1
|
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
|
2
|
Andrews PLR, Golding JF, Sanger GJ. An assessment of the effects of neurokinin 1 receptor antagonism against nausea and vomiting: Relative efficacy, sites of action and lessons for future drug development. Br J Clin Pharmacol 2023; 89:3468-3490. [PMID: 37452618 DOI: 10.1111/bcp.15852] [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: 05/12/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
A broad-spectrum anti-vomiting effect of neurokinin1 receptor antagonists (NK1 RA), shown in pre-clinical animal studies, has been supported by a more limited range of clinical studies in different indications. However, this review suggests that compared with vomiting, the self-reported sensation of nausea is less affected or possibly unaffected (depending on the stimulus) by NK1 receptor antagonism, a common finding for anti-emetics. The stimulus-independent effects of NK1 RAs against vomiting are explicable by actions within the central pattern generator (ventral brainstem) and the nucleus tractus solitarius (NTS; dorsal brainstem), with additional effects on vagal afferent activity for certain stimuli (e.g., highly emetogenic chemotherapy). The central pattern generator and NTS neurones are multifunctional so the notable lack of obvious effects of NK1 RAs on other reflexes mediated by the same neurones suggests that their anti-vomiting action is dependent on the activation state of the pathway leading to vomiting. Nausea requires activation of cerebral pathways by projection of information from the NTS. Although NK1 receptors are present in cerebral nuclei implicated in nausea, and imaging studies show very high receptor occupancy at clinically used doses, the variable or limited ability of NK1 RAs to inhibit nausea emphasizes: (i) our inadequate understanding of the mechanisms of nausea; and (ii) that classification of a drug as an anti-emetic may give a false impression of efficacy against nausea vs. vomiting. We discuss the potential mechanisms for the differential efficacy of NK1 RA and the implications for future development of drugs that can effectively treat nausea, an area of unmet clinical need.
Collapse
Affiliation(s)
- Paul L R Andrews
- Division of Biomedical Sciences, St George's University of London, London, UK
| | | | - Gareth J Sanger
- Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
3
|
Wang Q, Wang J, Tokhtaeva E, Li Z, Martín MG, Ling XB, Dunn JC. An Engineered Living Intestinal Muscle Patch Produces Macroscopic Contractions that can Mix and Break Down Artificial Intestinal Contents. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207255. [PMID: 36779454 PMCID: PMC10101936 DOI: 10.1002/adma.202207255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/13/2023] [Indexed: 06/03/2023]
Abstract
The intestinal muscle layers execute various gut wall movements to achieve controlled propulsion and mixing of intestinal content. Engineering intestinal muscle layers with complex contractile function is critical for developing bioartificial intestinal tissue to treat patients with short bowel syndrome. Here, the first demonstration of a living intestinal muscle patch capable of generating three distinct motility patterns and displaying multiple digesta manipulations is reported. Assessment of contractility, cellular morphology, and transcriptome profile reveals that successful generation of the contracting muscle patch relies on both biological factors in a serum-free medium and environmental cues from an elastic electrospun gelatin scaffold. By comparing gene-expression patterns among samples, it is shown that biological factors from the medium strongly affect ion-transport activities, while the scaffold unexpectedly regulates cell-cell communication. Analysis of ligandreceptor interactome identifies scaffold-driven changes in intercellular communication, and 78% of the upregulated ligand-receptor interactions are involved in the development and function of enteric neurons. The discoveries highlight the importance of combining biomolecular and biomaterial approaches for tissue engineering. The living intestinal muscle patch represents a pivotal advancement for building functional replacement intestinal tissue. It offers a more physiological model for studying GI motility and for preclinical drug discovery.
Collapse
Affiliation(s)
- Qianqian Wang
- Division of Pediatric Surgery, Departments of Surgery and Bioengineering, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Jiafang Wang
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Elmira Tokhtaeva
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Zhen Li
- Division of Pediatric Surgery, Departments of Surgery and Bioengineering, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Martín G. Martín
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Xuefeng B. Ling
- Division of Pediatric Surgery, Departments of Surgery and Bioengineering, Stanford University School of Medicine, Stanford, California 94305, USA
| | - James C.Y. Dunn
- Division of Pediatric Surgery, Departments of Surgery and Bioengineering, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
4
|
Foong D, Liyanage L, Zhou J, Zarrouk A, Ho V, O'Connor MD. Single-cell RNA sequencing predicts motility networks in purified human gastric interstitial cells of Cajal. Neurogastroenterol Motil 2022; 34:e14303. [PMID: 34913225 DOI: 10.1111/nmo.14303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/25/2021] [Accepted: 11/17/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Gastrointestinal (GI) motility disorders affect millions of people worldwide, yet they remain poorly treated in part due to insufficient knowledge of the molecular networks controlling GI motility. Interstitial cells of Cajal (ICC) are critical GI pacemaker cells, and abnormalities in ICC are implicated in GI motility disorders. Two cell surface proteins, KIT and ANO1, are used for identifying ICC. However, difficulties accessing human tissue and the low frequency of ICC in GI tissues have meant human ICC are insufficiently characterized. Here, a range of characterization assays including single-cell RNA sequencing (scRNA-seq) was performed using KIT+ CD45- CD11B- primary human gastric ICC to better understand networks controlling human ICC biology. METHODS Excess sleeve gastrectomy tissues were dissected; ICC were analyzed by immunofluorescence, fluorescence-activated cell sorting (FACSorting), real-time PCR, mass spectrometry, and scRNA-seq. KEY RESULTS Immunofluorescence identified ANO1+ /KIT+ cells throughout the gastric muscle. Compared to the FACSorted negative cells, PCR showed the KIT+ CD45- CD11B- ICC were enriched 28-fold in ANO1 expression (p < 0.01). scRNA-seq analysis of the KIT- CD45+ CD11B+ and KIT+ CD45- CD11B- ICC revealed separate clusters of immune cells and ICC (respectively); cells in the ICC cluster expressed critical GI motility genes (eg, CAV1 and PRKG1). The scRNA-seq data for these two cell clusters predicted protein interaction networks consistent with immune cell and ICC biology, respectively. CONCLUSIONS & INFERENCES The single-cell transcriptome of purified KIT+ CD45- CD11B- human gastric ICC presented here provides new molecular insights and hypotheses into evolving models of GI motility. This knowledge will provide an improved framework to investigate targeted therapies for GI motility disorders.
Collapse
Affiliation(s)
- Daphne Foong
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Liwan Liyanage
- School of Computing, Data and Mathematical Sciences, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Jerry Zhou
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Ali Zarrouk
- Campbelltown Private Hospital, Campbelltown, New South Wales, Australia
| | - Vincent Ho
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia.,Campbelltown Private Hospital, Campbelltown, New South Wales, Australia
| | - Michael D O'Connor
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| |
Collapse
|
5
|
Huizinga JD, Hussain A, Chen JH. Interstitial cells of Cajal and human colon motility in health and disease. Am J Physiol Gastrointest Liver Physiol 2021; 321:G552-G575. [PMID: 34612070 DOI: 10.1152/ajpgi.00264.2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Our understanding of human colonic motility, and autonomic reflexes that generate motor patterns, has increased markedly through high-resolution manometry. Details of the motor patterns are emerging related to frequency and propagation characteristics that allow linkage to interstitial cells of Cajal (ICC) networks. In studies on colonic motor dysfunction requiring surgery, ICC are almost always abnormal or significantly reduced. However, there are still gaps in our knowledge about the role of ICC in the control of colonic motility and there is little understanding of a mechanistic link between ICC abnormalities and colonic motor dysfunction. This review will outline the various ICC networks in the human colon and their proven and likely associations with the enteric and extrinsic autonomic nervous systems. Based on our extensive knowledge of the role of ICC in the control of gastrointestinal motility of animal models and the human stomach and small intestine, we propose how ICC networks are underlying the motor patterns of the human colon. The role of ICC will be reviewed in the autonomic neural reflexes that evoke essential motor patterns for transit and defecation. Mechanisms underlying ICC injury, maintenance, and repair will be discussed. Hypotheses are formulated as to how ICC dysfunction can lead to motor abnormalities in slow transit constipation, chronic idiopathic pseudo-obstruction, Hirschsprung's disease, fecal incontinence, diverticular disease, and inflammatory conditions. Recent studies on ICC repair after injury hold promise for future therapies.
Collapse
Affiliation(s)
- Jan D Huizinga
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Amer Hussain
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Ji-Hong Chen
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
6
|
Colmenares Aguilar MG, Mazzone A, Eisenman ST, Strege PR, Bernard CE, Holmes HL, Romero MF, Farrugia G, Gibbons SJ. Expression of the regulated isoform of the electrogenic Na +/HCO 3- cotransporter, NBCe1, is enriched in pacemaker interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 2021; 320:G93-G107. [PMID: 33112159 PMCID: PMC8112189 DOI: 10.1152/ajpgi.00255.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Interstitial cells of Cajal (ICCs) generate electrical slow waves, which are required for normal gastrointestinal motility. The mechanisms for generation of normal pacemaking are not fully understood. Normal gastrointestinal contractility- and electrical slow-wave activity depend on the presence of extracellular HCO3-. Previous transcriptional analysis identified enrichment of mRNA encoding the electrogenic Na+/HCO3- cotransporter (NBCe1) gene (Slc4a4) in pacemaker myenteric ICCs in mouse small intestine. We aimed to determine the distribution of NBCe1 protein in ICCs of the mouse gastrointestinal tract and to identify the transcripts of the Slc4a4 gene in mouse and human small intestinal tunica muscularis. We determined the distribution of NBCe1 immunoreactivity (NBCe1-IR) by immunofluorescent labeling in mouse and human tissues. In mice, NBCe1-IR was restricted to Kit-positive myenteric ICCs of the stomach and small intestine and submuscular ICCs of the large intestine, that is, the slow wave generating subset of ICCs. Other subtypes of ICCs were NBCe1-negative. Quantitative real-time PCR identified >500-fold enrichment of Slc4a4-207 and Slc4a4-208 transcripts ["IP3-receptor-binding protein released by IP3" (IRBIT)-regulated isoforms] in Kit-expressing cells isolated from KitcreERT2/+, Rpl22tm1.1Psam/Sj mice and from single GFP-positive ICCs from Kittm1Rosay mice. Human jejunal tunica muscularis ICCs were also NBCe1-positive, and SLC4A4-201 and SLC4A4-204 RNAs were >300-fold enriched relative to SLC4A4-202. In summary, NBCe1 protein expressed in ICCs with electrical pacemaker function is encoded by Slc4a4 gene transcripts that generate IRBIT-regulated isoforms of NBCe1. In conclusion, Na+/HCO3- cotransport through NBCe1 contributes to the generation of pacemaker activity in subsets of ICCs.NEW & NOTEWORTHY In this study, we show that the electrogenic Na+/HCO3- cotransporter, NBCe1/Slc4a4, is expressed in subtypes of interstitial cells of Cajal (ICCs) responsible for electrical slow wave generation throughout the mouse gastrointestinal tract and is absent in other types of ICCs. The transcripts of Slc4a4 expressed in mouse ICCs and human gastrointestinal smooth muscle are the regulated isoforms. This indicates a key role for HCO3- transport in generation of gastrointestinal motility patterns.
