
Gastrointestinal dysfunction in autism displayed by altered motility and achalasia in Foxp1 +/- mice. Proc Natl Acad Sci U S A 2019; 116:22237-22245. [PMID: 31611379 DOI: 10.1073/pnas.1911429116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal dysfunctions in individuals with autism spectrum disorder are poorly understood, although they are common among this group of patients. FOXP1 haploinsufficiency is characterized by autistic behavior, language impairment, and intellectual disability, but feeding difficulties and gastrointestinal problems have also been reported. Whether these are primary impairments, the result of altered eating behavior, or side effects of psychotropic medication remains unclear. To address this question, we investigated Foxp1 +/- mice reflecting FOXP1 haploinsufficiency. These animals show decreased body weight and altered feeding behavior with reduced food and water intake. A pronounced muscular atrophy was detected in the esophagus and colon, caused by reduced muscle cell proliferation. Nitric oxide-induced relaxation of the lower esophageal sphincter was impaired and achalasia was confirmed in vivo by manometry. Foxp1 targets (Nexn, Rbms3, and Wls) identified in the brain were dysregulated in the adult Foxp1 +/- esophagus. Total gastrointestinal transit was significantly prolonged due to impaired colonic contractility. Our results have uncovered a previously unknown dysfunction (achalasia and impaired gut motility) that explains the gastrointestinal disturbances in patients with FOXP1 syndrome, with potential wider relevance for autism.
Collapse
|
20
|
Drumm BT, Hwang SJ, Baker SA, Ward SM, Sanders KM. Ca 2+ signalling behaviours of intramuscular interstitial cells of Cajal in the murine colon. J Physiol 2019; 597:3587-3617. [PMID: 31124144 DOI: 10.1113/jp278036] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Colonic intramuscular interstitial cells of Cajal (ICC-IM) exhibit spontaneous Ca2+ transients manifesting as stochastic events from multiple firing sites with propagating Ca2+ waves occasionally observed. Firing of Ca2+ transients in ICC-IM is not coordinated with adjacent ICC-IM in a field of view or even with events from other firing sites within a single cell. Ca2+ transients, through activation of Ano1 channels and generation of inward current, cause net depolarization of colonic muscles. Ca2+ transients in ICC-IM rely on Ca2+ release from the endoplasmic reticulum via IP3 receptors, spatial amplification from RyRs and ongoing refilling of ER via the sarcoplasmic/endoplasmic-reticulum-Ca2+ -ATPase. ICC-IM are sustained by voltage-independent Ca2+ influx via store-operated Ca2+ entry. Some of the properties of Ca2+ in ICC-IM in the colon are similar to the behaviour of ICC located in the deep muscular plexus region of the small intestine, suggesting there are functional similarities between these classes of ICC. ABSTRACT A component of the SIP syncytium that regulates smooth muscle excitability in the colon is the intramuscular class of interstitial cells of Cajal (ICC-IM). All classes of ICC (including ICC-IM) express Ca2+ -activated Cl- channels, encoded by Ano1, and rely upon this conductance for physiological functions. Thus, Ca2+ handling in ICC is fundamental to colonic motility. We examined Ca2+ handling mechanisms in ICC-IM of murine proximal colon expressing GCaMP6f in ICC. Several Ca2+ firing sites were detected in each cell. While individual sites displayed rhythmic Ca2+ events, the overall pattern of Ca2+ transients was stochastic. No correlation was found between discrete Ca2+ firing sites in the same cell or in adjacent cells. Ca2+ transients in some cells initiated Ca2+ waves that spread along the cell at ∼100 µm s-1 . Ca2+ transients were caused by release from intracellular stores, but depended strongly on store-operated Ca2+ entry mechanisms. ICC Ca2+ transient firing regulated the resting membrane potential of colonic tissues as a specific Ano1 antagonist hyperpolarized colonic muscles by ∼10 mV. Ca2+ transient firing was independent of membrane potential and not affected by blockade of L- or T-type Ca2+ channels. Mechanisms regulating Ca2+ transients in the proximal colon displayed both similarities to and differences from the intramuscular type of ICC in the small intestine. Similarities and differences in Ca2+ release patterns might determine how ICC respond to neurotransmission in these two regions of the gastrointestinal tract.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sung J Hwang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
21
|
Generation of Spontaneous Tone by Gastrointestinal Sphincters. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019. [PMID: 31183822 DOI: 10.1007/978-981-13-5895-1_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
An important feature of the gastrointestinal (GI) muscularis externa is its ability to generate phasic contractile activity. However, in some GI regions, a more sustained contraction, referred to as "tone," also occurs. Sphincters are muscles oriented in an annular manner that raise intraluminal pressure, thereby reducing or blocking the movement of luminal contents from one compartment to another. Spontaneous tone generation is often a feature of these muscles. Four distinct smooth muscle sphincters are present in the GI tract: the lower esophageal sphincter (LES), the pyloric sphincter (PS), the ileocecal sphincter (ICS), and the internal anal sphincter (IAS). This chapter examines how tone generation contributes to the functional behavior of these sphincters. Historically, tone was attributed to contractile activity arising directly from the properties of the smooth muscle cells. However, there is increasing evidence that interstitial cells of Cajal (ICC) play a significant role in tone generation in GI muscles. Indeed, ICC are present in each of the sphincters listed above. In this chapter, we explore various mechanisms that may contribute to tone generation in sphincters including: (1) summation of asynchronous phasic activity, (2) partial tetanus, (3) window current, and (4) myofilament sensitization. Importantly, the first two mechanisms involve tone generation through summation of phasic events. Thus, the historical distinction between "phasic" versus "tonic" smooth muscles in the GI tract requires revision. As described in this chapter, it is clear that the unique functional role of each sphincter in the GI tract is accompanied by a unique combination of contractile mechanisms.
Collapse
|
22
|
Lin Q, Qin M, Zhao SG, Liu ZX, Dou WJ, Zhang R, Li YL, Xi XH, Xu JQ, Ma LT, Wang JJ. The roles of PDGFRα signaling in the postnatal development and functional maintenance of the SMC-ICC-PDGFRα+ cell (SIP) syncytium in the colon. Neurogastroenterol Motil 2019; 31:e13568. [PMID: 30848008 DOI: 10.1111/nmo.13568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND The SIP syncytium in the gut consists of smooth muscle cells, interstitial cells of Cajal, and PDGFRα+ cells. We studied the fate of SIP cells after blocking PDGFRα receptor to explore the roles of PDGFRα signaling in the postnatal development and functional maintenance of the SIP syncytium. METHODS Crenolanib was administered to mice from P0, P10, or P50. The morphological changes in SIP cells were examined by immunofluorescence. Protein expression in SIP cells was detected by Western blotting. Moreover, colonic transit was analyzed by testing the colonic bead expulsion time. KEY RESULTS A dose of 5 mg(kg•day)-1 crenolanib administered for 10 days beginning on P0 apparently hindered the development of PDGFRα+ cells in the colonic longitudinal muscularis and myenteric plexus without influencing their proliferative activity and apoptosis, but this result was not seen in the colonic circular muscularis. SMCs were also inhibited by crenolanib. A dose of 7.5 mg(kg•day)-1 crenolanib administered for 15 days beginning on P0 caused reductions in both PDGFRα+ cells and ICC in the longitudinal muscularis, myenteric plexus, and circular muscularis. However, when crenolanib was administered at a dose of 5 mg(kg•day)-1 beginning on P10 or P50, it only noticeably decreased the number of PDGFRα+ cells in the colonic longitudinal muscularis. Crenolanib also caused PDGFRα+ cells to transdifferentiate into SMC in adult mice. Colonic transit was delayed after administration of crenolanib. CONCLUSIONS & INFERENCES Therefore, PDGFRα signaling is essential for the development and functional maintenance of the SIP cells, especially PDGFRα+ cells.
