1
|
Choi NR, Ko SJ, Nam JH, Choi WG, Lee JH, Nah SY, Park JW, Kim BJ. Study on the Therapeutic Effects and Mechanisms of Gintonin in Irritable Bowel Syndrome and Its Relationship with TRPV1, TRPV4, and NaV1.5. Pharmaceuticals (Basel) 2024; 17:1170. [PMID: 39338333 PMCID: PMC11435028 DOI: 10.3390/ph17091170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Irritable bowel syndrome (IBS) is a gastrointestinal (GI) disease accompanied by changes in bowel habits without any specific cause. Gintonin is a newly isolated glycoprotein from ginseng that is a lysophosphatidic acid (LPA) receptor ligand. To investigate the efficacy and mechanisms of action of gintonin in IBS, we developed a zymosan-induced IBS murine model. In addition, electrophysiological experiments were conducted to confirm the relevance of various ion channels. In mice, gintonin restored colon length and weight to normal and decreased stool scores, whilst food intake remained constant. Colon mucosal thickness and inflammation-related tumor necrosis factor-α levels were decreased by gintonin, along with a reduction in pain-related behaviors. In addition, the fecal microbiota from gintonin-treated mice had relatively more Lactobacillaceae and Lachnospiraceae and less Bacteroidaceae than microbiota from the control mice. Moreover, gintonin inhibited transient receptor potential vanilloid (TRPV) 1 and TRPV4 associated with visceral hypersensitivity and voltage-gated Na+ 1.5 channels associated with GI function. These results suggest that gintonin may be one of the effective components in the treatment of IBS.
Collapse
Affiliation(s)
- Na-Ri Choi
- Department of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea; (N.-R.C.); (W.-G.C.)
- Department of Korean Medical Science, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea
| | - Seok-Jae Ko
- Department of Clinical Korean Medicine, Graduate School of Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joo-Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Kyungju 38066, Republic of Korea;
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Woo-Gyun Choi
- Department of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea; (N.-R.C.); (W.-G.C.)
| | - Jong-Hwan Lee
- Department of Biomedical Engineering, Dong-Eui University College of Engineering, Busan 47340, Republic of Korea;
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
| | - Jae-Woo Park
- Department of Clinical Korean Medicine, Graduate School of Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Byung-Joo Kim
- Department of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612, Republic of Korea; (N.-R.C.); (W.-G.C.)
| |
Collapse
|
2
|
Du P, Joshi V, Beyder A. Tracking Gut Motility in Organ and Cultures. Methods Mol Biol 2023; 2644:449-466. [PMID: 37142940 DOI: 10.1007/978-1-0716-3052-5_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Gastrointestinal (GI) motility is a key component of digestive health, and it is complex, involving a multitude of cell types and mechanisms to drive both rhythmic and arrhythmic activity. Tracking GI motility in organ and tissue cultures across multiple temporal (seconds, minutes, hours, days) scales can provide valuable information regarding dysmotility and to evaluate treatment options. Here, the chapter describes a simple method to monitor GI motility in organotypic cultures, using a single video camera is placed perpendicularly to the surface of the tissue. A cross-correlational analysis is used to track the relative movements of tissues between subsequent frames and subsequent fitting procedures to fit finite element functions to the deformed tissue to calculate the strain fields. Additional motility index measures from the displacement information are used to further quantify the behaviors of the tissues that are maintained in organotypic culture over days. The protocols presented in this chapter can be adapted to study organotypic cultures from other organs.
Collapse
Affiliation(s)
- Peng Du
- Auckland Bioengineering Institute, Department of Engineering Science and Biomedical Engineering, University of Auckland, Auckland, New Zealand.
| | - Vikram Joshi
- Department of Physiology and Biomedical Engineering, Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Arthur Beyder
- Department of Physiology and Biomedical Engineering, Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Yang H, Hou C, Xiao W, Qiu Y. The role of mechanosensitive ion channels in the gastrointestinal tract. Front Physiol 2022; 13:904203. [PMID: 36060694 PMCID: PMC9437298 DOI: 10.3389/fphys.2022.904203] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Mechanosensation is essential for normal gastrointestinal (GI) function, and abnormalities in mechanosensation are associated with GI disorders. There are several mechanosensitive ion channels in the GI tract, namely transient receptor potential (TRP) channels, Piezo channels, two-pore domain potassium (K2p) channels, voltage-gated ion channels, large-conductance Ca2+-activated K+ (BKCa) channels, and the cystic fibrosis transmembrane conductance regulator (CFTR). These channels are located in many mechanosensitive intestinal cell types, namely enterochromaffin (EC) cells, interstitial cells of Cajal (ICCs), smooth muscle cells (SMCs), and intrinsic and extrinsic enteric neurons. In these cells, mechanosensitive ion channels can alter transmembrane ion currents in response to mechanical forces, through a process known as mechanoelectrical coupling. Furthermore, mechanosensitive ion channels are often associated with a variety of GI tract disorders, including irritable bowel syndrome (IBS) and GI tumors. Mechanosensitive ion channels could therefore provide a new perspective for the treatment of GI diseases. This review aims to highlight recent research advances regarding the function of mechanosensitive ion channels in the GI tract. Moreover, it outlines the potential role of mechanosensitive ion channels in related diseases, while describing the current understanding of interactions between the GI tract and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Haoyu Yang
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Chaofeng Hou
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Army Medical University, Chongqing, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuan Qiu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
- *Correspondence: Yuan Qiu,
| |
Collapse
|
4
|
Cowan LM, Strege PR, Rusinova R, Andersen OS, Farrugia G, Beyder A. Capsaicin as an amphipathic modulator of Na V1.5 mechanosensitivity. Channels (Austin) 2022; 16:9-26. [PMID: 35412435 PMCID: PMC9009938 DOI: 10.1080/19336950.2022.2026015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
SCN5A-encoded NaV1.5 is a voltage-gated Na+ channel that drives the electrical excitability of cardiac myocytes and contributes to slow waves of the human gastrointestinal smooth muscle cells. NaV1.5 is mechanosensitive: mechanical force modulates several facets of NaV1.5’s voltage-gated function, and some NaV1.5 channelopathies are associated with abnormal NaV1.5 mechanosensitivity (MS). A class of membrane-active drugs, known as amphiphiles, therapeutically target NaV1.5’s voltage-gated function and produce off-target effects including alteration of MS. Amphiphiles may provide a novel option for therapeutic modulation of NaV1.5’s mechanosensitive operation. To more selectively target NaV1.5 MS, we searched for a membrane-partitioning amphipathic agent that would inhibit MS with minimal closed-state inhibition of voltage-gated currents. Among the amphiphiles tested, we selected capsaicin for further study. We used two methods to assess the effects of capsaicin on NaV1.5 MS: (1) membrane suction in cell-attached macroscopic patches and (2) fluid shear stress on whole cells. We tested the effect of capsaicin on NaV1.5 MS by examining macro-patch and whole-cell Na+ current parameters with and without force. Capsaicin abolished the pressure- and shear-mediated peak current increase and acceleration; and the mechanosensitive shifts in the voltage-dependence of activation (shear) and inactivation (pressure and shear). Exploring the recovery from inactivation and use-dependent entry into inactivation, we found divergent stimulus-dependent effects that could potentiate or mitigate the effect of capsaicin, suggesting that mechanical stimuli may differentially modulate NaV1.5 MS. We conclude that selective modulation of NaV1.5 MS makes capsaicin a promising candidate for therapeutic interventions targeting MS.
