1
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
2
|
Schneider KM, Kim J, Bahnsen K, Heuckeroth RO, Thaiss CA. Environmental perception and control of gastrointestinal immunity by the enteric nervous system. Trends Mol Med 2022; 28:989-1005. [PMID: 36208986 DOI: 10.1016/j.molmed.2022.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/25/2022] [Accepted: 09/07/2022] [Indexed: 12/12/2022]
Abstract
The enteric nervous system (ENS) forms a versatile sensory system along the gastrointestinal tract that interacts with most cell types in the bowel. Herein, we portray host-environment interactions at the intestinal mucosal surface through the lens of the enteric nervous system. We describe local cellular interactions as well as long-range circuits between the enteric, central, and peripheral nervous systems. Additionally, we discuss recently discovered mechanisms by which enteric neurons and glia respond to biotic and abiotic environmental changes and how they regulate intestinal immunity and inflammation. The enteric nervous system emerges as an integrative sensory system with manifold immunoregulatory functions under both homeostatic and pathophysiological conditions.
Collapse
Affiliation(s)
- Kai Markus Schneider
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Jihee Kim
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Klaas Bahnsen
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christoph A Thaiss
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA.
| |
Collapse
|
3
|
Salem M, Lecka J, Pelletier J, Gomes Marconato D, Dumas A, Vallières L, Brochu G, Robaye B, Jobin C, Sévigny J. NTPDase8 protects mice from intestinal inflammation by limiting P2Y 6 receptor activation: identification of a new pathway of inflammation for the potential treatment of IBD. Gut 2022; 71:43-54. [PMID: 33452178 DOI: 10.1136/gutjnl-2020-320937] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Nucleotides are danger signals that activate inflammatory responses via binding P2 receptors. The nucleoside triphosphate diphosphohydrolase-8 (NTPDase8) is an ectonucleotidase that hydrolyses P2 receptor ligands. We investigated the role of NTPDase8 in intestinal inflammation. DESIGN We generated NTPDase8-deficient (Entpd8-/-) mice to define the role of NTPDase8 in the dextran sodium sulfate (DSS) colitis model. To assess inflammation, colons were collected and analysed by histopathology, reverse transcriptase-quantitative real-time PCR (RT-qPCR) and immunohistochemistry. P2 receptor expression was analysed by RT-qPCR on primary intestinal epithelium and NTPDase8 activity by histochemistry. The role of intestinal P2Y6 receptors was assessed by bone marrow transplantation experiments and with a P2Y6 receptor antagonist. RESULTS NTPDase8 is the dominant enzyme responsible for the hydrolysis of nucleotides in the lumen of the colon. Compared with wild-type (WT) control mice, the colon of Entpd8-/- mice treated with DSS displayed significantly more histological damage, immune cell infiltration, apoptosis and increased expression of several proinflammatory cytokines. P2Y6 was the dominant P2Y receptor expressed at the mRNA level by the colonic epithelia. Irradiated P2ry6-/- mice transplanted with WT bone marrow were fully protected from DSS-induced intestinal inflammation. In agreement, the daily intrarectal injection of a P2Y6 antagonist protected mice from DSS-induced intestinal inflammation in a dose-dependent manner. Finally, human intestinal epithelial cells express NTPDase8 and P2Y6 similarly as in mice. CONCLUSION NTPDase8 protects the intestine from inflammation most probably by limiting the activation of P2Y6 receptors in colonic epithelial cells. This may provide a novel therapeutic strategy for the treatment of inflammatory bowel disease.
Collapse
Affiliation(s)
- Mabrouka Salem
- Dép de microbiologie-infectiologie et d'immunologie, fac de médecine, Université Laval, Quebec City, QC, Canada
- Axe Maladies infectieuses et immunitaires, CHU de Québec - Université Laval, Quebec City, QC, Canada
| | - Joanna Lecka
- Axe Maladies infectieuses et immunitaires, CHU de Québec - Université Laval, Quebec City, QC, Canada
| | - Julie Pelletier
- Axe Maladies infectieuses et immunitaires, CHU de Québec - Université Laval, Quebec City, QC, Canada
| | - Danielle Gomes Marconato
- Dép de microbiologie-infectiologie et d'immunologie, fac de médecine, Université Laval, Quebec City, QC, Canada
- Axe Maladies infectieuses et immunitaires, CHU de Québec - Université Laval, Quebec City, QC, Canada
| | - Aline Dumas
- Axe Neurosciences, CHU de Québec - Université Laval, Quebec city, QC, Canada
| | - Luc Vallières
- Axe Neurosciences, CHU de Québec - Université Laval, Quebec city, QC, Canada
- Dép de médecine moléculaire, fac de médecine, Université Laval, Quebec City, QC, Canada
| | - Gaetan Brochu
- CHU de Québec - Université Laval, Quebec City, QC, Canada
- Dept. of Surgery, Université Laval, Quebec City, QC, Canada
| | - Bernard Robaye
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Christian Jobin
- Dept of Infectious Diseases & Pathology, University of Florida, Gainesville, Florida, USA
| | - Jean Sévigny
- Dép de microbiologie-infectiologie et d'immunologie, fac de médecine, Université Laval, Quebec City, QC, Canada
- Axe Maladies infectieuses et immunitaires, CHU de Québec - Université Laval, Quebec City, QC, Canada
| |
Collapse
|
4
|
Jakob MO, Kofoed-Branzk M, Deshpande D, Murugan S, Klose CSN. An Integrated View on Neuronal Subsets in the Peripheral Nervous System and Their Role in Immunoregulation. Front Immunol 2021; 12:679055. [PMID: 34322118 PMCID: PMC8312561 DOI: 10.3389/fimmu.2021.679055] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022] Open
Abstract
The peripheral nervous system consists of sensory circuits that respond to external and internal stimuli and effector circuits that adapt physiologic functions to environmental challenges. Identifying neurotransmitters and neuropeptides and the corresponding receptors on immune cells implies an essential role for the nervous system in regulating immune reactions. Vice versa, neurons express functional cytokine receptors to respond to inflammatory signals directly. Recent advances in single-cell and single-nuclei sequencing have provided an unprecedented depth in neuronal analysis and allowed to refine the classification of distinct neuronal subsets of the peripheral nervous system. Delineating the sensory and immunoregulatory capacity of different neuronal subsets could inform a better understanding of the response happening in tissues that coordinate physiologic functions, tissue homeostasis and immunity. Here, we summarize current subsets of peripheral neurons and discuss neuronal regulation of immune responses, focusing on neuro-immune interactions in the gastrointestinal tract. The nervous system as a central coordinator of immune reactions and tissue homeostasis may predispose for novel promising therapeutic approaches for a large variety of diseases including but not limited to chronic inflammation.
Collapse
Affiliation(s)
- Manuel O Jakob
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michael Kofoed-Branzk
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Divija Deshpande
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Shaira Murugan
- Department of BioMedical Research, Group of Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Christoph S N Klose
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
5
|
Kong Q, Quan Y, Tian G, Zhou J, Liu X. Purinergic P2 Receptors: Novel Mediators of Mechanotransduction. Front Pharmacol 2021; 12:671809. [PMID: 34025431 PMCID: PMC8138185 DOI: 10.3389/fphar.2021.671809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/26/2021] [Indexed: 02/05/2023] Open
Abstract
Mechanosensing and mechanotransduction are vital processes in mechanobiology and play critical roles in regulating cellular behavior and fate. There is increasing evidence that purinergic P2 receptors, members of the purinergic family, play a crucial role in cellular mechanotransduction. Thus, information on the specific mechanism of P2 receptor-mediated mechanotransduction would be valuable. In this review, we focus on purinergic P2 receptor signaling pathways and describe in detail the interaction of P2 receptors with other mechanosensitive molecules, including transient receptor potential channels, integrins, caveolae-associated proteins and hemichannels. In addition, we review the activation of purinergic P2 receptors and the role of various P2 receptors in the regulation of various pathophysiological processes induced by mechanical stimuli.
Collapse
Affiliation(s)
- Qihang Kong
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Quan
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Geer Tian
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junteng Zhou
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Buddington RK, Wong T, Howard SC. Paracellular Filtration Secretion Driven by Mechanical Force Contributes to Small Intestinal Fluid Dynamics. Med Sci (Basel) 2021; 9:medsci9010009. [PMID: 33572202 PMCID: PMC7931054 DOI: 10.3390/medsci9010009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/26/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
Studies of fluid secretion by the small intestine are dominated by the coupling with ATP-dependent generation of ion gradients, whereas the contribution of filtration secretion has been overlooked, possibly by the lack of a known mechanistic basis. We measured apical fluid flow and generation of hydrostatic pressure gradients by epithelia of cultured mouse enterocytes, Caco-2 and T-84 cells, and fibroblasts exposed to mechanical force provided by vigorous aeration and in response to ion gradients, inhibitors of ion channels and transporters and in vitro using intact mouse and rat small intestine. We describe herein a paracellular pathway for unidirectional filtration secretion that is driven by mechanical force, requires tight junctions, is independent of ionic and osmotic gradients, generates persistent hydrostatic pressure gradients, and would contribute to the fluid shifts that occur during digestion and diarrhea. Zinc inhibits the flow of fluid and the paracellular marker fluorescein isothyocyanate conjugated dextran (MW = 4 kD) across epithelia of cultured enterocytes (>95%; p < 0.001) and intact small intestine (>40%; p = 0.03). We propose that mechanical force drives fluid secretion through the tight junction complex via a “one-way check valve” that can be regulated. This pathway of filtration secretion complements chloride-coupled fluid secretion during high-volume fluid flow. The role of filtration secretion in the genesis of diarrhea in intact animals needs further study. Our findings may explain a potential linkage between intestinal motility and intestinal fluid dynamics.
Collapse
Affiliation(s)
- Randal K. Buddington
- School of Health Studies, University of Memphis, Memphis, TN 38152, USA;
- Babies Taking Flight, Memphis, TN 38117, USA
- Correspondence: ; Tel.: +1-662-418-2666
| | - Thomas Wong
- School of Health Studies, University of Memphis, Memphis, TN 38152, USA;
| | - Scott C. Howard
- Department of Acute and Tertiary Care, University of Tennessee Health Sciences Center, Memphis, TN 38163, USA;
| |
Collapse
|
7
|
Fung C, Vanden Berghe P. Functional circuits and signal processing in the enteric nervous system. Cell Mol Life Sci 2020; 77:4505-4522. [PMID: 32424438 PMCID: PMC7599184 DOI: 10.1007/s00018-020-03543-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/13/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
The enteric nervous system (ENS) is an extensive network comprising millions of neurons and glial cells contained within the wall of the gastrointestinal tract. The major functions of the ENS that have been most studied include the regulation of local gut motility, secretion, and blood flow. Other areas that have been gaining increased attention include its interaction with the immune system, with the gut microbiota and its involvement in the gut-brain axis, and neuro-epithelial interactions. Thus, the enteric circuitry plays a central role in intestinal homeostasis, and this becomes particularly evident when there are faults in its wiring such as in neurodevelopmental or neurodegenerative disorders. In this review, we first focus on the current knowledge on the cellular composition of enteric circuits. We then further discuss how enteric circuits detect and process external information, how these signals may be modulated by physiological and pathophysiological factors, and finally, how outputs are generated for integrated gut function.