Collapse
Affiliation(s)
- Qiang Lin
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ming Qin
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Shu-Guang Zhao
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen-Xiong Liu
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei-Jia Dou
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Rong Zhang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu-Long Li
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Xiao-Hou Xi
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jia-Qiao Xu
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Li-Tian Ma
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing-Jie Wang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
23
|
Sergeant GP, Hollywood MA, Thornbury KD. Spontaneous Activity in Urethral Smooth Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:149-167. [DOI: 10.1007/978-981-13-5895-1_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
24
|
Sanders KM, Ward SM. Nitric oxide and its role as a non-adrenergic, non-cholinergic inhibitory neurotransmitter in the gastrointestinal tract. Br J Pharmacol 2019; 176:212-227. [PMID: 30063800 PMCID: PMC6295421 DOI: 10.1111/bph.14459] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/06/2018] [Accepted: 07/12/2018] [Indexed: 12/19/2022] Open
Abstract
NO is a neurotransmitter released from enteric inhibitory neurons and responsible for modulating gastrointestinal (GI) motor behaviour. Enteric neurons express nNOS (NOS1) that associates with membranes of nerve varicosities. NO released from neurons binds to soluble guanylate cyclase in post-junctional cells to generate cGMP. cGMP-dependent protein kinase type 1 (PKG1) is a major mediator but perhaps not the only pathway involved in cGMP-mediated effects in GI muscles based on gene deletion studies. NOS1+ neurons form close contacts with smooth muscle cells (SMCs), interstitial cells of Cajal (ICC) and PDGFRα+ cells, and these cells are electrically coupled (SIP syncytium). Cell-specific gene deletion studies have shown that nitrergic responses are due to mechanisms in SMCs and ICC. Controversy exists about the ion channels and other post-junctional mechanisms that mediate nitrergic responses in GI muscles. Reduced nNOS expression in enteric inhibitory motor neurons and/or reduced connectivity between nNOS+ neurons and the SIP syncytium appear to be responsible for motor defects that develop in diabetes. An overproduction of NO in some inflammatory conditions also impairs normal GI motor activity. This review summarizes recent findings regarding the role of NO as an enteric inhibitory neurotransmitter. LINKED ARTICLES: This article is part of a themed section on Nitric Oxide 20 Years from the 1998 Nobel Prize. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.2/issuetoc.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell BiologyUniversity of Nevada, Reno, School of MedicineRenoNVUSA
| | - Sean M Ward
- Department of Physiology and Cell BiologyUniversity of Nevada, Reno, School of MedicineRenoNVUSA
| |
Collapse
|
25
|
Beck K, Friebe A, Voussen B. Nitrergic signaling via interstitial cells of Cajal and smooth muscle cells influences circular smooth muscle contractility in murine colon. Neurogastroenterol Motil 2018; 30:e13300. [PMID: 29377328 DOI: 10.1111/nmo.13300] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/03/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND Regulation of gastrointestinal motility involves excitatory and inhibitory neurotransmission. Nitric oxide (NO), the major inhibitory neurotransmitter, acts via its receptor NO-sensitive guanylyl cyclase (NO-GC). In the GI tract, NO-GC is expressed in several cell types such as smooth muscle cells (SMC) and interstitial cells of Cajal (ICC). Using cell-specific knockout mice, we have previously shown that NO-GC modulates spontaneous contractions in colonic longitudinal smooth muscle. However, its detailed role in the colonic circular smooth muscle is still unclear. METHODS Myography was performed to evaluate spontaneous contractions in rings of proximal colon (2.5 mm) from global (GCKO) and cell-specific knockout mice for NO-GC. Immunohistochemistry and in situ hybridization were used to specify NO-GC expression. KEY RESULTS Colonic circular smooth muscle showed three different contraction patterns: high-frequency ripples, slow phasic contractions, and large contractions. Ripples formed independently of NO-GC. Slow phasic contractions occurred intermittently in WT, SMC-GCKO, and ICC-GCKO tissue, whereas they were more prominent and prolonged in GCKO and SMC/ICC-GCKO tissue. Tetrodotoxin and the NO-GC inhibitor ODQ transformed slow phasic contractions of WT and single cell-specific knockout into GCKO-like contractions. ODQ increased the frequency of large contractions in WT and ICC-GCKO colon but not in GCKO, SMC-GCKO, and SMC/ICC-GCKO preparations. Tetrodotoxin and hexamethonium abolished large contractions. CONCLUSIONS AND INFERENCES We conclude that short rings of murine colon can be effectively used to record spontaneous contractions. Although NO-GC in SMC determines smooth muscle tone, concerted action of NO-GC in both SMC and ICC modulates slow phasic contractions and large contractions.
Collapse
Affiliation(s)
- K Beck
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - A Friebe
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - B Voussen
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
26
|
Veličkov A, Radenković G, Petrović V, Veličkov A. DIABETIC ALTERATIONS OF INTERSTITIAL CELLS OF CAJAL. ACTA MEDICA MEDIANAE 2017. [DOI: 10.5633/amm.2017.0416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
27
|
Radenkovic G, Radenkovic D, Velickov A. Development of interstitial cells of Cajal in the human digestive tract as the result of reciprocal induction of mesenchymal and neural crest cells. J Cell Mol Med 2017; 22:778-785. [PMID: 29193736 PMCID: PMC5783873 DOI: 10.1111/jcmm.13375] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/08/2017] [Indexed: 01/02/2023] Open
Abstract
Neural crest cells (NCC) can migrate into different parts of the body and express their strong inductive potential. In addition, they are multipotent and are able to differentiate into various cell types with diverse functions. In the primitive gut, NCC induce differentiation of muscular structures and interstitial cells of Cajal (ICC), and they themselves differentiate into the elements of the enteric nervous system (ENS), neurons and glial cells. ICC develop by way of mesenchymal cell differentiation in the outer parts of the primitive gut wall around the myenteric plexus (MP) ganglia, with the exception of colon, where they appear simultaneously also at the submucosal border of the circular muscular layer around the submucosal plexus (SMP) ganglia. However, in a complex process of reciprocal induction of NCC and local mesenchyma, c‐kit positive precursors are the first to differentiate, representing probably the common precursors of ICC and smooth muscle cells (SMC). C‐kit positive precursors could represent a key impact factor regarding the final differentiation of NCC into neurons and glial cells with neurons subsequently excreting stem cell factor (SCF) and other signalling molecules. Under the impact of SCF, a portion of c‐kit positive precursors lying immediately around the ganglia differentiate into ICC, while the rest differentiate into SMC.