Collapse
Affiliation(s)
| | - Amelia Mazzone
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Seth T. Eisenman
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Peter R. Strege
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Cheryl E. Bernard
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| | - Heather L. Holmes
- 2Physiology and Biomedical Engineering, Nephrology and
Hypertension, Mayo Clinic College of Medicine and
Science, Rochester, Minnesota
| | - Michael F. Romero
- 2Physiology and Biomedical Engineering, Nephrology and
Hypertension, Mayo Clinic College of Medicine and
Science, Rochester, Minnesota
| | - Gianrico Farrugia
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota,3Department of Physiology and Biomedical Engineering,
Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- 1Enteric NeuroScience Program, Division of
Gastroenterology and Hepatology, Mayo Clinic,
Rochester, Minnesota
| |
Collapse
|
7
|
Foong D, Zhou J, Zarrouk A, Ho V, O’Connor MD. Understanding the Biology of Human Interstitial Cells of Cajal in Gastrointestinal Motility. Int J Mol Sci 2020; 21:ijms21124540. [PMID: 32630607 PMCID: PMC7352366 DOI: 10.3390/ijms21124540] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Millions of patients worldwide suffer from gastrointestinal (GI) motility disorders such as gastroparesis. These disorders typically include debilitating symptoms, such as chronic nausea and vomiting. As no cures are currently available, clinical care is limited to symptom management, while the underlying causes of impaired GI motility remain unaddressed. The efficient movement of contents through the GI tract is facilitated by peristalsis. These rhythmic slow waves of GI muscle contraction are mediated by several cell types, including smooth muscle cells, enteric neurons, telocytes, and specialised gut pacemaker cells called interstitial cells of Cajal (ICC). As ICC dysfunction or loss has been implicated in several GI motility disorders, ICC represent a potentially valuable therapeutic target. Due to their availability, murine ICC have been extensively studied at the molecular level using both normal and diseased GI tissue. In contrast, relatively little is known about the biology of human ICC or their involvement in GI disease pathogenesis. Here, we demonstrate human gastric tissue as a source of primary human cells with ICC phenotype. Further characterisation of these cells will provide new insights into human GI biology, with the potential for developing novel therapies to address the fundamental causes of GI dysmotility.
Collapse
Affiliation(s)
- Daphne Foong
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (D.F.); (J.Z.); (V.H.)
| | - Jerry Zhou
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (D.F.); (J.Z.); (V.H.)
| | - Ali Zarrouk
- Campbelltown Private Hospital, Campbelltown, NSW 2560, Australia;
| | - Vincent Ho
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (D.F.); (J.Z.); (V.H.)
- Campbelltown Private Hospital, Campbelltown, NSW 2560, Australia;
| | - Michael D. O’Connor
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia; (D.F.); (J.Z.); (V.H.)
- Correspondence:
| |
Collapse
|
8
|
Dal Ben D, Antonioli L, Lambertucci C, Spinaci A, Fornai M, D'Antongiovanni V, Pellegrini C, Blandizzi C, Volpini R. Approaches for designing and discovering purinergic drugs for gastrointestinal diseases. Expert Opin Drug Discov 2020; 15:687-703. [PMID: 32228110 DOI: 10.1080/17460441.2020.1743673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Purines finely modulate physiological motor, secretory, and sensory functions in the gastrointestinal tract. Their activity is mediated by the purinergic signaling machinery, including receptors and enzymes regulating their synthesis, release, and degradation. Several gastrointestinal dysfunctions are characterized by alterations affecting the purinergic system. AREAS COVERED The authors provide an overview on the purinergic receptor signaling machinery, the molecules and proteins involved, and a summary of medicinal chemistry efforts aimed at developing novel compounds able to modulate the activity of each player involved in this machinery. The involvement of purinergic signaling in gastrointestinal motor, secretory, and sensory functions and dysfunctions, and the potential therapeutic applications of purinergic signaling modulators, are then described. EXPERT OPINION A number of preclinical and clinical studies demonstrate that the pharmacological manipulation of purinergic signaling represents a viable way to counteract several gastrointestinal diseases. At present, the paucity of purinergic therapies is related to the lack of receptor-subtype-specific agonists and antagonists that are effective in vivo. In this regard, the development of novel therapeutic strategies should be focused to include tools able to control the P1 and P2 receptor expression as well as modulators of the breakdown or transport of purines.
Collapse
Affiliation(s)
- Diego Dal Ben
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Catia Lambertucci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| | - Andrea Spinaci
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Vanessa D'Antongiovanni
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | | | - Corrado Blandizzi
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Rosaria Volpini
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino , Camerino, Italy
| |
Collapse
|
9
|
Grubišić V, Perez-Medina AL, Fried DE, Sévigny J, Robson SC, Galligan JJ, Gulbransen BD. NTPDase1 and -2 are expressed by distinct cellular compartments in the mouse colon and differentially impact colonic physiology and function after DSS colitis. Am J Physiol Gastrointest Liver Physiol 2019; 317:G314-G332. [PMID: 31188623 PMCID: PMC6774087 DOI: 10.1152/ajpgi.00104.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
ATP is both an important mediator of physiological gut functions such as motility and epithelial function, and a key danger signal that mediates cell death and tissue damage. The actions of extracellular ATP are regulated through the catalytic functions extracellular nucleoside triphosphate diphosphohydrolase-1 (NTPDase1), -2, -3, and -8, which ultimately generate nucleosides. Ectonucleotidases have distinct cellular associations, but the specific locations and functional roles of individual NTPDases in the intestine are still poorly understood. Here, we tested the hypothesis that differential and cell-selective regulation of purine hydrolysis by NTPDase1 and -2 plays important roles in gut physiology and disease. We studied Entpd1 and Entpd2 null mice in health and following colitis driven by 2% dextran sulfate sodium (DSS) administration using functional readouts of gut motility, epithelial barrier function, and neuromuscular communication. NTPDase1 is expressed by immune cells, and the ablation of Entpd1 altered glial numbers in the myenteric plexus. NTPDase2 is expressed by enteric glia, and the ablation of Entpd2 altered myenteric neuron numbers. Mice lacking either NTPDase1 or -2 exhibited decreased inhibitory neuromuscular transmission and altered components of inhibitory junction potentials. Ablation of Entpd2 increased gut permeability following inflammation. In conclusion, the location- and context-dependent extracellular nucleotide phosphohydrolysis by NTPDase1 and -2 substantially impacts gut function in health and disease.NEW & NOTEWORTHY Purines are important mediators of gastrointestinal physiology and pathophysiology. Nucleoside triphosphate diphosphohydrolases (NTPDases) regulate extracellular purines, but the roles of specific NTPDases in gut functions are poorly understood. Here, we used Entpd1- and Entpd2-deficient mice to show that the differential and cell-selective regulation of purine hydrolysis by NTPDase1 and -2 plays important roles in barrier function, gut motility, and neuromuscular communication in health and disease.
Collapse
Affiliation(s)
- Vladimir Grubišić
- 1Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Alberto L. Perez-Medina
- 2Department of Pharmacology and Toxicology and Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - David E. Fried
- 1Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Jean Sévigny
- 3Centre de recherche du CHU de Québec–Université Laval, Québec City, Quebec, Canada,4Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, Quebec, Canada
| | - Simon C. Robson
- 5Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - James J. Galligan
- 2Department of Pharmacology and Toxicology and Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Brian D. Gulbransen
- 1Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, Michigan
| |
Collapse
|
10
|
Park IK, Kim JH, Park CG, Kim MY, Parajuli SP, Hong CS, Choi S, Jun JY. Effects of ATP on Pacemaker Activity of Interstitial Cells of Cajal from the Mouse Small Intestine. Chonnam Med J 2018; 54:63-71. [PMID: 29399568 PMCID: PMC5794481 DOI: 10.4068/cmj.2018.54.1.63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 01/23/2023] Open
Abstract
Purinergic receptors play an important role in regulating gastrointestinal (GI) motility. Interstitial cells of Cajal (ICCs) are pacemaker cells that regulate GI smooth muscle activity. We studied the functional roles of external adenosine 5′-triphosphate (ATP) on pacemaker activity in cultured ICCs from mouse small intestines by using the whole-cell patch clamp technique and intracellular Ca2+ ([Ca2+]i) imaging. External ATP dose-dependently depolarized the resting membrane and produced tonic inward pacemaker currents, and these effects were antagonized by suramin, a purinergic P2 receptor antagonist. ATP-induced effects on pacemaker currents were suppressed by an external Na+-free solution and inhibited by the nonselective cation channel blockers, flufenamic acid and niflumic acid. The removal of external Ca2+ or treatment with thapsigargin (inhibitor of Ca2+ uptake into endoplasmic reticulum) inhibited the ATP-induced effects on pacemaker currents. Spontaneous [Ca2+]i oscillations were enhanced by external ATP. These results suggest that external ATP modulates pacemaker activity by activating nonselective cation channels via external Ca2+ influx and [Ca2+]i release from the endoplasmic reticulum. Thus, it seems that activating the purinergic P2 receptor may modulate GI motility by acting on ICCs in the small intestine.