Collapse
Affiliation(s)
- Candice Fung
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - Pieter Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium.
| |
Collapse
|
8
|
Adenosine triphosphate is co-secreted with glucagon-like peptide-1 to modulate intestinal enterocytes and afferent neurons. Nat Commun 2019; 10:1029. [PMID: 30833673 PMCID: PMC6399286 DOI: 10.1038/s41467-019-09045-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 02/01/2019] [Indexed: 02/02/2023] Open
Abstract
Enteroendocrine cells are specialised sensory cells located in the intestinal epithelium and generate signals in response to food ingestion. Whilst traditionally considered hormone-producing cells, there is evidence that they also initiate activity in the afferent vagus nerve and thereby signal directly to the brainstem. We investigate whether enteroendocrine L-cells, well known for their production of the incretin hormone glucagon-like peptide-1 (GLP-1), also release other neuro-transmitters/modulators. We demonstrate regulated ATP release by ATP measurements in cell supernatants and by using sniffer patches that generate electrical currents upon ATP exposure. Employing purinergic receptor antagonists, we demonstrate that evoked ATP release from L-cells triggers electrical responses in neighbouring enterocytes through P2Y2 and nodose ganglion neurones in co-cultures through P2X2/3-receptors. We conclude that L-cells co-secrete ATP together with GLP-1 and PYY, and that ATP acts as an additional signal triggering vagal activation and potentially synergising with the actions of locally elevated peptide hormone concentrations.
Collapse
|
9
|
Sheikh IA, Ammoury R, Ghishan FK. Pathophysiology of Diarrhea and Its Clinical Implications. PHYSIOLOGY OF THE GASTROINTESTINAL TRACT 2018:1669-1687. [DOI: 10.1016/b978-0-12-809954-4.00068-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Liñán-Rico A, Ochoa-Cortes F, Zuleta-Alarcon A, Alhaj M, Tili E, Enneking J, Harzman A, Grants I, Bergese S, Christofi FL. UTP - Gated Signaling Pathways of 5-HT Release from BON Cells as a Model of Human Enterochromaffin Cells. Front Pharmacol 2017; 8:429. [PMID: 28751862 PMCID: PMC5508028 DOI: 10.3389/fphar.2017.00429] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/15/2017] [Indexed: 12/30/2022] Open
Abstract
Background: Enterochromaffin cells (EC) synthesize and release 5-HT and ATP to trigger or modulate gut neural reflexes and transmit information about visceral/pain sensation. Alterations in 5-HT signaling mechanisms may contribute to the pathogenesis of IBD or IBS, but the pharmacologic or molecular mechanisms modulating Ca2+-dependent 5-HT release are not understood. Previous studies indicated that purinergic signaling via ATP and ADP is an important mechanism in modulation of 5-HT release. However, EC cells also respond to UTP and UDP suggesting uridine triphosphate receptor and signaling pathways are involved as well. We tested the hypothesis that UTP is a regulator of 5-HT release in human EC cells. Methods: UTP signaling mechanisms were studied in BON cells, a human EC model, using Fluo-4/Ca2+imaging, patch-clamp, pharmacological analysis, immunohistochemistry, western blots and qPCR. 5-HT release was monitored in BON or EC isolated from human gut surgical specimens (hEC). Results: UTP, UTPγS, UDP or ATP induced Ca2+oscillations in BON. UTP evoked a biphasic concentration-dependent Ca2+response. Cells responded in the order of UTP, ATP > UTPγS > UDP >> MRS2768, BzATP, α,β-MeATP > MRS2365, MRS2690, and NF546. Different proportions of cells activated by UTP and ATP also responded to UTPγS (P2Y4, 50% cells), UDP (P2Y6, 30%), UTPγS and UDP (14%) or MRS2768 (<3%). UTP Ca2+responses were blocked with inhibitors of PLC, IP3R, SERCA Ca2+pump, La3+sensitive Ca2+channels or chelation of intracellular free Ca2+ by BAPTA/AM. Inhibitors of L-type, TRPC, ryanodine-Ca2+pools, PI3-Kinase, PKC or SRC-Kinase had no effect. UTP stimulated voltage-sensitive Ca2+currents (ICa), Vm-depolarization and inhibited IK (not IA) currents. An IKv7.2/7.3 K+ channel blocker XE-991 mimicked UTP-induced Vm-depolarization and blocked UTP-responses. XE-991 blocked IK and UTP caused further reduction. La3+ or PLC inhibitors blocked UTP depolarization; PKC inhibitors, thapsigargin or zero Ca2+buffer did not. UTP stimulated 5-HT release in hEC expressing TPH1, 5-HT, P2Y4/P2Y6R. Zero-Ca2+buffer augmented Ca2+responses and 5-HT release. Conclusion: UTP activates a predominant P2Y4R pathway to trigger Ca2+oscillations via internal Ca2+mobilization through a PLC/IP3/IP3R/SERCA Ca2+signaling pathway to stimulate 5-HT release; Ca2+influx is inhibitory. UTP-induced Vm-depolarization depends on PLC signaling and an unidentified K channel (which appears independent of Ca2+oscillations or Ica/VOCC). UTP-gated signaling pathways triggered by activation of P2Y4R stimulate 5-HT release.
Collapse
Affiliation(s)
- Andromeda Liñán-Rico
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Fernando Ochoa-Cortes
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Alix Zuleta-Alarcon
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Mazin Alhaj
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Esmerina Tili
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
- Molecular Virology, Immunology and Medical Genetics, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Josh Enneking
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Alan Harzman
- Department of Surgery, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Iveta Grants
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Sergio Bergese
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| | - Fievos L. Christofi
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, ColumbusOH, United States
| |
Collapse
|
11
|
Linan-Rico A, Ochoa-Cortes F, Beyder A, Soghomonyan S, Zuleta-Alarcon A, Coppola V, Christofi FL. Mechanosensory Signaling in Enterochromaffin Cells and 5-HT Release: Potential Implications for Gut Inflammation. Front Neurosci 2016; 10:564. [PMID: 28066160 PMCID: PMC5165017 DOI: 10.3389/fnins.2016.00564] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 11/22/2016] [Indexed: 12/12/2022] Open
Abstract
Enterochromaffin (EC) cells synthesize 95% of the body 5-HT and release 5-HT in response to mechanical or chemical stimulation. EC cell 5-HT has physiological effects on gut motility, secretion and visceral sensation. Abnormal regulation of 5-HT occurs in gastrointestinal disorders and Inflammatory Bowel Diseases (IBD) where 5-HT may represent a key player in the pathogenesis of intestinal inflammation. The focus of this review is on mechanism(s) involved in EC cell "mechanosensation" and critical gaps in our knowledge for future research. Much of our knowledge and concepts are from a human BON cell model of EC, although more recent work has included other cell lines, native EC cells from mouse and human and intact mucosa. EC cells are "mechanosensors" that respond to physical forces generated during peristaltic activity by translating the mechanical stimulus (MS) into an intracellular biochemical response leading to 5-HT and ATP release. The emerging picture of mechanosensation includes Piezo 2 channels, caveolin-rich microdomains, and tight regulation of 5-HT release by purines. The "purinergic hypothesis" is that MS releases purines to act in an autocrine/paracrine manner to activate excitatory (P2Y1, P2Y4, P2Y6, and A2A/A2B) or inhibitory (P2Y12, A1, and A3) receptors to regulate 5-HT release. MS activates a P2Y1/Gαq/PLC/IP3-IP3R/SERCA Ca2+signaling pathway, an A2A/A2B-Gs/AC/cAMP-PKA signaling pathway, an ATP-gated P2X3 channel, and an inhibitory P2Y12-Gi/o/AC-cAMP pathway. In human IBD, P2X3 is down regulated and A2B is up regulated in EC cells, but the pathophysiological consequences of abnormal mechanosensory or purinergic 5-HT signaling remain unknown. EC cell mechanosensation remains poorly understood.
Collapse
Affiliation(s)
- Andromeda Linan-Rico
- Department of Anesthesiology, Wexner Medical Center at Ohio State UniversityColumbus, OH, USA; CONACYT-Centro Universitario de Investigaciones Biomedicas, University of ColimaColima, Mexico
| | - Fernando Ochoa-Cortes
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| | - Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic Rochester, MN, USA
| | - Suren Soghomonyan
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| | - Alix Zuleta-Alarcon
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| | - Vincenzo Coppola
- SBS-Cancer Biology and Genetics, Ohio State University Columbus, OH, USA
| | - Fievos L Christofi
- Department of Anesthesiology, Wexner Medical Center at Ohio State University Columbus, OH, USA
| |
Collapse
|
12
|
Durnin L, Moreland N, Lees A, Mutafova-Yambolieva VN. A commonly used ecto-ATPase inhibitor, ARL-67156, blocks degradation of ADP more than the degradation of ATP in murine colon. Neurogastroenterol Motil 2016; 28:1370-81. [PMID: 27060478 PMCID: PMC5002237 DOI: 10.1111/nmo.12836] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/14/2016] [Indexed: 01/24/2023]
Abstract
BACKGROUND Adenosine 5'-triphosphate (ATP) is released extracellularly as a neurotransmitter and an autocrine or paracrine mediator in numerous systems, including the gastrointestinal tract. It is rapidly degraded to active and inactive metabolites by membrane-bound enzymes. Investigators frequently use inhibitors of ATP hydrolysis such as ARL-67156 and POM-1 to suppress the catabolism of ATP and prolong its effects in pharmacological studies. Our aim was to investigate directly the effects of ARL-67156 and POM-1 on the degradation of ATP and adenosine 5'-diphosphate (ADP) in mouse colonic muscles. METHODS The degradation of ATP and ADP was evaluated by superfusing tissues with 1,N(6) -etheno-ATP (eATP) and 1,N(6) -etheno-ADP (eADP) as substrates and monitoring the decrease in substrate and increase in products (i.e., eADP, eAMP, and e-adenosine) by high-performance liquid chromatography techniques with fluorescence detection. Relaxation responses to etheno-derivatized and non-derivatized ATP and ADP were examined in isometric tension experiments. KEY RESULTS ARL-67156 inhibits the degradation of ADP but not of ATP, whereas POM-1 inhibits the degradation of ATP but not of ADP in murine colonic muscles. Consequently, ARL-67156 enhances relaxation responses to both ATP and ADP, whereas POM-1 reduces relaxation to ATP and does not affect relaxation to ADP. CONCLUSIONS & INFERENCES Studies that use ARL-67156 to inhibit ATP degradation in smooth muscle likely evaluate responses to accumulated ADP rather than ATP. POM-1 appears to be a more selective inhibitor of ATP degradation in the mouse colon. The choice of pharmacological tools in studies on extracellular ATP signaling may affect the interpretation of experimental data in functional studies.
Collapse
Affiliation(s)
- L. Durnin
- Department of Physiology and Cell Biology; University of Nevada School of Medicine; Reno NV USA
| | - N. Moreland
- Department of Physiology and Cell Biology; University of Nevada School of Medicine; Reno NV USA
| | - A. Lees
- Department of Physiology and Cell Biology; University of Nevada School of Medicine; Reno NV USA
| | | |
Collapse
|
13
|
Mañé N, Jiménez-Sábado V, Jiménez M. BPTU, an allosteric antagonist of P2Y1 receptor, blocks nerve mediated inhibitory neuromuscular responses in the gastrointestinal tract of rodents. Neuropharmacology 2016; 110:376-385. [PMID: 27496690 DOI: 10.1016/j.neuropharm.2016.07.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/28/2016] [Accepted: 07/26/2016] [Indexed: 12/28/2022]
Abstract
P2Y1 receptors mediate nerve mediated purinergic inhibitory junction potentials (IJP) and relaxations in the gastrointestinal (GI) tract in a wide range of species including rodents and humans. A new P2Y1 antagonist, with a non-nucleotide structure, BPTU, has recently been described using X-ray crystallography as the first allosteric G-protein-coupled receptor antagonist located entirely outside of the helical bundle. In this study, we tested its effect on purinergic responses in the gastrointestinal tract of rodents using electrophysiological and myographic techniques. BPTU concentration dependently inhibited purinergic inhibitory junction potentials and inhibition of spontaneous motility induced by electrical field stimulation in the colon of rats (EC50 = 0.3 μM) and mice (EC50 = 0.06 μM). Mechanical inhibitory responses were also concentration-dependently blocked in the stomach of both species. Compared to MRS2500, BPTU displays a lower potency. In the rat colon nicotine induced relaxation was also blocked by BPTU. BPTU also blocked the cessation of spontaneous contractility elicited by ADPβS and the P2Y1 agonist MRS2365. We conclude that BPTU is a novel antagonist with different structural and functional properties than nucleotidic antagonists that is able to block the P2Y1 receptor located at the neuromuscular junction of the GI tract.