Collapse
Affiliation(s)
- Luke M Cowan
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Peter R Strege
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Radda Rusinova
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Olaf S Andersen
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| | - Arthur Beyder
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program (ENSP), Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Mn, USA
| |
Collapse
|
5
|
Huizinga JD, Hussain A, Chen JH. Interstitial cells of Cajal and human colon motility in health and disease. Am J Physiol Gastrointest Liver Physiol 2021; 321:G552-G575. [PMID: 34612070 DOI: 10.1152/ajpgi.00264.2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Our understanding of human colonic motility, and autonomic reflexes that generate motor patterns, has increased markedly through high-resolution manometry. Details of the motor patterns are emerging related to frequency and propagation characteristics that allow linkage to interstitial cells of Cajal (ICC) networks. In studies on colonic motor dysfunction requiring surgery, ICC are almost always abnormal or significantly reduced. However, there are still gaps in our knowledge about the role of ICC in the control of colonic motility and there is little understanding of a mechanistic link between ICC abnormalities and colonic motor dysfunction. This review will outline the various ICC networks in the human colon and their proven and likely associations with the enteric and extrinsic autonomic nervous systems. Based on our extensive knowledge of the role of ICC in the control of gastrointestinal motility of animal models and the human stomach and small intestine, we propose how ICC networks are underlying the motor patterns of the human colon. The role of ICC will be reviewed in the autonomic neural reflexes that evoke essential motor patterns for transit and defecation. Mechanisms underlying ICC injury, maintenance, and repair will be discussed. Hypotheses are formulated as to how ICC dysfunction can lead to motor abnormalities in slow transit constipation, chronic idiopathic pseudo-obstruction, Hirschsprung's disease, fecal incontinence, diverticular disease, and inflammatory conditions. Recent studies on ICC repair after injury hold promise for future therapies.
Collapse
Affiliation(s)
- Jan D Huizinga
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Amer Hussain
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Ji-Hong Chen
- Division of Gastroenterology, Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
6
|
Zhao W, Zhang L, Ermilov LG, Colmenares Aguilar MG, Linden DR, Eisenman ST, Romero MF, Farrugia G, Sha L, Gibbons SJ. Bicarbonate ion transport by the electrogenic Na + /HCO 3- cotransporter, NBCe1, is required for normal electrical slow-wave activity in mouse small intestine. Neurogastroenterol Motil 2021; 33:e14149. [PMID: 33837991 PMCID: PMC8485339 DOI: 10.1111/nmo.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/26/2021] [Accepted: 03/21/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Normal gastrointestinal motility depends on electrical slow-wave activity generated by interstitial cells of Cajal (ICC) in the tunica muscularis of the gastrointestinal tract. A requirement for HCO3- in extracellular solutions used to record slow waves indicates a role for HCO3- transport in ICC pacemaking. The Slc4a4 gene transcript encoding the electrogenic Na+ /HCO3- cotransporter, NBCe1, is enriched in mouse small intestinal myenteric region ICC (ICC-MY) that generate slow waves. This study aimed to determine how extracellular HCO3- concentrations affect electrical activity in mouse small intestine and to determine the contribution of NBCe1 activity to these effects. METHODS Immunohistochemistry and sharp electrode electrical recordings were used. KEY RESULTS The NBCe1 immunoreactivity was localized to ICC-MY of the tunica muscularis. In sharp electrode electrical recordings, removal of HCO3- from extracellular solutions caused significant, reversible, depolarization of the smooth muscle and a reduction in slow-wave amplitude and frequency. In 100 mM HCO3- , the muscle hyperpolarized and slow wave amplitude and frequency increased. The effects of replacing extracellular Na+ with Li+ , an ion that does not support NBCe1 activity, were similar to, but larger than, the effects of removing HCO3- . There were no additional changes to electrical activity when HCO3- was removed from Li+ containing solutions. The Na+ /HCO3- cotransport inhibitor, S-0859 (30µM) significantly reduced the effect of removing HCO3- on electrical activity. CONCLUSIONS & INFERENCES These studies demonstrate a major role for Na+ /HCO3- cotransport by NBCe1 in electrical activity of mouse small intestine and indicated that regulation of intracellular acid:base homeostasis contributes to generation of normal pacemaker activity in the gastrointestinal tract.
Collapse
Affiliation(s)
- Wenchang Zhao
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Liwen Zhang
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Leonid G. Ermilov
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Maria Gabriela Colmenares Aguilar
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - David R. Linden
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Seth T. Eisenman
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Michael F. Romero
- Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Lei Sha
- Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China.,Corresponding Authors: Simon J Gibbons, Ph.D., Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA. . Telephone: +1 507 284 9652, Lei Sha, M.D., China Medical University, 77 Pu He Road, Shenbei New District, Shenyang, Liaoning Province, P. R. China, 110122, , . Telephone: +86 18900911003
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Corresponding Authors: Simon J Gibbons, Ph.D., Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA. . Telephone: +1 507 284 9652, Lei Sha, M.D., China Medical University, 77 Pu He Road, Shenbei New District, Shenyang, Liaoning Province, P. R. China, 110122, , . Telephone: +86 18900911003
| |
Collapse
|
7
|
Joshi V, Strege PR, Farrugia G, Beyder A. Mechanotransduction in gastrointestinal smooth muscle cells: role of mechanosensitive ion channels. Am J Physiol Gastrointest Liver Physiol 2021; 320:G897-G906. [PMID: 33729004 PMCID: PMC8202201 DOI: 10.1152/ajpgi.00481.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mechanosensation, the ability to properly sense mechanical stimuli and transduce them into physiologic responses, is an essential determinant of gastrointestinal (GI) function. Abnormalities in this process result in highly prevalent GI functional and motility disorders. In the GI tract, several cell types sense mechanical forces and transduce them into electrical signals, which elicit specific cellular responses. Some mechanosensitive cells like sensory neurons act as specialized mechanosensitive cells that detect forces and transduce signals into tissue-level physiological reactions. Nonspecialized mechanosensitive cells like smooth muscle cells (SMCs) adjust their function in response to forces. Mechanosensitive cells use various mechanoreceptors and mechanotransducers. Mechanoreceptors detect and convert force into electrical and biochemical signals, and mechanotransducers amplify and direct mechanoreceptor responses. Mechanoreceptors and mechanotransducers include ion channels, specialized cytoskeletal proteins, cell junction molecules, and G protein-coupled receptors. SMCs are particularly important due to their role as final effectors for motor function. Myogenic reflex-the ability of smooth muscle to contract in response to stretch rapidly-is a critical smooth muscle function. Such rapid mechanotransduction responses rely on mechano-gated and mechanosensitive ion channels, which alter their ion pores' opening in response to force, allowing fast electrical and Ca2+ responses. Although GI SMCs express a variety of such ion channels, their identities remain unknown. Recent advancements in electrophysiological, genetic, in vivo imaging, and multi-omic technologies broaden our understanding of how SMC mechano-gated and mechanosensitive ion channels regulate GI functions. This review discusses GI SMC mechanosensitivity's current developments with a particular emphasis on mechano-gated and mechanosensitive ion channels.