Collapse
Affiliation(s)
- Il Koo Park
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Jin Ho Kim
- Department of Neurology, College of Medicine, Chosun University, Gwangju, Korea
| | - Chan Guk Park
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Man Yoo Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | | | - Chan Sik Hong
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Seok Choi
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Jae Yeoul Jun
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| |
Collapse
|
11
|
Lee SM, Kim N, Jo HJ, Park JH, Nam RH, Lee HS, Kim HJ, Lee MY, Kim YS, Lee DH. Comparison of Changes in the Interstitial Cells of Cajal and Neuronal Nitric Oxide Synthase-positive Neuronal Cells With Aging Between the Ascending and Descending Colon of F344 Rats. J Neurogastroenterol Motil 2017; 23:592-605. [PMID: 28774159 PMCID: PMC5628993 DOI: 10.5056/jnm17061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/20/2017] [Accepted: 07/09/2017] [Indexed: 12/22/2022] Open
Abstract
Background/Aims Neuronal degeneration and changes in interstitial cells of Cajal (ICCs) are important mechanisms of age-related constipation. This study aims to compare the distribution of ICCs and neuronal nitric oxide synthase (nNOS) with regard to age-related changes between the ascending colon (AC) and descending colon (DC) in 6-, 31-, and 74-week old and 2-year old male Fischer-344 rats. Methods The amount of fecal pellet and the bead expulsion times were measured. Fat proportion in the muscle layer of the colon was analyzed by hematoxylin and eosin staining. Proto-oncogene receptor tyrosine kinase (KIT) and neuronal nitric oxide synthase (nNOS) expression were analyzed with Western blotting and immunohistochemistry. Isovolumetric contractile measurements and electrical field stimulation were used to assess smooth muscle contractility. Results Colon transit and bead expulsion slowed with senescence. Fat in the muscle layer accumulated with age in the AC, but not in the DC. The proportion of KIT-immunoreactive ICCs in the submucosal and myenteric plexus was higher in the DC than in the AC, and it declined with age, especially in the AC. In contrast, the proportion of NOS-immunoreactive neurons in the myenteric plexus was higher in the AC than in the DC, and both decreased in older rats. Nitric oxide levels declined with age in the DC. Muscle strip experiments showed that the inhibitory response mediated by nitric oxide in the circular direction of the DC was reduced in 2-year old rats. Conclusion The AC and DC differ in their distribution of ICCs and nNOS, and age-related loss of nitrergic neurons more severely affects the DC than the AC.
Collapse
Affiliation(s)
- Sun Min Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Jin Jo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Hyun Jin Kim
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Jinju, Gyeongsangnam-do, Korea
| | - Moon Young Lee
- Department of Physiology and Institute of Wonkwang Medical Science, Wonkwang University College of Medicine, Iksan, Jeollabuk-do, Korea
| | - Yong Sung Kim
- Division of Gastroenterology and Wonkwang Digestive Disease Research Institute, Department of Internal Medicine, Wonkwang University Sanbon Hospital, Gunpo, Gyeonggi-do, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| |
Collapse
|
12
|
Zhou J, O'Connor MD, Ho V. The Potential for Gut Organoid Derived Interstitial Cells of Cajal in Replacement Therapy. Int J Mol Sci 2017; 18:ijms18102059. [PMID: 28954442 PMCID: PMC5666741 DOI: 10.3390/ijms18102059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/15/2017] [Accepted: 09/24/2017] [Indexed: 12/24/2022] Open
Abstract
Effective digestion requires propagation of food along the entire length of the gastrointestinal tract. This process involves coordinated waves of peristalsis produced by enteric neural cell types, including different categories of interstitial cells of Cajal (ICC). Impaired food transport along the gastrointestinal tract, either too fast or too slow, causes a range of gut motility disorders that affect millions of people worldwide. Notably, loss of ICC has been shown to affect gut motility. Patients that suffer from gut motility disorders regularly experience diarrhoea and/or constipation, insomnia, anxiety, attention lapses, irritability, dizziness, and headaches that greatly affect both physical and mental health. Limited treatment options are available for these patients, due to the scarcity of human gut tissue for research and transplantation. Recent advances in stem cell technology suggest that large amounts of rudimentary, yet functional, human gut tissue can be generated in vitro for research applications. Intriguingly, these stem cell-derived gut organoids appear to contain functional ICC, although their frequency and functional properties are yet to be fully characterised. By reviewing methods of gut organoid generation, together with what is known of the molecular and functional characteristics of ICC, this article highlights short- and long-term goals that need to be overcome in order to develop ICC-based therapies for gut motility disorders.
Collapse
Affiliation(s)
- Jerry Zhou
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia.
- Medical Sciences Research Group, Western Sydney University, Campbelltown, NSW 2560, Australia.
| | - Michael D O'Connor
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia.
- Medical Sciences Research Group, Western Sydney University, Campbelltown, NSW 2560, Australia.
| | - Vincent Ho
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia.
- Medical Sciences Research Group, Western Sydney University, Campbelltown, NSW 2560, Australia.
| |
Collapse
|
13
|
Słupecka M, Grzesiak P, Kwiatkowski J, Gajewska M, Kuwahara A, Kato I, Woliński J. The influence of enteral obestatin administration to suckling rats on intestinal contractility. Gen Comp Endocrinol 2017; 248:69-78. [PMID: 28212895 DOI: 10.1016/j.ygcen.2017.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 01/24/2017] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
This study investigated the effect of enteral administration of obestatin on the contractility of whole-thickness preparations of duodenum and middle jejunum, as well as on the morphology of the enteric nervous system (ENS). Suckling rats were assigned to 3 groups (n=12) treated with: C-saline solution; LO-obestatin (125nmol/kgb.wt); HO-obestatin (250nmol/kgb.wt). Saline solution or obestatin were administered twice daily, from the 14th to the 21st day of life. Sections were studied in an organ bath, for isometric recording in the presence of acetylocholine (ACh), atropine (ATR) and tetradotoxin (TTX). Thickness of intestinal muscularis layer, the number of interstitial cells of Cajal (ICC) were measured in the paraffin sections. The immunodetection of Muscarinic Acetylocholine Receptor 2 (M2 receptor) was performed in the intestinal segments. In both intestinal segments HO treatment decreased the amplitude of spontaneous contraction compared to that observed in the C group. In the middle jejunum, the LO treatment also decreased the amplitude. TTX and ATR had no effect on amplitude of spontaneous contraction in the jejunum of LO and HO-treated animals. Compared to the C group, duodenal sections from HO animals and middle jejunum sections from LO and HO groups displayed a lower amplitude in response to ACh and EFS evoked contraction. An increase in the thickness of the muscularis layer was observed in the duodenum of LO and HO groups whereas the number ICC did not change significantly after treatment with obestatin. Moreover, the enteral administration of obestatin did not effect significantly on the cytoplasmic expression of M2 receptor in the jejunum. Our study demonstrated that enteral administration of obestatin to suckling rats influences small intestine contractility in the segment specific manner.
Collapse
Affiliation(s)
- M Słupecka
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland.
| | - P Grzesiak
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - J Kwiatkowski
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - M Gajewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - A Kuwahara
- Laboratory of Physiology, Institute for Environmental Sciences and Graduate School of Nutritional and Enviromental Science, University of Shizuoka, Japan
| | - I Kato
- Department of Medical Biochemistry, Kobe Pharmaceutical University, Japan
| | - J Woliński
- Department of Animal Physiology, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| |
Collapse
|
14
|
Purinergic Signalling in the Gut. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 891:91-112. [PMID: 27379638 DOI: 10.1007/978-3-319-27592-5_10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The article will begin with the discovery of purinergic inhibitory neuromuscular transmission in the 1960s/1970s, the proposal for purinergic cotransmission in 1976 and the recognition that sympathetic nerves release adenosine 5'-triphosphate (ATP), noradrenaline and neuropeptide Y, while non-adrenergic, non-cholinergic inhibitory nerve cotransmitters are ATP, nitric oxide and vasoactive intestinal polypeptide in variable proportions in different regions of the gut. Later, purinergic synaptic transmission in the myenteric and submucosal plexuses was established and purinergic receptors expressed by both glial and interstitial cells. The focus will then be on purinergic mechanosensory transduction involving release of ATP from mucosal epithelial cells during distension to activate P2X3 receptors on submucosal sensory nerve endings. The responses of low threshold fibres mediate enteric reflex activity via intrinsic sensory nerves, while high threshold fibres initiate pain via extrinsic sensory nerves. Finally, the involvement of purinergic signalling in an animal model of colitis will be presented, showing that during distension there is increased ATP release, increased P2X3 receptor expression on calcitonin gene-related peptide-labelled sensory neurons and increased sensory nerve activity.
Collapse
|
15
|
Change in the Interstitial Cells of Cajal and nNOS Positive Neuronal Cells with Aging in the Stomach of F344 Rats. PLoS One 2017; 12:e0169113. [PMID: 28045993 PMCID: PMC5207530 DOI: 10.1371/journal.pone.0169113] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/11/2016] [Indexed: 12/22/2022] Open
Abstract
The gastric accommodation reflex is an important mechanism in gastric physiology. However, the aging-associated structural and functional changes in gastric relaxation have not yet been established. Thus, we evaluated the molecular changes of interstitial cell of Cajal (ICC) and neuronal nitric oxide synthase (nNOS) and the function changes in the corpus of F344 rats at different ages (6-, 31-, 74-wk and 2-yr). The proportion of the c-Kit-positive area in the submucosal border (SMB) and myenteric plexus (MP) layer was significantly lower in the older rats, as indicated by immunohistochemistry. The density of the nNOS-positive immunoreactive area also decreased with age in the SMB, circular muscle (CM), and MP. Similarly, the percent of nNOS-positive neuronal cells per total neuronal cells and the proportion of nNOS immunoreactive area of MP also decreased in aged rats. In addition, the mRNA and protein expression of c-Kit and nNOS significantly decreased with age. Expression of stem cell factor (SCF) and the pan-neuronal marker PGP 9.5 mRNA was significantly lower in the older rats than in the younger rats. Barostat studies showed no difference depending on age. Instead, the change of volume was significantly decreased by L-NG63-nitroarginine methyl ester in the 2-yr-old rats compared with the 6-wk-old rats (P = 0.003). Taken together, the quantitative and molecular nNOS changes in the stomach might play a role in the decrease of gastric accommodation with age.