Collapse
Affiliation(s)
- Noemí Mañé
- Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Verónica Jiménez-Sábado
- Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Marcel Jiménez
- Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain.
| |
Collapse
|
14
|
Liñán-Rico A, Wunderlich JE, Enneking JT, Tso DR, Grants I, Williams KC, Otey A, Michel K, Schemann M, Needleman B, Harzman A, Christofi FL. Neuropharmacology of purinergic receptors in human submucous plexus: Involvement of P2X₁, P2X₂, P2X₃ channels, P2Y and A₃ metabotropic receptors in neurotransmission. Neuropharmacology 2015; 95:83-99. [PMID: 25724083 DOI: 10.1016/j.neuropharm.2015.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 02/11/2015] [Accepted: 02/15/2015] [Indexed: 02/08/2023]
Abstract
RATIONALE The role of purinergic signaling in human ENS is not well understood. We sought to further characterize the neuropharmacology of purinergic receptors in human ENS and test the hypothesis that endogenous purines are critical regulators of neurotransmission. EXPERIMENTAL APPROACH LSCM-Fluo-4/(Ca(2+))-imaging of postsynaptic Ca(2+) transients (PSCaTs) was used as a reporter of synaptic transmission evoked by fiber tract electrical stimulation in human SMP surgical preparations. Pharmacological analysis of purinergic signaling was done in 1,556 neurons (identified by HuC/D-immunoreactivity) in 235 ganglia from 107 patients; P2XR-immunoreactivity was evaluated in 19 patients. Real-time MSORT (Di-8-ANEPPS) imaging tested effects of adenosine on fast excitatory synaptic potentials (fEPSPs). RESULTS Synaptic transmission is sensitive to pharmacological manipulations that alter accumulation of extracellular purines: Apyrase blocks PSCaTs in a majority of neurons. An ecto-NTPDase-inhibitor 6-N,N-diethyl-D-β,γ-dibromomethyleneATP or adenosine deaminase augments PSCaTs. Blockade of reuptake/deamination of eADO inhibits PSCaTs. Adenosine inhibits fEPSPs and PSCaTs (IC50 = 25 µM), sensitive to MRS1220-antagonism (A3AR). A P2Y agonist ADPβS inhibits PSCaTs (IC50 = 111 nM) in neurons without stimulatory ADPbS responses (EC50 = 960 nM). ATP or a P2X1,2,2/3 (α,β-MeATP) agonist evokes fast, slow, biphasic Ca(2+) transients or Ca(2+) oscillations (ATP,EC50 = 400 mM). PSCaTs are sensitive to P2X1 antagonist NF279. Low (20 nM) or high (5 µM) concentrations of P2X antagonist TNP-ATP block PSCaTs in different neurons; proportions of neurons with P2XR-immunoreactivity follow the order P2X2 > P2X1 >> P2X3; P2X1 + P2X2 and P2X3 + P2X2 are co-localized. RT-PCR identified mRNA-transcripts for P2X1-7, P2Y1,2,12-14R. CONCLUSIONS Purines are critical regulators of neurotransmission in human ENS. Purinergic signaling involves P2X1, P2X2, P2X3 channels, P2X1 + P2X2 co-localization and inhibitory P2Y or A3 receptors. These are potential novel therapeutic targets for neurogastroenterology.
Collapse
Affiliation(s)
- A Liñán-Rico
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - J E Wunderlich
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - J T Enneking
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - D R Tso
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - I Grants
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - K C Williams
- Division of Pediatric Gastroenterology, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - A Otey
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - K Michel
- Human Biology, Technische Universität München, Freising, Germany
| | - M Schemann
- Human Biology, Technische Universität München, Freising, Germany
| | - B Needleman
- Department of Surgery, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - A Harzman
- Department of Surgery, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA
| | - F L Christofi
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
15
|
Ochoa-Cortes F, Liñán-Rico A, Jacobson KA, Christofi FL. Potential for developing purinergic drugs for gastrointestinal diseases. Inflamm Bowel Dis 2014; 20:1259-87. [PMID: 24859298 PMCID: PMC4340257 DOI: 10.1097/mib.0000000000000047] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Treatments for inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), functional dyspepsia, or motility disorders are not adequate, and purinergic drugs offer exciting new possibilities. Gastrointestinal symptoms that could be targeted for therapy include visceral pain, inflammatory pain, dysmotility, constipation, and diarrhea. The focus of this review is on the potential for developing purinergic drugs for clinical trials to treat gastrointestinal symptoms. Purinergic receptors are divided into adenosine P1 (A(1), A(2A), A(2B), A(3)), ionotropic ATP-gated P2X ion channel (P2X(1-7)), or metabotropic P2Y(1,2,4,6,11-14) receptors. There is good experimental evidence for targeting A(2A), A(2B), A(3), P2X(7), and P2X(3) receptors or increasing endogenous adenosine levels to treat IBD, inflammatory pain, IBS/visceral pain, inflammatory diarrhea, and motility disorders. Purine genes are also potential biomarkers of disease. Advances in medicinal chemistry have an accelerated pace toward clinical trials: Methotrexate and sulfasalazine, used to treat IBD, act by stimulating CD73-dependent adenosine production. ATP protects against NSAID-induced enteropathy and has pain-relieving properties in humans. A P2X(7)R antagonist AZD9056 is in clinical trials for Crohn's disease. A(3) adenosine receptor drugs target inflammatory diseases (e.g., CF101, CF102). Dipyridamole, a nucleoside uptake inhibitor, is in trials for endotoxemia. Drugs for pain in clinical trials include P2X(3)/P2X(2/3) (AF-219) and P2X(7) (GSK1482160) antagonists and A(1) (GW493838) or A(2A) (BVT.115959) agonists. Iberogast is a phytopharmacon targeting purine mechanisms with efficacy in IBS and functional dyspepsia. Purinergic drugs have excellent safety/efficacy profile for prospective clinical trials in IBD, IBS, functional dyspepsia, and inflammatory diarrhea. Genetic polymorphisms and caffeine consumption may affect susceptibility to treatment. Further studies in animals can clarify mechanisms and test new generation drugs. Finally, there is still a huge gap in our knowledge of human pathophysiology of purinergic signaling.
Collapse
Affiliation(s)
- Fernando Ochoa-Cortes
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, Ohio
| | - Andromeda Liñán-Rico
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, Ohio
| | - Kenneth A. Jacobson
- Laboratory of Bioorganic Chemistry & Molecular Recognition Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health
| | - Fievos L. Christofi
- Department of Anesthesiology, The Wexner Medical Center at The Ohio State University, Columbus, Ohio
| |
Collapse
|
16
|
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: 85] [Impact Index Per Article: 7.7] [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
|
17
|
Foong JPP, Tough IR, Cox HM, Bornstein JC. Properties of cholinergic and non-cholinergic submucosal neurons along the mouse colon. J Physiol 2013; 592:777-93. [PMID: 24344165 DOI: 10.1113/jphysiol.2013.265686] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Submucosal neurons are vital regulators of water and electrolyte secretion and local blood flow in the gut. Due to the availability of transgenic models for enteric neuropathies, the mouse has emerged as the research model of choice, but much is still unknown about the murine submucosal plexus. The progeny of choline acetyltransferase (ChAT)-Cre × ROSA26(YFP) reporter mice, ChAT-Cre;R26R-yellow fluorescent protein (YFP) mice, express YFP in every neuron that has ever expressed ChAT. With the aid of the robust YFP staining in these mice, we correlated the neurochemistry, morphology and electrophysiology of submucosal neurons in distal colon. We also examined whether there are differences in neurochemistry along the colon and in neurally mediated vectorial ion transport between the proximal and distal colon. All YFP(+) submucosal neurons also contained ChAT. Two main neurochemical but not electrophysiological groups of neurons were identified: cholinergic (containing ChAT) or non-cholinergic. The vast majority of neurons in the middle and distal colon were non-cholinergic but contained vasoactive intestinal peptide. In the distal colon, non-cholinergic neurons had one or two axons, whereas the cholinergic neurons examined had only one axon. All submucosal neurons exhibited S-type electrophysiology, shown by the lack of long after-hyperpolarizing potentials following their action potentials and fast excitatory postsynaptic potentials (EPSPs). Fast EPSPs were predominantly nicotinic, and somatic action potentials were mediated by tetrodotoxin-resistant voltage-gated channels. The size of submucosal ganglia decreased but the proportion of cholinergic neurons increased distally along the colon. The distal colon had a significantly larger nicotinic ion transport response than the proximal colon. This work shows that the properties of murine submucosal neurons and their control of epithelial ion transport differ between colonic regions. There are several key differences between the murine submucous plexus and that of other animals, including a lack of conventional intrinsic sensory neurons, which suggests there is an incomplete neuronal circuitry within the murine submucous plexus.
Collapse
Affiliation(s)
- Jaime Pei Pei Foong
- Department of Physiology, University of Melbourne, Parkville, Vic. 3010, Australia.
| | | | | | | |
Collapse
|
18
|
Purinergic autocrine regulation of mechanosensitivity and serotonin release in a human EC model: ATP-gated P2X3 channels in EC are downregulated in ulcerative colitis. Inflamm Bowel Dis 2013; 19:2366-79. [PMID: 23917247 PMCID: PMC4037929 DOI: 10.1097/mib.0b013e31829ecf4d] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alterations in 5-hydroxytryptamine (HT) signaling in inflamed gut may contribute to pathogenesis of inflammatory bowel diseases. Adenosine 5'-triphosphate (ATP) regulates mucosal-mechanosensory reflexes and ATP receptors are sensitive to mucosal inflammation. Yet, it remains unknown whether ATP can modulate 5-HT signaling in enterochromaffin cells (EC). We tested the novel purinergic hypothesis that ATP is a critical autocrine regulator of EC mechanosensitivity and whether EC expression of ATP-gated P2X3-ion channels is altered in inflammatory bowel diseases. METHODS Laser confocal (fluo-4) Ca imaging was performed in 1947 BON cells. Chemical stimulation or mechanical stimulation (MS) was used to study 5-HT or ATP release in human BON or surgical mucosal specimens, and purine receptors by reverse transcription-polymerase chain reaction, Western Blot, or P2X3-immunoreactivity in BON or 5-HT human EC (hEC) in 11 control and 10 severely inflamed ulcerative colitis (UC) cases. RESULTS ATP or MS triggered Ca-transients or 5-HT release in BON. ATP or adenosine diphosphate increased 5-HT release 5-fold. MS caused ATP release, detected after 5'ecto-ATPase inhibition by ARL67156. ARL67156 augmented and apyrase blocked Ca/5-HT mechanosensitive responses. 2-Methyl-thio-adenosine diphosphate 5'-monophosphate-evoked (P2Y1,12) or mechanically-evoked responses were blocked or augmented by a P2Y1,12 antagonist, MRS2179, in different cells or inhibited by U73122. A P2Y12 antagonist, 2MeSAMP, augmented responses. A P2X1,3 agonist, α,β-MeATP, triggered Ca responses, whereas a P2X1,2/3,3 antagonist, 2',3'-O-(2,4,6-trinitrophenyl)-ATP, blocked mechanical responses or cell-surface 5'ATP- labeling. In hEC, α,β-MeATP stimulated 5-HT release. In UC, P2X3-immunoreactivity decreased from 15% to 0.2% of 5-HThECs. Human mucosa and BON expressed P2X1, P2X3, P2X4, P2X5, P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, and P2Y12R-messenger RNA transcripts. CONCLUSIONS ATP is a critical determinant of mechanosensation and 5-HT release via autocrine activation of slow P2Y1-phospholipase C/inositol-1,4,5-triphosphate-Ca or inhibitory P2Y12-purinergic pathways, and fast ATP-gated P2X3-channels. UC downregulation of P2X3-channels (or A2B) is postulated to mediate abnormal 5-HT signaling.