Collapse
Affiliation(s)
- Vikram Joshi
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota
| | - Peter R. Strege
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota,2Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- 1Division of Gastroenterology & Hepatology, Enteric NeuroScience Program (ENSP), Mayo Clinic, Rochester, Minnesota,2Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
8
|
Mazzone A, Strege PR, Gibbons SJ, Alcaino C, Joshi V, Haak AJ, Tschumperlin DJ, Bernard CE, Cima RR, Larson DW, Chua HK, Graham RP, El Refaey M, Mohler PJ, Hayashi Y, Ordog T, Calder S, Du P, Farrugia G, Beyder A. microRNA overexpression in slow transit constipation leads to reduced Na V1.5 current and altered smooth muscle contractility. Gut 2020; 69:868-876. [PMID: 31757880 PMCID: PMC7147984 DOI: 10.1136/gutjnl-2019-318747] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 10/16/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study was designed to evaluate the roles of microRNAs (miRNAs) in slow transit constipation (STC). DESIGN All human tissue samples were from the muscularis externa of the colon. Expression of 372 miRNAs was examined in a discovery cohort of four patients with STC versus three age/sex-matched controls by a quantitative PCR array. Upregulated miRNAs were examined by quantitative reverse transcription qPCR (RT-qPCR) in a validation cohort of seven patients with STC and age/sex-matched controls. The effect of a highly differentially expressed miRNA on a custom human smooth muscle cell line was examined in vitro by RT-qPCR, electrophysiology, traction force microscopy, and ex vivo by lentiviral transduction in rat muscularis externa organotypic cultures. RESULTS The expression of 13 miRNAs was increased in STC samples. Of those miRNAs, four were predicted to target SCN5A, the gene that encodes the Na+ channel NaV1.5. The expression of SCN5A mRNA was decreased in STC samples. Let-7f significantly decreased Na+ current density in vitro in human smooth muscle cells. In rat muscularis externa organotypic cultures, overexpression of let-7f resulted in reduced frequency and amplitude of contraction. CONCLUSIONS A small group of miRNAs is upregulated in STC, and many of these miRNAs target the SCN5A-encoded Na+ channel NaV1.5. Within this set, a novel NaV1.5 regulator, let-7f, resulted in decreased NaV1.5 expression, current density and reduced motility of GI smooth muscle. These results suggest NaV1.5 and miRNAs as novel diagnostic and potential therapeutic targets in STC.
Collapse
Affiliation(s)
- Amelia Mazzone
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter R Strege
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Simon J Gibbons
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Constanza Alcaino
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Vikram Joshi
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Cheryl E Bernard
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert R Cima
- Department of Colon and Rectal Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - David W Larson
- Department of Colon and Rectal Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Heidi K Chua
- Department of Colon and Rectal Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Mona El Refaey
- Departments of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA,Department of Internal Medicine, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Peter J Mohler
- Departments of Physiology and Cell Biology, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Yujiro Hayashi
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamas Ordog
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Stefan Calder
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Peng Du
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| | - Gianrico Farrugia
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA .,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Arthur Beyder
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA .,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Strege PR, Mazzone A, Bernard CE, Neshatian L, Gibbons SJ, Saito YA, Tester DJ, Calvert ML, Mayer EA, Chang L, Ackerman MJ, Beyder A, Farrugia G. Irritable bowel syndrome patients have SCN5A channelopathies that lead to decreased Na V1.5 current and mechanosensitivity. Am J Physiol Gastrointest Liver Physiol 2018; 314:G494-G503. [PMID: 29167113 PMCID: PMC5966747 DOI: 10.1152/ajpgi.00016.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The SCN5A-encoded voltage-gated mechanosensitive Na+ channel NaV1.5 is expressed in human gastrointestinal smooth muscle cells and interstitial cells of Cajal. NaV1.5 contributes to smooth muscle electrical slow waves and mechanical sensitivity. In predominantly Caucasian irritable bowel syndrome (IBS) patient cohorts, 2-3% of patients have SCN5A missense mutations that alter NaV1.5 function and may contribute to IBS pathophysiology. In this study we examined a racially and ethnically diverse cohort of IBS patients for SCN5A missense mutations, compared them with IBS-negative controls, and determined the resulting NaV1.5 voltage-dependent and mechanosensitive properties. All SCN5A exons were sequenced from somatic DNA of 252 Rome III IBS patients with diverse ethnic and racial backgrounds. Missense mutations were introduced into wild-type SCN5A by site-directed mutagenesis and cotransfected with green fluorescent protein into HEK-293 cells. NaV1.5 voltage-dependent and mechanosensitive functions were studied by whole cell electrophysiology with and without shear force. Five of 252 (2.0%) IBS patients had six rare SCN5A mutations that were absent in 377 IBS-negative controls. Six of six (100%) IBS-associated NaV1.5 mutations had voltage-dependent gating abnormalities [current density reduction (R225W, R433C, R986Q, and F1293S) and altered voltage dependence (R225W, R433C, R986Q, G1037V, and F1293S)], and at least one kinetic parameter was altered in all mutations. Four of six (67%) IBS-associated SCN5A mutations (R225W, R433C, R986Q, and F1293S) resulted in altered NaV1.5 mechanosensitivity. In this racially and ethnically diverse cohort of IBS patients, we show that 2% of IBS patients harbor SCN5A mutations that are absent in IBS-negative controls and result in NaV1.5 channels with abnormal voltage-dependent and mechanosensitive function. NEW & NOTEWORTHY The voltage-gated Na+ channel NaV1.5 contributes to smooth muscle physiology and electrical slow waves. In a racially and ethnically mixed irritable bowel syndrome cohort, 2% had mutations in the NaV1.5 gene SCN5A. These mutations were absent in irritable bowel syndrome-negative controls. Most mutant NaV1.5 channels were loss of function in voltage dependence or mechanosensitivity.
Collapse
Affiliation(s)
- Peter R. Strege
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Amelia Mazzone
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Cheryl E. Bernard
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Leila Neshatian
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Yuri A. Saito
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - David J. Tester
- 2Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Melissa L. Calvert
- 2Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Emeran A. Mayer
- 3Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California Los Angeles, Los Angeles, California
| | - Lin Chang
- 3Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California Los Angeles, Los Angeles, California
| | - Michael J. Ackerman
- 2Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- 1Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
10
|
Erickson A, Deiteren A, Harrington AM, Garcia‐Caraballo S, Castro J, Caldwell A, Grundy L, Brierley SM. Voltage-gated sodium channels: (Na V )igating the field to determine their contribution to visceral nociception. J Physiol 2018; 596:785-807. [PMID: 29318638 PMCID: PMC5830430 DOI: 10.1113/jp273461] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic visceral pain, altered motility and bladder dysfunction are common, yet poorly managed symptoms of functional and inflammatory disorders of the gastrointestinal and urinary tracts. Recently, numerous human channelopathies of the voltage-gated sodium (NaV ) channel family have been identified, which induce either painful neuropathies, an insensitivity to pain, or alterations in smooth muscle function. The identification of these disorders, in addition to the recent utilisation of genetically modified NaV mice and specific NaV channel modulators, has shed new light on how NaV channels contribute to the function of neuronal and non-neuronal tissues within the gastrointestinal tract and bladder. Here we review the current pre-clinical and clinical evidence to reveal how the nine NaV channel family members (NaV 1.1-NaV 1.9) contribute to abdominal visceral function in normal and disease states.
Collapse
Affiliation(s)
- Andelain Erickson
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Annemie Deiteren
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Andrea M. Harrington
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Sonia Garcia‐Caraballo
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Joel Castro
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Ashlee Caldwell
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Luke Grundy
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Stuart M. Brierley
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| |
Collapse
|
11
|
Yeoh JW, Corrias A, Buist ML. Modelling Human Colonic Smooth Muscle Cell Electrophysiology. Cell Mol Bioeng 2017; 10:186-197. [PMID: 31719859 DOI: 10.1007/s12195-017-0479-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 01/28/2017] [Indexed: 12/13/2022] Open
Abstract
The colon is a digestive organ that is subject to a wide range of motility disorders. However, our understanding of the etiology of these disorders is far from complete. In this study, a quantitative single cell model has been developed to describe the electrical behaviour of a human colonic smooth muscle cell (hCSMC). This model includes the pertinent ionic channels and intracellular calcium homoeostasis. These components are believed to contribute significantly to the electrical response of the hCSMC during a slow wave. The major ion channels were constructed based on published data recorded from isolated human colonic myocytes. The whole cell model is able to reproduce experimentally recorded slow waves from human colonic muscles. This represents the first biophysically-detailed model of a hCSMC and provides a means to better understand colonic disorders.