Collapse
|
16
|
Zhu YF, Wang XY, Parsons SP, Huizinga JD. Stimulus-induced pacemaker activity in interstitial cells of Cajal associated with the deep muscular plexus of the small intestine. Neurogastroenterol Motil 2016; 28:1064-74. [PMID: 26968691 DOI: 10.1111/nmo.12808] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 02/01/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND The ICC-DMP have been proposed to generate stimulus-dependent pacemaker activity, rhythmic transient depolarizations, that take part in orchestrating segmentation and clustered propulsive motor patterns in the small intestine. However, little is known about the fundamental properties of ICC-DMP. METHODS This study was undertaken to increase our understanding of intrinsic properties of the ICC-DMP through calcium imaging and intracellular electrical recordings. KEY RESULTS Without stimulation, most ICC-DMP were quiescent. In some preparations ICC-DMP generated rhythmic low-frequency calcium oscillations (<10 cpm) with or without high frequency activity superimposed (>35 cpm). Immunohistochemistry proved the existence of NK1R on the ICC-DMP and close contacts between ICC-DMP and substance P-positive nerves. Substance P (25 nM) induced low-frequency calcium oscillations that were synchronized across the ICC-DMP network. Substance P also induced low frequency rhythmic transient depolarizations (<10cpm) in circular muscle cells close to the ICC-DMP. An intracellular recording from a positively identified ICC-DMP showed rhythmic transient depolarizations with superimposed high frequency activity. To investigate if quiescent ICC-DMP were chronically inhibited by nitrergic activity, nNOS was inhibited, but without effect. CONCLUSIONS & INFERENCES Substance P changes non-synchronized high frequency flickering or quiescence in ICC-DMP into strong rhythmic calcium transients that are synchronized within the network; they are associated with rhythmic transient depolarizations within the same frequency range. We hypothesize that Substance P, released from nerves, can evoke rhythmicity in ICC-DMP, thereby providing it with potential pacemaker activity.
Collapse
Affiliation(s)
- Y F Zhu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - X-Y Wang
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - S P Parsons
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - J D Huizinga
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
17
|
Baker SA, Drumm BT, Saur D, Hennig GW, Ward SM, Sanders KM. Spontaneous Ca(2+) transients in interstitial cells of Cajal located within the deep muscular plexus of the murine small intestine. J Physiol 2016; 594:3317-38. [PMID: 26824875 DOI: 10.1113/jp271699] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/24/2016] [Indexed: 01/13/2023] Open
Abstract
KEY POINTS Interstitial cells of Cajal at the level of the deep muscular plexus (ICC-DMP) in the small intestine generate spontaneous Ca(2+) transients that consist of localized Ca(2+) events and limited propagating Ca(2+) waves. Ca(2+) transients in ICC-DMP display variable characteristics: from discrete, highly localized Ca(2+) transients to regionalized Ca(2+) waves with variable rates of occurrence, amplitude, duration and spatial spread. Ca(2+) transients fired stochastically, with no cellular or multicellular rhythmic activity being observed. No correlation was found between the firing sites in adjacent cells. Ca(2+) transients in ICC-DMP are suppressed by the ongoing release of inhibitory neurotransmitter(s). Functional intracellular Ca(2+) stores are essential for spontaneous Ca(2+) transients, and the sarco/endoplasmic reticulum Ca(2+) -ATPase (SERCA) pump is necessary for maintenance of spontaneity. Ca(2+) release mechanisms involve both ryanodine receptors (RyRs) and inositol triphosphate receptors (InsP3 Rs). Release from these channels is interdependent. ICC express transcripts of multiple RyRs and InsP3 Rs, with Itpr1 and Ryr2 subtypes displaying the highest expression. ABSTRACT Interstitial cells of Cajal in the deep muscular plexus of the small intestine (ICC-DMP) are closely associated with varicosities of enteric motor neurons and generate responses contributing to neural regulation of intestinal motility. Responses of ICC-DMP are mediated by activation of Ca(2+) -activated Cl(-) channels; thus, Ca(2+) signalling is central to the behaviours of these cells. Confocal imaging was used to characterize the nature and mechanisms of Ca(2+) transients in ICC-DMP within intact jejunal muscles expressing a genetically encoded Ca(2+) indicator (GCaMP3) selectively in ICC. ICC-DMP displayed spontaneous Ca(2+) transients that ranged from discrete, localized events to waves that propagated over variable distances. The occurrence of Ca(2+) transients was highly variable, and it was determined that firing was stochastic in nature. Ca(2+) transients were tabulated in multiple cells within fields of view, and no correlation was found between the events in adjacent cells. TTX (1 μm) significantly increased the occurrence of Ca(2+) transients, suggesting that ICC-DMP contributes to the tonic inhibition conveyed by ongoing activity of inhibitory motor neurons. Ca(2+) transients were minimally affected after 12 min in Ca(2+) free solution, indicating these events do not depend immediately upon Ca(2+) influx. However, inhibitors of sarco/endoplasmic reticulum Ca(2+) -ATPase (SERCA) pump and blockers of inositol triphosphate receptor (InsP3 R) and ryanodine receptor (RyR) channels blocked ICC Ca(2+) transients. These data suggest an interdependence between RyR and InsP3 R in the generation of Ca(2+) transients. Itpr1 and Ryr2 were the dominant transcripts expressed by ICC. These findings provide the first high-resolution recording of the subcellular Ca(2+) dynamics that control the behaviour of ICC-DMP in situ.
Collapse
Affiliation(s)
- Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Dieter Saur
- II. Medizinische Klinik und Poliklinik, Klinikum rechts der Isar der TU München, München, Germany
| | - Grant W Hennig
- 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
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
18
|
Sanders KM, Ward SM, Friebe A. Rebuttal from Kenton M. Sanders, Sean M. Ward and Andreas Friebe. J Physiol 2016; 594:1515. [PMID: 26842221 DOI: 10.1113/jp271971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Kenton M Sanders
- 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
| | - Andreas Friebe
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
19
|
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: 347] [Impact Index Per Article: 31.5] [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
|
20
|
Kurahashi M, Mutafova-Yambolieva V, Koh SD, Sanders KM. Platelet-derived growth factor receptor-α-positive cells and not smooth muscle cells mediate purinergic hyperpolarization in murine colonic muscles. Am J Physiol Cell Physiol 2014; 307:C561-70. [PMID: 25055825 DOI: 10.1152/ajpcell.00080.2014] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Enteric inhibitory neurotransmission is an important feature of the neural regulation of gastrointestinal motility. Purinergic neurotransmission, via P2Y1 receptors, mediates one phase of inhibitory neural control. For decades, ATP has been assumed to be the purinergic neurotransmitter and smooth muscle cells (SMCs) have been considered the primary targets for inhibitory neurotransmission. Recent experiments have cast doubt on both of these assumptions and suggested that another cell type, platelet-derived growth factor receptor-α-positive (PDGFRα(+)) cells, is the target for purinergic neurotransmission. We compared responses of PDGFRα(+) cells and SMCs to several purine compounds to determine if these cells responded in a manner consistent with enteric inhibitory neurotransmission. ATP hyperpolarized PDGFRα(+) cells but depolarized SMCs. Only part of the ATP response in PDGFRα(+) cells was blocked by MRS 2500, a P2Y1 antagonist. ADP, MRS 2365, β-NAD, and adenosine 5-diphosphate-ribose, P2Y1 agonists, hyperpolarized PDGFRα(+) cells, and these responses were blocked by MRS 2500. Adenosine 5-diphosphate-ribose was more potent in eliciting hyperpolarization responses than β-NAD. P2Y1 agonists failed to elicit responses in SMCs. Small hyperpolarization responses were elicited in SMCs by a small-conductance Ca(2+)-activated K(+) channel agonist, cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine, consistent with the low expression and current density of small-conductance Ca(2+)-activated K(+) channels in these cells. Large-amplitude hyperpolarization responses, elicited in PDGFRα(+) cells, but not SMCs, by P2Y1 agonists are consistent with the generation of inhibitory junction potentials in intact muscles in response to purinergic neurotransmission. The responses of PDGFRα(+) cells and SMCs to purines suggest that SMCs are unlikely targets for purinergic neurotransmission in colonic muscles.
Collapse
Affiliation(s)
- Masaaki Kurahashi
- 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
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
21
|
Márquez S, Galotta JM, Gálvez GA, Portiansky E, Barbeito CG. Presence of c-kit positive cells in fetal and adult bovine forestomachs. Biotech Histochem 2014; 89:591-601. [DOI: 10.3109/10520295.2014.919023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
22
|
Jo HJ, Kim N, Nam RH, Kang JM, Kim JH, Choe G, Lee HS, Park JH, Chang H, Kim H, Lee MY, Kim YS, Kim JS, Jung HC. Fat deposition in the tunica muscularis and decrease of interstitial cells of Cajal and nNOS-positive neuronal cells in the aged rat colon. Am J Physiol Gastrointest Liver Physiol 2014; 306:G659-69. [PMID: 24525022 DOI: 10.1152/ajpgi.00304.2012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Little is known about the time course of aging on interstitial cells of Cajal (ICC) of colon. The aim of this study was to investigate the change of morphology, ICC, and neuronal nitric oxide synthase (nNOS)-immunoreactive cells in the aged rat. The proximal colon of 344 Fischer rats at four different ages (6, 31, 74 wk, and 2 yr) were studied. The immunoreactivity of c-Kit, nNOS, anti-protein gene product 9.5, and synaptophysin were counted after immunohistochemistry. The c-kit, stem cell factor (ligand of Kit), and nNOS mRNA were measured by real-time PCR. c-Kit and nNOS protein were assessed by Western blot. Isovolumetric contractile force measurement and electrical field stimulation (EFS) were conducted. The area of intramuscular fat deposition significantly increased with age after 31 wk. c-Kit-immunoreactive ICC and nNOS-immunoreactive neurons and nerve fibers significantly declined with age. mRNA and protein expression of c-kit and nNOS decreased with aging. The functional study showed that the spontaneous contractility was decreased in aged rat, whereas EFS responses in the presence of atropine and L-NG-Nitroarginine methyl ester were increased in aged rat. In conclusion, the decrease of proportion of proper smooth muscle, the density of ICC and nNOS-immunoreactive neuronal fibers, and the number of nNOS-immunoreactive neurons during the aging process may explain the aging-associated colonic dysmotility.