Collapse
Affiliation(s)
- Goran Radenkovic
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Dina Radenkovic
- UCL Medical School, University College London (UCL), London, UK
| | - Aleksandra Velickov
- Department of Histology and Embryology, Faculty of Medicine, University of Nis, Nis, Serbia
| |
Collapse
|
28
|
Cobine CA, Hannah EE, Zhu MH, Lyle HE, Rock JR, Sanders KM, Ward SM, Keef KD. ANO1 in intramuscular interstitial cells of Cajal plays a key role in the generation of slow waves and tone in the internal anal sphincter. J Physiol 2017; 595:2021-2041. [PMID: 28054347 DOI: 10.1113/jp273618] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS The internal anal sphincter develops tone important for maintaining high anal pressure and continence. Controversy exists regarding the mechanisms underlying tone development. We examined the hypothesis that tone depends upon electrical slow waves (SWs) initiated in intramuscular interstitial cells of Cajal (ICC-IM) by activation of Ca2+ -activated Cl- channels (ANO1, encoded by Ano1) and voltage-dependent L-type Ca2+ channels (CavL , encoded by Cacna1c). Measurement of membrane potential and contraction indicated that ANO1 and CavL have a central role in SW generation, phasic contractions and tone, independent of stretch. ANO1 expression was examined in wildtype and Ano1/+egfp mice with immunohistochemical techniques. Ano1 and Cacna1c expression levels were examined by quantitative PCR in fluorescence-activated cell sorting. ICC-IM were the predominant cell type expressing ANO1 and the most likely candidate for SW generation. SWs in ICC-IM are proposed to conduct to smooth muscle where Ca2+ entry via CavL results in phasic activity that sums to produce tone. ABSTRACT The mechanism underlying tone generation in the internal anal sphincter (IAS) is controversial. We examined the hypothesis that tone depends upon generation of electrical slow waves (SWs) initiated in intramuscular interstitial cells of Cajal (ICC-IM) by activation of Ca2+ -activated Cl- channels (encoded by Ano1) and voltage-dependent L-type Ca2+ channels (encoded by Cacna1c). Phasic contractions and tone in the IAS were nearly abolished by ANO1 and CavL antagonists. ANO1 antagonists also abolished SWs as well as transient depolarizations that persisted after addition of CavL antagonists. Tone development in the IAS did not require stretch of muscles, and the sensitivity of contraction to ANO1 antagonists was the same in stretched versus un-stretched muscles. ANO1 expression was examined in wildtype and Ano1/+egfp mice with immunohistochemical techniques. Dual labelling revealed that ANO1 expression could be resolved in ICC but not smooth muscle cells (SMCs) in the IAS and rectum. Ano1, Cacna1c and Kit gene expression were the same in extracts of IAS and rectum muscles. In IAS cells isolated with fluorescence-activated cell sorting, Ano1 expression was 26.5-fold greater in ICC than in SMCs while Cacna1c expression was only 2-fold greater in SMCs than in ICC. These data support a central role for ANO1 and CavL in the generation of SWs and tone in the IAS. ICC-IM are the probable cellular candidate for ANO1 currents and SW generation. We propose that ANO1 and CavL collaborate to generate SWs in ICC-IM followed by conduction to adjacent SMCs where phasic calcium entry through CavL sums to produce tone.
Collapse
Affiliation(s)
- C A Cobine
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - E E Hannah
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - M H Zhu
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - H E Lyle
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - J R Rock
- Department of Anatomy, UCSF School of Medicine, San Francisco, CA, 94143, USA
| | - K M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - S M Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - K D Keef
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
29
|
Gallego D, Mañé N, Gil V, Martínez-Cutillas M, Jiménez M. Mechanisms responsible for neuromuscular relaxation in the gastrointestinal tract. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2016; 108:721-731. [DOI: 10.17235/reed.2016.4058/2015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
Lies B, Beck K, Keppler J, Saur D, Groneberg D, Friebe A. Nitrergic signalling via interstitial cells of Cajal regulates motor activity in murine colon. J Physiol 2015; 593:4589-601. [PMID: 26227063 DOI: 10.1113/jp270511] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/16/2015] [Indexed: 12/11/2022] Open
Abstract
In the enteric nervous systems, NO is released from nitrergic neurons as a major inhibitory neurotransmitter. NO acts via NO-sensitive guanylyl cyclase (NO-GC), which is found in different gastrointestinal (GI) cell types including smooth muscle cells (SMCs) and interstitial cells of Cajal (ICC). The precise mechanism of nitrergic signalling through these two cell types to regulate colonic spontaneous contractions is not fully understood yet. In the present study we investigated the impact of endogenous and exogenous NO on colonic contractile motor activity using mice lacking nitric oxide-sensitive guanylyl cyclase (NO-GC) globally and specifically in SMCs and ICC. Longitudinal smooth muscle of proximal colon from wild-type (WT) and knockout (KO) mouse strains exhibited spontaneous contractile activity ex vivo. WT and smooth muscle-specific guanylyl cyclase knockout (SMC-GCKO) colon showed an arrhythmic contractile activity with varying amplitudes and frequencies. In contrast, colon from global and ICC-specific guanylyl cyclase knockout (ICC-GCKO) animals showed a regular contractile rhythm with constant duration and amplitude of the rhythmic contractions. Nerve blockade (tetrodotoxin) or specific blockade of NO signalling (L-NAME, ODQ) did not significantly affect contractions of GCKO and ICC-GCKO colon whereas the arrhythmic contractile patterns of WT and SMC-GCKO colon were transformed into uniform motor patterns. In contrast, the response to electric field-stimulated neuronal NO release was similar in SMC-GCKO and global GCKO. In conclusion, our results indicate that basal enteric NO release acts via myenteric ICC to influence the generation of spontaneous contractions whereas the effects of elevated endogenous NO are mediated by SMCs in the murine proximal colon.
Collapse
Affiliation(s)
- Barbara Lies
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Katharina Beck
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Jonas Keppler
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Dieter Saur
- II. Medizinische Klinik und Poliklinik, Technische Universität München, München, Germany
| | - Dieter Groneberg
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Andreas Friebe
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
31
|
Park JH, Kwon JG, Kim SJ, Song DK, Lee SG, Kim ES, Kim ES, Cho KB, Jang BI, Kim DH, Sin JI, Kim TW, Song IH, Park KS. Alterations of colonic contractility in an interleukin-10 knockout mouse model of inflammatory bowel disease. J Neurogastroenterol Motil 2015; 21:51-61. [PMID: 25537671 PMCID: PMC4288096 DOI: 10.5056/jnm14008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 09/25/2014] [Accepted: 09/27/2014] [Indexed: 12/18/2022] Open
Abstract
Background/Aims Inflammatory bowel disease is commonly accompanied by colonic dysmotility and causes changes in intestinal smooth muscle contractility. In this study, colonic smooth muscle contractility in a chronic inflammatory condition was investigated using smooth muscle tissues prepared from interleukin-10 knockout (IL-10−/−) mice. Methods Prepared smooth muscle sections were placed in an organ bath system. Cholinergic and nitrergic neuronal responses were observed using carbachol and electrical field stimulation with L-NG-nitroarginine methyl ester (L-NAME). The expression of interstitial cells of Cajal (ICC) networks, muscarinic receptors, neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS) was observed via immunofluorescent staining. Results The spontaneous contractility and expression of ICC networks in the proximal and distal colon was significantly decreased in IL-10−/− mice compared to IL-10+/+ mice. The contractility in response to carbachol was significantly decreased in the proximal colon of IL-10−/− mice compared to IL-10+/+ mice, but no significant difference was found in the distal colon. In addition, the expression of muscarinic receptor type 2 was reduced in the proximal colon of IL-10−/− mice. The nictric oxide-mediated relaxation after electrical field stimulation was significantly decreased in the proximal and distal colon of IL-10−/− mice. In inflamed colon, the expression of nNOS decreased, whereas the expression of iNOS increased. Conclusions These results suggest that damage to the ICC network and NOS system in the proximal and distal colon, as well as damage to the smooth muscle cholinergic receptor in the proximal colon may play an important role in the dysmotility of the inflamed colon.