Collapse
Affiliation(s)
- Jing Wui Yeoh
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Block E4, #04-08, 4 Engineering Drive 3, Singapore, 117583 Singapore
| | - Alberto Corrias
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Block E4, #04-08, 4 Engineering Drive 3, Singapore, 117583 Singapore
| | - Martin L Buist
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Block E4, #04-08, 4 Engineering Drive 3, Singapore, 117583 Singapore
| |
Collapse
|
12
|
Beyder A, Gibbons SJ, Mazzone A, Strege PR, Saravanaperumal SA, Sha L, Higgins S, Eisenman ST, Bernard CE, Geurts A, Kline CF, Mohler PJ, Farrugia G. Expression and function of the Scn5a-encoded voltage-gated sodium channel NaV 1.5 in the rat jejunum. Neurogastroenterol Motil 2016; 28:64-73. [PMID: 26459913 PMCID: PMC4688076 DOI: 10.1111/nmo.12697] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/05/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND The SCN5A-encoded voltage-gated sodium channel NaV 1.5 is expressed in human jejunum and colon. Mutations in NaV 1.5 are associated with gastrointestinal motility disorders. The rat gastrointestinal tract expresses voltage-gated sodium channels, but their molecular identity and role in rat gastrointestinal electrophysiology are unknown. METHODS The presence and distribution of Scn5a-encoded NaV 1.5 was examined by PCR, Western blotting and immunohistochemistry in rat jejunum. Freshly dissociated smooth muscle cells were examined by whole cell electrophysiology. Zinc finger nuclease was used to target Scn5a in rats. Lentiviral-mediated transduction with shRNA was used to target Scn5a in rat jejunum smooth muscle organotypic cultures. Organotypic cultures were examined by sharp electrode electrophysiology and RT-PCR. KEY RESULTS We found NaV 1.5 in rat jejunum and colon smooth muscle by Western blot. Immunohistochemistry using two other antibodies of different portions of NaV 1.5 revealed the presence of the ion channel in rat jejunum. Whole cell voltage-clamp in dissociated smooth muscle cells from rat jejunum showed fast activating and inactivating voltage-dependent inward current that was eliminated by Na(+) replacement by NMDG(+) . Constitutive rat Scn5a knockout resulted in death in utero. NaV 1.5 shRNA delivered by lentivirus into rat jejunum smooth muscle organotypic culture resulted in 57% loss of Scn5a mRNA and several significant changes in slow waves, namely 40% decrease in peak amplitude, 30% decrease in half-width, and 7 mV hyperpolarization of the membrane potential at peak amplitude. CONCLUSIONS & INFERENCES Scn5a-encoded NaV 1.5 is expressed in rat gastrointestinal smooth muscle and it contributes to smooth muscle electrophysiology.
Collapse
Affiliation(s)
- A Beyder
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S J Gibbons
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A Mazzone
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - P R Strege
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S A Saravanaperumal
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - L Sha
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S Higgins
- College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - S T Eisenman
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - C E Bernard
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - A Geurts
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - C F Kline
- The Dorothy M. Davis Heart and Lung Research Institute and Departments of Physiology & Cell Biology and Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - P J Mohler
- The Dorothy M. Davis Heart and Lung Research Institute and Departments of Physiology & Cell Biology and Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - G Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
13
|
Neshatian L, Strege PR, Rhee PL, Kraichely RE, Mazzone A, Bernard CE, Cima RR, Larson DW, Dozois EJ, Kline CF, Mohler PJ, Beyder A, Farrugia G. Ranolazine inhibits voltage-gated mechanosensitive sodium channels in human colon circular smooth muscle cells. Am J Physiol Gastrointest Liver Physiol 2015; 309:G506-12. [PMID: 26185330 PMCID: PMC4572410 DOI: 10.1152/ajpgi.00051.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/09/2015] [Indexed: 01/31/2023]
Abstract
Human jejunum smooth muscle cells (SMCs) and interstitial cells of Cajal (ICCs) express the SCN5A-encoded voltage-gated, mechanosensitive sodium channel NaV1.5. NaV1.5 contributes to small bowel excitability, and NaV1.5 inhibitor ranolazine produces constipation by an unknown mechanism. We aimed to determine the presence and molecular identity of Na(+) current in the human colon smooth muscle and to examine the effects of ranolazine on Na(+) current, mechanosensitivity, and smooth muscle contractility. Inward currents were recorded by whole cell voltage clamp from freshly dissociated human colon SMCs at rest and with shear stress. SCN5A mRNA and NaV1.5 protein were examined by RT-PCR and Western blots, respectively. Ascending human colon strip contractility was examined in a muscle bath preparation. SCN5A mRNA and NaV1.5 protein were identified in human colon circular muscle. Freshly dissociated human colon SMCs had Na(+) currents (-1.36 ± 0.36 pA/pF), shear stress increased Na(+) peaks by 17.8 ± 1.8% and accelerated the time to peak activation by 0.7 ± 0.3 ms. Ranolazine (50 μM) blocked peak Na(+) current by 43.2 ± 9.3% and inhibited shear sensitivity by 25.2 ± 3.2%. In human ascending colon strips, ranolazine decreased resting tension (31%), reduced the frequency of spontaneous events (68%), and decreased the response to smooth muscle electrical field stimulation (61%). In conclusion, SCN5A-encoded NaV1.5 is found in human colonic circular smooth muscle. Ranolazine blocks both peak amplitude and mechanosensitivity of Na(+) current in human colon SMCs and decreases contractility of human colon muscle strips. Our data provide a likely mechanistic explanation for constipation induced by ranolazine.
Collapse
Affiliation(s)
- Leila Neshatian
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Peter R. Strege
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Poong-Lyul Rhee
- 4Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea; and
| | - Robert E. Kraichely
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Amelia Mazzone
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Cheryl E. Bernard
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Robert R. Cima
- 3Department of Colon and Rectal Surgery, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - David W. Larson
- 3Department of Colon and Rectal Surgery, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Eric J. Dozois
- 3Department of Colon and Rectal Surgery, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Crystal F. Kline
- 5The Dorothy M. Davis Heart and Lung Research Institute and Departments of Internal Medicine and Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Peter J. Mohler
- 5The Dorothy M. Davis Heart and Lung Research Institute and Departments of Internal Medicine and Physiology & Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Arthur Beyder
- 1Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; ,2Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Mayo Clinic College of Medicine, Rochester, Minnesota; Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota;
| |
Collapse
|
14
|
Verstraelen TE, Ter Bekke RMA, Volders PGA, Masclee AAM, Kruimel JW. The role of the SCN5A-encoded channelopathy in irritable bowel syndrome and other gastrointestinal disorders. Neurogastroenterol Motil 2015; 27:906-13. [PMID: 25898860 DOI: 10.1111/nmo.12569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 03/17/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Gastrointestinal functional and motility disorders, like irritable bowel syndrome (IBS), have a high prevalence in the Western population and cause significant morbidity and loss of quality of life leading to considerable costs for health care. A decade ago, it has been demonstrated that interstitial cells of Cajal and intestinal smooth muscle cells, cells important for gastrointestinal motility, express the sodium channel alpha subunit Nav 1.5. In the heart, aberrant variants in this sodium channel, encoded by SCN5A, are linked to inherited arrhythmia syndromes, like the long-QT syndrome type 3 and Brugada syndrome. Mounting data show a possible contribution of SCN5A mutants to gastrointestinal functional and motility disorders. Two percent of IBS patients harbor SCN5A mutations with electrophysiological evidence of loss- and gain-of-function. In addition, gastrointestinal symptoms are more prevalent in cardiac SCN5A-mutation positive patients. PURPOSE This review firstly describes the Nav 1.5 channel and its physiological role in ventricular cardiomyocytes and gastrointestinal cells, then we focus on the involvement of mutant Nav 1.5 in gastrointestinal functional and motility disorders. Future research might uncover novel mutation-specific treatment strategies for SCN5A-encoded gastrointestinal channelopathies.