Collapse
|
23
|
Burnstock G. Purinergic signalling in the gastrointestinal tract and related organs in health and disease. Purinergic Signal 2014; 10:3-50. [PMID: 24307520 PMCID: PMC3944042 DOI: 10.1007/s11302-013-9397-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/24/2013] [Indexed: 01/04/2023] Open
Abstract
Purinergic signalling plays major roles in the physiology and pathophysiology of digestive organs. Adenosine 5'-triphosphate (ATP), together with nitric oxide and vasoactive intestinal peptide, is a cotransmitter in non-adrenergic, non-cholinergic inhibitory neuromuscular transmission. P2X and P2Y receptors are widely expressed in myenteric and submucous enteric plexuses and participate in sympathetic transmission and neuromodulation involved in enteric reflex activities, as well as influencing gastric and intestinal epithelial secretion and vascular activities. Involvement of purinergic signalling has been identified in a variety of diseases, including inflammatory bowel disease, ischaemia, diabetes and cancer. Purinergic mechanosensory transduction forms the basis of enteric nociception, where ATP released from mucosal epithelial cells by distension activates nociceptive subepithelial primary afferent sensory fibres expressing P2X3 receptors to send messages to the pain centres in the central nervous system via interneurons in the spinal cord. Purinergic signalling is also involved in salivary gland and bile duct secretion.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| |
Collapse
|
24
|
Tanahashi Y, Ichimura Y, Kimura K, Matsuyama H, Iino S, Komori S, Unno T. Cholinergic neuromuscular transmission mediated by interstitial cells of Cajal in the myenteric layer in mouse ileal longitudinal smooth muscles. Naunyn Schmiedebergs Arch Pharmacol 2013; 387:377-88. [PMID: 24322587 DOI: 10.1007/s00210-013-0944-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/21/2013] [Indexed: 01/15/2023]
Abstract
To elucidate the roles played by the interstitial cells of Cajal in the myenteric layer (ICC-MY) in cholinergic neuromuscular transmission, we recorded mechanical and electrical activities in response to electrical field stimulation (EFS) of the ileal longitudinal muscle strips from WBB6F1-W/W(V) (W/W(V)) mutant mice, that lacked ICC-MY and compared with those in WBB6F1-+/+ (+/+) control mice. In +/+ muscle strips, EFS induced phasic contractions, which were abolished or strongly attenuated by atropine or tetrodotoxin. In W/W(V) preparations, EFS induced similar phasic contractions, but the cholinergic component was smaller than that in +/+ strips. This was despite of the fact that the contractions because of exogenous applications of carbachol and high K(+) solution in W/W(V) strips were comparable to or rather greater than those in the +/+ preparations. EFS induced atropine-sensitive excitatory junction potentials (EJPs) in the +/+ longitudinal smooth muscle cells but not in W/W(V) cells. In the presence of eserine, EFS induced atropine-sensitive EJPs in W/W(V) cells. These results suggest that ICC-MY mediate the cholinergic neuromuscular transmission in mouse ileal longitudinal smooth muscles. In addition, the other pathway in which ICC-MY are not involved can operate concomitantly.
Collapse
Affiliation(s)
- Yasuyuki Tanahashi
- Department of Animal Medical Sciences, Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-Ku, Kyoto, 603-8555, Japan,
| | | | | | | | | | | | | |
Collapse
|
25
|
Matsumoto K, Nakajima T, Sakai H, Kato S, Sagara A, Arakawa K, Tashima K, Narita M, Horie S. Increased expression of 5-HT3 and NK 1 receptors in 5-fluorouracil-induced mucositis in mouse jejunum. Dig Dis Sci 2013; 58:3440-51. [PMID: 23695873 DOI: 10.1007/s10620-013-2709-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 04/26/2013] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Although 5-fluorouracil (5-FU) is a widely used as chemotherapy agent, severe mucositis develops in approximately 80% of patients. 5-FU-induced small intestinal mucositis can cause nausea and vomiting. The current study was designed to investigate peripheral alterations due to the 5-FU-induced mucositis of neuronal and non-neuronal 5-HT3 and NK1 receptor expression by immunohistochemical analysis. METHODS 5-FU was administered by i.p. injection to C57BL/6 mice. After 4 days, segments of the jejunum were removed. The specimens were analyzed by immunohistochemistry, real-time PCR, and enzyme immunoassay. RESULTS The numbers of 5-HT3 receptor immunopositive cells and nerve fibers in mucosa were increased by 5-FU treatment. The 5-HT3 receptor immunopositive cell bodies were found only in jejunal submucosa and myenteric plexus in the 5-FU-treated mice. The numbers of NK1 receptor cells in mucosa and immunopositive expression of NK1 receptors in deep muscular plexus were dramatically increased in 5-FU-treated mice. Real-time PCR demonstrated that 5-FU treatment significantly increased mRNA levels of 5-HT3A, 5-HT3B, and NK1 receptors. The amounts of 5-HT and substance P increased after 5-FU treatment. The 5-HT3 or NK1 receptor immunopositive cells colocalized with both 5-HT and substance P. Furthermore, 5-HT3 and NK1 receptors colocalized with CD11b. CONCLUSIONS The 5-HT3 and NK1 immunopositive macrophages and mucosal mast cells in lamina propria release 5-HT and substance P, which in turn activate their corresponding receptors on mucosal cells in autocrine and paracrine manners. It is assumed to result in the release of 5-HT and substance P in mucosa.
Collapse
Affiliation(s)
- Kenjiro Matsumoto
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba, 283-8555, Japan,
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Interstitial cells of Cajal integrate excitatory and inhibitory neurotransmission with intestinal slow-wave activity. Nat Commun 2013; 4:1630. [PMID: 23535651 DOI: 10.1038/ncomms2626] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/20/2013] [Indexed: 12/27/2022] Open
Abstract
The enteric nervous system contains excitatory and inhibitory neurons, which control contraction and relaxation of smooth muscle cells as well as gastrointestinal motor activity. Little is known about the exact cellular mechanisms of neuronal signal transduction to smooth muscle cells in the gut. Here we generate a c-Kit(CreERT2) knock-in allele to target a distinct population of pacemaker cells called interstitial cells of Cajal. By genetic loss-of-function studies, we show that interstitial cells of Cajal, which generate spontaneous electrical slow waves and thus rhythmic contractions of the smooth musculature, are essential for transmission of signals from enteric neurons to gastrointestinal smooth muscle cells. Interstitial cells of Cajal, therefore, integrate excitatory and inhibitory neurotransmission with slow-wave activity to orchestrate peristaltic motor activity of the gut. Impairment of the function of interstitial cells of Cajal causes severe gastrointestinal motor disorders. The results of our study show at the genetic level that these disorders are not only due to loss of slow-wave activity but also due to disturbed neurotransmission.
Collapse
|
27
|
Koh SD, Rhee PL. Ionic Conductance(s) in Response to Post-junctional Potentials. J Neurogastroenterol Motil 2013; 19:426-32. [PMID: 24199003 PMCID: PMC3816177 DOI: 10.5056/jnm.2013.19.4.426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 08/28/2013] [Accepted: 08/30/2013] [Indexed: 01/29/2023] Open
Abstract
The gastrointestinal motility is regulated by extrinsic and intrinsic neural regulation. Intrinsic neural pathways are controlled by sensory input, inter-neuronal relay and motor output. Enteric motor neurons release many transmitters which affect post-junctional responses. Post-junctional responses can be excitatory and inhibitory depending on neurotransmitters. Excitatory neurotransmitters induce depolarization and contraction. In contrast, inhibitory neurotransmitters hyperpolarize and relaxe the gastrointestinal smooth muscle. Smooth muscle syncytium is composed of smooth muscle cells, interstitial cells of Cajal and platelet-derived growth factor receptor α-positive (PDGFRα(+)) cells (SIP syncytium). Specific expression of receptors and ion channels in these cells can be affected by neurotransmitters. In recent years, molecular reporter expression techniques are able to study the properties of ion channels and receptors in isolated specialized cells. In this review, we will discuss the mechanisms of ion channels to interpret the post-junctional responses in the gastrointestinal smooth muscles.
Collapse
Affiliation(s)
- Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada, School of Medicine, Reno, NV, USA
| | | |
Collapse
|
28
|
Al-Shboul OA. The importance of interstitial cells of cajal in the gastrointestinal tract. Saudi J Gastroenterol 2013; 19:3-15. [PMID: 23319032 PMCID: PMC3603487 DOI: 10.4103/1319-3767.105909] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Accepted: 10/09/2012] [Indexed: 12/13/2022] Open
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.
| |
Collapse
|
29
|
Furuya S, Furuya K. Roles of substance P and ATP in the subepithelial fibroblasts of rat intestinal villi. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 304:133-89. [PMID: 23809436 DOI: 10.1016/b978-0-12-407696-9.00003-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ingestion of food and water induces chemical and mechanical signals that trigger peristaltic reflexes and also villous movement in the gut. In the intestinal villi, subepithelial fibroblasts under the epithelium form contractile cellular networks and closely contact to the varicosities of substance P and nonsubstance P afferent neurons. Subepithelial fibroblasts of the duodenal villi possess purinergic receptor P2Y1 and tachykinin receptor NK1. ATP and substance P induce increase in intracellular Ca(2+) and cell contraction in subepithelial fibroblasts. They are highly mechanosensitive and release ATP by mechanical stimuli. Released ATP spreads to form an ATP "cloud" with nearly 1μM concentration and activates the surroundings via P2Y1 and afferent neurons via P2X receptors. These findings suggest that villous subepithelial fibroblasts and afferent neurons interact via ATP and substance P. This mutual interaction may play important roles in the signal transduction of mechano reflex pathways including a coordinate villous movement and also in the maturation of the structure and function of the intestinal villi.
Collapse
Affiliation(s)
- Sonoko Furuya
- Section of Brain Structure Information, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan.
| | | |
Collapse
|
30
|
Yu W, Zeidel ML, Hill WG. Cellular expression profile for interstitial cells of cajal in bladder - a cell often misidentified as myocyte or myofibroblast. PLoS One 2012; 7:e48897. [PMID: 23145014 PMCID: PMC3492220 DOI: 10.1371/journal.pone.0048897] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 10/03/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Interstitial cells of Cajal (ICC) have been identified in urinary bladder of several species, but their presence in mice remains uncertain. Meanwhile, dozens of reports indicate that dysregulation of connexin 43 plays an important role in bladder overactivity, but its localization has not been clearly defined, with reports of expression in either the smooth muscle or in myofibroblasts. We recently identified a population of ectonucleoside triphosphate diphosphohydrolase 2 (NTPDase2) positive cells that resemble ICC and are distinct from smooth muscle, fibroblasts, myofibroblasts and neurons. Thus we sought to define more clearly the molecular signature of ICC and in doing so resolve some of these uncertainties. PRINCIPLE FINDINGS Immunofluorescent localization revealed that NTPDase2-positive cells lie closely adjacent to smooth muscle but are separate from them. NTPDase2 positive cells exhibited co-localization with the widely accepted ICC marker - c-kit. They were further shown to co-localize with other ICC markers CD34 and Ano1, but not with mast cell marker tryptase. Significantly, they show convincing co-localization with connexin 43, which was not present in smooth muscle. The identity of these cells as ICC was further confirmed by the presence of three mesenchymal markers - vimentin, desmin, and PDGFβ receptor, which indicates their mesenchymal origin. Finally, we observed for the first time, the presence of merlin/neurofibromin 2 in ICC. Normally considered a neuronal protein, the presence of merlin suggests ICC in bladder may have a role in neurotransmission. CONCLUSIONS NTPDase2 positive cells in mice bladder are ICC, which can be defined by the presence of c-Kit, CD34, Ano1, NTPDase2, connexin 43, vimentin, desmin, PDGFβ receptor and merlin/NF2. These data establish a definitive molecular expression profile, which can be used to assist in explorations of their functional roles, and the presence of NTPDase2 suggests that purinergic signaling plays a role in regulation of ICC function.