Collapse
Affiliation(s)
- Jae Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Joong Goo Kwon
- Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - Sun Joo Kim
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Dae Kyu Song
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Seok Guen Lee
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | | | - Eun Su Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Kwang Bum Cho
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Byung Ik Jang
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Dae Hwan Kim
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jeong-Im Sin
- Department of Microbiology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Tae Wan Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Kyungpook National University, Daegu, Korea
| | - In Hwan Song
- Department of Anatomy, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyung Sik Park
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
32
|
van der Pol RJ, Benninga MA, Magré J, Van Maldergem L, Rotteveel J, van der Knaap MS, de Meij TG. Berardinelli-Seip syndrome and achalasia: a shared pathomechanism? Eur J Pediatr 2015; 174:975-80. [PMID: 25994244 PMCID: PMC4475245 DOI: 10.1007/s00431-015-2556-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 04/23/2015] [Accepted: 04/26/2015] [Indexed: 10/27/2022]
Abstract
UNLABELLED Berardinelli-Seip congenital lipodystrophy (BSCL) is an uncommon autosomal recessive disorder. Patients with BSCL present with a distinct phenotype since subcutaneous fat is largely lacking and musculature has become more prominent. During childhood, diabetes and acanthosis nigricans evolve and female patients may develop hirsutism. Different genes encoding this entity have been described. Achalasia is a rare esophageal motility disorder, characterized by its distinct motility pattern with absent or incomplete lower esophageal sphincter (LES) relaxations. The exact cause of achalasia is yet unknown. Here, we describe a patient with achalasia in the context of BSCL, which might be linked by a shared pathophysiologic background, as evaluated in this case report. CONCLUSION In a BSCL patient presenting with gastrointestinal symptoms, a motility disorder of the gastrointestinal tract should be considered. WHAT IS KNOWN • Berardinelli-Seip congenital lipodystrophy (BSCL) and achalasia are both disorders characterized by low prevalence. What is New: • Co-existence of both diseases is described in this report. Linkage by a potential common pathophysiologic background is discussed in this paper.
Collapse
Affiliation(s)
- Rachel J. van der Pol
- grid.5650.60000000404654431Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital AMC, Amsterdam, The Netherlands
| | - Marc A. Benninga
- grid.5650.60000000404654431Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital AMC, Amsterdam, The Netherlands
| | - Jocelyne Magré
- grid.5805.80000000119553500INSERM UMR_S938, Université Pierre et Marie Curie, Paris, France ,grid.462318.aINSERM UMR_S1087, L’Institut du Thorax, Nantes, France
| | - Lionel Van Maldergem
- grid.7459.f0000000121883779Centre de Génétique Humaine, Université de Franche-Comté, Besançon, France
| | - Joost Rotteveel
- grid.16872.3a000000040435165XDepartment of Pediatric Endocrinology, VU University Medical Center, Amsterdam, The Netherlands
| | - Marjo S. van der Knaap
- grid.16872.3a000000040435165XDepartment of Pediatric Neurology, VU University Medical Center, Amsterdam, The Netherlands
| | - Tim G. de Meij
- grid.16872.3a000000040435165XDepartment of Pediatric Gastroenterology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
33
|
Groneberg D, Zizer E, Lies B, Seidler B, Saur D, Wagner M, Friebe A. Dominant role of interstitial cells of Cajal in nitrergic relaxation of murine lower oesophageal sphincter. J Physiol 2014; 593:403-14. [PMID: 25630261 DOI: 10.1113/jphysiol.2014.273540] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 10/24/2014] [Indexed: 12/20/2022] Open
Abstract
Oesophageal achalasia is a disease known to result from reduced relaxation of the lower oesophageal sphincter (LES). Nitric oxide (NO) is one of the main inhibitory transmitters. NO-sensitive guanylyl cyclase (NO-GC) acts as the key target of NO and, by the generation of cGMP, mediates nitrergic relaxation in the LES. To date, the exact mechanism of nitrergic LES relaxation is still insufficiently elucidated. To clarify the role of NO-GC in LES relaxation, we used cell-specific knockout (KO) mouse lines for NO-GC. These include mice lacking NO-GC in smooth muscle cells (SMC-GCKO), in interstitial cells of Cajal (ICC-GCKO) and in both SMC/ICC (SMC/ICC-GCKO). We applied oesophageal manometry to study the functionality of LES in vivo. Isometric force studies were performed to monitor LES responsiveness to exogenous NO and electric field stimulation of intrinsic nerves in vitro. Cell-specific expression/deletion of NO-GC was monitored by immunohistochemistry. Swallowing-induced LES relaxation is strongly reduced by deletion of NO-GC in ICC. Basal LES tone is affected by NO-GC deletion in either SMC or ICC. Lack of NO-GC in both cells leads to a complete interruption of NO-induced relaxation and, therefore, to an achalasia-like phenotype similar to that seen in global GCKO mice. Our data indicate that regulation of basal LES tone is based on a dual mechanism mediated by NO-GC in SMC and ICC whereas swallow-induced LES relaxation is mainly regulated by nitrergic mechanisms in ICC.
Collapse
Affiliation(s)
- Dieter Groneberg
- Physiologisches Institut I, Universität Würzburg, Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Müller M, Colcuc S, Drescher DG, Eckardt AJ, von Pein H, Taube C, Schumacher J, Gockel HR, Schimanski CC, Lang H, Gockel I. Murine genetic deficiency of neuronal nitric oxide synthase (nNOS(-/-) ) and interstitial cells of Cajal (W/W(v) ): Implications for achalasia? J Gastroenterol Hepatol 2014; 29:1800-7. [PMID: 24720557 DOI: 10.1111/jgh.12600] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIM Nitric oxide (NO) is an important inhibitory mediator of esophageal function, and its lack leads to typical features of achalasia. In contrast, the role of intramuscular interstitial cells of Cajal (ICC-IM) and vasoactive intestinal peptide (VIP) in lower esophageal sphincter (LES) function is still controversial. Therefore, we examined the function and morphology of the LES in vivo in NO-deficient (nNOS(-/-) ), ICC-IM-deficient (W/W(v) )-, and wild-type (WT) mice. METHODS Esophageal manometry was performed with a micro-sized transducer catheter to quantify LES pressure, swallow evoked LES relaxation, and esophageal body motility. The LES morphology was examined by semiquantitative analysis of the immunoreactivity (reduction grade I-IV) of neuronal NOS (nNOS), ICC-IM, and VIP and their correlation with esophageal function. RESULTS nNOS(-/-) in comparison to WT mice showed a significantly higher LES mean resting pressure with an impaired swallow induced relaxation, whereas W/W(v) mice had a hypotensive LES with decreased relaxation. W/W(v) and nNOS(-/-) mice demonstrated differing degrees of tubular esophageal dysfunction. The reduced immunoreactivity of nNOS correlated with an increased LES pressure and decreased LES relaxation, respectively. Cajal-cell reduction correlated with impaired LES relaxation, whereas VIP reduction revealed no correlation with esophageal function. CONCLUSIONS The reduction of ICC-IM and nNOS can cause dysfunction of the LES and esophageal peristalsis, whereas VIP reduction seems to have no effect. ICC-IM and nNOS deficiency might be independent relevant causes of esophageal dysfunction similar to that seen in human achalasia.