Collapse
Affiliation(s)
- T E Verstraelen
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - R M A Ter Bekke
- Department of Cardiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - P G A Volders
- Department of Cardiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - A A M Masclee
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - J W Kruimel
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
15
|
Mazet B. Gastrointestinal motility and its enteric actors in mechanosensitivity: past and present. Pflugers Arch 2014; 467:191-200. [PMID: 25366494 DOI: 10.1007/s00424-014-1635-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 10/14/2014] [Accepted: 10/19/2014] [Indexed: 12/14/2022]
Abstract
Coordinated contractions of the smooth muscle layers of the gastrointestinal (GI) tract are required to produce motor patterns that ensure normal GI motility. The crucial role of the enteric nervous system (ENS), the intrinsic ganglionated network located within the GI wall, has long been recognized in the generation of the main motor patterns. However, devising an appropriate motility requires the integration of informations emanating from the lumen of the GI tract. As already found more than half a century ago, the ability of the GI tract to respond to mechanical forces such as stretch is not restricted to neuronal mechanisms. Instead, mechanosensitivity is now recognized as a property of several non-neuronal cell types, the excitability of which is probably involved in shaping the motor patterns. This brief review gives an overview on how mechanosensitivity of different cell types in the GI tract has been established and, whenever available, on what ionic conductances are involved in mechanotransduction and their potential impact on normal GI motility.
Collapse
Affiliation(s)
- Bruno Mazet
- Aix Marseille Université, CNRS, CRN2M UMR 7286, CS80011 Bd Pierre Dramard, 13344, Marseille Cedex 15, France,
| |
Collapse
|
16
|
Youm JB, Leem CH, Lee SR, Song IS, Kim HK, Heo HJ, Kim BJ, Kim N, Han J. Modeling of stochastic behavior of pacemaker potential in interstitial cells of Cajal. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2014; 116:56-69. [PMID: 25238716 DOI: 10.1016/j.pbiomolbio.2014.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 08/28/2014] [Accepted: 09/06/2014] [Indexed: 01/20/2023]
Abstract
It is widely accepted that interstitial cells of Cajal (ICCs) generate pacemaker potentials to propagate slow waves along the whole gastrointestinal tract. Previously, we constructed a biophysically based model of ICCs in mouse small intestine to explain the pacemaker mechanism. Our previous model, however, could not explain non-uniformity of pacemaker potentials and random occurrence of unitary potentials, thus we updated our model. The inositol 1,4,5-trisphosphate (IP3)-mediated Ca(2+) mobilization is a key event to drive the cycle of pacemaker activity and was updated to reproduce its stochastic behavior. The stochasticity was embodied by simulating random opening and closing of individual IP3-mediated Ca(2+) channel. The updated model reproduces the stochastic features of pacemaker potentials in ICCs. Reproduced pacemaker potentials are not uniform in duration and interval. The resting and peak potentials are -75.5 ± 1.1 mV and -0.8 ± 0.5 mV, respectively (n = 55). Frequency of pacemaker potential is 14.3 ± 0.4 min(-1) (n = 10). Width at half-maximal amplitude of pacemaker potential is 902 ± 6 ms (n = 55). There are random events of unitary potential-like depolarization. Finally, we compared our updated model with a recently published model to speculate which ion channel is the best candidate to drive pacemaker depolarization. In conclusion, our updated mathematical model could now reproduce stochastic features of pacemaker activity in ICCs.
Collapse
Affiliation(s)
- Jae Boum Youm
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea.
| | - Chae Hun Leem
- Department of Physiology University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung Ryul Lee
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - In-Sung Song
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - Hyoung Kyu Kim
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - Hye Jin Heo
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan, South Korea
| | - Nari Kim
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan, South Korea
| |
Collapse
|
17
|
Beyder A, Mazzone A, Strege PR, Tester DJ, Saito YA, Bernard CE, Enders FT, Ek WE, Schmidt PT, Dlugosz A, Lindberg G, Karling P, Ohlsson B, Gazouli M, Nardone G, Cuomo R, Usai-Satta P, Galeazzi F, Neri M, Portincasa P, Bellini M, Barbara G, Camilleri M, Locke GR, Talley NJ, D'Amato M, Ackerman MJ, Farrugia G. Loss-of-function of the voltage-gated sodium channel NaV1.5 (channelopathies) in patients with irritable bowel syndrome. Gastroenterology 2014; 146:1659-1668. [PMID: 24613995 PMCID: PMC4096335 DOI: 10.1053/j.gastro.2014.02.054] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 02/06/2014] [Accepted: 02/26/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS SCN5A encodes the α-subunit of the voltage-gated sodium channel NaV1.5. Many patients with cardiac arrhythmias caused by mutations in SCN5A also have symptoms of irritable bowel syndrome (IBS). We investigated whether patients with IBS have SCN5A variants that affect the function of NaV1.5. METHODS We performed genotype analysis of SCN5A in 584 persons with IBS and 1380 without IBS (controls). Mutant forms of SCN5A were expressed in human embryonic kidney-293 cells, and functions were assessed by voltage clamp analysis. A genome-wide association study was analyzed for an association signal for the SCN5A gene, and replicated in 1745 patients in 4 independent cohorts of IBS patients and controls. RESULTS Missense mutations were found in SCN5A in 13 of 584 patients (2.2%, probands). Diarrhea-predominant IBS was the most prevalent form of IBS in the overall study population (25%). However, a greater percentage of individuals with SCN5A mutations had constipation-predominant IBS (31%) than diarrhea-predominant IBS (10%; P < .05). Electrophysiologic analysis showed that 10 of 13 detected mutations disrupted NaV1.5 function (9 loss-of-function and 1 gain-of-function function). The p. A997T-NaV1.5 had the greatest effect in reducing NaV1.5 function. Incubation of cells that expressed this variant with mexiletine restored their sodium current and administration of mexiletine to 1 carrier of this mutation (who had constipation-predominant IBS) normalized their bowel habits. In the genome-wide association study and 4 replicated studies, the SCN5A locus was strongly associated with IBS. CONCLUSIONS About 2% of patients with IBS carry mutations in SCN5A. Most of these are loss-of-function mutations that disrupt NaV1.5 channel function. These findings provide a new pathogenic mechanism for IBS and possible treatment options.