Collapse
|
19
|
Fei G, Fang X, Wang GD, Liu S, Wang XY, Xia Y, Wood JD. Neurogenic mucosal bicarbonate secretion in guinea pig duodenum. Br J Pharmacol 2013; 168:880-90. [PMID: 22994306 DOI: 10.1111/j.1476-5381.2012.02218.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 08/24/2012] [Accepted: 08/31/2012] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE To test a hypothesis that: (i) duodenal pH and osmolarity are individually controlled at constant set points by negative feedback control centred in the enteric nervous system (ENS); (ii) the purinergic P2Y(1) receptor subtype is expressed by non-cholinergic secretomotor/vasodilator neurons, which represent the final common excitatory pathway from the ENS to the bicarbonate secretory glands. EXPERIMENTAL APPROACH Ussing chamber and pH-stat methods investigated involvement of the P2Y(1) receptor in neurogenic stimulation of mucosal bicarbonate (HCO(3)(-)) secretion in guinea pig duodenum. KEY RESULTS ATP increased HCO(3)(-) secretion with an EC(50) of 160 nM. MRS2179, a selective P2Y(1) purinergic receptor antagonist, suppressed ATP-evoked HCO(3)(-) secretion by 47% and Cl(-) secretion by 63%. Enteric neuronal blockade by tetrodotoxin or exposure to a selective vasoactive intestinal peptide (VIP, VPAC(1)) receptor antagonist suppressed ATP-evoked HCO(3)(-) secretion by 61 and 41%, respectively, and Cl- by 97 and 70% respectively. Pretreatment with the muscarinic antagonist, scopolamine did not alter ATP-evoked HCO3(-) or Cl(-) secretion. CONCLUSION AND IMPLICATIONS Whereas acid directly stimulates the mucosa to release ATP and stimulate HCO(3)(-) secretion in a cytoprotective manner, neurogenically evoked HCO(3)(-) secretion accounts for feedback control of optimal luminal pH for digestion. ATP stimulates duodenal HCO(3)(-) secretion through an excitatory action at purinergic P2Y(1) receptors on neurons in the submucosal division of the ENS. Stimulation of the VIPergic non-cholinergic secretomotor/vasodilator neurons, which are one of three classes of secretomotor neurons, accounts for most, if not all, of the neurogenic secretory response evoked by ATP.
Collapse
Affiliation(s)
- G Fei
- Department of Physiology and Cell Biology, Ohio State University College of Medicine, Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Antonioli L, Colucci R, Pellegrini C, Giustarini G, Tuccori M, Blandizzi C, Fornai M. The role of purinergic pathways in the pathophysiology of gut diseases: pharmacological modulation and potential therapeutic applications. Pharmacol Ther 2013; 139:157-88. [PMID: 23588157 DOI: 10.1016/j.pharmthera.2013.04.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 03/15/2013] [Indexed: 02/08/2023]
Abstract
Gut homeostasis results from complex neuro-immune interactions aimed at triggering stereotypical and specific programs of coordinated mucosal secretion and powerful motor propulsion. A prominent role in the regulation of this highly integrated network, comprising a variety of immune/inflammatory cells and the enteric nervous system, is played by purinergic mediators. The cells of the digestive tract are literally plunged into a "biological sea" of functionally active nucleotides and nucleosides, which carry out the critical task of driving regulatory interventions on cellular functions through the activation of P1 and P2 receptors. Intensive research efforts are being made to achieve an integrated view of the purinergic system, since it is emerging that the various components of purinergic pathways (i.e., enzymes, transporters, mediators and receptors) are mutually linked entities, deputed to finely modulating the magnitude and the duration of purinergic signaling, and that alterations occurring in this balanced network could be intimately involved in the pathophysiology of several gut disorders. This review article intends to provide a critical appraisal of current knowledge on the purinergic system role in the regulation of gastrointestinal functions, considering these pathways as a whole integrated network, which is capable of finely controlling the levels of bioactive nucleotides and nucleosides in the biophase of their respective receptors. Special attention is paid to the mechanisms through which alterations in the various compartments of the purinergic system could contribute to the pathophysiology of gut disorders, and to the possibility of counteracting such dysfunctions by means of pharmacological interventions on purinergic molecular targets.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Italy.
| | | | | | | | | | | | | |
Collapse
|
21
|
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
|
22
|
Ammoury RF, Ghishan FK. Pathophysiology of Diarrhea and its Clinical Implications. PHYSIOLOGY OF THE GASTROINTESTINAL TRACT 2012:2183-2197. [DOI: 10.1016/b978-0-12-382026-6.00082-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
23
|
Ren T, Grants I, Alhaj M, McKiernan M, Jacobson M, Hassanain HH, Frankel W, Wunderlich J, Christofi FL. Impact of disrupting adenosine A₃ receptors (A₃⁻/⁻ AR) on colonic motility or progression of colitis in the mouse. Inflamm Bowel Dis 2011; 17:1698-713. [PMID: 21744424 PMCID: PMC3116114 DOI: 10.1002/ibd.21553] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Accepted: 10/01/2010] [Indexed: 01/24/2023]
Abstract
BACKGROUND Pharmacological studies suggest that adenosine A₃AR influences motility and colitis. Functional A₃⁻/⁻AR knockout mice were used to prove whether A₃AR activation is involved in modulating either motility or colitis. METHODS A₃AR was probed by polymerase chain reaction (PCR) genotyping, Western blot, and immunochemistry. Motility was assessed in vivo by artificial bead-expulsion, stool-frequency, and FITC-dextran transit. Colitis was induced with dextran sodium sulfate (DSS) in A₃⁻/⁻AR or wildtype (WT) age- and sex-matched controls. Progression of colitis was evaluated by histopathology, changes in myeloperoxidase (MPO), colon length, CD4(+) -cells, weight-loss, diarrhea, and the guaiac test. RESULTS Goat anti-hu-A₃ antiserum identified a 66 kDa immunogenic band in colon. A₃AR-immunoreactivity is expressed in SYN(+) -nerve varicosities, s-100(+) -glia, and crypt cells, but not 5-HT(+) (EC), CD4(+) (T), tryptase(+) (MC), or muscle cells. A₃AR immunoreactivity in myenteric ganglia of distal colon >> proximal colon by a ratio of 2:1. Intestinal transit and bead expulsion were accelerated in A₃⁻/⁻AR mice compared to WT; stool retention was lower by 40%-60% and stool frequency by 67%. DSS downregulated A₃AR in epithelia. DSS histopathology scores indicated less mucosal damage in AA₃⁻/⁻AR mice than WT. A₃⁻/⁻AR phenotype protected against DSS-induced weight loss, neutrophil (MPO), or CD4(+) -T cell infiltration, colon shortening, change in splenic weight, diarrhea, or occult-fecal blood. CONCLUSIONS Functional disruption of A₃AR in A₃⁻/⁻AR mice alters intestinal motility. We postulate that ongoing release of adenosine and activation of presynaptic-inhibitory A₃AR can slow down transit and inhibit the defecation reflex. A₃AR may be involved in gliotransmission. In separate studies, A₃⁻/⁻AR protects against DSS colitis, consistent with a novel hypothesis that A₃AR activation contributes to development of colitis.
Collapse
Affiliation(s)
- Tianhua Ren
- The Ohio State University, Dept of Anesthesiology, Columbus, Ohio, 43210
| | - Iveta Grants
- The Ohio State University, Dept of Anesthesiology, Columbus, Ohio, 43210
| | - Mazin Alhaj
- The Ohio State University, Dept of Anesthesiology, Columbus, Ohio, 43210
| | - Matt McKiernan
- The Ohio State University, Dept of Anesthesiology, Columbus, Ohio, 43210
| | | | - Hamdy H. Hassanain
- The Ohio State University, Dept of Anesthesiology, Columbus, Ohio, 43210
| | - Wendy Frankel
- The Ohio State University, Dept of Pathology, Columbus, Ohio, 43210
| | | | - Fievos L. Christofi
- The Ohio State University, Dept of Anesthesiology, Columbus, Ohio, 43210,Correspondence to: Fievos L. Christofi, Ph.D., Professor and Vice Chair of Research, Department of Anesthesiology, Professor of Physiology & Cell Biology, College of Medicine and Public Health, The Ohio State University, 226 Tzagournis Medical Research Facility, 420 West 12 Avenue, Columbus, OH, U.S.A. 43210, Phone: 614-688-3802, Fax: 614-688-4894,
| |
Collapse
|
24
|
Foong JPP, Parry LJ, Bornstein JC. Activation of neuronal SST₁ and SST₂ receptors decreases neurogenic secretion in the guinea-pig jejunum. Neurogastroenterol Motil 2010; 22:1209-16, e317. [PMID: 20626789 DOI: 10.1111/j.1365-2982.2010.01566.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Vasoactive intestinal peptide (VIP) submucosal neurons, the main regulators of gut secretion, display inhibitory postsynaptic potentials mediated by somatostatin (SOM) acting on SST(1) and SST(2) receptors (SSTR(1), SSTR(2)) in the guinea-pig small intestine. We investigated the implications of this for neurally-evoked mucosal secretion. METHODS Mucosal-submucosal preparations from guinea-pig jejunum were mounted in Ussing chambers to measure Cl(-) secretion, measured by short circuit current (I(sc)). All drugs were added serosally. Veratridine (1 μmol L(-1)) was used to stimulate neurons and provide a robust secretory response for pharmacological testing.5-hydroxytrptamine (5-HT, 300 nmol L(-1)) was used to specifically activate non-cholinergic secretomotor neurons, while 1,1-dimethyl-4-phenylpiperazinium (DMPP, 10 μmol L(-1)) was used to stimulate all secretomotor neurons. KEY RESULTS Somatostatin (50 nmol L(-1)) induced a tetrodotoxin (TTX, 1 μmol L(-1))-sensitive decrease in secretion. Somatostatin also reduced the veratridine-induced increase in I(sc). The effects of SOM were significantly reduced by blocking SSTR(1) and SSTR(2) individually or together. Blocking SSTR(1) abolished the inhibition produced by SOM. Quantitative PCR demonstrated that SSTR(1) and SSTR(2) were much more highly expressed in the submucosa than the mucosa. Submucosal SSTR(1) expression was several fold higher than SSTR(2). Responses to DMPP (biphasic) and 5-HT (monophasic) were TTX-sensitive. Somatostatin significantly reduced the 5-HT-induced increase in I(sc), and the second, more sustained phase evoked by DMPP. CONCLUSIONS & INFERENCES These data suggest that SOM exerts its antisecretory effects by suppressing firing of VIP secretomotor neurons, rather than via a direct action on mucosal enterocytes.