Collapse
Affiliation(s)
- Michaela Müller
- Department of Gastroenterology, German Diagnostic Clinic, Wiesbaden, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Sanders KM, Ward SM, Koh SD. Interstitial cells: regulators of smooth muscle function. Physiol Rev 2014; 94:859-907. [PMID: 24987007 DOI: 10.1152/physrev.00037.2013] [Citation(s) in RCA: 321] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
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 connectivity 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 PDGFRα(+) 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 PDGFRα(+) 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 mechanosensitivity. 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.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
36
|
Blair PJ, Rhee PL, Sanders KM, Ward SM. The significance of interstitial cells in neurogastroenterology. J Neurogastroenterol Motil 2014; 20:294-317. [PMID: 24948131 PMCID: PMC4102150 DOI: 10.5056/jnm14060] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/21/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Peter J Blair
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| |
Collapse
|
37
|
Park KS. Is this the era of interstitial cells of cajal transplantation? J Neurogastroenterol Motil 2014; 20:281-3. [PMID: 24931082 PMCID: PMC4102163 DOI: 10.5056/jnm14064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 05/30/2014] [Accepted: 05/31/2014] [Indexed: 01/20/2023] Open
Affiliation(s)
- Kyung Sik Park
- Department of Internal Medicine and Institute for Medical Science, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
38
|
Blair PJ, Rhee PL, Sanders KM, Ward SM. The significance of interstitial cells in neurogastroenterology. J Neurogastroenterol Motil 2014. [PMID: 24948131 DOI: 10.5056/jnm140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Peter J Blair
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
39
|
Lies B, Gil V, Groneberg D, Seidler B, Saur D, Wischmeyer E, Jiménez M, Friebe A. Interstitial cells of Cajal mediate nitrergic inhibitory neurotransmission in the murine gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2014; 307:G98-106. [PMID: 24833707 DOI: 10.1152/ajpgi.00082.2014] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Nitric oxide (NO) is a major inhibitory neurotransmitter in the gastrointestinal (GI) tract. Its main effector, NO-sensitive guanylyl cyclase (NO-GC), is expressed in several GI cell types, including smooth muscle cells (SMC), interstitial cells of Cajal (ICC), and fibroblast-like cells. Up to date, the interplay between neurons and these cells to initiate a nitrergic inhibitory junction potential (IJP) is unclear. Here, we investigate the origin of the nitrergic IJP in murine fundus and colon. IJPs were determined in fundus and colon SMC of mice lacking NO-GC globally (GCKO) and specifically in SMC (SM-GCKO), ICC (ICC-GCKO), and both SMC/ICC (SM/ICC-GCKO). Nitrergic IJP was abolished in ICC-GCKO fundus and reduced in SM-GCKO fundus. In the colon, the amplitude of nitrergic IJP was reduced in ICC-GCKO, whereas nitrergic IJP in SM-GCKO was reduced in duration. These results were corroborated by loss of the nitrergic IJP in global GCKO. In conclusion, our results prove the obligatory role of NO-GC in ICC for the initiation of an IJP. NO-GC in SMC appears to enhance the nitrergic IJP, resulting in a stronger and prolonged hyperpolarization in fundus and colon SMC, respectively. Thus NO-GC in both cell types is mandatory to induce a full nitrergic IJP. Our data from the colon clearly reveal the nitrergic IJP to be biphasic, resulting from individual inputs of ICC and SMC.
Collapse
Affiliation(s)
- Barbara Lies
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Víctor Gil
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, CIBERehd, Instituto de Salud Carlos III, Barcelona, Spain; and
| | - Dieter Groneberg
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| | - Barbara Seidler
- II. Medizinische Klinik und Poliklinik, Technische Universität München, München, Germany
| | - Dieter Saur
- II. Medizinische Klinik und Poliklinik, Technische Universität München, München, Germany
| | | | - Marcel Jiménez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, CIBERehd, Instituto de Salud Carlos III, Barcelona, Spain; and
| | - Andreas Friebe
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany;
| |
Collapse
|
40
|
Lies B, Groneberg D, Friebe A. Toward a better understanding of gastrointestinal nitrergic neuromuscular transmission. Neurogastroenterol Motil 2014; 26:901-12. [PMID: 24827638 DOI: 10.1111/nmo.12367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 04/21/2014] [Indexed: 12/30/2022]
Abstract
BACKGROUND Nitric oxide (NO) is an important inhibitory neurotransmitter in the gastrointestinal (GI) tract. The majority of nitrergic effects are transduced by NO-sensitive guanylyl cyclase (NO-GC) as the receptor for NO, and, thus, mediated by cGMP-dependent mechanisms. Work carried out during the past years has demonstrated NO to be largely involved in GI smooth muscle relaxation and motility. However, detailed investigation of nitrergic signaling has turned out to be complicated as NO-GC was identified in several different GI cell types such as smooth muscle cells, interstitial cells of Cajal and fibroblast-like cells. With regards to nitrergic neurotransmission, special focus has been placed on the role of interstitial cells of Cajal using mutant mice with reduced populations of ICC. Recently, global and cell-specific knockout mice for enzymes participating in nitrergic signaling have been generated providing a suitable approach to further examine the role of NO-mediated signaling in GI smooth muscle. PURPOSE This review discusses the current knowledge on nitrergic mechanisms in gastrointestinal neuromuscular transmission with a focus on genetic models and outlines possible further investigations to gain better understanding on NO-mediated effects in the GI tract.
Collapse
Affiliation(s)
- B Lies
- Physiologisches Institut I, Universität Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
41
|
Yarandi SS, Srinivasan S. Diabetic gastrointestinal motility disorders and the role of enteric nervous system: current status and future directions. Neurogastroenterol Motil 2014; 26:611-24. [PMID: 24661628 PMCID: PMC4104990 DOI: 10.1111/nmo.12330] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 02/18/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Gastrointestinal manifestations of diabetes are common and a source of significant discomfort and disability. Diabetes affects almost every part of gastrointestinal tract from the esophagus to the rectum and causes a variety of symptoms including heartburn, nausea, vomiting, abdominal pain, diarrhea and constipation. Understanding the underlying mechanisms of diabetic gastroenteropathy is important to guide development of therapies for this common problem. Over recent years, the data regarding the pathophysiology of diabetic gastroenteropathy is expanding. In addition to autonomic neuropathy causing gastrointestinal disturbances the role of enteric nervous system is becoming more evident. PURPOSE In this review, we summarize the reported alterations in enteric nervous system including enteric neurons, interstitial cells of Cajal and neurotransmission in diabetic animal models and patients. We also review the possible underlying mechanisms of these alterations, with focus on oxidative stress, growth factors and diabetes induced changes in gastrointestinal smooth muscle. Finally, we will discuss recent advances and potential areas for future research related to diabetes and the ENS such as gut microbiota, micro-RNAs and changes in the microvasculature and endothelial dysfunction.