Collapse
Affiliation(s)
- Weiqun Yu
- Laboratory of Voiding Dysfunction, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
31
|
Gulbransen BD, Sharkey KA. Novel functional roles for enteric glia in the gastrointestinal tract. Nat Rev Gastroenterol Hepatol 2012; 9:625-32. [PMID: 22890111 DOI: 10.1038/nrgastro.2012.138] [Citation(s) in RCA: 271] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Enteric glia are a unique class of peripheral glial cells within the gastrointestinal tract. Major populations of enteric glia are found in enteric ganglia in the myenteric and submucosal plexuses of the enteric nervous system (ENS); these cells are also found outside of the ENS, within the circular muscle and in the lamina propria of the mucosa. These different populations of cells probably represent unique classes of glial cells with differing functions. In the past few years, enteric glia have been found to be involved in almost every gut function including motility, mucosal secretion and host defence. Subepithelial glia seem to have a trophic and supporting relationship with intestinal epithelial cells, but the necessity of these roles in the maintenance of normal epithelial functions remains to be shown. Likewise, glia within enteric ganglia are activated by synaptic stimulation, suggesting an active role in synaptic transmission, but the precise role of glial activation in normal enteric network activity is unclear. Excitingly, enteric glia can also give rise to new neurons, but seemingly only under limited circumstances. In this Review, we discuss the current body of evidence supporting functional roles of enteric glia and identify key gaps in our understanding of the physiology of these unique cells.
Collapse
Affiliation(s)
- Brian D Gulbransen
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive North West Calgary, AB T2N 4N1, Canada
| | | |
Collapse
|
32
|
Sanders KM, Koh SD, Ro S, Ward SM. Regulation of gastrointestinal motility--insights from smooth muscle biology. Nat Rev Gastroenterol Hepatol 2012; 9:633-645. [PMID: 22965426 PMCID: PMC4793911 DOI: 10.1038/nrgastro.2012.168] [Citation(s) in RCA: 298] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gastrointestinal motility results from coordinated contractions of the tunica muscularis, the muscular layers of the alimentary canal. Throughout most of the gastrointestinal tract, smooth muscles are organized into two layers of circularly or longitudinally oriented muscle bundles. Smooth muscle cells form electrical and mechanical junctions between cells that facilitate coordination of contractions. Excitation-contraction coupling occurs by Ca(2+) entry via ion channels in the plasma membrane, leading to a rise in intracellular Ca(2+). Ca(2+) binding to calmodulin activates myosin light chain kinase; subsequent phosphorylation of myosin initiates cross-bridge cycling. Myosin phosphatase dephosphorylates myosin to relax muscles, and a process known as Ca(2+) sensitization regulates the activity of the phosphatase. Gastrointestinal smooth muscles are 'autonomous' and generate spontaneous electrical activity (slow waves) that does not depend upon input from nerves. Intrinsic pacemaker activity comes from interstitial cells of Cajal, which are electrically coupled to smooth muscle cells. Patterns of contractile activity in gastrointestinal muscles are determined by inputs from enteric motor neurons that innervate smooth muscle cells and interstitial cells. Here we provide an overview of the cells and mechanisms that generate smooth muscle contractile behaviour and gastrointestinal motility.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Anderson Medical Sciences, Reno, NV 89557, USA.
| | | | | | | |
Collapse
|
33
|
Ball ER, Matsuda MM, Dye L, Hoffmann V, Zerfas PM, Szarek E, Rich A, Chitnis AB, Stratakis CA. Ultra-structural identification of interstitial cells of Cajal in the zebrafish Danio rerio. Cell Tissue Res 2012; 349:483-91. [PMID: 22628160 PMCID: PMC3674513 DOI: 10.1007/s00441-012-1434-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 04/12/2012] [Indexed: 11/27/2022]
Abstract
The interstitial cells of Cajal (ICCs) are important mediators of gastrointestinal (GI) motility because of their role as pacemakers in the GI tract. In addition to their function, ICCs are also structurally distinct cells most easily identified by their ultra-structural features and expression of the tyrosine kinase receptor c-KIT. ICCs have been described in mammals, rodents, birds, reptiles, and amphibians, but there are no reports at the ultra-structural level of ICCs within the GI tract of an organism from the teleost lineage. We describe the presence of cells in the muscularis of the zebrafish intestine; these cells have similar features to ICCs in other vertebrates. The ICC-like cells are associated with the muscularis, are more electron-dense than surrounding smooth muscle cells, possess long cytoplasmic processes and mitochondria, and are situated opposing enteric nervous structures. In addition, immunofluorescent and immunoelectron-microscopic studies with antibodies targeting the zebrafish ortholog of a putative ICC marker, c-KIT (kita), showed c-kit immunoreactivity in zebrafish ICCs. Taken together, these data represent the first ultra-structural characterization of cells in the muscularis of the zebrafish Danio rerio and suggest that ICC differentiation in vertebrate evolution dates back to the teleost lineage.
Collapse
Affiliation(s)
- Evan R Ball
- Section on Endocrinology & Genetics, Program on Developmental Endocrinology & Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Building 10, CRC, Room 1-3330, 10 Center Drive, MSC1103, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cole WC. ANO1-ther brick in the wall--role of Ca2+-activated Cl- channels of interstitial cells of Cajal in cholinergic motor control of gastrointestinal smooth muscle. J Physiol 2012; 589:4641-2. [PMID: 21965629 DOI: 10.1113/jphysiol.2011.218453] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- William C Cole
- The Smooth Muscle Research Group, Department of Physiology & Pharmacology,Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
35
|
Xu WD, Jiang X, Lan L, Wang CH, Tong HX, Wang BX. Long-term culture and cryopreservation of interstitial cells of Cajal. Scand J Gastroenterol 2012; 47:89-98. [PMID: 22050097 DOI: 10.3109/00365521.2011.627445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Interstitial cells of Cajal (ICCs) in the gastrointestinal tract generate and propagate slow waves and mediate neuromuscular neurotransmission. Damage to ICCs has been described in several gastrointestinal motor disorders, and although many studies have examined ICCs in culture, they have been largely limited to freshly dissociated cells or short-term cultures. An efficient and reliable method to establish a source of ICCs is much needed. The aim of this study was to investigate methods for culturing, subculturing, cryopreservation, and recovery of ICCs. METHODS ICCs were derived from intestinal segments of domestic rabbits, and immunohistochemistry for c-Kit was used to identify ICCs in culture and after recovery. Recovered ICCs were also examined for motilin receptor expression. RESULTS Optimal conditions for ICC culture and cryopreservation were based on cell growth curves and MTT assay. On the basis of these findings, recovered cells were cultured for 7 days and then sorted via flow cytometry based on c-Kit immunoreactivity. The percent of c-Kit positive cells was 64.3%, and the number of ICCs sorted was 6.7 × 10(5). Reverse-transcription polymerase chain reaction and western blotting verified motilin receptor expression in c-Kit-positive ICCs. CONCLUSIONS This is the first study to describe the culture, passage, and recovery of ICCs and to show motilin receptor expression. Our results suggest that ICCs play an important role, at least in some species, in initiating the migrating myoelectric complex induced by motilin.
Collapse
Affiliation(s)
- Wen-Da Xu
- Department of Pediatrics, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shanxi Province, China
| | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Rusu M, Pop F, Hostiuc S, Dermengiu D, Lală A, Ion D, Mănoiu V, Mirancea N. The human trigeminal ganglion: c-kit positive neurons and interstitial cells. Ann Anat 2011; 193:403-11. [DOI: 10.1016/j.aanat.2011.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 06/26/2011] [Accepted: 06/28/2011] [Indexed: 10/18/2022]
|
38
|
Kashyap P, Gomez-Pinilla PJ, Pozo MJ, Cima RR, Dozois EJ, Larson DW, Ordog T, Gibbons SJ, Farrugia G. Immunoreactivity for Ano1 detects depletion of Kit-positive interstitial cells of Cajal in patients with slow transit constipation. Neurogastroenterol Motil 2011; 23:760-5. [PMID: 21585622 PMCID: PMC3138829 DOI: 10.1111/j.1365-2982.2011.01729.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Depletion of interstitial cells of Cajal (ICC) is associated with several gastrointestinal (GI) motility disorders. Changes in ICC networks are usually detected by immunolabeling for the receptor tyrosine kinase Kit. Ano1 (DOG1 or TMEM16A) was recently described as a marker of ICC in GI tract. Our aim was to determine whether Ano1 immunoreactivity can be used as a reliable marker for ICC in tissues from patients with motility disorders. METHODS Four tissues from patients with normal ICC numbers and four tissues from patients with slow transit constipation and loss of Kit-positive ICC were studied. Interstitial cells of Cajal were detected by double labeling using antisera to Kit and Ano1. KEY RESULTS Both the processes and cell bodies of ICC in tissue from controls and slow transit constipation were immunoreactive for Ano1. There was a near complete overlap between Kit and Ano1 immunoreactivity. Tissues from patients with slow transit constipation contained significantly fewer Ano1-positive ICC than control tissues. The numbers of ICC identified by Ano1 and Kit immunoreactivity were nearly identical across the range of ICC numbers from an average of 1.64 to 7.05 cells per field and correlated with an R(2) value of 0.99. CONCLUSIONS & INFERENCES Ano1 is a reliable and sensitive marker for detecting changes in ICC networks in humans. Labeling with antibodies selective for Ano1 reproducibly detects depletion of Kit-positive ICC in tissues from patients with slow transit constipation.