Collapse
Affiliation(s)
- Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Amelia Mazzone
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Peter R. Strege
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - David J. Tester
- Departments of Medicine (Cardiovascular Diseases), Pediatrics (Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics and the Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Yuri A. Saito
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Cheryl E. Bernard
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Felicity T. Enders
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Weronica E Ek
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Peter T Schmidt
- Department of Gastroenterology and Hepatology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Aldona Dlugosz
- Department of Gastroenterology and Hepatology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Greger Lindberg
- Department of Gastroenterology and Hepatology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Bodil Ohlsson
- Department of Clinical Sciences, Skånes University Hospital, Malmoe, Sweden
| | - Maria Gazouli
- Laboratory of Biology, School of Medicine, University of Athens, Athens, Greece
| | - Gerardo Nardone
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Rosario Cuomo
- Digestive Motility Diseases, Department of Clinical Medicine and Surgery, Federico II University Hospital , Naples, Italy
| | - Paolo Usai-Satta
- S.C. Gastroenterologia, Azienda Ospedaliera G. Brotzu, Cagliari, Italy
| | | | - Matteo Neri
- Department of Medicine and Aging Sciences and CESI, G. D'Annunzio University & Foundation, Chieti, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology (DIMO), Clinica Medica ‘A. Murri', University of Bari Medical School, Bari, Italy
| | - Massimo Bellini
- Gastroenterology Unit, Department of Gastroenterology, University of Pisa, Pisa, Italy
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, St. Orsola - Malpighi Hospital, Bologna, Italy
| | - Michael Camilleri
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - G. Richard Locke
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Nicholas J. Talley
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Mauro D'Amato
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Michael J. Ackerman
- Departments of Medicine (Cardiovascular Diseases), Pediatrics (Pediatric Cardiology), and Molecular Pharmacology & Experimental Therapeutics and the Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, Minnesota;,Corresponding authors: Gianrico Farrugia, MD and Michael J. Ackerman MD, PhD Mayo Clinic 200 First Street SW, Rochester, Minnesota 55905 Phone: 507-284-4695. Fax: 507-284-0266.
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Division of Gastroenterology &Hepatology, Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Corresponding authors: Gianrico Farrugia, MD and Michael J. Ackerman MD, PhD Mayo Clinic 200 First Street SW, Rochester, Minnesota 55905 Phone: 507-284-4695. Fax: 507-284-0266.
| |
Collapse
|
18
|
Bradley E, Webb TI, Hollywood MA, Sergeant GP, McHale NG, Thornbury KD. The cardiac sodium current Na(v)1.5 is functionally expressed in rabbit bronchial smooth muscle cells. Am J Physiol Cell Physiol 2013; 305:C427-35. [PMID: 23784541 DOI: 10.1152/ajpcell.00034.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A collagenase-proteinase mixture was used to isolate airway smooth muscle cells (ASMC) from rabbit bronchi, and membrane currents were recorded using the whole cell patch-clamp technique. Stepping from -100 mV to a test potential of -40 mV evoked a fast voltage-dependent Na(+) current, sometimes with an amplitude of several nanoamperes. The current disappeared within 15 min of exposure to papain + DTT (n = 6). Comparison of the current in ASMC with current mediated by NaV1.5 α-subunits expressed in human embryonic kidney cells revealed similar voltage dependences of activation (V1/2 = -42 mV for NaV1.5) and sensitivities to TTX (IC50 = 1.1 and 1.2 μM for ASMC and NaV1.5, respectively). The current in ASMC was also blocked by lidocaine (IC50 = 160 μM). Although veratridine, an agonist of voltage-gated Na(+) channels, reduced the peak current by 33%, it slowed inactivation, resulting in a fourfold increase in sustained current (measured at 25 ms after onset). In current-clamp mode, veratridine prolonged evoked action potentials from 37 ± 9 to 1,053 ± 410 ms (n = 8). Primers for NaV1.2-1.9 were used to amplify mRNA from groups of ∼20 isolated ASMC and from whole bronchial tissue by RT-PCR. Transcripts for NaV1.2, NaV1.3, and NaV1.5-1.9 were detected in whole tissue, but only NaV1.2 and NaV1.5 were detected in single cells. We conclude that freshly dispersed rabbit ASMC express a fast voltage-gated Na(+) current that is mediated mainly by the NaV1.5 subtype.
Collapse
Affiliation(s)
- E Bradley
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | | | | | | | | | | |
Collapse
|
19
|
Beyder A, Strege PR, Bernard C, Farrugia G. Membrane permeable local anesthetics modulate Na(V)1.5 mechanosensitivity. Channels (Austin) 2012; 6:308-16. [PMID: 22874086 PMCID: PMC3508909 DOI: 10.4161/chan.21202] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Voltage-gated sodium selective ion channel NaV1.5 is expressed in the heart and the gastrointestinal tract, which are mechanically active organs. NaV1.5 is mechanosensitive at stimuli that gate other mechanosensitive ion channels. Local anesthetic and antiarrhythmic drugs act upon NaV1.5 to modulate activity by multiple mechanisms. This study examined whether NaV1.5 mechanosensitivity is modulated by local anesthetics. NaV1.5 channels wereexpressed in HEK-293 cells, and mechanosensitivity was tested in cell-attached and excised inside-out configurations. Using a novel protocol with paired voltage ladders and short pressure pulses, negative patch pressure (-30 mmHg) in both configurations produced a hyperpolarizing shift in the half-point of the voltage-dependence of activation (V1/2a) and inactivation (V1/2i) by about -10 mV. Lidocaine (50 µM) inhibited the pressure-induced shift of V1/2a but not V1/2i. Lidocaine inhibited the tonic increase in pressure-induced peak current in a use-dependence protocol, but it did not otherwise affect use-dependent block. The local anesthetic benzocaine, which does not show use-dependent block, also effectively blocked a pressure-induced shift in V1/2a. Lidocaine inhibited mechanosensitivity in NaV1.5 at the local anesthetic binding site mutated (F1760A). However, a membrane impermeable lidocaine analog QX-314 did not affect mechanosensitivity of F1760A NaV1.5 when applied from either side of the membrane. These data suggest that the mechanism of lidocaine inhibition of the pressure-induced shift in the half-point of voltage-dependence of activation is separate from the mechanisms of use-dependent block. Modulation of NaV1.5 mechanosensitivity by the membrane permeable local anesthetics may require hydrophobic access and may involve membrane-protein interactions.
Collapse
Affiliation(s)
- Arthur Beyder
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | | | | | | |
Collapse
|
20
|
Beyder A, Farrugia G. Targeting ion channels for the treatment of gastrointestinal motility disorders. Therap Adv Gastroenterol 2012; 5:5-21. [PMID: 22282704 PMCID: PMC3263980 DOI: 10.1177/1756283x11415892] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gastrointestinal (GI) functional and motility disorders are highly prevalent and responsible for long-term morbidity and sometimes mortality in the affected patients. It is estimated that one in three persons has a GI functional or motility disorder. However, diagnosis and treatment of these widespread conditions remains challenging. This partly stems from the multisystem pathophysiology, including processing abnormalities in the central and peripheral (enteric) nervous systems and motor dysfunction in the GI wall. Interstitial cells of Cajal (ICCs) are central to the generation and propagation of the cyclical electrical activity and smooth muscle cells (SMCs) are responsible for electromechanical coupling. In these and other excitable cells voltage-sensitive ion channels (VSICs) are the main molecular units that generate and regulate electrical activity. Thus, VSICs are potential targets for intervention in GI motility disorders. Research in this area has flourished with advances in the experimental methods in molecular and structural biology and electrophysiology. However, our understanding of the molecular mechanisms responsible for the complex and variable electrical behavior of ICCs and SMCs remains incomplete. In this review, we focus on the slow waves and action potentials in ICCs and SMCs. We describe the constituent VSICs, which include voltage-gated sodium (Na(V)), calcium (Ca(V)), potassium (K(V), K(Ca)), chloride (Cl(-)) and nonselective ion channels (transient receptor potentials [TRPs]). VSICs have significant structural homology and common functional mechanisms. We outline the approaches and limitations and provide examples of targeting VSICs at the pores, voltage sensors and alternatively spliced sites. Rational drug design can come from an integrated view of the structure and mechanisms of gating and activation by voltage or mechanical stress.