Collapse
Affiliation(s)
- S. S. Yarandi
- Division of Digestive Diseases; Emory University; Atlanta GA
- Atlanta VA Medical Center; Decatur Georgia USA
| | - S. Srinivasan
- Division of Digestive Diseases; Emory University; Atlanta GA
- Atlanta VA Medical Center; Decatur Georgia USA
| |
Collapse
|
42
|
Winston JH, Chen J, Shi XZ, Sarna SK. Inflammation induced by mast cell deficiency rather than the loss of interstitial cells of Cajal causes smooth muscle dysfunction in W/W(v) mice. Front Physiol 2014; 5:22. [PMID: 24550836 PMCID: PMC3912454 DOI: 10.3389/fphys.2014.00022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 01/09/2014] [Indexed: 01/19/2023] Open
Abstract
The initial hypothesis suggested that the interstitial cells of Cajal (ICC) played an essential role in mediating enteric neuronal input to smooth muscle cells. Much information for this hypothesis came from studies in W/Wv mice lacking ICC. However, mast cells, which play critical roles in regulating inflammation in their microenvironment, are also absent in W/Wv mice. We tested the hypothesis that the depletion of mast cells in W/Wv mice generates inflammation in fundus muscularis externa (ME) that impairs smooth muscle reactivity to Ach, independent of the depletion of ICC. We performed experiments on the fundus ME from wild type (WT) and W/Wv mice before and after reconstitution of mast cells by bone marrow transplant. We found that mast cell deficiency in W/Wv mice significantly increased COX-2 and iNOS expression and decreased smooth muscle reactivity to Ach. Mast cell reconstitution or concurrent blockade of COX-2 and iNOS restored smooth muscle contractility without affecting the suppression of c-kit in W/Wv mice. The expression of nNOS and ChAT were suppressed in W/Wv mice; mast cell reconstitution did not restore them. We conclude that innate inflammation induced by mast cell deficiency in W/Wv mice impairs smooth muscle contractility independent of ICC deficiency. The impairment of smooth muscle contractility and the suppression of the enzymes regulating the synthesis of Ach and NO in W/Wv mice need to be considered in evaluating the role of ICC in regulating smooth muscle and enteric neuronal function in W/Wv mice.
Collapse
Affiliation(s)
- John H Winston
- Division of Gastroenterology, Department of Internal Medicine, Enteric Neuromuscular Disorders and Visceral Pain Center, The University of Texas Medical Branch at Galveston Galveston, TX, USA
| | - Jinghong Chen
- Division of Gastroenterology, Department of Internal Medicine, Enteric Neuromuscular Disorders and Visceral Pain Center, The University of Texas Medical Branch at Galveston Galveston, TX, USA
| | - Xuan-Zheng Shi
- Division of Gastroenterology, Department of Internal Medicine, Enteric Neuromuscular Disorders and Visceral Pain Center, The University of Texas Medical Branch at Galveston Galveston, TX, USA
| | - Sushil K Sarna
- Division of Gastroenterology, Department of Internal Medicine, Enteric Neuromuscular Disorders and Visceral Pain Center, The University of Texas Medical Branch at Galveston Galveston, TX, USA
| |
Collapse
|
43
|
Lentle RG, Reynolds GW, Janssen PWM. Gastrointestinal tone; its genesis and contribution to the physical processes of digestion. Neurogastroenterol Motil 2013; 25:931-42. [PMID: 24028606 DOI: 10.1111/nmo.12223] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/08/2013] [Indexed: 12/24/2022]
Abstract
BACKGROUND Myogenic tone has long been recognised as an important component of gastrointestinal motility. Recent work has clarified the cellular mechanisms that engender tone and the neurogenic and mechanical stimuli that modulate it but has also highlighted cellular and regional specialisation in these mechanisms within the GI tract. Smooth muscle in all segments of the gut has the capability of latching, i.e. can generate ongoing specific rather than tetanic tone. This is likely modulated by both direct and indirect input from agonists such as acetylcholine and mechanoreceptors, the latter originating in ICC-IM, smooth muscle cells or elements of the ENS. Tonic contraction can occur in the absence of phasic contractions or concurrent with them, and it can modulate wall compliance and the capacity of particular segments, thereby affecting the level of on-flow and mixing, both luminal and adjacent to the mucosa. PURPOSE The review seeks to provide an overview of our understanding of the mechanism by which tone is generated and maintained, highlighting its modulation by neurogenic and mechanical stimuli, its mechanical consequences in the walls of the various segments of the gastrointestinal tract and its contribution to flow and mixing of contained digesta.
Collapse
Affiliation(s)
- R G Lentle
- Institute of Food, Nutrition and Human Health, Massey University, Palmerston North, New Zealand
| | | | | |
Collapse
|
44
|
Baker SA, Hennig GW, Salter AK, Kurahashi M, Ward SM, Sanders KM. Distribution and Ca(2+) signalling of fibroblast-like (PDGFR(+)) cells in the murine gastric fundus. J Physiol 2013; 591:6193-208. [PMID: 24144881 DOI: 10.1113/jphysiol.2013.264747] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Platelet-derived growth factor receptor α positive (PDGFRα(+)) cells are suggested to mediate purinergic inputs in GI muscles, but the responsiveness of these cells to purines in situ has not been evaluated. We developed techniques to label and visualize PDGFRα(+) cells in murine gastric fundus, load cells with Ca(2+) indicators, and follow their activity via digital imaging. Immunolabelling demonstrated a high density of PDGFRα(+) cells in the fundus. Cells were isolated and purified by fluorescence-activated cell sorting (FACS) using endogenous expression of enhanced green fluorescent protein (eGFP) driven off the Pdgfra promoter. Quantitative PCR showed high levels of expression of purinergic P2Y1 receptors and SK3 K(+) channels in PDGFRα(+) cells. Ca(2+) imaging was used to characterize spontaneous Ca(2+) transients and responses to purines in PDGFRα(+) cells in situ. ATP, ADP, UTP and β-NAD elicited robust Ca(2+) transients in PDGFRα(+) cells. Ca(2+) transients were also elicited by the P2Y1-specific agonist (N)-methanocarba-2MeSADP (MRS-2365), and inhibited by MRS-2500, a P2Y1-specific antagonist. Responses to ADP, MRS-2365 and β-NAD were absent in PDGFRα(+) cells from P2ry1((-/-)) mice, but responses to ATP were retained. Purine-evoked Ca(2+) transients were mediated through Ca(2+) release mechanisms. Inhibitors of phospholipase C (U-73122), IP3 (2-APB), ryanodine receptors (Ryanodine) and SERCA pump (cyclopiazonic acid and thapsigargin) abolished Ca(2+) transients elicited by purines. This study provides a link between purine binding to P2Y1 receptors and activation of SK3 channels in PDGFRα(+) cells. Activation of Ca(2+) release is likely to be the signalling mechanism in PDGFRα(+) cells responsible for the transduction of purinergic enteric inhibitory input in gastric fundus muscles.