Collapse
Affiliation(s)
- J P P Foong
- Department of Anatomy & Cell Biology, University of Melbourne, Parkville, Victoria, Australia.
| | | | | |
Collapse
|
25
|
Furuya S, Furuya K, Shigemoto R, Sokabe M. Localization of NK1 receptors and roles of substance-P in subepithelial fibroblasts of rat intestinal villi. Cell Tissue Res 2010; 342:243-59. [PMID: 20967467 DOI: 10.1007/s00441-010-1056-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 09/10/2010] [Indexed: 11/28/2022]
Abstract
Subepithelial fibroblasts of the intestinal villi, which form a contractile cellular network beneath the epithelium, are in close contact with epithelial cells, nerve varicosities, capillaries, smooth muscles and immune cells, and secrete extracellular matrix molecules, growth factors and cytokines, etc. Cultured subepithelial fibroblasts of the rat duodenal villi display various receptors such as endothelins, ATP, substance-P and bradykinin, and release ATP in response to mechanical stimulation. In this study, the presence of functional NK1 receptors (NK1R) was pharmacologically confirmed in primary culture by Ca(2+) measurement, and the effects of substance-P were measured in an acute preparation of epithelium-free duodenal villi from 2- to 3-week-old rats using a two-photon laser microscope. Substance-P elicited an increase in the intracellular Ca(2+) concentration and contraction of the subepithelial fibroblasts in culture and the isolated villi. The localization of NK1R and substance-P in the villi was examined by light and electron microscopic immunohistochemistry. NK1R-like immunoreactivity was intensely localized on the plasma membrane of villous subepithelial fibroblasts in 10-day- to 4-week-old rats and mice and was decreased or absent in adulthood. The pericryptal fibroblasts of the small and large intestine were NK1R immuno-negative. These villous subepithelial fibroblasts form synapse-like structures with both substance-P-immunopositive and -immunonegative nerve varicosities. Here, we propose that the mutual interaction between villous subepithelial fibroblasts and afferent neurons via substance-P and ATP plays important roles in the maturation of the structure and function of the small intestine.
Collapse
Affiliation(s)
- Sonoko Furuya
- Section of Brain Structure, Center for Brain Research, National Institute for Physiological Sciences, Myodaiji, Okazaki, 444-8585, Japan.
| | | | | | | |
Collapse
|
26
|
Karaki SI, Kuwahara A. Propionate-induced epithelial K(+) and Cl(-)/HCO3(-) secretion and free fatty acid receptor 2 (FFA2, GPR43) expression in the guinea pig distal colon. Pflugers Arch 2010; 461:141-52. [PMID: 20945073 DOI: 10.1007/s00424-010-0889-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 08/30/2010] [Accepted: 09/28/2010] [Indexed: 02/07/2023]
Abstract
Propionate, a fermented product in the lumen of the large intestine, is a short-chain fatty acid (SCFA) known to have a variety of localized physiological and pathophysiological functions (e.g., luminal fluid secretion and anti-inflammatory response). In the present study, we investigated propionate-induced transepithelial ion transport and the expression of SCFA receptor, free fatty acid receptor 2 (FFA2, otherwise known as GPR43) in the guinea pig distal colon utilizing the Ussing chamber technique and immunohistochemistry. The addition of propionate to the luminal bathing solution concentration-dependently induced transient K(+) and Cl(-) and/or bicarbonate secretion within approximately 30 s and long-lasting Cl(-) secretion for approximately 60 min was first identified in the present study. The transient anion secretion was tetrodotoxin (TTX)-sensitive and mediated through the cholinergic (both nicotinic and muscarinic) neural pathway, but the transient K(+) and long-lasting Cl(-) secretion were due to TTX-insensitive mechanism. Immunohistochemistry studies showed that some chromogranin A-immunoreactive enteroendocrine cells were also immunoreactive for FFA2 but not colocalized with 5-hydroxytryptamine. In conclusion, the propionate-induced secretion consisted of the neural and non-neural three-phase secretory manner possibly mediated by the stimulation of FFA2 expressed by enteroendocrine cells.
Collapse
Affiliation(s)
- Shin-ichiro Karaki
- Laboratory of Physiology, Graduate School of Nutritional and Environmental Sciences, Institute for Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | | |
Collapse
|
27
|
Schulzke JD, Pfaffenbach S, Fromm A, Epple HJ, Troeger H, Fromm M. Prostaglandin I(2) sensory input into the enteric nervous system during distension-induced colonic chloride secretion in rat colon. Acta Physiol (Oxf) 2010; 199:305-16. [PMID: 20136796 DOI: 10.1111/j.1748-1716.2010.02096.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
AIM Intestinal pressure differences or experimental distension induce ion secretion via the enteric nervous system, the sensorial origin of which is only poorly understood. This study aimed to investigate sensorial inputs and the role of afferent and interneurones in mechanically activated submucosal secretory reflex circuits. METHODS Distension-induced rheogenic chloride secretion was measured as increase in short-circuit current 10 min after distension (DeltaI(SC)(10); distension parameters +/- 100 microL, 2 Hz, 20 s) in partially stripped rat distal colon in the Ussing-chamber in vitro. PGE(2) and PGI(2) were measured by radioimmunoassay. RESULTS DeltaI(SC)(10) was 2.0 +/- 0.2 micromol h(-1) cm(-2) and could be attenuated by lobeline, mecamylamine and dimethylphenylpiperazine, indicating an influence of nicotinergic interneurones. Additionally, a contribution of afferent neurones was indicated from the short-term potentiation of DeltaI(SC)(10) by capsaicin (1 microm). As evidence for its initial event, indomethacin (1 microm) inhibited distension-induced secretion and the release of PGI(2) was directly detected after distension. Furthermore, serotoninergic mediation was confirmed by granisetron (100 microm) which was functionally localized distally to PGI(2) in this reflex circuit, as granisetron inhibited an iloprost-induced I(SC), while indomethacin did not affect serotonin-activated ion secretion. CONCLUSIONS Distension-induced active electrogenic chloride secretion in rat colon is mediated by a neuronal reflex circuit which includes afferent neurones and nicotinergic interneurones. It is initiated by distension-induced PGI(2) release from subepithelial cells triggering this reflex via serotoninergic 5-HT(3) receptor transmission. Functionally, this mechanism may help to protect against intestinal stasis but could also contribute to luminal fluid loss, e.g. during intestinal obstruction.
Collapse
|
28
|
Rybaczyk L, Rozmiarek A, Circle K, Grants I, Needleman B, Wunderlich JE, Huang K, Christofi FL. New bioinformatics approach to analyze gene expressions and signaling pathways reveals unique purine gene dysregulation profiles that distinguish between CD and UC. Inflamm Bowel Dis 2009; 15:971-84. [PMID: 19253308 PMCID: PMC2697273 DOI: 10.1002/ibd.20893] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Expression of purine genes is modulated by inflammation or experimental colitis and altered expression leads to disrupted gut function. We studied purine gene dysregulation profiles in inflammatory bowel disease (IBD) and determined whether they can distinguish between Crohn's disease (CD) and ulcerative colitis (UC) using Pathway Analysis and a new Comparative Analysis of Gene Expression and Selection (CAGES) method. METHODS Raw datasets for 22 purine genes and 36 probe-sets from National Center for Biotechnology Information (NCBI) GEO (Gene Expression Omnibus) (http://www.ncbi.nlm.nih.gov/projects/geo/) were analyzed by National Cancer Institute (NCI) Biological Resources Branch (BRB) array tools for random-variance of multiple/36 t-tests in colonic mucosal biopsies or peripheral blood mononuclear cells (PBMCs) of CD, UC or control subjects. Dysregulation occurs in 59% of purine genes in IBD including ADORA3, CD73, ADORA2A, ADORA2B, ADAR, AMPD2, AMPD3, DPP4, P2RY5, P2RY6, P2RY13, P2RY14, and P2RX5. RESULTS In CD biopsies, expression of ADORA3, AMPD3, P2RY13, and P2RY5 were negatively correlated with acute inflammatory score, Crohn's Disease Activity Index (CDAI) or disease chronicity; P2RY14 was positively correlated in UC. In mucosal biopsies or PBMCs, CD and UC were distinguished by unique patterns of dysregulation (up- or downregulation) in purine genes. Purine gene dysregulation differs between PBMCs and biopsies and possibly between sexes for each disease. Ingenuity Pathway Analysis (IPA) revealed significant associations between alterations in the expression of CD73 (upregulation) or ADORA3 (downregulation) and inflammatory or purine genes (<or=10% of 57 genes) as well as G-protein coupled receptors, cAMP-dependent, and inflammatory pathways; IPA distinguishes CD from UC. CONCLUSION CAGES and Pathway Analysis provided novel evidence that UC and CD have distinct purine gene dysregulation signatures in association with inflammation, cAMP, or other signaling pathways. Disease-specific purine gene signature profiles and pathway associations may be of therapeutic, diagnostic, and functional relevance.
Collapse
Affiliation(s)
- Leszek Rybaczyk
- Dept of Bioinformatics, The Ohio State University, Columbus, Ohio, 43210
| | - Andrew Rozmiarek
- Dept of Anesthesiology, The Ohio State University, Columbus, Ohio, 43210
| | - Kristin Circle
- Dept of Bioinformatics, The Ohio State University, Columbus, Ohio, 43210
| | - Iveta Grants
- Dept of Anesthesiology, The Ohio State University, Columbus, Ohio, 43210
| | | | - Jacqueline E Wunderlich
- Dept of Anesthesiology, The Ohio State University, Columbus, Ohio, 43210,Dept of Surgery, The Ohio State University, Columbus, Ohio, 43210
| | - Kun Huang
- Dept of Bioinformatics, The Ohio State University, Columbus, Ohio, 43210
| | - Fievos L Christofi
- Dept of Anesthesiology, The Ohio State University, Columbus, Ohio, 43210
| |
Collapse
|
29
|
Purinergic signalling in autonomic control. Trends Neurosci 2009; 32:241-8. [PMID: 19359051 DOI: 10.1016/j.tins.2009.03.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Revised: 03/16/2009] [Accepted: 03/17/2009] [Indexed: 02/07/2023]
Abstract
Intercellular purinergic signalling, which utilizes ATP as a transmitter, is fundamental for the operation of the autonomic nervous system. ATP is released together with 'classical' transmitters from sympathetic and parasympathetic nerves supplying various peripheral targets, modulates neurotransmission in autonomic ganglia, has an important role in local enteric neural control and coordination of intestinal secretion and motility, and acts as a common mediator for several distinct sensory modalities. Recently, the role of ATP-mediated signalling in the central nervous control of autonomic function has been addressed. Emerging data demonstrate that in the brain ATP is involved in the operation of several key cardiorespiratory reflexes, contributes to central processing of viscerosensory information, mediates central CO(2) chemosensory transduction and triggers adaptive changes in breathing, and modulates the activities of the brainstem vagal preganglionic, presympathetic and respiratory neural networks.