Collapse
Affiliation(s)
- Purna Kashyap
- Enteric NeuroScience Program and Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Pedro Julian Gomez-Pinilla
- Enteric NeuroScience Program and Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Maria J. Pozo
- Dept of Physiology, Nursing School, University of Extremadura, Caceres, Spain
| | - Robert R. Cima
- Department of Colorectal Surgery, Mayo Clinic, Rochester, MN
| | - Eric J. Dozois
- Department of Colorectal Surgery, Mayo Clinic, Rochester, MN
| | - David W. Larson
- Department of Colorectal Surgery, Mayo Clinic, Rochester, MN
| | - Tamas Ordog
- Enteric NeuroScience Program and Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Simon J. Gibbons
- Enteric NeuroScience Program and Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Gianrico Farrugia
- Enteric NeuroScience Program and Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| |
Collapse
|
39
|
Gallego D, Gil V, Aleu J, Martinez-Cutillas M, Clavé P, Jimenez M. Pharmacological characterization of purinergic inhibitory neuromuscular transmission in the human colon. Neurogastroenterol Motil 2011; 23:792-e338. [PMID: 21585621 DOI: 10.1111/j.1365-2982.2011.01725.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND In the present study, we further characterize the purinergic receptors mediating the inhibitory junction potential (IJP) and smooth muscle relaxation in the human colon using a new, potent and selective agonist (MRS2365), and antagonists (MR2279 and MRS2500) of the P2Y(1) receptor. The P2Y(12) antagonist AR-C66096 was tested as well. Using this pharmacological approach, we tested whether β-nicotinamide adenine dinucleotide (β-NAD) fulfilled the criteria to be considered an inhibitory neurotransmitter in the human colon. METHODS We carried out muscle bath and microelectrode experiments on circular strips from the human colon and calcium imaging recordings on HEK293 cells, which constitutively express the human P2Y(1) receptor. KEY RESULTS Both the fast component of IJP and non-nitrergic relaxation was concentration-dependently inhibited by MRS2279 and MRS2500. This antagonism was confirmed in HEK293 cells. However, AR-C66096 did not modify either inhibitory response. Adenosine 5'-Ο-2-thiodiphosphate and MRS2365 caused a smooth muscle hyperpolarization and transient inhibition of spontaneous motility that was antagonized by MRS2279 and MRS2500. β-Nicotinamide adenine dinucleotide inhibited the spontaneous motility (IC(50) = 3.3 mmol L(-1) ). Nevertheless, this effect was not antagonized by high concentrations of P2Y(1) antagonists. CONCLUSIONS & INFERENCES Inhibitory purinergic neuromuscular transmission in the human colon was pharmacologically assessed by the use of new P2Y(1) receptor antagonists MRS2179, MRS2279, and MRS2500. The rank order of potency of the P2Y(1) antagonists is MRS2500 > MRS2279 > MRS2179. We found that β-NAD partially fulfills the criteria to be considered an inhibitory neurotransmitter in the human colon, but the relative contribution of each purine (ATP/ADP vsβ-NAD) requires further studies.
Collapse
Affiliation(s)
- D Gallego
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
40
|
Zhu MH, Sung IK, Zheng H, Sung TS, Britton FC, O'Driscoll K, Koh SD, Sanders KM. Muscarinic activation of Ca2+-activated Cl- current in interstitial cells of Cajal. J Physiol 2011; 589:4565-82. [PMID: 21768263 DOI: 10.1113/jphysiol.2011.211094] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Interstitial cells of Cajal (ICC) provide pacemaker activity and functional bridges between enteric motor nerve terminals and gastrointestinal smooth muscle cells. The ionic conductance(s) in ICC that are activated by excitatory neural inputs are unknown. Transgenic mice (Kit(copGFP/+)) with constitutive expression of a bright green fluorescent protein were used to investigate cellular responses of ICC to cholinergic stimulation. ICC displayed spontaneous transient inward currents (STICs) under voltage clamp that corresponded to spontaneous transient depolarizations (STDs) under current clamp. STICs reversed at 0 mV when E(Cl) = 0 mV and at -40 mV when E(Cl) was -40 mV, suggesting the STICs were due to a chloride conductance. Carbachol (CCh, 100 nm and 1 μm) induced a sustained inward current (depolarization in current clamp) and increased the amplitude and frequency of STICs and STDs. CCh responses were blocked by atropine (10 μm) or 4-DAMP (100 nm), an M(3) receptor antagonist. STDs were blocked by niflumic acid and 5-nitro-2-(3-phenylpropylamino)-benzoic acid (both 100 μm), and CCh had no effect in the presence of these drugs. The responses of intact circular muscles to CCh and stimulation of intrinsic excitatory nerves by electrical field stimulation (EFS) were also compared. CCh (1 μm) caused atropine-sensitive depolarization and increased the maximum depolarization of slow waves. Similar atropine-sensitive responses were elicited by stimulation of intrinsic excitatory neurons. Niflumic acid (100 μm) blocked responses to EFS but had minor effect on responses to exogenous CCh. These data suggest that different ionic conductances are responsible for electrical responses elicited by bath-applied CCh and cholinergic nerve stimulation.
Collapse
Affiliation(s)
- Mei Hong Zhu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Rusu MC, Pop F, Hostiuc S, Curcă GC, Streinu-Cercel A. Extrahepatic and intrahepatic human portal interstitial Cajal cells. Anat Rec (Hoboken) 2011; 294:1382-92. [PMID: 21714117 DOI: 10.1002/ar.21441] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Accepted: 05/16/2011] [Indexed: 01/18/2023]
Abstract
Portal interstitial cells of Cajal (PICCs), acting as vascular pacemakers, were previously only identified in nonhumans. Moreover, there is no evidence available about the presence of such cells within the liver. The objective of the study is to evaluate whether or not PICCs are identifiable in humans and, if they are, whether or not they are following the scaffold of portal vein (PV) branches within the liver. We obtained extrahepatic PVs and liver samples from six adult human cadavers, negative for liver disease, in accordance with ethical rules. They were stained with hematoxylin-eosin (HE) and Giemsa, and then we performed immunohistochemistry on formalin-fixed paraffin-embedded specimens for CD117/c-kit, a marker of the Cajal's cells. Immune labeling was also performed for S-100 protein, desmin, glial fibrillary acidic protein (GFAP), neurofilaments, α-smooth muscle actin (α-SMA), and CD34. c-kit-Positive PICCs were identified within the extrahepatic PV, in portal spaces, and septa. On adjacent sections, these PICCs were negative for all the other antibodies used. In conclusion, our study confirms the presence of extrahepatic PICCs on humans, which may act as a possible intrinsic pacemaker in the human PV. However, the intrahepatic PICCs, which were evidenced here for the first time, are in need for further experimental studies to evaluate their functional role. A promising further direction of the study is the PICCs role in the idiopathic portal hypertension.
Collapse
Affiliation(s)
- M C Rusu
- Discipline of Anatomy, Faculty of Dental Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.
| | | | | | | | | |
Collapse
|
42
|
Ultrastructural features and possible functional role of kit-positive interstitial cells in the guinea pig corpus cavernosum. Int J Impot Res 2011; 23:173-9. [PMID: 21654812 DOI: 10.1038/ijir.2011.27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The objective of the present study was to identify kit-positive interstitial cells (ICs) in guinea pig corpus cavernosum and examine their relationships with adjacent smooth muscle cells (SMCs) and intramural nerves. In addition, we investigated the possible involvement of ICs in nitric oxide (NO)-mediated relaxation of corpus cavernosum smooth muscle (CCSM). ICs were identified by their immunoreactivity to the kit receptor, a cell surface marker encoded by c-kit proto-oncogene and specific for interstitial cells of Cajal. ICs were abundantly distributed in guinea pig corporal tissues. Ultrastructural investigation by conventional transmission electron microscopy revealed the ultrastructural features of ICs and gap junctions located between ICs and adjacent SMCs, furthermore, a close contact between ICs and intramural nerves for the first time. Western blot analysis of purified ICs by fluorescence-activated cell sorting revealed coexpression of soluble guanylate cyclase (sGC)α1, sGCβ1 and kit receptor tyrosine kinase protein in them. These observations imply that ICs express the NO-sensitive sGC molecule and may be involved in the NO-mediated relaxation of CCSM in the guinea pig corpus cavernosum.
Collapse
|
43
|
Sanders KM, Hwang SJ, Ward SM. Neuroeffector apparatus in gastrointestinal smooth muscle organs. J Physiol 2010; 588:4621-39. [PMID: 20921202 DOI: 10.1113/jphysiol.2010.196030] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Control of gastrointestinal (GI) movements by enteric motoneurons is critical for orderly processing of food, absorption of nutrients and elimination of wastes. Work over the past several years has suggested that motor neurotransmission is more complicated than simple release of transmitter from nerve terminals and binding of receptors on smooth muscle cells. In fact the 'neuro-effector' junction in the tunica muscularis might consist of synaptic-like connectivity with specialized cells, and contributions from multiple cell types in integrated post-junctional responses. Interstitial cells of Cajal (ICC) were proposed as potential mediators in motor neurotransmission based on reduced post-junctional responses observed in W mutants that have reduced populations of ICC. More recent studies on W mutants have contradicted the original findings, and suggested that ICC may not be significant players in motor neurotransmission. This review examines the evidence for and against the role of ICC in motor neurotransmission and outlines areas for additional investigation that would help further resolve this controversy.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA.
| | | | | |
Collapse
|
44
|
Takaki M, Suzuki H, Nakayama S. Recent advances in studies of spontaneous activity in smooth muscle: ubiquitous pacemaker cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2010; 102:129-35. [PMID: 20553741 DOI: 10.1016/j.pbiomolbio.2010.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 05/19/2010] [Indexed: 02/08/2023]
Abstract
The general and specific properties of pacemaker cells, including Kit-negative cells, that are distributed in gastrointestinal, urethral and uterine smooth muscle tissues, are discussed herein. In intestinal tissues, interstitial cells of Cajal (ICC) are heterogeneous in both their forms and roles. ICC distributed in the myenteric layer (ICC-MY) act as primary pacemaker cells for intestinal mechanical and electrical activity. ICC distributed in muscle bundles play a role as mediators of signals from autonomic nerves to smooth muscle cells. A group of ICC also appears to act as a stretch sensor. Intracellular Ca2+ dynamics play a crucial role in ICC-MY pacemaking; intracellular Ca2+ ([Ca2+](i)) oscillations periodically activate plasmalemmal Ca2+-activated ion channels, such as Ca2+-activated Cl(-) channels and/or non-selective cation channels, although the relative contributions of these channels are not defined. With respect to gut motility, both the ICC network and enteric nervous system, including excitatory and inhibitory enteric neurons, play an essential role in producing highly coordinated peristalsis.