Collapse
Affiliation(s)
- Salah A Baker
- K. M. Sanders: Department of Physiology and Cell Biology, University of Nevada School of Medicine, MS 352, Reno, NV 89557, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Bautista-Cruz F, Nair DG, Lourenssen S, Miller DV, Blennerhassett MG, Paterson WG. Impaired platelet-derived growth factor receptor expression and function in cultured lower esophageal sphincter circular smooth muscle cells from W/W(v) mutant mice. Can J Physiol Pharmacol 2013; 92:34-41. [PMID: 24383871 DOI: 10.1139/cjpp-2013-0254] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously demonstrated that lower esophageal sphincter (LES) circular smooth muscle (CSM) is functionally impaired in W/W(v) mutant mice that lack interstitial cells of Cajal, and speculated that this could be due to altered smooth muscle differentiation. Platelet-derived growth factor (PDGF) is involved in the maturation and differentiation of smooth muscle. To determine whether PDGF expression and (or) function is altered in W/W(v) mutant mice, PDGF-Rβ expression was measured using RT-PCR, qPCR, and immunocytochemistry, and Ca(2+) imaging and perforated patch clamp recordings performed in isolated LES CSM cells. RT-PCR and immunocytochemistry showed significantly reduced PDGF-Rβ expression in the LES from mutant as opposed to wild-type mice. Quantitative comparison of CSM cell numbers in histological specimens revealed a significantly increased average cell size in the mutant tissue. The specific PDGF-Rβ ligand, PDGF-BB, caused a significant increase in intracellular Ca(2+) in cells from the wild-type mice compared with the mutants. Using a ramp protocol, PDGF-BB caused a 2-fold increase in outward K(+) currents in cells from the wild-type mice, whereas no significant increase was measured in the cells from the mutants. We conclude that the expression and function of PDGF-Rβ in LES CSM from W/W(v) mice is impaired, providing further evidence that LES CSM is abnormal in W/W(v) mutants.
Collapse
Affiliation(s)
- Francisco Bautista-Cruz
- a Gastrointestinal Disease Research Unit, Kingston General Hospital, 76 Stuart Street, Kingston ON K7L 2V7, Canada
| | | | | | | | | | | |
Collapse
|
46
|
Groneberg D, Lies B, König P, Jäger R, Seidler B, Klein S, Saur D, Friebe A. Cell-specific deletion of nitric oxide-sensitive guanylyl cyclase reveals a dual pathway for nitrergic neuromuscular transmission in the murine fundus. Gastroenterology 2013; 145:188-196. [PMID: 23528627 DOI: 10.1053/j.gastro.2013.03.042] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 02/26/2013] [Accepted: 03/18/2013] [Indexed: 01/29/2023]
Abstract
BACKGROUND & AIMS It is not clear how nitric oxide (NO) released from enteric neurons relaxes gastrointestinal (GI) smooth muscle. In analogy to the vascular system, NO might directly induce relaxation of smooth muscle cells (SMCs) by acting on its receptor, NO-sensitive guanylyl cyclase (NO-GC). Alternatively, intermediate cells, such as the interstitial cells of Cajal (ICCs), might detect nitrergic signals to indirectly regulate smooth muscle tone, and thereby regulate the motor function of the GI tract. We investigated the role of ICCs and SMCs in nitrergic relaxation using mice with cell-specific disruption of the gene encoding the β1 subunit of NO-GC (GUCY1B3). METHODS We created mice that lack NO-GC specifically in SMCs (SM-guanylyl cyclase knockout [GCKO]), ICCs (ICC-GCKO), or both (SM/ICC-GCKO). We investigated the effects of exogenous and endogenous NO on murine fundus using isometric force studies. Total gut transit time was measured to monitor the functional consequences of NO-GC deletion on GI motility in vivo. RESULTS NO-GC is expressed in ICC and SMC. Deletion of the NO receptor from SMCs incompletely reduced NO-induced fundus relaxation, which was hardly affected after ICC-specific deletion. Gut transit time did not change in SM-GCKO or ICC-GCKO mice compared with control mice. However, nitrergic relaxation was not observed in SM/ICC-GCKO mice, which had increased gut transit time compared with controls. CONCLUSIONS In mice, NO-GC is the only NO receptor to relax the fundus; deletion of NO-GC from the combination of SMCs and ICCs blocks nitrergic signaling. Therefore, ICCs and SMCs jointly mediate the relaxant effect of enteric NO.
Collapse
Affiliation(s)
- Dieter Groneberg
- Physiologisches Institut I, Universität Würzburg, Würzburg, Germany
| | - Barbara Lies
- Physiologisches Institut I, Universität Würzburg, Würzburg, Germany
| | - Peter König
- Institut für Anatomie, Zentrum für medizinische Struktur- und Zellbiologie, Universität zu Lübeck, Lübeck, Germany
| | - Ronald Jäger
- Physiologisches Institut I, Universität Würzburg, Würzburg, Germany
| | - Barbara Seidler
- II. Medizinische Klinik und Poliklinik, Technische Universität München, München, Germany
| | - Sabine Klein
- II. Medizinische Klinik und Poliklinik, Technische Universität München, München, Germany
| | - Dieter Saur
- II. Medizinische Klinik und Poliklinik, Technische Universität München, München, Germany
| | - Andreas Friebe
- Physiologisches Institut I, Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
47
|
Chen JH, Wang XY, Liu LWC, Yu W, Yu Y, Zhao L, Huizinga JD. On the origin of rhythmic contractile activity of the esophagus in early achalasia, a clinical case study. Front Neurosci 2013; 7:77. [PMID: 23734090 PMCID: PMC3659367 DOI: 10.3389/fnins.2013.00077] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 04/30/2013] [Indexed: 01/26/2023] Open
Abstract
A patient with early achalasia presented spontaneous strong rhythmic non-propulsive contractions at ~7/min, independent of swallows. Our aim was to evaluate characteristics of the rhythmic contractions, provide data on the structure of pacemaker cells in the esophagus and discuss a potential role for interstitial cells of Cajal (ICC) in the origin of rhythmicity. We hypothesize that intramuscular ICC (ICC-IM) are the primary pacemaker cells. The frequency but not the amplitude of the rhythmic contractions was inhibited by the phosphodiesterase inhibitor drotaverine consistent with cAMP inhibiting pacemaker currents in ICC-IM. The frequency increased by wet swallows but not dry swallows, consistent with stretch causing increase in slow wave frequency in ICC-IM. New studies on archival material showed that ICC-IM were present throughout the human esophageal musculature and were not diminished in early achalasia. Although ICC-IM exhibited a low density, they were connected to PDGFRα-positive fibroblast-like cells with whom they formed a dense gap junction coupled network. Nitrergic innervation of ICC was strongly diminished in early achalasia because of the loss of nitrergic nerves. It therefore appears possibly that ICC-IM function as pacemaker cells in the esophagus and that the network of ICC and PDGFRα-positive cells allows for coupling and propagation of the pacemaker activity. Loss of nitrergic innervation to ICC in achalasia may render them more excitable such that its pacemaker activity is more easily expressed. Loss of propagation in achalasia may be due to loss of contraction-induced aboral nitrergic inhibition.