Collapse
|
30
|
Abstract
Changes in primary sensory neurons are likely to contribute to the emergence of chronic visceral pain. An important step in understanding visceral pain is the development of comprehensive phenotypes that combines functional and anatomical properties for these neurons. We developed a novel ex vivo physiology preparation in mice that allows intracellular recording from colon sensory neurons during colon distension, in the presence and absence of pharmacologic agents. This preparation also allows recovery of functionally characterized afferents for histochemical analysis. Recordings obtained from L6 dorsal root ganglion cells in C57BL/6 mice identified two distinct populations of distension-responsive colon afferents: high-firing frequency (HF) and low-firing frequency (LF) cells. Fluid distension of the colon elicited rapid firing (>20 Hz) in HF cells, whereas LF cells seldom fired >5 Hz. Distension response thresholds were significantly lower in HF cells (LF, 17.5 +/- 1.1 cmH(2)O; HF, 2.6 +/- 1.0 cmH(2)O). Responses of most LF afferents to colon distension were sensitized by luminal application of capsaicin (1 microm; 8 of 9 LF cells), mustard oil (100 microm; 10 of 12 LF cells), and low pH (pH 4.0; 5 of 6 LF cells). In contrast, few HF afferents were sensitized by capsaicin (3 of 9), mustard oil (2 of 7), or low pH (1 of 6) application. Few HF afferents (4 of 23) expressed the capsaicin receptor, TRPV1. In contrast, 87% (25 of 29) of LF afferents expressed TRPV1. TRPV1 has been shown to be required for development of inflammatory hyperalgesia. These results suggest a unique functional role of TRPV1-positive colon afferents that could be exploited to design specific therapies for visceral hypersensitivity.
Collapse
|
31
|
Purinergic receptors and gastrointestinal secretomotor function. Purinergic Signal 2008; 4:213-36. [PMID: 18604596 DOI: 10.1007/s11302-008-9104-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2007] [Accepted: 04/07/2008] [Indexed: 02/06/2023] Open
Abstract
Secretomotor reflexes in the gastrointestinal (GI) tract are important in the lubrication and movement of digested products, absorption of nutrients, or the diarrhea that occurs in diseases to flush out unwanted microbes. Mechanical or chemical stimulation of mucosal sensory enterochromaffin (EC) cells triggers release of serotonin (5-HT) (among other mediators) and initiates local reflexes by activating intrinsic primary afferent neurons of the submucous plexus. Signals are conveyed to interneurons or secretomotor neurons to stimulate chloride and fluid secretion. Inputs from myenteric neurons modulate secretory rates and reflexes, and special neural circuits exist to coordinate secretion with motility. Cellular components of secretomotor reflexes variably express purinergic receptors for adenosine (A1, A2a, A2b, or A3 receptors) or the nucleotides adenosine 5'-triphosphate (ATP), adenosine diphosphate (ADP), uridine 5'-triphosphate (UTP), or uridine diphosphate (UDP) (P2X(1-7), P2Y(2), P2Y(4), P2Y(6), P2Y(12) receptors). This review focuses on the emerging concepts in our understanding of purinergic regulation at these receptors, and in particular of mechanosensory reflexes. Purinergic inhibitory (A(1), A(3), P2Y(12)) or excitatory (A(2), P2Y(1)) receptors modulate mechanosensitive 5-HT release. Excitatory (P2Y(1), other P2Y, P2X) or inhibitory (A(1), A(3)) receptors are involved in mechanically evoked secretory reflexes or "neurogenic diarrhea." Distinct neural (pre- or postsynaptic) and non-neural distribution profiles of P2X(2), P2X(3), P2X(5), P2Y(1), P2Y(2), P2Y(4), P2Y(6), or P2Y(12) receptors, and for some their effects on neurotransmission, suggests their role in GI secretomotor function. Luminal A(2b), P2Y(2), P2Y(4), and P2Y(6) receptors are involved in fluid and Cl(-), HCO(3) (-), K(+), or mucin secretion. Abnormal receptor expression in GI diseases may be of clinical relevance. Adenosine A(2a) or A(3) receptors are emerging as therapeutic targets in inflammatory bowel diseases (IBD) and gastroprotection; they can also prevent purinergic receptor abnormalities and diarrhea. Purines are emerging as fundamental regulators of enteric secretomotor reflexes in health and disease.
Collapse
|
32
|
Wunderlich JE, Needleman BJ, Chen Z, Yu JG, Wang Y, Grants I, Mikami DJ, Melvin WS, Cooke HJ, Christofi FL. Dual purinergic synaptic transmission in the human enteric nervous system. Am J Physiol Gastrointest Liver Physiol 2008; 294:G554-66. [PMID: 18079280 DOI: 10.1152/ajpgi.00500.2007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Based on findings in rodents, we sought to test the hypothesis that purinergic modulation of synaptic transmission occurs in the human intestine. Time series analysis of intraneuronal free Ca(2+) levels in submucosal plexus (SMP) from Roux-en-Y specimens was done using Zeiss LSM laser-scanning confocal fluo-4 AM Ca(2+) imaging. A 3-s fiber tract stimulation (FTS) was used to elicit a synaptic Ca(2+) response. Short-circuit current (I(sc) = chloride secretion) was recorded in mucosa-SMP in flux chambers. A distension reflex or electrical field stimulation was used to study I(sc) responses. Ca(2+) imaging was done in 1,222 neurons responding to high-K(+) depolarization from 61 surgical cases. FTS evoked synaptic Ca(2+) responses in 62% of recorded neurons. FTS caused frequency-dependent Ca(2+) responses (0.1-100 Hz). FTS Ca(2+) responses were inhibited by Omega-conotoxin (70%), hexamethonium (50%), TTX, high Mg(2+)/low Ca(2+) (< or = 100%), or capsaicin (25%). A P2Y(1) receptor (P2Y(1)R) antagonist, MRS-2179 or PLC inhibitor U-73122, blocked FTS responses (75-90%). P2Y(1)R-immunoreactivity occurred in 39% of vasoactive intestinal peptide-positive neurons. The selective adenosine A(3) receptor (AdoA(3)R) agonist 2-chloro-N(6)-(3-iodobenzyl)adenosine-5'-N-methylcarboxamide (2-Cl-IBMECA) caused concentration- and frequency-dependent inhibition of FTS Ca(2+) responses (IC(50) = 8.5 x 10(-8) M). The AdoA(3)R antagonist MRS-1220 augmented such Ca(2+) responses; 2-Cl-IBMECA competed with MRS-1220. Knockdown of AdoA(1)R with 8-cyclopentyl-3-N-(3-{[3-(4-fluorosulphonyl)benzoyl]-oxy}-propyl)-1-N-propyl-xanthine did not prevent 2-Cl-IBMECA effects. MRS-1220 caused 31% augmentation of TTX-sensitive distension I(sc) responses. The SMP from Roux-en-Y patients is a suitable model to study synaptic transmission in human enteric nervous system (huENS). The P2Y(1)/Galphaq/PLC/inositol 1,3,5-trisphosphate/Ca(2+) signaling pathway, N-type Ca(2+) channels, nicotinic receptors, and extrinsic nerves contribute to neurotransmission in huENS. Inhibitory AdoA(3)R inhibit nucleotide or cholinergic transmission in the huENS.
Collapse
Affiliation(s)
- J E Wunderlich
- Department of Anesthesiology, College of Medicine and Public Health, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Gallego D, Vanden Berghe P, Farré R, Tack J, Jiménez M. P2Y1 receptors mediate inhibitory neuromuscular transmission and enteric neuronal activation in small intestine. Neurogastroenterol Motil 2008; 20:159-68. [PMID: 17971025 DOI: 10.1111/j.1365-2982.2007.01004.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
There is increasing evidence that adenosine 5'-triphosphate or a related purine plays a crucial role in smooth muscle relaxation and enteric synaptic neurotransmission. Accordingly, the aim of the present work is to investigate the role P2Y(1) receptors in purinergic inhibitory neurotransmission (pig ileum) and enteric neuronal activation in the small intestine (guinea-pig ileum). Using contractility measurements, micro-electrode recordings and Ca(2+) imaging we found that (i) adenosine 5'-Omicron-2-thiodiphosphate (ADPbetaS) (10 micromol L(-1)) caused smooth muscle relaxation and hyperpolarization that was antagonized by MRS2179 (10 micromol L(-1)) a P2Y(1) receptor antagonist and apamin (1 micromol L(-1)); (ii) electrical field stimulation (EFS) caused a non-nitrergic inhibitory junction potential (IJP) and relaxation that was antagonized by MRS2179 (10 micromol L(-1)); (iii) P2Y(1) receptors were immunolocalized in smooth muscle cells and enteric neurons; (i.v.) superfusion of ADPbetaS (1 micromol L(-1)) induced Ca(2+) transients in myenteric neurons that were inhibited by MRS2179 (1 micromol L(-1)), but not by tetrodotoxin (1 micromol L(-1)); and (v) EFS induced calcium transients were partially inhibited by MRS2179 (1 micromol L(-1)). We conclude that in the small intestine purinergic neuromuscular transmission responsible for the IJP and non-nitrergic relaxation is mediated by P2Y(1) receptors located in smooth muscle cells. Functional P2Y(1) receptors are also present in guinea-pig myenteric neurons. Therefore, P2Y(1) receptors might be an important pharmacological target to modulate gastrointestinal functions.
Collapse
Affiliation(s)
- D Gallego
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Bellaterra, Barcelona, Spain
| | | | | | | | | |
Collapse
|
34
|
Wang GD, Wang XY, Hu HZ, Liu S, Gao N, Fang X, Xia Y, Wood JD. Inhibitory neuromuscular transmission mediated by the P2Y1 purinergic receptor in guinea pig small intestine. Am J Physiol Gastrointest Liver Physiol 2007; 292:G1483-9. [PMID: 17322065 DOI: 10.1152/ajpgi.00450.2006] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
ATP is a putative inhibitory neurotransmitter responsible for inhibitory junction potentials (IJPs) at neuromuscular junctions (IJPs) in the intestine. This study tested the hypothesis that the purinergic P2Y(1) receptor subtype mediates the IJPs. IJPs were evoked by focal electrical stimulation in the myenteric plexus and recorded with "sharp" intracellular microelectrodes in the circular muscle coat. Stimulation evoked three categories of IJPs: 1) purely purinergic IJPs, 2) partially purinergic IJPs, and 3) nonpurinergic IJPs. Purely purinergic IJPs were suppressed by the selective P2Y(1) purinergic receptor antagonist MRS2179. Purely purinergic IJPs comprised 26% of the IJPs. Partially purinergic IJPs (72% of the IJPs) consisted of a component that was abolished by MRS2179 and a second unaffected component. The MRS2179-insensitive component was suppressed or abolished by inhibition of formation of nitric oxide by N(omega)-nitro-l-arginine methyl ester (l-NAME) in some, but not all, IJPs. An unidentified neurotransmitter, different from nitric oxide, mediated the second component in these cases. Nonpurinergic IJPs were a small third category (4%) of IJPs that were abolished by l-NAME and unaffected by MRS2179. Exogenous application of ATP evoked IJP-like hyperpolarizing responses, which were blocked by MRS2179. Application of apamin, which suppresses opening of small-conductance Ca(2+)-operated K(+) channels in the muscle, decreased the amplitude of the purinergic IJPs and the amplitude of IJP-like responses to ATP. The results support ATP as a neurotransmitter for IJPs in the intestine and are consistent with the hypothesis that the P2Y(1) purinergic receptor subtype mediates the action of ATP.