Collapse
Affiliation(s)
- Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
21
|
Squecco R, Garella R, Luciani G, Francini F, Baccari MC. Muscular effects of orexin A on the mouse duodenum: mechanical and electrophysiological studies. J Physiol 2011; 589:5231-46. [PMID: 21911618 DOI: 10.1113/jphysiol.2011.214940] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Orexin A (OXA) has been reported to influence gastrointestinal motility, acting at both central and peripheral neural levels. The aim of the present study was to evaluate whether OXA also exerts direct effects on the duodenal smooth muscle. The possible mechanism of action involved was investigated by employing a combined mechanical and electrophysiological approach. Duodenal segments were mounted in organ baths for isometric recording of the mechanical activity. Ionic channel activity was recorded in current- and voltage-clamp conditions by a single microelectrode inserted in a duodenal longitudinal muscle cell. In the duodenal preparations, OXA (0.3 μM) caused a TTX-insensitive transient contraction. Nifedipine (1 μM), as well as 2-aminoethyl diphenyl borate (10 μM), reduced the amplitude and shortened the duration of the response to OXA, which was abolished by Ni(2+) (50 μM) or TEA (1 mM). Electrophysiological studies in current-clamp conditions showed that OXA caused an early depolarization, which paralleled in time the contractile response, followed by a long-lasting depolarization. Such a depolarization was triggered by activation of receptor-operated Ca(2+) channels and enhanced by activation of T- and L-type Ca(2+) channels and store-operated Ca(2+) channels and by inhibition of K(+) channels. Experiments in voltage-clamp conditions demonstrated that OXA affects not only receptor-operated Ca(2+) channels, but also the maximal conductance and kinetics of activation and inactivation of Na(+), T- and L-type Ca(2+) voltage-gated channels. The results demonstrate, for the first time, that OXA exerts direct excitatory effects on the mouse duodenal smooth muscle. Finally, this work demonstrates new findings related to the expression and kinetics of the voltage-gated channel types, as well as store-operated Ca(2+) channels.
Collapse
Affiliation(s)
- Roberta Squecco
- Dipartimento di Scienze Fisiologiche, Università di Firenze, Firenze, Italy
| | | | | | | | | |
Collapse
|
22
|
Lees-Green R, Du P, O'Grady G, Beyder A, Farrugia G, Pullan AJ. Biophysically based modeling of the interstitial cells of cajal: current status and future perspectives. Front Physiol 2011; 2:29. [PMID: 21772822 PMCID: PMC3131535 DOI: 10.3389/fphys.2011.00029] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 06/13/2011] [Indexed: 12/29/2022] Open
Abstract
Gastrointestinal motility research is progressing rapidly, leading to significant advances in the last 15 years in understanding the cellular mechanisms underlying motility, following the discovery of the central role played by the interstitial cells of Cajal (ICC). As experimental knowledge of ICC physiology has expanded, biophysically based modeling has become a valuable tool for integrating experimental data, for testing hypotheses on ICC pacemaker mechanisms, and for applications in in silico studies including in multiscale models. This review is focused on the cellular electrophysiology of ICC. Recent evidence from both experimental and modeling domains have called aspects of the existing pacemaker theories into question. Therefore, current experimental knowledge of ICC pacemaker mechanisms is examined in depth, and current theories of ICC pacemaking are evaluated and further developed. Existing biophysically based ICC models and their physiological foundations are then critiqued in light of the recent advances in experimental knowledge, and opportunities to improve these models are identified. The review concludes by examining several potential clinical applications of biophysically based ICC modeling from the subcellular through to the organ level, including ion channelopathies and ICC network degradation.
Collapse
Affiliation(s)
- Rachel Lees-Green
- Auckland Bioengineering Institute, The University of Auckland Auckland, New Zealand
| | | | | | | | | | | |
Collapse
|
23
|
Strege PR, Bernard CE, Kraichely RE, Mazzone A, Sha L, Beyder A, Gibbons SJ, Linden DR, Kendrick ML, Sarr MG, Szurszewski JH, Farrugia G. Hydrogen sulfide is a partially redox-independent activator of the human jejunum Na+ channel, Nav1.5. Am J Physiol Gastrointest Liver Physiol 2011; 300:G1105-14. [PMID: 21393430 PMCID: PMC3119119 DOI: 10.1152/ajpgi.00556.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hydrogen sulfide (H(2)S) is produced endogenously by L-cysteine metabolism. H(2)S modulates several ion channels with an unclear mechanism of action. A possible mechanism is through reduction-oxidation reactions attributable to the redox potential of the sulfur moiety. The aims of this study were to determine the effects of the H(2)S donor NaHS on Na(V)1.5, a voltage-dependent sodium channel expressed in the gastrointestinal tract in human jejunum smooth muscle cells and interstitial cells of Cajal, and to elucidate whether H(2)S acts on Na(V)1.5 by redox reactions. Whole cell Na(+) currents were recorded in freshly dissociated human jejunum circular myocytes and Na(V)1.5-transfected human embryonic kidney-293 cells. RT-PCR amplified mRNA for H(2)S enzymes cystathionine β-synthase and cystathionine γ-lyase from the human jejunum. NaHS increased native Na(+) peak currents and shifted the half-point (V(1/2)) of steady-state activation and inactivation by +21 ± 2 mV and +15 ± 3 mV, respectively. Similar effects were seen on the heterologously expressed Na(V)1.5 α subunit with EC(50)s in the 10(-4) to 10(-3) M range. The reducing agent dithiothreitol (DTT) mimicked in part the effects of NaHS by increasing peak current and positively shifting steady-state activation. DTT together with NaHS had an additive effect on steady-state activation but not on peak current, suggesting that the latter may be altered via reduction. Pretreatment with the Hg(2+)-conjugated oxidizer thimerosal or the alkylating agent N-ethylmaleimide inhibited or decreased NaHS induction of Na(V)1.5 peak current. These studies show that H(2)S activates the gastrointestinal Na(+) channel, and the mechanism of action of H(2)S is partially redox independent.
Collapse
Affiliation(s)
- Peter R. Strege
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Cheryl E. Bernard
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Robert E. Kraichely
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Amelia Mazzone
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Lei Sha
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - David R. Linden
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Michael L. Kendrick
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Michael G. Sarr
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Joseph H. Szurszewski
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
24
|
Du P, O'Grady G, Davidson JB, Cheng LK, Pullan AJ. Multiscale modeling of gastrointestinal electrophysiology and experimental validation. Crit Rev Biomed Eng 2011; 38:225-54. [PMID: 21133835 DOI: 10.1615/critrevbiomedeng.v38.i3.10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Normal gastrointestinal (GI) motility results from the coordinated interplay of multiple cooperating mechanisms, both intrinsic and extrinsic to the GI tract. A fundamental component of this activity is an omnipresent electrical activity termed slow waves, which is generated and propagated by the interstitial cells of Cajal (ICCs). The role of ICC loss and network degradation in GI motility disorders is a significant area of ongoing research. This review examines recent progress in the multiscale modeling framework for effectively integrating a vast range of experimental data in GI electrophysiology, and outlines the prospect of how modeling can provide new insights into GI function in health and disease. The review begins with an overview of the GI tract and its electrophysiology, and then focuses on recent work on modeling GI electrical activity, spanning from cell to body biophysical scales. Mathematical cell models of the ICCs and smooth muscle cell are presented. The continuum framework of monodomain and bidomain models for tissue and organ models are then considered, and the forward techniques used to model the resultant body surface potential and magnetic field are discussed. The review then outlines recent progress in experimental support and validation of modeling, and concludes with a discussion on potential future research directions in this field.