Collapse
Affiliation(s)
- Miyako Takaki
- Department of Physiology II, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan.
| | | | | |
Collapse
|
45
|
Garcia-Lopez P, Garcia-Marin V, Martínez-Murillo R, Freire M. Updating old ideas and recent advances regarding the Interstitial Cells of Cajal. ACTA ACUST UNITED AC 2009; 61:154-69. [PMID: 19520112 DOI: 10.1016/j.brainresrev.2009.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 05/30/2009] [Accepted: 06/01/2009] [Indexed: 12/11/2022]
Abstract
Since their discovery by Cajal in 1889, the Interstitial Cells of Cajal (ICC) have generated much controversy in the scientific community. Indeed, the nervous, muscle or fibroblastic nature of the ICC has remained under debate for more than a century, as has their possible physiological function. Cajal and his colleagues considered them to be neurons, while contemporary histologists like Kölliker and Dogiel categorized these cells as fibroblasts. More recently, the role of ICC in the origin of slow-wave peristaltism has been elucidated, and several studies have shown that they participate in neurotransmission (intercalation theory). The fact that ICC assemble in the circular muscular layer and that they originate from cells which emerge from the ventral neural tube (VENT cells), a source of neurons, glia and ICC precursors other than the neural crest, suggests a neural origin for this particular subset of ICC. The discovery that ICC express the Kit protein, a type III tyrosine kinase receptor encoded by the proto-oncogene c-kit, has helped better understand their physiological role and implication in pathological conditions. Gleevec, a novel molecule designed to inhibit the mutant activated version of c-Kit receptors, is the drug of choice to treat the so-called gastrointestinal stromal tumours (GIST), the most common non-epithelial neoplasm of the gastrointestinal tract. Here we review Cajal's original contributions with the aid of unique images taken from Cajal's histological slides (preserved at the Cajal Museum, Cajal Institute, CSIC). In addition, we present a historical review of the concepts associated with this particular cell type, emphasizing current data that has advanced our understanding of the role these intriguing cells fulfil.
Collapse
Affiliation(s)
- P Garcia-Lopez
- Cajal Institute, CSIC, Avda Doctor Arce 37, 28002 - Madrid, Spain
| | | | | | | |
Collapse
|
46
|
Gomez-Pinilla PJ, Gibbons SJ, Bardsley MR, Lorincz A, Pozo MJ, Pasricha PJ, de Rijn MV, West RB, Sarr MG, Kendrick ML, Cima RR, Dozois EJ, Larson DW, Ordog T, Farrugia G. Ano1 is a selective marker of interstitial cells of Cajal in the human and mouse gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1370-81. [PMID: 19372102 PMCID: PMC2697941 DOI: 10.1152/ajpgi.00074.2009] [Citation(s) in RCA: 310] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Populations of interstitial cells of Cajal (ICC) are altered in several gastrointestinal neuromuscular disorders. ICC are identified typically by ultrastructure and expression of Kit (CD117), a protein that is also expressed on mast cells. No other molecular marker currently exists to independently identify ICC. The expression of ANO1 (DOG1, TMEM16A), a Ca(2+)-activated Cl(-) channel, in gastrointestinal stromal tumors suggests it may be useful as an ICC marker. The aims of this study were therefore to determine the distribution of Ano1 immunoreactivity compared with Kit and to establish whether Ano1 is a reliable marker for human and mouse ICC. Expression of Ano1 in human and mouse stomach, small intestine, and colon was investigated by immunofluorescence labeling using antibodies to Ano1 alone and in combination with antibodies to Kit. Colocalization of immunoreactivity was demonstrated by epifluorescence and confocal microscopy. In the muscularis propria, Ano1 immunoreactivity was restricted to cells with the morphology and distribution of ICC. All Ano1-positive cells in the muscularis propria were also Kit positive. Kit-expressing mast cells were not Ano1 positive. Some non-ICC in the mucosa and submucosa of human tissues were Ano1 positive but Kit negative. A few (3.2%) Ano1-positive cells in the human gastric muscularis propria were labeled weakly for Kit. Ano1 labels all classes of ICC and represents a highly specific marker for studying the distribution of ICC in mouse and human tissues with an advantage over Kit since it does not label mast cells.
Collapse
Affiliation(s)
- Pedro J. Gomez-Pinilla
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Michael R. Bardsley
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Andrea Lorincz
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Maria J. Pozo
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Pankaj J. Pasricha
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Matt Van de Rijn
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Robert B. West
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Michael G. Sarr
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Michael L. Kendrick
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Robert R. Cima
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Eric J. Dozois
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - David W. Larson
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Tamas Ordog
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases and Department of Physiology and Biomedical Engineering, Mayo Clinic; Department of Physiology, Nursing School, University of Extremadura and RETICEF, Caceres, Spain; Department of Surgery, Mayo Clinic, Rochester, Minnesota; Division of Gastroenterology, Department of Medicine, Stanford University School of Medicine; and Department of Pathology, Stanford University Medical Center, Stanford, California
| |
Collapse
|
47
|
Kim YC, Suzuki H, Xu WX, Choi W, Kim SH, Lee SJ. Ca2+-activated K+ current in freshly isolated c-Kit positive cells in guinea-pig stomach. J Korean Med Sci 2009; 24:384-91. [PMID: 19543421 PMCID: PMC2698181 DOI: 10.3346/jkms.2009.24.3.384] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Accepted: 01/29/2008] [Indexed: 11/22/2022] Open
Abstract
This study was designed to isolate Ca2+-activated K+ current (I(KCa)) and elucidate its physiological significance in freshly isolated interstitial cells of Cajal (ICCs) of guinea-pig stomach. Single ICC was freshly isolated by enzymatically dissociating from myenteric border of gastric antrum free of circular muscles, and conventional whole-cell voltage clamp technique including immunohistochemical techniques were employed to characterize the cells: In myenteric border of gastric antrum, ICC-MY (ICCs from myenteric border) were detected by immunohistochemical reactivity, and single ICC-MY which has many branches was immunohistochemically c-Kit positive. Under K+-rich and 0.1 mM ethylene glycol-bis (2-aminoethyl ether)-N,N,N',N'-tetraacetic acid pipette solution, ICC produced spontaneous inward current (-256 +/- 92.2 pA). When step-depolarizing pulse from -80 to +80 mV was applied at holding potential (V(h)) of -80 mV, voltage-dependent outward currents were recorded with superimposed spontaneous transient outward currents (STOCs). Both STOCs and outward currents were reversibly affected by tetraethylammonium chloride (TEA) and iberiotoxin (IbTX); 2 mM TEA and 200 nM IbTX completely abolished STOCs and significantly inhibited outward K+ current over the whole potential range tested for current/voltage (I/V) relationship. In addition, TEA delayed repolarization phase of spontaneous inward current. The present results indicate the presence of I(KCa) in a single ICC, and it might be involved in regulation of repolarizing phase of spontaneous inward current in guinea-pig stomach.
Collapse
Affiliation(s)
- Young Chul Kim
- Department of Physiology, Chungbuk National University, College of Medicine, Cheongju, Korea.
| | | | | | | | | | | |
Collapse
|
48
|
Gibbons SJ, Strege PR, Lei S, Roeder JL, Mazzone A, Ou Y, Rich A, Farrugia G. The alpha1H Ca2+ channel subunit is expressed in mouse jejunal interstitial cells of Cajal and myocytes. J Cell Mol Med 2008; 13:4422-31. [PMID: 19413888 PMCID: PMC2855776 DOI: 10.1111/j.1582-4934.2008.00623.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
T-type Ca2+ currents have been detected in cells from the external muscular layers of gastrointestinal smooth muscles and appear to contribute to the generation of pacemaker potentials in interstitial cells of Cajal from those tissues. However, the Ca2+ channel α subunit responsible for these currents has not been determined. We established that the α subunit of the α1H Ca2+ channel is expressed in single myocytes and interstitial cells of Cajal using reverse transcription and polymerase chain reaction from whole tissue, laser capture microdissected tissue and single cells isolated from the mouse jejunum. Whole-cell voltage clamp recordings demonstrated that a nifedipine and Cd2+ resistant, mibefradil-sensitive current is present in myocytes dissociated from the jejunum. Electrical recordings from the circular muscle layer demonstrated that mibefradil reduced the frequency and initial rate of rise of the electrical slow wave. Gene targeted knockout of both alleles of the cacna1h gene, which encodes the α1H Ca2+ channel subunit, resulted in embryonic lethality because of death of the homozygous knockouts prior to E13.5 days in utero. We conclude that a channel with the pharmacological and molecular characteristics of the α1H Ca2+ channel subunit is expressed in interstitial cells of Cajal and myocytes from the mouse jejunum, and that ionic conductances through the α1H Ca2+ channel contribute to the upstroke of the pacemaker potential. Furthermore, the survival of mice that do not express the α1H Ca2+ channel protein is dependent on the genetic background and targeting approach used to generate the knockout mice.
Collapse
Affiliation(s)
- Simon J Gibbons
- Enteric Neuroscience Program, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Identification and distribution of interstitial Cajal cells in human pulmonary veins. Heart Rhythm 2008; 5:1063-7. [DOI: 10.1016/j.hrthm.2008.03.057] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2007] [Accepted: 03/27/2008] [Indexed: 01/08/2023]
|
50
|
Abstract
Smooth muscle cells (SMC) make up the muscular portion of the gastrointestinal (GI) tract from the distal oesophagus to the internal anal sphincter. Coordinated contractions of these cells produce the motor patterns of GI motility. Considerable progress was made during the last 20 years to understand the basic mechanisms controlling excitation-contraction (E-C) coupling. The smooth muscle motor is now understood in great molecular detail, and much has been learned about the mechanisms that deliver and recover Ca2+ during contractions. The majority of Ca2+ that initiates contractions comes from the external solution and is supplied by voltage-dependent Ca2+ channels (VDCC). VDCC are regulated largely by the effects of K+ and non-selective cation conductances (NSCC) on cell membrane potential and excitability. Ca2+ entry is supplemented by release of Ca2+ from IP(3) receptor-operated stores and by mechanisms that alter the sensitivity of the contractile apparatus to changes in cytoplasmic Ca2+. Molecular studies of the regulation of smooth muscle have been complicated by the plasticity of SMC and difficulties in culturing these cells without dramatic phenotypic changes. Major questions remain to be resolved regarding the details of E-C coupling in human GI smooth muscles. New discoveries regarding molecular expression that give GI smooth muscle their unique properties, the phenotypic changes that occur in SMC in GI motor disorders, tissue engineering approaches to repair or replace defective muscular regions, and molecular manipulations of GI smooth muscles in animals models and in cell culture will be topics for exciting investigations in the future.
Collapse
Affiliation(s)
- K M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA.
| |
Collapse
|