Collapse
Affiliation(s)
- Guo-Du Wang
- Dept. of Physiology and Cell Biology, The Ohio State Univ., College of Medicine and Public Health, 304 Hamilton Hall, 1645 Neil Ave., Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Kim M, Christofi FL, Xue J, Robinson JM, Cooke HJ. Mechanically evoked 5-hydroxytryptamine release is mediated by caveolin-associated cholesterol rich membrane domains. Neurogastroenterol Motil 2007; 19:309-17. [PMID: 17391247 DOI: 10.1111/j.1365-2982.2007.00912.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
5-Hydroxytryptamine (5-HT) from enterochromaffin cells activates neural reflexes that govern intestinal motility and secretion. Mechanical stimulation of human enterochromaffin cell-derived BON cells activates a G alpha q-signalling pathway coupled to 5-HT release. Molecular mechanisms identifying elements of mechanosensory transduction are unknown. The aim of this study was to determine the role of caveolin and caveolin-associated cholesterol rich microdomains in mechanically stimulated 5-HT release from BON cells. Caveolin-1 transcripts and immunofluorescence were found in BON cells. In the static state, caveolins-1 and -2 co-precipitated with G alpha q in cholesterol rich cell fractions, but not with G alpha s, G alpha i/o and G beta. Mechanical stimulation transiently uncoupled G alpha q from caveolin-1 and increased 5-HT release. Disassembly of caveolin-associated membrane microdomains by filipin or by cholesterol depletion with methyl-beta-cyclodextrin decreased mechanically evoked 5-HT release. These results suggest that caveolin and caveolin-associated cholesterol rich membrane microdomains are key regulators in mechanically evoked 5-HT release from enterochromaffin cells.
Collapse
Affiliation(s)
- M Kim
- Department of Surgery, Rhode Island Hospital, Brown Medical School, Providence, Rhode Island, USA
| | | | | | | | | |
Collapse
|
36
|
Xue J, Askwith C, Javed NH, Cooke HJ. Autonomic nervous system and secretion across the intestinal mucosal surface. Auton Neurosci 2007; 133:55-63. [PMID: 17336595 PMCID: PMC1936976 DOI: 10.1016/j.autneu.2007.02.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2006] [Revised: 01/30/2007] [Accepted: 02/01/2007] [Indexed: 01/27/2023]
Abstract
Chloride secretion is important because it is the driving force for fluid movement into the intestinal lumen. The flow of accumulated fluid flushes out invading micro-organisms in defense of the host. Chloride secretion is regulated by neurons in the submucosal plexus of the enteric nervous system. Mechanosensitive enterochromaffin cells that release 5-hydroxytryptamine (5-HT) and activate intrinsic afferent neurons in the submucosal plexus and initiate chloride secretion. Mechanical stimulation by distention may also trigger reflexes by a direct action on intrinsic afferent neurons. Dysregulation of 5-HT release or altered activity of intrinsic afferents is likely to occur in states of inflammation and other disorders.
Collapse
Affiliation(s)
- Jianjing Xue
- Department of Neuroscience, 333 West 10th Avenue, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | |
Collapse
|
37
|
Gao N, Hu HZ, Liu S, Gao C, Xia Y, Wood JD. Stimulation of adenosine A1 and A2A receptors by AMP in the submucosal plexus of guinea pig small intestine. Am J Physiol Gastrointest Liver Physiol 2007; 292:G492-500. [PMID: 17023550 DOI: 10.1152/ajpgi.00257.2006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Actions of adenosine 5'-monophosphate (AMP) on electrical and synaptic behavior of submucosal neurons in guinea pig small intestine were studied with "sharp" intracellular microelectrodes. Application of AMP (0.3-100 microM) evoked slowly activating depolarizing responses associated with increased excitability in 80.5% of the neurons. The responses were concentration dependent with an EC(50) of 3.5 +/- 0.5 microM. They were abolished by the adenosine A(2A) receptor antagonist ZM-241385 but not by pyridoxal-phosphate-6-azophenyl-2,4-disulfonic acid, trinitrophenyl-ATP, 8-cyclopentyl-1,3-dimethylxanthine, suramin, or MRS-12201220. The AMP-evoked responses were insensitive to AACOCF3 or ryanodine. They were reduced significantly by 1) U-73122, which is a phospholipase C inhibitor; 2) cyclopiazonic acid, which blocks the Ca(2+) pump in intraneuronal membranes; and 3) 2-aminoethoxy-diphenylborane, which is an inositol (1,4,5)-trisphosphate receptor antagonist. Inhibitors of PKC or calmodulin-dependent protein kinase also suppressed the AMP-evoked excitatory responses. Exposure to AMP suppressed fast nicotinic ionotropic postsynaptic potentials, slow metabotropic excitatory postsynaptic potentials, and slow noradrenergic inhibitory postsynaptic potentials in the submucosal plexus. Inhibition of each form of synaptic transmission reflected action at presynaptic inhibitory adenosine A(1) receptors. Slow excitatory postsynaptic potentials, which were mediated by the release of ATP and stimulation of P2Y(1) purinergic receptors in the submucosal plexus, were not suppressed by AMP. The results suggest an excitatory action of AMP at adenosine A(2A) receptors on neuronal cell bodies and presynaptic inhibitory actions mediated by adenosine A(1) receptors for most forms of neurotransmission in the submucosal plexus, with the exception of slow excitatory purinergic transmission mediated by the P2Y(1) receptor subtype.
Collapse
Affiliation(s)
- Na Gao
- Dept of Physiology and Cell Biology, Columbus, OH 43210-1218, USA
| | | | | | | | | | | |
Collapse
|
38
|
Furuya S, Furuya K. Subepithelial fibroblasts in intestinal villi: roles in intercellular communication. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 264:165-223. [PMID: 17964923 DOI: 10.1016/s0074-7696(07)64004-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ingestion of food and water induces chemical and mechanical signals that trigger peristaltic reflexes in the gut. Intestinal villi are motile, equipped with chemosensors and mechanosensors, and transduce signaling to sensory neurons, but the exact mechanisms have not yet been elucidated. Subepithelial fibroblasts located under the villous epithelium form contractile cellular networks via gap junctions. The networks ensheathe lamina propria and are in close contact with epithelium, neural and capillary networks, smooth muscles, and immune cells. Unique characteristics of subepithelial fibroblasts have been revealed by primary cultures isolated from rat duodenal villi. They include rapid reversal changes in cell shape by cAMP reagents and endothelins, cell shape-dependent mechanosensitivity that induces ATP release as a paracrine mediator, contractile ability, and expression of various receptors for vasoactive and neuroactive substances. Herein, we review these characteristics that play a key role in the villi. They serve as a barrier/sieve, flexible mechanical frame, mechanosensor, and signal transduction machinery in the intestinal villi, which are regulated locally and dynamically by rapid cell shape conversion.
Collapse
Affiliation(s)
- Sonoko Furuya
- Section of Brain Structure, Center for Brain Experiment, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | | |
Collapse
|
39
|
Van Nassauw L, Costagliola A, Van Op den Bosch J, Cecio A, Vanderwinden JM, Burnstock G, Timmermans JP. Region-specific distribution of the P2Y4 receptor in enteric glial cells and interstitial cells of Cajal within the guinea-pig gastrointestinal tract. Auton Neurosci 2006; 126-127:299-306. [PMID: 16616701 DOI: 10.1016/j.autneu.2006.02.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Revised: 02/16/2006] [Accepted: 02/27/2006] [Indexed: 01/01/2023]
Abstract
Although there is pharmacological evidence to assume that the P2Y4 receptor is a regulator of epithelial ion transport, no detailed data about its distribution within the gut are available. Therefore, this study, using whole mounts and cryosections, aimed to reveal the expression pattern of P2Y4 along the entire guinea-pig gastrointestinal tract. P2Y4 immunoreactivity was absent from enteric neurons but present in enteric glial cells of the stomach, small and large intestine. In the esophagus, P2Y4 appeared to be exclusively located within striated muscle cells. P2Y4 showed also a region dependency regarding its presence in different subpopulations of interstitial cells of Cajal: in myenteric interstitial cells of Cajal in the stomach and ileum; in some intramuscular interstitial cells in the stomach and cecum; in some deep muscular plexus interstitial cells in the ileum; and in some submucosal surface interstitial cells in the colon. These results and the knowledge that P2Y4 activation causes intracellular Ca2+ recruitment led us to suggest that P2Y4 in enteric glia plays a modulatory role in intercellular Ca2+ waves, while P2Y4 in interstitial cells of Cajal modulates intracellular Ca2+ oscillations.
Collapse
Affiliation(s)
- Luc Van Nassauw
- Laboratory of Cell Biology and Histology, University of Antwerp, Groenenborgerlaan 171, B-2020 Antwerpen, Belgium
| | | | | | | | | | | | | |
Collapse
|
40
|
Grundy D, Al-Chaer ED, Aziz Q, Collins SM, Ke M, Taché Y, Wood JD. Fundamentals of neurogastroenterology: basic science. Gastroenterology 2006; 130:1391-411. [PMID: 16678554 DOI: 10.1053/j.gastro.2005.11.060] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2005] [Accepted: 11/03/2005] [Indexed: 02/06/2023]
Abstract
The focus of neurogastroenterology in Rome II was the enteric nervous system (ENS). To avoid duplication with Rome II, only advances in ENS neurobiology after Rome II are reviewed together with stronger emphasis on interactions of the brain, spinal cord, and the gut in terms of relevance for abdominal pain and disordered gastrointestinal function. A committee with expertise in selective aspects of neurogastroenterology was invited to evaluate the literature and provide a consensus overview of the Fundamentals of Neurogastroenterology textbook as they relate to functional gastrointestinal disorders (FGIDs). This review is an abbreviated version of a fuller account that appears in the forthcoming book, Rome III. This report reviews current basic science understanding of visceral sensation and its modulation by inflammation and stress and advances in the neurophysiology of the ENS. Many of the concepts are derived from animal studies in which the physiologic mechanisms underlying visceral sensitivity and neural control of motility, secretion, and blood flow are examined. Impact of inflammation and stress in experimental models relative to FGIDs is reviewed as is human brain imaging, which provides a means for translating basic science to understanding FGID symptoms. Investigative evidence and emerging concepts implicate dysfunction in the nervous system as a significant factor underlying patient symptoms in FGIDs. Continued focus on neurogastroenterologic factors that underlie the development of symptoms will lead to mechanistic understanding that is expected to directly benefit the large contingent of patients and care-givers who deal with FGIDs.
Collapse
Affiliation(s)
- David Grundy
- Department of Biomedical Sciences, University of Sheffield, Sheffield, England
| | | | | | | | | | | | | |
Collapse
|
41
|
Fang X, Hu HZ, Gao N, Liu S, Wang GD, Wang XY, Xia Y, Wood JD. Neurogenic secretion mediated by the purinergic P2Y1 receptor in guinea-pig small intestine. Eur J Pharmacol 2006; 536:113-22. [PMID: 16566916 DOI: 10.1016/j.ejphar.2006.02.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2005] [Revised: 02/07/2006] [Accepted: 02/21/2006] [Indexed: 11/29/2022]
Abstract
We tested the hypothesis that ATP is an enteric neurotransmitter that acts at P2Y1 excitatory purinergic receptors on intestinal secretomotor neurons to evoke neurogenic mucosal secretion in the guinea pig. Ussing chamber methods for studying neurogenic intestinal secretion were used to test the hypothesis. Application of ATP evoked concentration-dependent increases in short circuit current (Isc) indicative of stimulation of electrolyte secretion. MRS2179, a selective P2Y1 purinergic receptor antagonist, suppressed the ATP-evoked responses in a concentration-dependent manner with an IC50 of 0.9+/-0.1 microM. Tetrodotoxin or a selective vasoactive intestinal peptide (VPAC1) receptor antagonist suppressed or abolished the ATP-evoked responses. A selective VPAC1 receptor antagonist also suppressed Isc responses evoked by electrical field stimulation of the secretomotor neurons. Secretory responses to ATP were not suppressed by scopolamine, piroxicam nor selective adenosine receptor antagonists. Region-specific differences in responses to ATP corresponded to regional differences in the expression of mRNA transcripts for the P2Y1 receptor. Post-receptor signal transduction for the P2Y1-evoked responses involved stimulation of phospholipase C and an IP3/Ca2+-calmodulin/protein kinase C signaling cascade. Our evidence suggests that ATP is released as a neurotransmitter to stimulate neurogenic mucosal secretion by binding to P2Y1 receptors expressed by VIP-ergic secretomotor neurons.