Collapse
Affiliation(s)
- Peng Du
- Auckland Bioengineering Institute, The University of Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
25
|
Böttner M, Bär F, Von Koschitzky H, Tafazzoli K, Roblick UJ, Bruch HP, Wedel T. Laser microdissection as a new tool to investigate site-specific gene expression in enteric ganglia of the human intestine. Neurogastroenterol Motil 2010; 22:168-72, e52. [PMID: 19863635 DOI: 10.1111/j.1365-2982.2009.01424.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Myenteric ganglia are key-structures for the control of intestinal motility and their mRNA expression profiles might be altered under pathological conditions. A drawback of conventional RT-PCR from full-thickness specimens is that gene expression analysis is based on heterogeneously composed tissues. To overcome this problem, laser microdissection combined with real-time RT-PCR can be used to detect and quantify low levels of gene expression in isolated enteric ganglia. METHODS Fresh unfixed full-thickness specimens of sigmoid colon were obtained from patients (n = 8) with diseases unrelated to intestinal motility disorders. 10 microm cryo-sections were mounted on membrane-coated slides and ultra-rapidly stained with toluidine blue. Myenteric ganglia were isolated by laser microdissection and catapulting for mRNA isolation. Real-time RT-PCR was performed for selected growth factors, neurotransmitter receptors and specific cell type markers. KEY RESULTS Collection of 0.5 mm(2) of ganglionic tissue was sufficient to obtain positive RT-PCR results. Collection of 4 mm(2) resulted in ct-values allowing a reliable quantitative comparison of gene expression levels. mRNA analysis revealed that neurotrophic growth factor, neurotrophin-3, serotonin receptor 3A, PGP 9.5 and S100 beta are specifically expressed in myenteric ganglia of the human colon. CONCLUSIONS & INFERENCES Laser microdissection combined with real-time RT-PCR is a novel technique to reliably detect and quantify site-specific expression of low-abundance mRNAs (e.g. growth factors, neurotransmitter receptors) related to the human enteric nervous system. This technical approach expands the spectrum of available tools to characterize enteric neuropathologies underlying human gastrointestinal motility disorders at the molecular biological level.
Collapse
Affiliation(s)
- M Böttner
- Institute of Anatomy, University of Kiel, Kiel, Germany.
| | | | | | | | | | | | | |
Collapse
|
26
|
Kraichely RE, Strege PR, Sarr MG, Kendrick ML, Farrugia G. Lysophosphatidyl choline modulates mechanosensitive L-type Ca2+ current in circular smooth muscle cells from human jejunum. Am J Physiol Gastrointest Liver Physiol 2009; 296:G833-9. [PMID: 19179622 PMCID: PMC2670668 DOI: 10.1152/ajpgi.90610.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The L-type Ca2+ channel expressed in gastrointestinal smooth muscle is mechanosensitive. Direct membrane stretch and shear stress result in increased Ca2+ entry into the cell. The mechanism for mechanosensitivity is not known, and mechanosensitivity is not dependent on an intact cytoskeleton. The aim of this study was to determine whether L-type Ca2+ channel mechanosensitivity is dependent on tension in the lipid bilayer in human jejunal circular layer myocytes. Whole cell currents were recorded in the amphotericin-perforated-patch configuration, and lysophosphatidyl choline (LPC), lysophosphatidic acid (LPA), and choline were used to alter differentially the tension in the lipid bilayer. Shear stress (perfusion at 10 ml/min) was used to mechanostimulate L-type Ca2+ channels. The increase in L-type Ca2+ current induced by shear stress was greater in the presence of LPC (large head-to-tail proportions), but not LPA or choline, than in the control perfusion. The increased peak Ca2+ current also did not return to baseline levels as in control conditions. Furthermore, steady-state inactivation kinetics were altered in the presence of LPC, leading to a change in window current. These findings suggest that changes in tension in the plasmalemmal membrane can be transmitted to the mechanosensitive L-type Ca2+ channel, leading to altered activity and Ca2+ entry in the human jejunal circular layer myocyte.
Collapse
Affiliation(s)
- Robert E. Kraichely
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, and Gastroenterologic and General Surgery, Mayo Clinic, Rochester, Minnesota
| | - Peter R. Strege
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, and Gastroenterologic and General Surgery, Mayo Clinic, Rochester, Minnesota
| | - Michael G. Sarr
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, and Gastroenterologic and General Surgery, Mayo Clinic, Rochester, Minnesota
| | - Michael L. Kendrick
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, and Gastroenterologic and General Surgery, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Miles and Shirley Fiterman Center for Digestive Diseases, and Gastroenterologic and General Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
27
|
Abstract
The gastrointestinal tract serves the physiological function of digesting and absorbing nutrients from food and physically mixing and propelling these contents in an oral to anal direction. These functions require the coordinated interaction of several cell types, including enteric nerves, immune cells and smooth muscle. Interstitial cells of Cajal (ICC) are now recognized as another cell type that are required for the normal functioning of the gastrointestinal tract. Abnormalities in ICC numbers and networks are associated with several gastrointestinal motility disorders. This review will describe what is known about the function and role of ICC both in health and in a variety of motility disorders with a focus on unresolved issues pertaining to their role in the control of gastrointestinal motility.
Collapse
Affiliation(s)
- G Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
28
|
Mazzone A, Strege PR, Tester DJ, Bernard CE, Faulkner G, De Giorgio R, Makielski JC, Stanghellini V, Gibbons SJ, Ackerman MJ, Farrugia G. A mutation in telethonin alters Nav1.5 function. J Biol Chem 2008; 283:16537-44. [PMID: 18408010 DOI: 10.1074/jbc.m801744200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Excitable cells express a variety of ion channels that allow rapid exchange of ions with the extracellular space. Opening of Na(+) channels in excitable cells results in influx of Na(+) and cellular depolarization. The function of Na(v)1.5, an Na(+) channel expressed in the heart, brain, and gastrointestinal tract, is altered by interacting proteins. The pore-forming alpha-subunit of this channel is encoded by SCN5A. Genetic perturbations in SCN5A cause type 3 long QT syndrome and type 1 Brugada syndrome, two distinct heritable arrhythmia syndromes. Mutations in SCN5A are also associated with increased prevalence of gastrointestinal symptoms, suggesting that the Na(+) channel plays a role in normal gastrointestinal physiology and that alterations in its function may cause disease. We collected blood from patients with intestinal pseudo-obstruction (a disease associated with abnormal motility in the gut) and screened for mutations in SCN5A and ion channel-interacting proteins. A 42-year-old male patient was found to have a mutation in the gene TCAP, encoding for the small protein telethonin. Telethonin was found to be expressed in the human gastrointestinal smooth muscle, co-localized with Na(v)1.5, and co-immunoprecipitated with sodium channels. Expression of mutated telethonin, when co-expressed with SCN5A in HEK 293 cells, altered steady state activation kinetics of SCN5A, resulting in a doubling of the window current. These results suggest a new role for telethonin, namely that telethonin is a sodium channel-interacting protein. Also, mutations in telethonin can alter Na(v)1.5 kinetics and may play a role in intestinal pseudo-obstruction.
Collapse
Affiliation(s)
- Amelia Mazzone
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
|