Collapse
Affiliation(s)
- Ji-Hong Chen
- Department of Gastroenterology and Hepatology, Renmin Hospital of Wuhan University and Wuhan University Institute of Digestive and Liver diseases Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
48
|
Wang JP, Ding GF, Wang QZ. Interstitial cells of Cajal mediate excitatory sympathetic neurotransmission in guinea pig prostate. Cell Tissue Res 2013; 352:479-86. [PMID: 23411811 DOI: 10.1007/s00441-013-1572-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/17/2013] [Indexed: 01/26/2023]
Abstract
Morphological and functional studies have confirmed that interstitial cells of Cajal (ICCs) are involved in many enteric motor neurotransmission pathways. Recent investigations have demonstrated that human and guinea pig prostate glands possess a distinct cell type with morphological and immunological similarities to ICCs. These prostate ICCs have a close relationship with nerve bundles and smooth muscle cells. Prostate smooth muscle tone is largely induced by stimulation from the sympathetic nervous system, which releases excitatory norepinephrine (NE) to act on the α1-adrenoceptor. We have performed morphological and functional experiments to determine the role of ICCs in sympathetic neurotransmission in the guinea pig prostate based on the hypothesis that prostate ICCs act as mediators of sympathetic neurotransmission. Immunohistochemistry revealed many close points of contact between ICCs and sympathetic nerve bundles and smooth muscle cells. Double-labeled sections revealed that α1-adrenoceptor and the gap junction protein connexin 43 were expressed in prostate ICCs. Surprisingly, prostate ICCs co-expressed tyrosine hydroxylase and dopamine β-hydroxylase, two markers of sympathetic neurons. Functionally, the application of NE evoked a large single inward current in isolated prostate ICCs in a dose-dependent manner. The inward current evoked by NE was mediated via the activation of α1-adrenoceptors, because it was abolished by the non-specific α-adrenoceptor antagonist, phentolamine and the specific α1-adrenoceptor antagonist, prazosin. Thus, ICCs in the guinea pig prostate are target cells for prostate sympathetic nerves and possess the morphological and functional characteristics required to mediate sympathetic signals.
Collapse
Affiliation(s)
- Jiang-ping Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | | |
Collapse
|
49
|
Abstract
Gastrointestinal (GI) motility function and its regulation is a complex process involving collaboration and communication of multiple cell types such as enteric neurons, interstitial cells of Cajal (ICC), and smooth muscle cells. Recent advances in GI research made a better understanding of ICC function and their role in the GI tract, and studies based on different types of techniques have shown that ICC, as an integral part of the GI neuromuscular apparatus, transduce inputs from enteric motor neurons, generate intrinsic electrical rhythmicity in phasic smooth muscles, and have a mechanical sensation ability. Absence or improper function of these cells has been linked to some GI tract disorders. This paper provides a general overview of ICC; their discovery, subtypes, function, locations in the GI tract, and some disorders associated with their loss or disease, and highlights some controversial issues with regard to the importance of ICC in the GI tract.
Collapse
Affiliation(s)
- Othman A. Al-Shboul
- Department of Physiology, Jordan University of Science and Technology, Irbid, Jordan,Address for correspondence: Dr. Othman Abdullah Al-Shboul, Department of Physiology, Jordan University of Science and Technology, P.O. Box 3030, Irbid - 22110, Jordan. E-mail:
| |
Collapse
|
50
|
Gong YY, Si XM, Lin L, Lu J. Mechanisms of cholecystokinin-induced calcium mobilization in gastric antral interstitial cells of Cajal. World J Gastroenterol 2012; 18:7184-7193. [PMID: 23326123 PMCID: PMC3544020 DOI: 10.3748/wjg.v18.i48.7184] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Revised: 10/09/2012] [Accepted: 11/06/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of sulfated cholecystokinin-8 (CCK-8S) on calcium mobilization in cultured murine gastric antral interstitial cells of Cajal (ICC) and its possible mechanisms.
METHODS: ICC were isolated from the gastric antrum of mice and cultured. Immunofluorescence staining with a monoclonal antibody for c-Kit was used to identify ICC. The responsiveness of ICC to CCK-8S was measured using Fluo-3/AM based digital microfluorimetric measurement of intracellular Ca2+ concentration ([Ca2+]i). A confocal laser scanning microscope was used to monitor [Ca2+]i changes. The selective CCK1 receptor antagonist lorglumide, the intracellular Ca2+-ATPase inhibitor thapsigargin, the type III inositol 1,4,5-triphosphate (InsP3) receptor blocker xestospongin C and the L-type voltage-operated Ca2+ channel inhibitor nifedipine were used to examine the mechanisms of [Ca2+]i elevation caused by CCK-8S. Immunoprecipitation and Western blotting were used to determine the regulatory effect of PKC on phosphorylation of type III InsP3 receptor (InsP3R3) in ICC. Protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA) and inhibitor chelerythrine were used to assess the role of PKC in the CCK-8S-evoked [Ca2+]i increment of ICC.
RESULTS: ICC were successfully isolated from the gastric antrum of mice and cultured. Cultured ICC were identified by immunofluorescence staining. When given 80 nmol/L or more than 80 nmol/L CCK-8S, the [Ca2+]i in ICC increased and 100 nmol/L CCK-8S significantly increased the mean [Ca2+]i by 59.30% ± 4.85% (P < 0.01). Pretreatment of ICC with 5 μmol/L lorglumide inhibited 100 nmol/L CCK-8S-induced [Ca2+]i increment from 59.30% ± 4.85% to 14.97% ± 9.05% (P < 0.01), suggesting a CCK1R-mediated event. Emptying of intracellular calcium stores by thapsigargin (5 μmol/L) prevented CCK-8S (100 nmol/L) from inducing a [Ca2+]i increase. Moreover, pretreatment with xestospongin C (1 μmol/L) could also abolish the CCK-8S-induced effect, indicating that Ca2+ release from InsP3R-operated stores appeared to be a major mechanism responsible for CCK-8S-induced calcium mobilization in ICC. On the other hand, by removing extracellular calcium or blocking the L-type voltage-operated calcium channel with nifedipine, a smaller but significant rise in the [Ca2+]i could be still elicited by CCK-8S. These data suggest that the [Ca2+]i release is not stimulated or activated by the influx of extracellular Ca2+ in ICC, but the influx of extracellular Ca2+ can facilitate the [Ca2+]i increase evoked by CCK-8S. CCK-8S increased the phosphorylation of InsP3R3, which could be prevented by chelerythrine. Pretreatment with lorglumide (5 μmol/L) could significantly reduce the CCK-8S intensified phosphorylation of InsP3R3. In the positive control group, treatment of cells with PMA also resulted in an enhanced phosphorylation of InsP3R3. Pretreatment with various concentrations of PMA (10 nmol/L-10 μmol/L) apparently inhibited the effect of CCK-8S and the effect of 100 nmol/L PMA was most obvious. Likewise, the effect of CCK-8S was augmented by the pretreatment with chelerythrine (10 nmol/L-10 μmol/L) and 100 nmol/L chelerythrine exhibited the maximum effect.
CONCLUSION: CCK-8S increases [Ca2+]i in ICC via the CCK1 receptor. This effect depends on the release of InsP3R-operated Ca2+ stores, which is negatively regulated by PKC-mediated phosphorylation of InsP3R3.
Collapse
|