Collapse
MESH Headings
- Adenosine Diphosphate/analogs & derivatives
- Adenosine Diphosphate/pharmacology
- Adenosine Triphosphate/pharmacology
- Animals
- Bumetanide/pharmacology
- Chlorides/metabolism
- Dose-Response Relationship, Drug
- Electric Stimulation
- Electrolytes/metabolism
- Gene Expression
- Guinea Pigs
- In Vitro Techniques
- Intestine, Small/drug effects
- Intestine, Small/innervation
- Intestine, Small/metabolism
- Male
- Purinergic P1 Receptor Agonists
- Purinergic P1 Receptor Antagonists
- Purinergic P2 Receptor Antagonists
- Receptors, Purinergic P2/genetics
- Receptors, Purinergic P2/physiology
- Receptors, Purinergic P2Y1
- Receptors, Vasoactive Intestinal Polypeptide, Type I/agonists
- Receptors, Vasoactive Intestinal Polypeptide, Type I/antagonists & inhibitors
- Signal Transduction/drug effects
- Sodium Potassium Chloride Symporter Inhibitors/pharmacology
- Tetrodotoxin/pharmacology
- Theophylline/analogs & derivatives
- Theophylline/pharmacology
- Triazines/pharmacology
- Triazoles/pharmacology
- Vasoactive Intestinal Peptide/pharmacology
Collapse
Affiliation(s)
- Xiucai Fang
- Department of Physiology and Cell Biology, College of Medicine and Public Health, The Ohio State University, 304 Hamilton Hall, 1645 Neil Avenue, Columbus, OH 43210-1218, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Xiang Z, Burnstock G. Distribution of P2Y2 receptors in the guinea pig enteric nervous system and its coexistence with P2X2 and P2X3 receptors, neuropeptide Y, nitric oxide synthase and calretinin. Histochem Cell Biol 2005; 124:379-90. [PMID: 16136347 DOI: 10.1007/s00418-005-0043-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2005] [Indexed: 12/14/2022]
Abstract
The distribution of P2Y2 receptor-immunoreactive (ir) neurons and fibers and coexistence of P2Y2 with P2X2 and P2X3 receptors, neuropeptide Y (NPY), calretinin (CR), calbindin (CB) and nitric oxide synthase (NOS) was investigated with immunostaining methods. The results showed that P2Y2-ir neurons and fibers were distributed widely in myenteric and submucous plexuses of the guinea pig stomach corpus, jejunum, ileum and colon. The typical morphology of P2Y2-ir neurons was a long process with strong positive staining on the same side of the cell body. The P2Y2-ir neurons could be Dogiel type 1. About 40-60% P2X3-ir neurons were immunoreactive for P2Y2 in the myenteric plexus and all the P2X3-ir neurons expressed the P2Y2 receptor in the submucosal plexus; almost all the NPY-ir neurons and the majority of CR-ir neurons were also immunoreactive for P2Y2, especially in the myenteric plexus of the small intestine; no P2Y2-ir neurons were immunoreactive for P2X2 receptors, CB and NOS. It is shown for the first time that S type/Dogiel type 1 neurons with fast P2X and slow P2Y receptor-mediated depolarizations could be those neurons expressing both P2Y2-ir and P2X3-ir and that they are widely distributed in myenteric and submucosal plexuses of guinea pig gut.
Collapse
Affiliation(s)
- Zhenghua Xiang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, 200433, Shanghai, People's Republic of China
| | | |
Collapse
|
43
|
Xiang Z, Burnstock G. Distribution of P2Y6 and P2Y12 receptor: their colocalization with calbindin, calretinin and nitric oxide synthase in the guinea pig enteric nervous system. Histochem Cell Biol 2005; 125:327-36. [PMID: 16195889 DOI: 10.1007/s00418-005-0071-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2005] [Indexed: 12/31/2022]
Abstract
The distribution of P2Y(6) and P2Y(12) receptor-immunoreactive (ir) neurons and fibers and their coexistence with calbindin, calretinin and nitric oxide synthase (NOS) has been investigated with single and double labeling immunostaining methods. The results showed that 30-36% of the ganglion cells in the myenteric plexus are strongly P2Y(6) receptor-ir neurons; they are distributed widely in the myenteric plexus of stomach, jejunum, ileum and colon, but not in the submucosal plexus, with a typical morphology of multipolar neurons with a long axon-like process. About 42-46% of ganglion cells in both the myenteric and submucosal plexuses show P2Y(12) receptor-ir. About 28-35% of P2Y(6) receptor-ir neurons were found to coexist with NOS and 41-47% of them coexist with calretinin, but there was no coexistence of P2Y(6) receptor-ir with calbindin. In contrast, all P2Y(12) receptor-ir neurons were immunopositive for calbindin, although occasionally P2Y(12) receptor-ir neurons without calbindin immunoreactivity were found, while none of the P2Y(12) receptor-ir neurons were found to coexist with calretinin or NOS in the gastrointestinal system of guinea pig. The P2Y(12) receptor-ir neurons coexpressing calbindin-ir in the small intestine are Dogiel type II/AH, intrinsic primary afferent neurons.
Collapse
Affiliation(s)
- Zhenghua Xiang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, 200433, Shanghai, People's Republic of China
| | | |
Collapse
|
44
|
Furuya K, Sokabe M, Furuya S. Characteristics of subepithelial fibroblasts as a mechano-sensor in the intestine: cell-shape-dependent ATP release and P2Y1 signaling. J Cell Sci 2005; 118:3289-304. [PMID: 16030139 DOI: 10.1242/jcs.02453] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Subepithelial fibroblasts form a cellular network just under the epithelium of the gastrointestinal tract. Using primary cultured cells isolated from rat duodenal villi, we previously found that subepithelial fibroblasts reversibly changed cell morphology between flat and stellate-shape depending on intracellular cAMP levels. In this paper, we examined cell-cell communication via released ATP and Ca2+ signaling in the cellular network. Subepithelial fibroblasts were sensitive to mechanical stress such as ;touching' a cell with a fine glass rod and ;stretching' cells cultured on elastic silicone chamber. Mechanical stimulations evoked Ca2+-increase in the cells and ATP-release from the cells. The released ATP activated P2Y receptors on the surrounding cells and propagated Ca2+-waves through the network. Concomitant with Ca2+-waves, a transient contraction of the network was observed. Histochemical, RT-PCR, western blotting and Ca2+ response analyses indicated P2Y1 is a dominant functional subtype. ATP-release and Ca2+ signaling were cell-shape dependent, i.e. they were abolished in stellate-shaped cells treated with dBcAMP, and recovered or further enhanced in re-flattened cells treated with endothelin. The response to ATP also decreased in stellate-shaped cells. These findings indicate cAMP-mediated intracellular signaling causes cell-shape change, which accompanies the changes in mechano- and ATP sensitivities. Using a co-culture system of neuronal cells (NG108-15) with subepithelial fibroblasts, we confirmed that mechanically induced Ca2+-waves propagated to neurons. From these findings we propose that subepithelial fibroblasts work as a mechanosensor in the intestine. Uptake of food, water and nutrients may cause mechanical stress on subepithelial fibroblasts in the villi. The ATP released by mechanical stimulation elicits Ca2+-wave propagation through the network via P2Y1 activation and also activates P2X on terminals of mucosal sensory neurons to regulate peristaltic motility.
Collapse
Affiliation(s)
- Kishio Furuya
- Cell Mechano-Sensing Project, ICORP and SORST, Japan Science and Technology Agency, Nagoya, 466-8550, Japan.
| | | | | |
Collapse
|
45
|
Christofi FL, Kim M, Wunderlich JE, Xue J, Suntres Z, Cardounel A, Javed NH, Yu JG, Grants I, Cooke HJ. Endogenous adenosine differentially modulates 5-hydroxytryptamine release from a human enterochromaffin cell model. Gastroenterology 2004; 127:188-202. [PMID: 15236185 DOI: 10.1053/j.gastro.2004.04.070] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The aim was to determine whether adenosine receptors modulate cAMP, intracellular free calcium ([Ca(2+)](i)), and 5-hydroxytryptamine (5-HT) release in human carcinoid BON cells. METHODS Adenosine receptor (R) mRNA, proteins, and function were identified by Western blots, immunofluorescent labeling, Fluo-4/AM [Ca(2+)](i) imaging, and pharmacologic/physiologic techniques. RESULTS A1, A2, and A3Rs were present in BON cells and carcinoid tumors. Baseline 5-HT levels increased with adenosine deaminase, activation of A2Rs, and inhibition of A3Rs, whereas A3R activation decreased 5-HT. A2R antagonists or blockade of adenosine reuptake that elevates extracellular adenosine reduced mechanically evoked 5-HT release. In single BON cells, touch elevated [Ca(2+)](i) responses were augmented by adenosine deaminase, A1, and A3R antagonists. CONCLUSIONS Tonic or mechanically evoked release of endogenous adenosine is a critical determinant of differential activation of adenosine receptors and may have important implications for gut mechanosensory reflexes.
Collapse
Affiliation(s)
- Fievos L Christofi
- Department of Anesthesiology, The Ohio State University, Columbus 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Monro RL, Bertrand PP, Bornstein JC. ATP participates in three excitatory postsynaptic potentials in the submucous plexus of the guinea pig ileum. J Physiol 2004; 556:571-84. [PMID: 14966305 PMCID: PMC1664948 DOI: 10.1113/jphysiol.2004.060848] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Synaptic transmission between neurones intrinsic to the wall of the intestine involves multiple neurotransmitters. This study aimed to identify neurotransmitters responsible for non-cholinergic excitatory synaptic transmission in the submucous plexus of the guinea pig ileum. Intracellular recordings were made from secretomotor and vasodilator neurones. A single electrical stimulus to a fibre tract evoked excitatory postsynaptic potentials (EPSPs) with three different time courses - fast, slow and an EPSP with an intermediate time course (latency 96 ms, duration 1.2 s). In all neurones, blocking nicotinic receptors reduced fast EPSPs, but they were abolished in only 57 of 78 neurones. Fast EPSPs were also reduced by P2 purinoceptor blockade (5 of 27 neurones) or 5-HT(3) receptor blockade (3 of 20 neurones). The intermediate EPSP was abolished by P2 receptor blockade (13 of 13 neurones) or by the specific P2Y(1) receptor antagonist MRS 2179 (5 of 5 neurones) and was always preceded by a nicotinic or mixed nicotinic/purinergic fast EPSP. Intermediate EPSPs were observed in over half of all neurones including most non-cholinergic secretomotor neurones identified by immunoreactivity for vasoactive intestinal peptide. The slow EPSP evoked by a single pulse stimulus was also abolished by P2 receptor blockade (5 of 5 neurones) or by MRS 2179 (3 of 3 neurones). We conclude that fast EPSPs in submucous neurones are mediated by acetylcholine acting at nicotinic receptors, ATP acting at P2X receptors and 5-HT acting at 5-HT(3) receptors. Both the intermediate EPSP and the single stimulus slow EPSP are mediated by ATP acting at P2Y(1) receptors.
Collapse
Affiliation(s)
- R L Monro
- Department of Physiology, University of Melbourne, Parkville 3010, Victoria, Australia.
| | | | | |
Collapse
|